Abstract

Gametogenesis represents the most dramatic cellular differentiation pathways in both female and male flies. At the genome level, meiosis ensures that diploid germ cells become haploid gametes. At the epigenome level, extensive changes are required to turn on and shut off gene expression in a precise spatiotemporally controlled manner. Research applying conventional molecular genetics and cell biology, in combination with rapidly advancing genomic tools have helped us to investigate (1) how germ cells maintain lineage specificity throughout their adult reproductive lifetime; (2) what molecular mechanisms ensure proper oogenesis and spermatogenesis, as well as protect genome integrity of the germline; (3) how signaling pathways contribute to germline-soma communication; and (4) if such communication is important. In this chapter, we highlight recent discoveries that have improved our understanding of these questions. On the other hand, restarting a new life cycle upon fertilization is a unique challenge faced by gametes, raising questions that involve intergenerational and transgenerational epigenetic inheritance. Therefore, we also discuss new developments that link changes during gametogenesis to early embryonic development—a rapidly growing field that promises to bring more understanding to some fundamental questions regarding metazoan development.

GAMETOGENESIS produces the only cell types within an organism that contribute genetic, as well as epigenetic, material to the offspring. Germ cells are distinct from the mortal somatic cells in their ability to differentiate into gametes that regain totipotency to produce an entire organism upon fertilization (Cinalli et al. 2008). Gametes undergo an extraordinary cellular differentiation process to produce morphologically and functionally distinct gametes, i.e., oocytes and sperm. Studying gametogenesis in Drosophila allows us to follow the linear organization of germ cells in adult ovaries and testes. Such organization ensures that all stages of oogenesis and spermatogenesis can be recognized in a highly orchestrated manner (Figure 1) (Fuller 1993; Spradling 1993).

Anatomy of Drosophila female and male gonads. (A) Anatomy of germarium and oogenesis. The Drosophila ovaries are made up of 16–20 tubule structures, called ovarioles, that resemble linear assembly lines of progressively differentiating egg chambers to produce eggs. The germanium, designated by the dashed rectangular outline, where the egg chamber originates, is located at the anterior tip of each ovariole. The germarium consists of the GSC niche and the proliferative germ cells that remain active, producing eggs throughout adulthood. At the anterior tip of each germanium resides the niche, which consists of a stack of 8–10 postmitotic somatic cells, called the terminal filament (light green), five to seven squamous epithelial cells, and cap cells (dark green) that literally cap the underlying two to three GSCs (red). Female GSCs divide asymmetrically such that the anteriorly positioned daughter cell remains in contact with the cap cells and maintains GSC identity, while the posteriorly displaced daughter cell leaves the niche and differentiates into cystoblasts (CBs) (pink). Following the asymmetric cell division, the daughter CB undergoes four rounds of synchronous mitotic cell divisions with incomplete cytokinesis to give rise to 16 interconnected cystocytes (pink). During early germ cell development, early germ cells associate intimately with neighboring somatic cells, including escort cells and follicle cells (yellow). Interspersed between the GSCs are four to six escort cells (blue cells), which cover most of the GSC and dividing CBs, isolating early germ cells from each other, but not from the cap cells. Next, the interconnected germ cell cyst associates with another somatic cell type, the somatic follicle cells. These somatic follicle cells are derived from two somatic follicle stem cells (FSCs) (orange), which are maintained at the boundary between escort cells and the follicle cells. When the 16-cell cyst is surrounded by follicle cells, it becomes an egg chamber, buds from the germanium, and continues to mature (Davring and Sunner 1973). One of the 16 cells will progress through meiosis and develop into the oocyte, while the other cells will develop into polyploid nurse cells that will support oocyte growth. A single egg chamber consists of the single oocyte connected to 15 nurse cells via a system of intercellular bridges and a surrounding monolayer of up to 650 somatic follicle cells (King 1970; Spradling 1993). The nurse cells deliver their cytoplasm into the oocytes and undergo apoptosis during the latest stage of oogenesis to produce a mature egg (Foley and Cooley 1998). Meiotic divisions in the egg are only accomplished after sperm entry, leading to one female pronucleus and three polar bodies, which subsequently undergo degeneration. The female and male pronuclei appose each other, followed by fusion, which labels the formation of a zygote and the onset of a new life cycle. (B) Anatomy of testis and spermatogenesis. The adult testis of Drosophila melanogaster is a pair of coiled tubes ∼2 mm in length, each composed of a single stem cell niche at the apical end designated by the dashed gray outline (Hardy et al. 1979). The apical cells are assembled into a centrally located structure having GSCs (dark blue), and CySCs (orange) are radially positioned around a cluster of 10–12 small densely packed somatic cells called the hub (Green). Six to nine GSCs are arranged around the hub, while two CySCs fully envelope one GSC such that GSC-to-GSC contact never occurs. Spermatogenesis initiates with the asymmetric division of the GSC to produce one self-renewed daughter cell and a gonialblast (GB) cell (light blue). Upon division, the GB is displaced from the hub and undergoes four transit-amplifying divisions with incomplete cytokinesis, generating a cyst of interconnected germ cells joined by cytoplasmic bridges (light blue). After transit-amplification, the cyst of 16 interconnected spermatogonia synchronously undergoes meiotic DNA synthesis. During meiotic prophase I as spermatocytes, each cell grows ∼25-fold and initiates a robust gene expression program that enables meiotic division and spermatid differentiation. After two meiotic divisions, 64 haploid spermatids are produced, as designated by the dashed blue outline.
Figure 1

Anatomy of Drosophila female and male gonads. (A) Anatomy of germarium and oogenesis. The Drosophila ovaries are made up of 16–20 tubule structures, called ovarioles, that resemble linear assembly lines of progressively differentiating egg chambers to produce eggs. The germanium, designated by the dashed rectangular outline, where the egg chamber originates, is located at the anterior tip of each ovariole. The germarium consists of the GSC niche and the proliferative germ cells that remain active, producing eggs throughout adulthood. At the anterior tip of each germanium resides the niche, which consists of a stack of 8–10 postmitotic somatic cells, called the terminal filament (light green), five to seven squamous epithelial cells, and cap cells (dark green) that literally cap the underlying two to three GSCs (red). Female GSCs divide asymmetrically such that the anteriorly positioned daughter cell remains in contact with the cap cells and maintains GSC identity, while the posteriorly displaced daughter cell leaves the niche and differentiates into cystoblasts (CBs) (pink). Following the asymmetric cell division, the daughter CB undergoes four rounds of synchronous mitotic cell divisions with incomplete cytokinesis to give rise to 16 interconnected cystocytes (pink). During early germ cell development, early germ cells associate intimately with neighboring somatic cells, including escort cells and follicle cells (yellow). Interspersed between the GSCs are four to six escort cells (blue cells), which cover most of the GSC and dividing CBs, isolating early germ cells from each other, but not from the cap cells. Next, the interconnected germ cell cyst associates with another somatic cell type, the somatic follicle cells. These somatic follicle cells are derived from two somatic follicle stem cells (FSCs) (orange), which are maintained at the boundary between escort cells and the follicle cells. When the 16-cell cyst is surrounded by follicle cells, it becomes an egg chamber, buds from the germanium, and continues to mature (Davring and Sunner 1973). One of the 16 cells will progress through meiosis and develop into the oocyte, while the other cells will develop into polyploid nurse cells that will support oocyte growth. A single egg chamber consists of the single oocyte connected to 15 nurse cells via a system of intercellular bridges and a surrounding monolayer of up to 650 somatic follicle cells (King 1970; Spradling 1993). The nurse cells deliver their cytoplasm into the oocytes and undergo apoptosis during the latest stage of oogenesis to produce a mature egg (Foley and Cooley 1998). Meiotic divisions in the egg are only accomplished after sperm entry, leading to one female pronucleus and three polar bodies, which subsequently undergo degeneration. The female and male pronuclei appose each other, followed by fusion, which labels the formation of a zygote and the onset of a new life cycle. (B) Anatomy of testis and spermatogenesis. The adult testis of Drosophila melanogaster is a pair of coiled tubes ∼2 mm in length, each composed of a single stem cell niche at the apical end designated by the dashed gray outline (Hardy et al. 1979). The apical cells are assembled into a centrally located structure having GSCs (dark blue), and CySCs (orange) are radially positioned around a cluster of 10–12 small densely packed somatic cells called the hub (Green). Six to nine GSCs are arranged around the hub, while two CySCs fully envelope one GSC such that GSC-to-GSC contact never occurs. Spermatogenesis initiates with the asymmetric division of the GSC to produce one self-renewed daughter cell and a gonialblast (GB) cell (light blue). Upon division, the GB is displaced from the hub and undergoes four transit-amplifying divisions with incomplete cytokinesis, generating a cyst of interconnected germ cells joined by cytoplasmic bridges (light blue). After transit-amplification, the cyst of 16 interconnected spermatogonia synchronously undergoes meiotic DNA synthesis. During meiotic prophase I as spermatocytes, each cell grows ∼25-fold and initiates a robust gene expression program that enables meiotic division and spermatid differentiation. After two meiotic divisions, 64 haploid spermatids are produced, as designated by the dashed blue outline.

Throughout oogenesis and spermatogenesis, germ cells are closely associated with somatic gonadal cells. In females, germline stem cells (GSCs) first interact with escort cells, followed by the follicle stem cell (FSC) lineage. FSC homeostasis depends on an epithelial niche structure that involves migration of FSCs across the ovariole (Nystul and Spradling 2007, 2010) (Figure 1A). In males, each GSC is enclosed by two cyst stem cells (CySCs). Through asymmetric cell division (Cheng et al. 2011), CySCs self-renew and give rise to differentiated cyst cells, which never divide again. Two cyst cells encapsulate synchronously dividing and differentiating germ cells, and form a distinct germ cell cyst (Figure 1B). Increasing evidence demonstrates that somatic gonadal cells are not only support cells, but also play important roles in instructing germ cell differentiation and maintaining germline identity. Drosophila gametogenesis studies have greatly benefited from enriched genetics tools, including many cell type- and stage-specific Gal4 drivers (Table 1) for performing targeted knockdown, rescue, or overexpression experiments.

Cell-type-specific Gal4 drivers in Drosophila gonads

Table 1
Cell-type-specific Gal4 drivers in Drosophila gonads
Cell-Type ExpressionNameDescriptionReferences
Male germline
 Germline expressionnos-Gal4-VP16GSCs and early germline cystsVan Doren et al. (1998)
bam-Gal4-VP16Initiates expression during transit-amplification divisions and expressed in early spermatocytesChen and McKearin (2003b)
vas-Gal4Most germline cellsZhao et al. (2013)
 Hub cellsupd-Gal4Hub cellsZeidler et al. (1999)
hh-Gal4Hub cellsTanimoto et al. (2000)
fasIII-Gal4Hub cellsWolfstetter and Holz (2012)
 CySCs and somatic cellstj-Gal4CySCs, early cyst cells, and hub cellsHayashi et al. (2002)
C587-Gal4CySCs, early cyst cells, and hub cellsKai and Spradling (2003), Zhu and Xie (2003)
ptc-Gal4CySCs and cyst cellsTazuke et al. (2002)
eya-gal4CySCs and cyst cells, weakly expressed in hub cellsLeatherman and Dinardo (2008)
 Somatic cellsarm-Gal4Most somatic cells including Hub cells, CySCs, and cyst cellsSanson et al. (1996)
 Germline and somatic cellsHsp83-Gal4Ubiquitously expressedArama et al. (2003)
Female germline
 Germline expressionnos-Gal4 [pBac(GreenEye.nosGal4-VP16)]Stem cells, young egg chambers, and increased expression at stage 5Holtzman et al. (2010)
nos-Gal4-VP16Stem cells, young egg chambers, and increased expression at stage 5Van Doren et al. (1998)
Maternal Triple Driver (MTD)-Gal4Uniform expression in the germarium and throughout oogenesis, including GSCsPetrella et al. (2007)
bam-Gal4-VP16Germ cell expression starting at the two-cell cyst stage or CB cellsChen and McKearin (2003b)
pCOG-Gal4-VP16Moderate levels throughout oogenesisRorth (1998)
 Escort cellsC587-Gal4Escort cellsX. Song et al. (2004)
 Terminal filament and Cap cellsbab1-Gal4Terminal filament and cap cellsCabrera et al. (2002)
hh-Gal4Terminal filament and cap cellsTanimoto et al. (2000)
 Follicle stem cells109-30-Gal4Follicle stem cells and early follicle lineageHartman et al. (2010)
 Follicle cellsCb16-Gal4All follicle cells starting in germariumWard et al. (2002)
 Somatic cellstj-Gal4All follicle, follicle stem cells, escort cells, and cap cellsHayashi et al. (2002)
 Ubiquitous expressiontub-Gal4Ubiquitous expression in all cellsLee and Luo (1999)
Cell-Type ExpressionNameDescriptionReferences
Male germline
 Germline expressionnos-Gal4-VP16GSCs and early germline cystsVan Doren et al. (1998)
bam-Gal4-VP16Initiates expression during transit-amplification divisions and expressed in early spermatocytesChen and McKearin (2003b)
vas-Gal4Most germline cellsZhao et al. (2013)
 Hub cellsupd-Gal4Hub cellsZeidler et al. (1999)
hh-Gal4Hub cellsTanimoto et al. (2000)
fasIII-Gal4Hub cellsWolfstetter and Holz (2012)
 CySCs and somatic cellstj-Gal4CySCs, early cyst cells, and hub cellsHayashi et al. (2002)
C587-Gal4CySCs, early cyst cells, and hub cellsKai and Spradling (2003), Zhu and Xie (2003)
ptc-Gal4CySCs and cyst cellsTazuke et al. (2002)
eya-gal4CySCs and cyst cells, weakly expressed in hub cellsLeatherman and Dinardo (2008)
 Somatic cellsarm-Gal4Most somatic cells including Hub cells, CySCs, and cyst cellsSanson et al. (1996)
 Germline and somatic cellsHsp83-Gal4Ubiquitously expressedArama et al. (2003)
Female germline
 Germline expressionnos-Gal4 [pBac(GreenEye.nosGal4-VP16)]Stem cells, young egg chambers, and increased expression at stage 5Holtzman et al. (2010)
nos-Gal4-VP16Stem cells, young egg chambers, and increased expression at stage 5Van Doren et al. (1998)
Maternal Triple Driver (MTD)-Gal4Uniform expression in the germarium and throughout oogenesis, including GSCsPetrella et al. (2007)
bam-Gal4-VP16Germ cell expression starting at the two-cell cyst stage or CB cellsChen and McKearin (2003b)
pCOG-Gal4-VP16Moderate levels throughout oogenesisRorth (1998)
 Escort cellsC587-Gal4Escort cellsX. Song et al. (2004)
 Terminal filament and Cap cellsbab1-Gal4Terminal filament and cap cellsCabrera et al. (2002)
hh-Gal4Terminal filament and cap cellsTanimoto et al. (2000)
 Follicle stem cells109-30-Gal4Follicle stem cells and early follicle lineageHartman et al. (2010)
 Follicle cellsCb16-Gal4All follicle cells starting in germariumWard et al. (2002)
 Somatic cellstj-Gal4All follicle, follicle stem cells, escort cells, and cap cellsHayashi et al. (2002)
 Ubiquitous expressiontub-Gal4Ubiquitous expression in all cellsLee and Luo (1999)
Table 1
Cell-type-specific Gal4 drivers in Drosophila gonads
Cell-Type ExpressionNameDescriptionReferences
Male germline
 Germline expressionnos-Gal4-VP16GSCs and early germline cystsVan Doren et al. (1998)
bam-Gal4-VP16Initiates expression during transit-amplification divisions and expressed in early spermatocytesChen and McKearin (2003b)
vas-Gal4Most germline cellsZhao et al. (2013)
 Hub cellsupd-Gal4Hub cellsZeidler et al. (1999)
hh-Gal4Hub cellsTanimoto et al. (2000)
fasIII-Gal4Hub cellsWolfstetter and Holz (2012)
 CySCs and somatic cellstj-Gal4CySCs, early cyst cells, and hub cellsHayashi et al. (2002)
C587-Gal4CySCs, early cyst cells, and hub cellsKai and Spradling (2003), Zhu and Xie (2003)
ptc-Gal4CySCs and cyst cellsTazuke et al. (2002)
eya-gal4CySCs and cyst cells, weakly expressed in hub cellsLeatherman and Dinardo (2008)
 Somatic cellsarm-Gal4Most somatic cells including Hub cells, CySCs, and cyst cellsSanson et al. (1996)
 Germline and somatic cellsHsp83-Gal4Ubiquitously expressedArama et al. (2003)
Female germline
 Germline expressionnos-Gal4 [pBac(GreenEye.nosGal4-VP16)]Stem cells, young egg chambers, and increased expression at stage 5Holtzman et al. (2010)
nos-Gal4-VP16Stem cells, young egg chambers, and increased expression at stage 5Van Doren et al. (1998)
Maternal Triple Driver (MTD)-Gal4Uniform expression in the germarium and throughout oogenesis, including GSCsPetrella et al. (2007)
bam-Gal4-VP16Germ cell expression starting at the two-cell cyst stage or CB cellsChen and McKearin (2003b)
pCOG-Gal4-VP16Moderate levels throughout oogenesisRorth (1998)
 Escort cellsC587-Gal4Escort cellsX. Song et al. (2004)
 Terminal filament and Cap cellsbab1-Gal4Terminal filament and cap cellsCabrera et al. (2002)
hh-Gal4Terminal filament and cap cellsTanimoto et al. (2000)
 Follicle stem cells109-30-Gal4Follicle stem cells and early follicle lineageHartman et al. (2010)
 Follicle cellsCb16-Gal4All follicle cells starting in germariumWard et al. (2002)
 Somatic cellstj-Gal4All follicle, follicle stem cells, escort cells, and cap cellsHayashi et al. (2002)
 Ubiquitous expressiontub-Gal4Ubiquitous expression in all cellsLee and Luo (1999)
Cell-Type ExpressionNameDescriptionReferences
Male germline
 Germline expressionnos-Gal4-VP16GSCs and early germline cystsVan Doren et al. (1998)
bam-Gal4-VP16Initiates expression during transit-amplification divisions and expressed in early spermatocytesChen and McKearin (2003b)
vas-Gal4Most germline cellsZhao et al. (2013)
 Hub cellsupd-Gal4Hub cellsZeidler et al. (1999)
hh-Gal4Hub cellsTanimoto et al. (2000)
fasIII-Gal4Hub cellsWolfstetter and Holz (2012)
 CySCs and somatic cellstj-Gal4CySCs, early cyst cells, and hub cellsHayashi et al. (2002)
C587-Gal4CySCs, early cyst cells, and hub cellsKai and Spradling (2003), Zhu and Xie (2003)
ptc-Gal4CySCs and cyst cellsTazuke et al. (2002)
eya-gal4CySCs and cyst cells, weakly expressed in hub cellsLeatherman and Dinardo (2008)
 Somatic cellsarm-Gal4Most somatic cells including Hub cells, CySCs, and cyst cellsSanson et al. (1996)
 Germline and somatic cellsHsp83-Gal4Ubiquitously expressedArama et al. (2003)
Female germline
 Germline expressionnos-Gal4 [pBac(GreenEye.nosGal4-VP16)]Stem cells, young egg chambers, and increased expression at stage 5Holtzman et al. (2010)
nos-Gal4-VP16Stem cells, young egg chambers, and increased expression at stage 5Van Doren et al. (1998)
Maternal Triple Driver (MTD)-Gal4Uniform expression in the germarium and throughout oogenesis, including GSCsPetrella et al. (2007)
bam-Gal4-VP16Germ cell expression starting at the two-cell cyst stage or CB cellsChen and McKearin (2003b)
pCOG-Gal4-VP16Moderate levels throughout oogenesisRorth (1998)
 Escort cellsC587-Gal4Escort cellsX. Song et al. (2004)
 Terminal filament and Cap cellsbab1-Gal4Terminal filament and cap cellsCabrera et al. (2002)
hh-Gal4Terminal filament and cap cellsTanimoto et al. (2000)
 Follicle stem cells109-30-Gal4Follicle stem cells and early follicle lineageHartman et al. (2010)
 Follicle cellsCb16-Gal4All follicle cells starting in germariumWard et al. (2002)
 Somatic cellstj-Gal4All follicle, follicle stem cells, escort cells, and cap cellsHayashi et al. (2002)
 Ubiquitous expressiontub-Gal4Ubiquitous expression in all cellsLee and Luo (1999)

It has been demonstrated that transcriptional changes at both local and global levels are robust throughout gametogenesis. Epigenetic mechanisms that modify chromatin state without altering primary DNA sequences have profound influence on regulating dynamic transcriptome changes in germ cells. Epigenetic regulation could act through modifications of DNA-associated proteins and/or RNAs, resulting in structural changes of chromatin or recruitment of effector proteins or RNAs, and leading, in turn, to activation or repression of target gene(s). The basic unit of chromatin is called a nucleosome, which contains 147 bp of DNA wrapped around a histone octamer composed of two copies each of H3, H4, H2A, and H2B—the core histones. The major epigenetic mechanisms known to orchestrate cell fate and function include (1) DNA methylation; (2) nucleosome repositioning driven by chromatin remodeling factors; (3) post-translational modifications (PTMs) of histones (e.g., methylation, acetylation, phosphorylation, and ubiquitination, etc.); (4) incorporation of histone variants; and (5) noncoding RNA-mediated chromatin regulation, including piRNA- and microRNA-mediated mechanisms.

As the germline genome is inherited across generations, it is threatened by transposons—genetic mobile elements parasitizing the genome. Transposons are discrete, autonomous, DNA sequences capable of moving from one place to another throughout the genome, or simply increasing their copies in the genome. Transposons constitute almost 23% of the Drosophila genome (Lander et al. 2001; Huang et al. 2012). In order to spread throughout the population, the transposon targets the germline genome, which carries the genetic information from one generation to another for species continuity. Active mobilization of transposons results in insertional mutations, leading to massive destruction of the genome, as well as sterility. Metazoans have evolved a small RNA-based repression system to combat a wide variety of transposons in gonads, called the PIWI-interacting RNA (piRNA) pathway, whose basic function and genes thereof are conserved from the lower invertebrates to mammals (Lim and Kai 2015).

Studies in recent years have shed light on how different mechanisms regulate extensive cellular differentiation during gametogenesis and protect germline identity. In this chapter, we focus on the most recent discoveries of epigenetic regulation and protection of the genome during Drosophila oogenesis and spermatogenesis. We start by discussing how known epigenetic mechanisms maintain GSC identity and activity. We next proceed to an examination of their roles in controlling mitotic germ cell proliferation, proper mitosis-to-meiosis transition, and meiotic maturation. Finally, the developing field of intergenerational and transgenerational epigenetic inheritance is explored.

Mechanisms Regulating GSC Self-Renewal vs. Differentiation

DNA methylation and demethylation

DNA methylation is a widely conserved epigenetic mechanism that functions through the covalent and heritable modification of genomic DNA at both cytosine and adenine residues (Suzuki and Bird 2008; Luo et al. 2015). DNA methylation of the fifth position of cytosine (5-methylcyosine, 5mC) is established and maintained by a conserved family of enzymes called DNA methyltransferases (DNMTs), which have been found to function in transcriptional silencing of promoters, transposable elements, and other repetitive sequences in most plant, animal, and fungal species (Wu and Zhang 2014). DNMTs are divided into three subfamilies based on sequence conservation and function. DNMT3 functions in the de novo methylation of cytosine. DNMT1 maintains DNA methylation postreplication on the newly synthesized DNA strands, and DNMT2 exhibits a weak catalytic activity on DNA compared to DNMT3 (Hermann et al. 2003). DNMT2-mediated methylation of multiple transfer ribonucleic acids (tRNAs) has also been documented (Goll et al. 2006; Schaefer et al. 2010). Drosophila belongs to the “DNMT2 only” category of organisms based on loss of the canonical DNA methyltransferases (DNMT1 and DNMT3), and retention of the DNMT2 homolog DNMT2/Mt2. In addition, 5mC levels in Drosophila have been found to be very low compared to other organisms (Lyko et al. 2000a; Phalke et al. 2009; Krauss and Reuter 2011; Raddatz et al. 2013; Capuano et al. 2014; Takayama et al. 2014; Zhang et al. 2015). For example, in mammalian DNA, between 2 and 10% of all cytosine residues are modified to 5mC, whereas Drosophila DNA contains only 0.1–0.6% of modified 5mC of all cytosine residues (Gowher et al. 2000; Zemach et al. 2010). While the mechanism by which DNMT2 functions in the germline remains obscure, recent studies have revealed that DNMT2 is involved in multiple processes, including sister chromatids' segregation in the male germline, retrotransposon silencing in the early embryo, and gene silencing (Phalke et al. 2009; Yadlapalli and Yamashita 2013). Expression of DNMT2 was first observed in the ovaries, as well as during early embryogenesis through RNA in situ hybridization, and more recently in the male germline (Lyko et al. 2000b; Gan et al. 2010a). DNMT2 function was found to be necessary for proper segregation of X and Y sister chromatids during asymmetric male GSC divisions (Yadlapalli and Yamashita 2013).

Until recently, DNA methylation of the sixth position of Adenine (6mA) was thought to be restricted to bacteria, archaea, protists, and fungi (Wion and Casadesus 2006). However, recent studies have identified 6mA to be present in 0.07–0.001% of all adenine residues in the Drosophila genome during early- and late-stage embryogenesis, respectively (Zhang et al. 2015). Demethylation of 6mA is regulated by the Drosophila Tet homolog, DNA 6mA demethylase (Dmad), during embryogenesis and oogenesis. During oogenesis, loss of Dmad results in an increase of 6mA in the ovaries and accumulation of GSC-like cells. On the other hand, overexpression of Dmad leads to a significant loss of germ cells, including GSCs. Furthermore, Dmad-mediated 6mA demethylation correlates with transposon suppression, indicating that Dmad actively removes 6mA to suppress transposon expression (Zhang et al. 2015). These results are consistent with the role Dmad plays in the demethylation of 6mA to promote GSC differentiation during oogenesis. Together, these results indicate that 5mC has a limited or spatiotemporally specific role in Drosophila, likely independent of DNMT2/Mt2 whose enzymatic role is yet to be defined. On the other hand, recent data demonstrate important roles of 6mA and its demethylase Dmad in Drosophila oogenesis and embryogenesis, even though the corresponding methyltransferase has not been characterized. Given the clear biological functions of 5mC in mammals, it is possible that fly and mammals use distinct DNA methylation mechanisms for their epigenomes.

Chromatin remodeling factors

The chromatin structure of GSCs and somatic stem cells (SSCs) is regulated by ATP-dependent chromatin remodeling enzymes in both males and females in order to maintain self-renewal and prevent differentiation. These enzymes utilize the energy of ATP hydrolysis to establish and maintain a particular chromatin state during development. The different subfamilies of chromatin-remodeling enzymes catalyze a remarkable range of chromatin modifications that include histone exchange, translocating the histone octamer and changing the conformation of nucleosomal DNA (Narlikar et al. 2013). Common across all ATP-dependent chromatin remodeling enzymes is the ATPase subunit belonging to the helicase superfamily 2 (SNF2) (Eisen et al. 1995). This SNF2 family of proteins can be further classified on the basis of distinct domains conserved among the subfamilies, such as the bromodomain shared by the SWI2/SNF2 (SWItch/Sucrose NonFermentable) family, the chromodomain shared by the CHD (Chromodomain-Helicase-DNA-binding protein) family, and the SANT domain shared by the ISWI (Imitation SWI) family (Hota and Bruneau 2016).

Imitation switch (ISWI):

In Drosophila, ISWI serves as an ATP-dependent motor that governs transcriptional regulation through catalyzing changes in nucleosomal assembly and composition (Deuring et al. 2000; Badenhorst et al. 2002; Corona et al. 2002). In both males and females, ISWI is essential for GSC maintenance, suggesting a common epigenetic mechanism employed by both sexes to maintain a chromatin configuration for stem cell maintenance. In females, ISWI is present at high levels in all cell types, including GSCs and FSCs (Xi and Xie 2005). Mitotic recombination techniques (Xu and Rubin 1993) were used to generate marked iswi mutant GSC clones, most of which were lost from the niche owing to premature differentiation. Similar to ISWI, bone morphogenetic protein (BMP) signal transduction is essential for GSC maintenance. Upon signal transduction, the BMP signaling cascade is mediated by phosphorylated MAD (pMAD), which activates the target Daughters against dpp (Dad) transcription, and results in transcriptional repression of the differentiation marker bag of marbles (bam) (Chen and McKearin 2003a; X. Song et al. 2004). Since cystoblasts (CBs) do not receive enough BMP ligand, they begin the differentiation process by the increased expression of bam (Y. Li et al. 2009). Significant premature upregulation of bam was found in iswi mutant GSCs when compared to wild-type GSC clones. Dad transcription was also aberrantly regulated in the absence of iswi. These results demonstrate that ISWI maintains GSC self-renewal through BMP signaling-mediated gene expression.

Nucleosome remodeling factor (NURF) complex:

In Drosophila, ISWI is a component of three chromatin remodeling complexes, including NURF (NUcleosome Remodeling Factor), ACF (ATP-utilizing Chromatin assembly and remodeling Factor), and CHRAC (CHRomatin Accessibility Complex). In males, GSC self-renewal is specifically regulated by the NURF complex (Cherry and Matunis 2010), which is composed of ISWI, NURF301, NURF55, and NURF38. Similar to the iswi mutant female GSC phenotype, inactivation of iswi and Nurf301 leads to loss of male GSCs from premature differentiation by precocious expression of Bam. Clonal analysis revealed that Nurf301 mutant CySCs, similar to GSCs, are lost rapidly as a result of premature differentiation.

Similar to the NURF complex, JAK/STAT signaling is also required for the maintenance of both GSCs and CySCs. In the male, the hub cells secrete the signaling ligand Unpaired (Upd) to support stem cell self-renewal of both GSCs and CySCs, as well as adhesion of GSCs to the hub cells (Kiger et al. 2001; Tulina and Matunis 2001; Leatherman and Dinardo 2008, 2010). Loss-of-function of either the Janus kinase (JAK), encoded by hopscotch (hop), or the signal transducer and activator of transcription (STAT), encoded by Stat92E, in the germline leads to rapid loss of GSCs and early germ cells (Kiger et al. 2001; Tulina and Matunis 2001). Consistent with the role of the JAK/STAT pathway in GSC self-renewal, ectopic expression of Upd in early germ cells leads to a dramatic increase in the number of GSC-like cells with a concomitant decrease in the number of cells undergoing differentiation. To test whether the NURF complex regulates the maintenance of GSCs and CySCs through mediating JAK-STAT signaling from the niche, JAK-STAT activity was monitored in Nurf301 null clones by measuring STAT92E expression levels. Loss of Nurf301 resulted in decreased STAT92E, suggesting that Nurf301 promotes the maintenance of GSCs, at least in part, through positively regulating the JAK-STAT pathway. Furthermore, suppressor of cytokine signaling 36E (Socs36E) is a conserved target of the JAK/STAT pathway in CySCs that functions in a negative feedback loop by downregulating JAK/STAT activity (Issigonis et al. 2009). Similar to loss of JAK/STAT signaling, downregulation of NURF301 partially rescued the Socs36E phenotype. These studies highlight that the chromatin remodeling complex, the NURF complex, functions as a positive regulator of JAK/STAT signaling in both GSCs and CySCs in the testis.

Recent studies have also revealed that the ecdysone steroid hormone pathway acts through the NURF complex in female GSCs and in male CySCs (Ables and Drummond-Barbosa 2010; Li et al. 2014). The ecdysone receptor (EcR) is expressed throughout the ovary in multiple cell types (Buszczak et al. 1999). Upon binding of ecdysone to EcR, EcR dimerization occurs with Ultraspiracle (Usp), initiating a transcriptional cascade that includes E74, E75, and broad (br) as targets (Riddiford et al. 2000). Analysis of GSC clones homozygous for usp and E74 in female and temperature-sensitive alleles of ecdysone and EcR demonstrated that ecdysone signaling promotes GSC maintenance. Interestingly, genetic interactions were discovered between the NURF complex genes iswi and Nurf301 and the Ecdysone pathways genes usp and E74. Additionally, loss-of-function mutations in usp and E74 result in reduced levels of nuclear ISWI. As mentioned earlier, iswi mutations result in aberrant BMP signaling and premature bam expression. Consistent with the decrease in ISWI, BMP signaling levels are reduced in usp and E74 null clones. The ecdysone signaling pathway acts with the NURF chromatin remodeling complex to promote female GSC maintenance. Intriguingly, purified NURF physically interacts with EcR in an ecydsone-dependent manner, and expression of EcR target genes is significantly reduced in Nurf mutants, suggesting that Nurf is a coactivator of EcR (Badenhorst et al. 2002).

In the male, ecdysone signaling components are expressed in both hub cells and the CySC lineage, and they are required for CySC maintenance (Li et al. 2014). Loss of ecdysone signaling in CySCs results in loss of GSCs, as well as CySCs, suggesting that EcR signaling contributes to both stem cell populations in the testis. It currently remains unknown whether GSC maintenance requires an ecdysone-dependent or -independent signal from CySCs.

Domino (dom):

This SWR1-like ATP-dependent chromatin remodeling factor functions in both male and female gonads for stem cell self-renewal (Xi and Xie 2005; Morillo Prado et al. 2013). Unlike the chromatin remodeling factor ISWI, which is essential for GSC self-renewal, DOM is only essential for FSC self-renewal in the female. Clonal analysis of dom mutant GSCs reported no change in their division rate. In contrast, female FSCs marked for loss of dom demonstrated that DOM specifically controls FSC self-renewal, but not survival (Xi and Xie 2005). In the female, these studies revealed that different stem cell types, GSCs and FSCs, depend on distinct chromatin remodeling factors, ISWI and DOM, respectively, to control their self-renewal.

In the male, clonal analysis revealed that DOM is required cell autonomously for both GSC and CySC maintenance, and may regulate the incorporation of the histone variant H2Av (Morillo Prado et al. 2013). H2Av is the Drosophila sole homolog of mammalian H2A.Z and H2A.X (Talbert and Henikoff 2010; Baldi and Becker 2013). Although the expression of H2Av is ubiquitous, its function is dispensable for germline and cyst cell differentiation, suggesting a specific role for maintaining the stem cell state in these lineages. H2Av, which is incorporated by SWR1-like remodeling complexes, regulates transcriptional control, formation of heterochromatin boundaries, lineage commitment, and DNA repair throughout development (Henikoff et al. 2004; Creyghton et al. 2008; Venkatesh and Workman 2015). Dom has been purified from S2 cells as part of a 16-subunit assembly, and this complex has been shown to exchange H2Av in vitro (Kusch et al. 2004). Because Dom is required for H2Av incorporation, loss of dom function reduced H2Av levels in male GSCs. Furthermore, a recent study has highlighted specific roles for a distinct dom splicing variant required for the incorporation and removal of H2Av during oogenesis (Borner and Becker 2016). Similar to dom, H2Av is required for both male GSC and CySC maintenance independent of the JAK/STAT pathway, and it has been implicated in both transcriptional repression and activation (Morillo Prado et al. 2013). Lack of H2Av does not result in global changes in H3K4me3 or H3K27me3 immunostaining pattern. However, it is possible that the H2Av mutation disrupts H3K9me2/3-enriched heterochromatin structure in GSCs, as previously shown in somatic cells (Swaminathan et al. 2005). Therefore, DOM and H2Av may be required to maintain GSC and CySCs by facilitating repression of differentiation genes and/or maintaining activation of genes necessary for GSC self-renewal. Finally, dom mutants can be partially rescued by the human ortholog, SRCAP (Eissenberg et al. 2005).

Brahma:

Brahma (Brm), a bromodomain protein, is the sole member of the Drosophila SWI/SNF-type ATPase chromatin remodeler. It has cell-autonomous, as well as non-cell-autonomous, roles in regulating female GSC self-renewal (Brizuela et al. 1994; Elfring et al. 1998; Zraly et al. 2003; He et al. 2014). Brahma is a member of two protein complexes, BAP and PBAP, and it is expressed in all cell types in the germarium and follicle cells. Both complexes share seven subunits, including Brm, and differ in three subunits. OSA is a member of the BAP complex, while Polybromo and BAP170 are members of the PBAP complex (Mohrmann et al. 2004). Using both clonal analysis of brm mutant and tissue-specific RNAi knockdown, a cell-autonomous role of brm in sustaining the GSC population has been revealed. Furthermore, knocking down brm in the niche cells showed a non-cell-autonomous role for brm in regulating GSC self-renewal. To distinguish whether a specific Brm complex, BAP or PBAP, regulates GSC self-renewal, loss of osa and polybromo/bap180 was tested individually. This revealed that mutations in polybromo/bap180, rather than osa, cause similar GSC loss phenotype. These studies indicate that Brm functions in the PBAP complex for GSC maintenance.

Nclb:

A novel chromatin factor encoded by no child left behind (nclb) specifically regulates male, but not female, GSC maintenance (Casper et al. 2011). Nclb is enriched at chromatin regions with active transcription. In nclb mutant GSCs, Stat92E has decreased transcription or protein accumulation (Casper et al. 2011), suggesting that Nclb acts via signaling pathways to determine GSC fate.

Histones

The principal components of epigenetic information, histones, are uniquely distributed with pre-existing (old) histone H3 segregating to the stem cell and newly synthesized (new) H3 localizing to the differentiating daughter cell during Drosophila male GSC asymmetric division [Figure 2, (Tran et al. 2012)]. The histone variant H3.3, which is incorporated in a replication-independent manner, does not exhibit such an asymmetric pattern. Therefore, it is likely that DNA replication plays an important role in establishing histone asymmetry between sister chromatids. Furthermore, asymmetric H3 inheritance occurs specifically in the asymmetrically dividing GSCs, but not in the symmetrically dividing progenitor germ cells, suggesting that polarized mitotic machinery could contribute to recognizing the sister chromatid asymmetry established by replication. Cellular specificity exhibited by H3 suggests that global asymmetric histone inheritance occurs uniquely in a cell type (GSC) where the mother cell must divide to produce two daughter cells, each with a unique cell fate. However, more research is required to investigate whether the observed H3 asymmetry occurs at all chromosomes, particular chromosomes, or at specific genomic regions. It has also been shown that differential phosphorylation at Threonine 3 of H3 (H3T3P) distinguishes old vs. new histones in dividing GSCs. The H3T3P is enriched at the pericentric region and is only detectable from prophase to early anaphase. The tight spatiotemporal regulation of this phosphorylation likely ensures that it acts at the right location and with the precise timing. Misregulation of this phosphorylation, using either a dominant negative mutant or a phosphomimetic form, leads to randomized segregation of old vs. new histones, as well as stem cell loss and germline tumors. This finding sheds light on the biological significance of asymmetric histone inheritance, which may help maintain GSC identity and reset chromatin structure in the other daughter cell for proper differentiation (Xie et al. 2015).

Asymmetric histone inheritance, Nonrandom segregation of sister chromatids, and asymmetric centrosome inheritance during male GSC asymmetric cell division. Investigations into asymmetric cell division using a dual-color labeling strategy to label pre-existing (green) vs. newly (red) synthesized canonical histone H3 have revealed that old histone H3 is selectively retained in the self-renewed GSC (green nuclei), whereas newly synthesized H3 is enriched in the differentiating daughter cell (red nuclei). More studies are needed to investigate whether the observed H3 asymmetry occurs at all chromosomes, particular chromosomes, or specific genomic regions. During this division, mother and daughter centrosomes with distinct microtubule nucleating capabilities are also observed to be asymmetric: the mother centrosome (yellow and green) remains in proximity to the stem cell niche, while the daughter centrosome (light-blue and turquoise) migrates to the distal side of the cell leading to a perpendicular spindle orientation relative to the niche and asymmetric centrosome inheritance. Using CO-FISH (Chromosome Orientation Fluorescence in situ Hybridization) combined with strand-specific probes to distinguish sister chromatids it has been shown that sex chromosomes (Purple outlined chromatid and Blue outlined chromatid), including both X and Y, exhibit an ∼85:15 biased segregation of sister chromatids during male GSC cell division.
Figure 2

Asymmetric histone inheritance, Nonrandom segregation of sister chromatids, and asymmetric centrosome inheritance during male GSC asymmetric cell division. Investigations into asymmetric cell division using a dual-color labeling strategy to label pre-existing (green) vs. newly (red) synthesized canonical histone H3 have revealed that old histone H3 is selectively retained in the self-renewed GSC (green nuclei), whereas newly synthesized H3 is enriched in the differentiating daughter cell (red nuclei). More studies are needed to investigate whether the observed H3 asymmetry occurs at all chromosomes, particular chromosomes, or specific genomic regions. During this division, mother and daughter centrosomes with distinct microtubule nucleating capabilities are also observed to be asymmetric: the mother centrosome (yellow and green) remains in proximity to the stem cell niche, while the daughter centrosome (light-blue and turquoise) migrates to the distal side of the cell leading to a perpendicular spindle orientation relative to the niche and asymmetric centrosome inheritance. Using CO-FISH (Chromosome Orientation Fluorescence in situ Hybridization) combined with strand-specific probes to distinguish sister chromatids it has been shown that sex chromosomes (Purple outlined chromatid and Blue outlined chromatid), including both X and Y, exhibit an ∼85:15 biased segregation of sister chromatids during male GSC cell division.

Utilizing CO-FISH (chromosome orientation fluorescence in situ hybridization) probes, which allow strand-specific hybridization of sister chromatids, it has been demonstrated that both X and Y sister chromatids exhibit an ∼85:15 bias during male GSC asymmetric division. Autosomes, specifically chromosome 2 and 3, display a random segregation pattern, but do show a remarkable cosegregation mode (i.e., WW: CC instead of WC: CW. W, Watson strand; C, Crick strand) [Figure 2, (Yadlapalli and Yamashita 2013)]. An earlier study using the nucleoside analog 4-bromo-2-deoxyuridine (BrdU) incorporation assay demonstrated that male GSCs do not follow the immortal strand model (Yadlapalli et al. 2011)—a model that hypothesizes that stem cells retain a template copy of DNA, specifically the sister chromatid that contains the oldest strand as a template, to avoid accumulation of DNA replication-induced mutations (Cairns 1975). Since sister chromatids are identical DNA copies of each other, the distribution of distinct information for asymmetric inheritance likely occurs through epigenetic mechanisms, which is consistent with an alternative hypothesis (Klar 2007; Lansdorp 2007).

Histone-modifying enzymes and factors that affect histone modification(s)

Cell autonomous mechanisms:

Recent research has identified a set of specific enzymes and factors that generate (“write”), recognize (“read”), and remove (“erase”) histone modifications, provoking studies of their in vivo functions during development (Sarmento et al. 2004; Seligson et al. 2005). Post-translational modifications that decorate canonical histones (i.e., H2A, H2B, H3, and H4), as well as histone variants, such as H3.3 and H2Av, can serve as molecular memory bookmarks to maintain, or reestablish, transcriptional activation or repression after mitosis. Indeed, different histone modifications are very robust in the male germline (Hennig and Weyrich 2013). The study of histone-modifying enzymes in Drosophila offers a great opportunity because many of them encode the sole ortholog, making interpretation of their endogenous roles unambiguous (Table 2).

PTMs and the corresponding enzymes in Drosophila gametogenesis
Table 2
PTMs and the corresponding enzymes in Drosophila gametogenesis
PTMWriterReaderEraserFunctionReference
H3K4(me)dset1, trr, trx, and ash1Phf7lid, Su(var)3-3 (lsd1)Commonly associated with promoters of actively transcribed geneBeisel et al. (2002), Sedkov et al. (2003), Di Stefano et al. (2007), Hallson et al. (2012), Yang et al. (2012), Tarayrah et al. (2015)
H3K9(me)Su(var)3-9, eggless/dSETDb1, dG9aRhino (HP1d), HP1a, HP1b, HP1edKDM4BA mark of constitute and facultative heterochromatin, functions in the maintenance of pericentric heterochromatinTschiersch et al. (1994), Ebert et al. (2006), Mis et al. (2006), Stabell et al. (2006), Levine et al. (2015)
H3K23(acetyl)enokN.D.N.D.Promotes gene expressionScott et al. (2001), Huang et al. (2014)
H3K27(me)E(z)PcdUTXAssociated with inactive gene promoters, indexing both pericentric heterochromatin and inactive euchromatic domainsMin et al. (2003), Ebert et al. (2004), Tarayrah et al. (2013)
H2A (Ub)dRingN.D.PR-DUB (calypso/Asx)Promotes repressive chromatinWang et al. (2004), Scheuermann et al. (2010)
H2B(Ub)dBre1N.D.ScrawnyEnhances nucleosome stability and mediates H3k4(me)Buszczak et al. (2009), Xuan et al. (2013)
PTMWriterReaderEraserFunctionReference
H3K4(me)dset1, trr, trx, and ash1Phf7lid, Su(var)3-3 (lsd1)Commonly associated with promoters of actively transcribed geneBeisel et al. (2002), Sedkov et al. (2003), Di Stefano et al. (2007), Hallson et al. (2012), Yang et al. (2012), Tarayrah et al. (2015)
H3K9(me)Su(var)3-9, eggless/dSETDb1, dG9aRhino (HP1d), HP1a, HP1b, HP1edKDM4BA mark of constitute and facultative heterochromatin, functions in the maintenance of pericentric heterochromatinTschiersch et al. (1994), Ebert et al. (2006), Mis et al. (2006), Stabell et al. (2006), Levine et al. (2015)
H3K23(acetyl)enokN.D.N.D.Promotes gene expressionScott et al. (2001), Huang et al. (2014)
H3K27(me)E(z)PcdUTXAssociated with inactive gene promoters, indexing both pericentric heterochromatin and inactive euchromatic domainsMin et al. (2003), Ebert et al. (2004), Tarayrah et al. (2013)
H2A (Ub)dRingN.D.PR-DUB (calypso/Asx)Promotes repressive chromatinWang et al. (2004), Scheuermann et al. (2010)
H2B(Ub)dBre1N.D.ScrawnyEnhances nucleosome stability and mediates H3k4(me)Buszczak et al. (2009), Xuan et al. (2013)
Table 2
PTMs and the corresponding enzymes in Drosophila gametogenesis
PTMWriterReaderEraserFunctionReference
H3K4(me)dset1, trr, trx, and ash1Phf7lid, Su(var)3-3 (lsd1)Commonly associated with promoters of actively transcribed geneBeisel et al. (2002), Sedkov et al. (2003), Di Stefano et al. (2007), Hallson et al. (2012), Yang et al. (2012), Tarayrah et al. (2015)
H3K9(me)Su(var)3-9, eggless/dSETDb1, dG9aRhino (HP1d), HP1a, HP1b, HP1edKDM4BA mark of constitute and facultative heterochromatin, functions in the maintenance of pericentric heterochromatinTschiersch et al. (1994), Ebert et al. (2006), Mis et al. (2006), Stabell et al. (2006), Levine et al. (2015)
H3K23(acetyl)enokN.D.N.D.Promotes gene expressionScott et al. (2001), Huang et al. (2014)
H3K27(me)E(z)PcdUTXAssociated with inactive gene promoters, indexing both pericentric heterochromatin and inactive euchromatic domainsMin et al. (2003), Ebert et al. (2004), Tarayrah et al. (2013)
H2A (Ub)dRingN.D.PR-DUB (calypso/Asx)Promotes repressive chromatinWang et al. (2004), Scheuermann et al. (2010)
H2B(Ub)dBre1N.D.ScrawnyEnhances nucleosome stability and mediates H3k4(me)Buszczak et al. (2009), Xuan et al. (2013)
PTMWriterReaderEraserFunctionReference
H3K4(me)dset1, trr, trx, and ash1Phf7lid, Su(var)3-3 (lsd1)Commonly associated with promoters of actively transcribed geneBeisel et al. (2002), Sedkov et al. (2003), Di Stefano et al. (2007), Hallson et al. (2012), Yang et al. (2012), Tarayrah et al. (2015)
H3K9(me)Su(var)3-9, eggless/dSETDb1, dG9aRhino (HP1d), HP1a, HP1b, HP1edKDM4BA mark of constitute and facultative heterochromatin, functions in the maintenance of pericentric heterochromatinTschiersch et al. (1994), Ebert et al. (2006), Mis et al. (2006), Stabell et al. (2006), Levine et al. (2015)
H3K23(acetyl)enokN.D.N.D.Promotes gene expressionScott et al. (2001), Huang et al. (2014)
H3K27(me)E(z)PcdUTXAssociated with inactive gene promoters, indexing both pericentric heterochromatin and inactive euchromatic domainsMin et al. (2003), Ebert et al. (2004), Tarayrah et al. (2013)
H2A (Ub)dRingN.D.PR-DUB (calypso/Asx)Promotes repressive chromatinWang et al. (2004), Scheuermann et al. (2010)
H2B(Ub)dBre1N.D.ScrawnyEnhances nucleosome stability and mediates H3k4(me)Buszczak et al. (2009), Xuan et al. (2013)
PcG and TrxG:

Increasing evidence indicates that the Polycomb group (PcG) and the Trithorax group (TrxG) complexes play critical roles for cells to decide between maintaining the proliferating precursor state, and initiating the terminal differentiation program (Ringrose and Paro 2004). It is generally agreed that both PcG and TrxG complexes employ epigenetic mechanisms that alter chromatin state to either repress or activate gene expression (Surface et al. 2010). PcG proteins act in at least two distinct, but interacting, protein complexes, Polycomb Repressive Complex 1 (PRC1) and PRC2 (Schwartz and Pirrotta 2007). PRC2 contains an enzymatic component, Enhancer of Zeste [E(z)], which methylates histone H3 at Lys27 (H3K27me3) (Cao et al. 2002; Czermin et al. 2002; Kuzmichev et al. 2002; Muller et al. 2002). This methylated histone recruits PRC1, which binds to the H3K27me3 epigenetic mark through the chromodomain of the Polycomb (Pc) protein (Fischle et al. 2003; Min et al. 2003), leading to the nucleation of the entire PcG complex. It has been shown that mutation on the H3 Lys27 residue phenocopies loss-of-function mutants of PRC2 components, indicating that H3K27 is indeed the critical in vivo substrate of PRC2 histone methyltransferase activity (Pengelly et al. 2013). In addition, the dRing in the PRC1 complex acts as an E3 ubiquitin ligase, which ubiquitylates histone H2A at Lys119 (H2AK119ub) (Wang et al. 2004). H2AK119ub may affect transcription by blocking efficient elongation (Stock et al. 2007). Phosphorylation of the H2A variant H2Av (γH2Av), which serves as a specific marker for double-strand DNA breaks, often indicates an early response to DNA damage. Consistently, it was reported that mutations in H2Av enhance Drosophila male germline defects caused by DNA damage that initially results from mutations in the PcG gene multi sex combs (mxc) (Landais et al. 2014).

On the other hand, the active H3K4me3 mark is generated by the TrxG complex (Byrd and Shearn 2003; Klymenko and Muller 2004; Ringrose and Paro 2004), and opposes the PcG function. In the female GSCs, it has recently been shown that the global level of H3K4me3 is decreased upon loss-of-function of the Drosophila ortholog of Ctr9, a component of the Paf1 complex normally required for transcriptional initiation and polyadenylation. However, the functional readout of this global H3K4me3 loss remains unclear (Chaturvedi et al. 2016).

In Drosophila, not only the maintenance of GSCs, but also the formation of primordial germline cells during embryogenesis, depends on the cell-autonomous function of Piwi (Megosh et al. 2006; Ma et al. 2014). Piwi promotes GSC differentiation nonautonomously through somatic gonadal cells in both male and female gonads (Ma et al. 2014; Gonzalez et al. 2015). The necessity of Piwi function for ovarian GSC maintenance, however, does not seem to be relevant to biogenesis of Piwi-interacting RNAs (piRNAs). The Piwi-piRNA complex is required for transposon silencing in the nucleus; however, Piwi in cytoplasm is shown to be sufficient for GSC maintenance (Klenov et al. 2011). The 3R-TAS1 piRNA is a specific Piwi-bound piRNA and is involved in female GSC maintenance. However, its role and the mode of action remain elusive (Yin and Lin 2007). Piwi function in GSC maintenance is conserved in other organisms, including mouse and zebrafish, where PIWI-family proteins are also involved in GSC maintenance and/or differentiation (Houwing et al. 2007, 2008; Unhavaithaya et al. 2009). Recently, it has been demonstrated that Piwi physically interacts with PRC2 components, and restricts the accessibility of PRC2 to chromatin (Peng et al. 2016).

Eggless:

eggless/dSETDB1 encodes a H3K9 methyltransferase which plays multiple roles in Drosophila oogenesis (Clough et al. 2014). The eggless mutants show female GSC self-renewal defects. However, the GSC loss defect is not caused by loss-of-function in the well-known BMP signaling pathway, suggesting a potential H3K9me3-dependent, but BMP-independent, mechanism for GSC maintenance (Wang et al. 2011). Another study showed that eggless mutant female flies have a defective egg chamber owing to its requirement for FSC proliferation and survival of both FSCs and germ cells (Clough et al. 2007). In addition, it has been shown that Eggless functions in the transcriptional regulation of piRNA clusters (Rangan et al. 2011). Activated piRNAs act with the PIWI protein to protect germline genome by preventing transposable element activity, which may be a conserved mechanism for germline genome integrity across species (Malone et al. 2009; Sienski et al. 2012; Huang et al. 2013; Le Thomas et al. 2013; Rozhkov et al. 2013).

Scrawny:

Scrawny encodes a deubiquitylating enzyme and targets the mono-ubiquitylation H2B, which normally serves as an active mark for transcription. Therefore, the normal function of Scrawny is to make chromatin more compact to repress gene expression. Interestingly, Scrawny is required for stem cell maintenance in multiple adult stem cell systems, including both female and male GSCs, as well as FSCs in the ovary, and intestinal stem cells, suggesting some common chromatin feature among different stem cell types. It is likely that Scrawny maintains stem cells by repressing transcription of the differentiation genes in the corresponding lineages. In addition, inactivation of Scrawny leads to global changes of the chromatin landscape, including increased levels of H3K4me3 and acetylated H3, suggesting crosstalk among different histone modifications, likely through their modifying enzymes. Another intriguing feature is that Scrawny has highly enriched nucleolar localization in both female and male germline (Buszczak et al. 2009). In another study, enhanced ribosomal RNA transcription at nucleolus was shown to maintain normal female GSC proliferation and avoid precocious differentiation (Q. Zhang et al. 2014). It is unclear whether Scrawny is required for proper chromatin structure at rDNA genes for their upregulated transcription.

Little imaginal disc (Lid):

In adult testis, the H3K4me3-specific histone demethylase Lid has cell-autonomous roles to maintain GSC self-renewal and prevent GSCs from undergoing precocious differentiation. When the function of Lid is compromised in early-stage germ cells, the niche is deprived of GSCs, but occupied by differentiating spermatogonial cysts. The key downstream effector of Lid is the Stat92E transcription factor of the JAK-STAT signaling pathway. Lid is required for both normal Stat92E transcript level and protein accumulation. Removing one copy of Stat92E greatly enhanced lid mutant phenotype, and expression of a Stat92E cDNA in early-stage germ cells rescued lid loss-of-function phenotype completely. Therefore, Lid acts through Stat92E in regulating male GSC activity (Tarayrah et al. 2015).

Stonewall (Stwl):

Stwl encodes a DNA-binding protein, which was originally predicted as a putative transcription factor. More recent work suggests that Stwl normally represses expression of many target genes, likely through making the chromatin structure more compact. Stwl is both necessary and sufficient for female GSC cell fate (Maines et al. 2007), as well as the transit amplification of CBs (Akiyama 2002). It has been shown that both H3K9me3 and H3K27me3, two histone modifications to silence gene expression, are decreased in Stwl mutants (Yi et al. 2009), suggesting that Stonewall maintains normal heterochromatin structure, as one of its functions.

longitudinals lacking (lola):

In the male GSC lineage, a transcriptional regulator of the BTB-Zinc finger family encoded by lola was reported to have pleiotropic roles in adult testis. Lola is ubiquitously expressed and is required cell-autonomously for both GSC and CySC maintenance, likely independent of the known JAK-STAT and BMP signaling pathways. In addition, lola is required for proper mitosis-to-meiosis transition, and lola mutant spermatogonial cysts have >16 cells, likely owing to faster cell cycle progression. Finally, lola is also needed for meiosis and terminal differentiation of sperm. Intriguingly, lola has 19 alternative spicing isoforms. With the possibility that Lola may act as a dimer, the combination among different isoforms could give rise to its pleiotropic roles (Davies et al. 2013). By contrast, in female GSCs, lola is repressed by Stwl and is dispensable for female GSC maintenance (Maines et al. 2007), suggesting sex-specific roles of Lola in gametogenesis.

Non-cell-autonomous mechanisms:

A fundamental question in stem cell biology is how extrinsic signaling pathways and intrinsic epigenetic mechanisms cooperate to determine and maintain stem cell fate. Recent findings provide new insights into the non-cell-autonomous roles of different histone-modifying enzymes, many of which are acting through signaling pathways and required for crosstalk among multiple cell types within the stem cell niche.

Lysine-specific histone demethylase 1 (Lsd1):

The lsd1 gene encodes the H3K4me1/2-demethylase in Drosophila (Di Stefano et al. 2007). Lsd1 acts in escort cells to regulate a diverse group of genes, including both BMP-related and BMP-unrelated genes (Eliazer et al. 2014). Lsd1 regulates germline differentiation by preventing ectopic BMP signaling outside of the niche (Eliazer et al. 2011, 2014), as well as using BMP-independent mechanisms.

Enhancer of zeste [E(z)]:

E(z) is a key PRC2 component, which is an H3K27me3-specific methyltransferase (Muller et al. 2002). In Drosophila testis, E(z) acts in somatic gonadal cells to prevent expression of a somatic lineage transcription factor encoded by zinc-finger homeodomain protein 1 (zfh-1) in the germline. Consensus holds that germ cells maintain their unique identity after being specified early in embryogenesis, which is essential for proper gametogenesis. Using complementary somatic and germline lineage-tracing experiments, Zfh-1 was shown to ectopically express in the germline in adult testes when E(z) is inactivated in the somatic cells, suggesting the importance of cell–cell communication in maintaining germ cell identity. Furthermore, only early-stage germ cells, including GSCs, retain the ability to express zfh-1. In contrast, further differentiated spermatogonial cells lose this ability, suggesting that chromatin undergoes structural changes during GSC differentiation that may lock their cell fate choice (Eun et al. 2014).

Posterior sex combs (Psc) and suppressor of zeste 2 [Su(z)2]:

Both Psc and Su(z)2 are PRC1 components which have some redundant functions. Loss of both genes in ovary leads to overproliferation and “metastasis” of FSCs, likely from misregulation of the canonical Wnt signaling and planar polarity pathways (Li et al. 2010). In testis, both Psc and Su(z)2 act in the CySC lineage to maintain their identity and restrict excess proliferation. Loss of both Psc and Su(z)2 leads to tumors that arise from overproliferative CySCs, which also physically displace GSCs from their niche (Morillo Prado et al. 2012).

Utx histone demethylase (dUTX):

The dUTX gene encodes an H3K27me3-specific histone demethylase (Herz et al. 2010). In the testis niche, dUTX removes the repressive H3K27me3 histone modification near the transcription start site of Socs36E and allows active transcription of Socs36E, which encodes an inhibitor of the JAK-STAT signaling pathway. JAK-STAT plays an essential role in the testis niche; as such, dUTX is critical for maintaining the balance between GSCs and CySCs. Loss of dUTX function in either GSCs or CySCs leads to niche cell identity and morphological defects in a non-cell-autonomous manner. These defects can be fully rescued by either overexpression of Socs36E or removal of one copy of the downstream transcription factor-encoding gene, Stat92E, of the JAK-STAT pathway. Therefore, through direct control of JAK-STAT signaling, dUTX coordinates crosstalk among different cell types within the Drosophila testis niche (Tarayrah et al. 2013).

Enoki mushroom (Enok):

The enok gene encodes a putative MYST family histone acetyltransferase that controls female GSC maintenance, both cell-autonomously and non-cell-autonomously (Xin et al. 2013). Loss of enok in female GSCs leads to rapid GSC loss. Enok maintains GSCs through regulating Bruno, which encodes an RNA-binding protein and targets mRNAs in the ovary for translational repression. Furthermore, compromised enok in cap cells impairs niche size and BMP signaling output, thereby causing defective GSC maintenance through a parallel non-cell-autonomous pathway.

dBre1:

dBre1 encodes an E3 ubiquitin ligase required for mono-ubiquitination of H2B. The dBre1 controls both GSC maintenance and germ cell differentiation via distinct mechanisms (Xuan et al. 2013). Loss of dBre1 leads to both GSC loss and a significant reduction in H3K4me3. Further analysis revealed that dBre1 regulates GSC maintenance through modulating BMP signaling response. In addition, dBre1 has a non-cell-autonomous role to maintain GSCs via DE-cadherin-mediated adhesion of GSCs to the niche, as well as the BMP signaling pathway. Finally, dBre1 functions in escort cells to control female germ cell differentiation in a non-cell-autonomous manner through limiting BMP signaling output by downregulating the BMP ligand (Dpp) and Dally—a regulator of BMP ligand diffusion. Interestingly, loss of dSet1—an H3K4 methyltransferase—results in phenotypes similar to those observed in dBre1 mutant ovaries. Genetic analysis suggests that dBre1 interacts with dSet1 to control both female GSC maintenance and germ cell differentiation.

RNA-binding proteins and noncoding RNAs

RNA-binding proteins, such as Musashi (Msi) (Siddall et al. 2006), Held-out-wings (HOW) (Monk et al. 2010), and the IGF-II mRNA-binding protein (Imp) (Toledano et al. 2012) are all required for male GSC maintenance, suggesting an important role of post-transcriptional regulation in the testis niche.

Epigenetic regulation is also controlled by noncoding RNAs, such as microRNAs (miRNAs) that regulate gene expression post-transcriptionally. Mature miRNAs are ∼22 nt, and they are processed from primary miRNAs by a set of evolutionally conserved enzymes, such as RNase III type endonucleases Partner of drosha (Pasha), Loquacious (Loqs), Dicer-1 (Dcr-1), and Argonaute-1 (Ago-1) (Filipowicz et al. 2008) in Drosophila. By base-pairing to the 3′ untranslated regions (3′UTR) of target mRNAs (Gu et al. 2009), mature miRNAs either control target mRNA stability or interfere with its translation (Vasudevan et al. 2007; Filipowicz et al. 2008). Mutations of miRNA biogenesis components dcr-1, loquacious, argonaut 1, and mei-P26 lead to the loss of female GSCs (Forstemann et al. 2005; Jin and Xie 2007; Park et al. 2007; Yang et al. 2007; Li et al. 2012, 2013). A couple of miRNAs have also been found to regulate GSC maintenance and differentiation in female gonads. For example, miR-184 (Iovino et al. 2009) and bantam (Yang et al. 2009) are both required to balance GSC maintenance vs. differentiation in ovaries. Another example is male germline-specific regulation of the Wnt signaling pathway by miRNAs. Both β-catenin and the downstream transcription factor TCF are downregulated by miRNAs in Drosophila. Loss of this antagonization leads to male germline differentiation defects and decreased fertility (Pancratov et al. 2013).

Another class of small noncoding RNAs, called piRNAs, is present in the both male and female gonads. The piRNAs are the most abundant class of small RNAs in gonads. The piRNAs repress transposons, provide immunity against transposons to protect the next generation, and function in maternal-to-zygotic transmission during early embryogenesis (Brennecke et al. 2008; Barckmann et al. 2015; Hermant et al. 2015; Iwasaki et al. 2015; Vourekas et al. 2016). Their biogenesis and functions will be described in detail in the following sections.

Mechanisms Controlling Mitotic Germ Cell Proliferation, Transition to Meiosis, and Dedifferentiation

Transit-amplification stage and mitosis-to-meiosis transition

The transit-amplification stage ensures that limited GSCs and their divisions have a high-throughput outcome for producing gametes. However, in both female and male germlines, this process needs to be tightly controlled since genetic lesions or epigenetic misregulation of gene expression may trap them as ever-dividing mitotic cells, and block entry into meiosis, leading, in turn, to either germline tumors or infertility (Clarke and Fuller 2006).

Cell-autonomous mechanisms:

A key differentiation factor encoded by the bam gene is expressed in transit-amplifying cells in both female and male gonads (McKearin and Spradling 1990; Gonczy et al. 1997). In GSCs from both sexes, BMP signaling activated by somatic cells in the niche represses bam transcription (Shivdasani and Ingham 2003; Kawase et al. 2004; Schulz et al. 2004). It is important that bam remain silenced in GSCs (Schulz et al. 2004; Insco et al. 2009; Monk et al. 2010). Ectopic expression of bam in GSCs induces precocious differentiation or cell death and, hence, loss of GSC phenotype (Ohlstein and McKearin 1997; Schulz et al. 2004; Sheng et al. 2009). A recent study shed light on the biochemical activity of Bam protein by showing that it assists a deubiquitinating enzyme and protects CycA from degradation (Ji et al. 2017).

In the female GSC lineage, Bam is absent in the GSC, but it is expressed in its immediate daughter cell CB. This abrupt change of Bam expression is regulated by a steep gradient of response to BMP signaling, both by a cis-acting transcriptional silencer repressing bam transcription in GSCs (Chen and McKearin 2003a,b; X. Song et al. 2004) and by a trans acting mechanism, such as the CB-specific degradation of Thickveins—a BMP signaling receptor (Xia et al. 2010, 2012). In addition, post-transcriptional regulation via different RNA-binding proteins contributes to sharpening the change of Bam expression and the decision between GSC self-renewal and CB differentiation (Chen and McKearin 2005). Further differentiation of the CB relies on the homolog of human Ataxin 2-Binding Protein 1 (A2BP1), which is expressed immediately after Bam in 4- to 16-cell germline cysts. Mutations in the A2BP1 gene lead to germline cyst differentiation defects, giving rise to germline tumors that result from mitosis-to-meiosis transition defects (Tastan et al. 2010). In addition, two H3K9 methyltransferases encoded by eggless/dSETDB1 and Su(var)3-9 act in a sequential manner with dSETDB1 in GSCs and early-stage germline, as discussed previously, while Su(var)3-9 mainly works in germ cells at the later stage. This temporal difference could underlie their distinct loss-of-function phenotypes with eggless mutants showing severe germline differentiation defects, whereas Su(var)3-9 mutants undergo normal oogenesis (Yoon et al. 2008).

In the male GSC lineage, Bam is required for the transition from mitotic spermatogonia to meiotic spermatocytes (McKearin and Spradling 1990; Gonczy et al. 1997). Bam protein is detectable in four- to 16-cell spermatogonia with a peak level in eight-cell spermatogonia (Gonczy et al. 1997). Examples of post-transcriptional regulation of bam include the HOW RNA-binding protein (Monk et al. 2010) and miR-7, both of which have been implicated in binding to bam mRNA and downregulating bam expression (Pek et al. 2009). Another RNA binding protein, Maelstrom (Mael), is required in spermatogonia to repress miR-7 and upregulate bam expression so that the transit-amplification can proceed normally (Pek et al. 2009). The transition from mitotic spermatogonia to meiotic spermatocyte is regulated by the accumulation of Bam to a threshold level. Expediting Bam accumulation or slowing down the transit-amplifying cell cycle results in insufficient proliferation before the transition to meiosis, as demonstrated by spermatocyte cysts with <16 cells. On the other hand, inhibition of Bam accumulation, or facilitating the transit-amplifying cell cycle, results in extra round(s) of mitosis before the transition to meiosis, as shown in spermatocyte cysts with >16 cells (Insco et al. 2009). Therefore, expression of Bam needs to be tightly controlled during the transit-amplification stage in the male germline.

Another differentiation gene, benign gonial cell neoplasm (bgcn), has mutant phenotypes similar to bam in both male and female germlines (Gonczy et al. 1997). Studies in the female germline demonstrate that Bam and Bgcn form a protein complex to antagonize GSC self-renewal factors, and promote differentiation gene expression in transit-amplifying cells (Y. Li et al. 2009). It has been demonstrated that the Trim-NHL tumor suppressor homolog Mei-P26 has a reciprocal regulation with Bam whereby Mei-P26 initially promotes Bam protein accumulation in early transit-amplifying cells. Increased Bam acts with Bgcn to bind the 3′UTR of Mei-P26, and, consequently, repress translation of Mei-P26 in late transit-amplifying cells (Insco et al. 2012). Recent studies have revealed another RNA-binding protein, Tumorous testis (Tut), that acts in synergy with Bam-Bgcn for the translational repression of Mei-P26 (Chen et al. 2014). Indeed, post-transcriptional regulation of gene expression is a widely used mechanism, particularly in the germline. A very recent study reports the generality of this mechanism in the male germline (Shan et al. 2017). In addition, post-translational regulation also directly, or indirectly, regulates Bam protein function. Specifically, a Drosophila homolog of the highly conserved LAMMER/Cdc2-like kinase (CLK), called Doa, has been shown to regulate the mitosis-to-meiosis switch in the male germline through regulating Bam protein (Zhao et al. 2013).

In order to study the transcriptional profile and chromatin state in transit-amplifying cells, bam or bgcn mutant testes were used for transcriptome profiling because they are enriched with overproliferative spermatogonial cells (Terry et al. 2006; Gan et al. 2010a; Chen et al. 2011). High-throughput mRNA sequencing (RNA-seq) studies reveal that both chromatin remodeling factors and histone-modifying enzymes have enriched transcription in bam testes compared to wild-type testes (Gan et al. 2010a). Furthermore, ChIP followed by high-throughput sequencing (ChIP-seq), revealed a distinct chromatin structure in bam testes (Gan et al. 2010b). In mouse embryonic stem cells, differentiation genes have both repressive H3K27me3 and active H3K4me3 modifications (i.e., “bivalent” chromatin signature), as well as stalled RNA polymerase II (Pol II, i.e., “poised” genes), at their promoter regions (Bernstein et al. 2006; Buszczak and Spradling 2006; Guenther et al. 2007). By contrast, differentiation genes required for spermatocyte maturation and spermiogenesis are either enriched with H3K27me3 only, or deprived of both H3K4me3 and H3K27me3, in bam testes, and they are not associated with stalled Pol II (Gan et al. 2010b). This distinct chromatin structure may prevent ectopic transcription of the differentiation genes in transit-amplifying cells. On the other hand, it suggests that dramatic changes at the promoter region of differentiation genes are needed to turn on their robust transcription in spermatocytes.

In addition to these genome-wide studies, it was reported that an epigenetic reader-encoding Plant Homeodomain Finger 7 (PHF7) gene is specifically expressed in GSCs and transit-amplifying cells. PHF7 recognizes active H3K4me2 histone modification and is required for GSC maintenance and proper spermatogonial differentiation (Yang et al. 2012). Further studies to identity the target genes of PHF7, which should be enriched with H3K4me2 or H3K4me3, will shed light on its in vivo roles.

Non-cell-autonomous mechanisms:

The Epidermal growth factor (Egf) signaling pathway plays an important role in the regulation of the mitosis-to-meiosis switch. The Egfr (Egf receptor) ligand Spitz is processed by Stet—a transmembrane protease—in germ cells (Schulz et al. 2002). Activated Spitz then acts on Egfr expressed in somatic cells (Kiger et al. 2000). Egf signaling acts through the guanine nucleotide exchange factor (GEF) Vav to activate Rac-type small GTPases, which are antagonized by the Rho-type small GTPases (Sarkar et al. 2007). Egfr signaling acts in cyst cells to restrict GSC self-renewal and spermatogonial proliferation, while promoting GSC-to-GB and spermatogonia-to-spermatocyte transitions (Kiger et al. 2000). Egfr signaling decreases the frequency of GSC divisions in the adult, but not larval, testes, suggesting a temporal mode of Egfr regulation (Parrott et al. 2012). In addition, mutations in a serine/threonine kinase signal transducer encoded by raf result in phenotypes similar to the Egfr mutant, suggesting that the receptor tyrosine kinase (RTK) pathway is, in general, required in cyst cells for proper transit-amplification (Tran et al. 2000). The direct target genes for the Egfr/Raf pathway have not been identified; however, because compromised Egf signaling leads to defects in germline-soma interaction and overproliferation of spermatogonial cells, it is possible that the target genes regulate proper encapsulation of germ cells by cyst cells (Schulz et al. 2002; Sarkar et al. 2007). A recent study has shown that a chromatin factor encoded by the Enhancer of Polycomb [E(Pc)] gene acts in the CySC lineage to regulate multiple signaling pathways, including both EGF and JAK-STAT pathways, in order to promote both CySC and GSC differentiation. In addition, consistent with biochemical data showing E(Pc) as a component of the NuA4 (nucleosome acetyltransferase of H4) histone acetyltransferase (HAT) complex (Galarneau et al. 2000; Boudreault et al. 2003; Chittuluru et al. 2011), inactivation of the Drosophila NuA4 homolog, Tip60, in the CySC lineage resembles E(Pc) loss-of-function phenotype, suggesting that they may act together in vivo (Feng et al. 2017). Another recent study revealed that the endocytic process in the CySC lineage is required to prevent overproliferation of transit-amplifying germ cells in testis, which is accomplished through both JNK and BMP signaling pathways (Tang et al. 2017).

Furthermore, a nuclear envelope component, Nucleoporin98-86, regulates proper GSC-to-GB and spermatogonia-to-spermatocyte transitions, and functions upstream of BMP, JAK-STAT, and Egfr signaling pathways (Parrott et al. 2011). Interestingly, another study showed that nuclear lamina regulates specific nucleoporin distributions and promotes nuclear localization of phosphorylated ERK—the downstream effector of the Egf pathway (Chen et al. 2013). These results highlight the importance of nuclear structure in regulating cellular differentiation during spermatogenesis.

Dedifferentiation

Cell-autonomous mechanisms:

In both female and male GSC lineages, partially differentiated mitotic germ cells could undergo a dedifferentiation process to return to the niche and become GSC-like cells (Brawley and Matunis 2004; Kai and Spradling 2004). During aging (Wallenfang et al. 2006; Cheng et al. 2008) and tissue regeneration (Sheng et al. 2009), lost GSCs could be replenished by dedifferentiation to maintain tissue homeostasis. However, once the meiotic program is initiated, as in spermatocytes, dedifferentiation could no longer be detected (Brawley and Matunis 2004; Wallenfang et al. 2006; Sheng et al. 2009), suggesting that the mitotic spermatogonial cells have unique characteristics permissive for dedifferentiation. Similar irreversible commitment may also apply to female meiotic germ cells because only four- to eight-cell transit-amplifying cells have been reported to undergo dedifferentiation in the ovary (Kai and Spradling 2004). It has recently been shown that RNA-binding Fox 1 (Rbfox1) represses pumilio mRNA translation. Because of the essential roles of Pumilio in early-stage germ cells, including GSCs, ectopic Pumilio is expressed in Rbfox1 mutants, and promotes dedifferentiation of germline cysts to become GSC-like cells in the ovary (Carreira-Rosario et al. 2016).

Non-cell-autonomous mechanisms:

Using live cell imaging, it has been observed that the dedifferentiated spermatogonial cyst undergoes fragmentation to become individual cells that form actin-based protrusions to make initial contact with the stem cell niche (Sheng et al. 2009), suggesting potential extrinsic cues from the niche to guide dedifferentiation. Indeed, it has been reported that the aminopeptidase Slamdance is highly expressed in the hub cells. Slamdance is both necessary and sufficient to promote dedifferentiation during homeostasis and regeneration, and such activity depends on its enzymatic function. These data showed that cells and molecules in the stem cell niche regulate the dedifferentiation process. Slamdance also has a cell-type-specific expression in the female GSC niche cells, and may play a similar role for the dedifferentiation process in the female germline (Lim et al. 2015).

Despite increasing knowledge about the intrinsic factors and extrinsic cues for dedifferentiation, the extent to which dedifferentiated GSC-like cells behave like bona fide GSCs remains to be elucidated. For example, it has been shown that dedifferentiated GSC-like cells tend to have misoriented centrosomes, which lead to cell cycle arrest because of a centrosome orientation checkpoint in male GSCs (Cheng et al. 2008; Inaba et al. 2010, 2015; Yuan et al. 2012). Therefore, it is unclear whether dedifferentiated GSC-like cells could reenter differentiation and give rise to fully functional gametes without any defects. Addressing this intriguing question would need double lineage tracing to trace those GSC-like cells arising from dedifferentiation and reentering differentiation, as well as functional analyses of those differentiated gametes from dedifferentiated GSC-like cells.

piRNAs and piRNA Pathway Function to Protect the Germline Genome

Transposons are autonomous elements present in all eukaryotic organisms. Their content in the genome of higher eukaryotes varies between 10 and 80%; they constitute 23% of the D. melanogaster genome. Transposons are involved in the regulation of gene expression, as well as both evolution and speciation. However, the ability of transposons to transpose from one site to another in the genome demands tight regulation of their movements. Transposition in the germline genome is particularly important for transposons to propagate in a population. Previously, piRNAs present in both the male and female gonads were introduced as another class of small noncoding RNAs. It is this unique class of small RNAs that carefully safeguard the germline genome, ensuring fitness of the offspring. Genome-wide screens in Drosophila have revealed that piRNA biogenesis requires 69 or more genes and that their distinct subsets are expressed in the germline and somatic gonadal cells (Czech et al. 2013; Handler et al. 2013). In this section, we focus on the biogenesis of piRNAs in germ cells, as well as somatic gonadal cells.

Transposons in Drosophila

D. melanogaster possesses >49 families of long terminal repeat (LTR) transposons (Kaminker et al. 2002). The transposition mechanisms for many transposon families have been studied. The most studied example among the LTR family members is gypsy, which is composed of three parts: a Gag-like protein containing a nucleocapsid region, a protease-polymerase fusion protein, and the envelope (Mejlumian et al. 2002). With the encoded coat proteins, gypsy is capable of exiting the follicle cells and infecting the neighboring oocyte. Another LTR retrotransposon called ZAM has a replicative cycle similar to that of gypsy, but it can be transmitted to the oocyte by the vitellogenin secretion pathway (Leblanc et al. 2000; Brasset et al. 2006). LTR families are often activated in ovarian somatic cells and transmitted to the oocyte, threatening the genome stability of the oocyte. By contrast, non-LTR families tend to be activated in nurse cells, and are deposited to the oocyte via the cytoplasmic bridges called ring canals (Chambeyron et al. 2008). Hence, active transposon mechanisms function both in the germline and somatic gonadal cells.

In Drosophila, though transposons threaten the genome, they are also essential for the integrity of both centromeres and telomeres (Pardue and DeBaryshe 2003; Wong and Choo 2004). In most other species, telomeres are composed of simple repeats and are maintained by telomerase, but, in Drosophila, telomeres consist of three non-LTR transposons, namely HeT-A, TART, and TAHRE (Pardue and DeBaryshe 2003, 2011; Abad et al. 2004). To properly maintain the telomere, the copy number of these transposons at telomeres is strictly regulated (Fanti et al. 1998; Perrini et al. 2004; Frydrychova et al. 2008; Pardue and DeBaryshe 2011).

piRNAs and PIWI proteins

Studies on the piRNA pathway in Drosophila ovaries have expanded our knowledge about piRNA pathway function (Iwasaki et al. 2015). The piRNAs in Drosophila are 23 to 29 nucleotides in length and are the most abundant small RNAs in gonads (Balakireva et al. 1992; Aravin et al. 2001, 2003, 2004). The piRNAs were first recognized for their role in suppressing the Stellate protein in the male germ cells (Aravin et al. 2001, 2004). Shortly thereafter, it was reported that Drosophila ovaries and embryos contain abundant repeat-associated small RNAs called rasiRNAs (Vagin et al. 2006). Later, they were renamed as piRNAs, as they are produced by and associate with PIWI-family proteins to suppress transposons (Saito et al. 2006; Brennecke et al. 2007; Gunawardane et al. 2007).

The piRNA pathway in Drosophila is active in both the germline and somatic gonadal cells to counter transposons that threaten to invade the germline through distinct routes. The piRNA pathway silences transposons post-transcriptionally by triggering degradation of their transcripts, as well as transcriptionally by silencing transposon loci (Vagin et al. 2006; Lim et al. 2009; Le Thomas et al. 2013; Rozhkov et al. 2013). Although a different subset of proteins function in piRNA biogenesis in germline and somatic cells, PIWI-family proteins—a subclade of the Argonaute family—are central in the piRNA pathway. Piwi, the founder member of PIWI-family proteins, is present in both germline and somatic cells. Two other PIWI-family proteins, namely Aubergine (Aub) and Argonaute3 (Ago3), are required for piRNA production in germ cells. These proteins contain PAZ (Piwi-Argonaute-Zwille) and PIWI domains. The PAZ domain also harbors an oligonucleotide/oligosaccharide-binding-fold, which binds to single-stranded nucleic acids (Lingel et al. 2003; Yan et al. 2003). The PIWI domain is structurally similar to that of RNase H enzymes (J. Song et al. 2004). Crystal structure analysis suggested that the PAZ domain forms a pocket for the target RNA, while the PIWI domain cleaves its bound RNA (Yan et al. 2003; J. Song et al. 2004).

The piRNAs are processed from longer precursor molecules. The piRNA precursor transcripts are produced from discrete loci, termed piRNA clusters (Brennecke et al. 2007). These clusters are composed of fragmented copies of transposons in the genome, and serve as heritable sequence repositories for transposon repression. In the gonadal somatic cells, piRNAs are processed from piRNA cluster transcripts, which are in antisense orientation, to active transposons, in a linear mode called primary processing. While in germ cells, piRNAs are generated from both cluster and transposon transcripts in two different modes: primary processing and secondary amplification cycle (Brennecke et al. 2007; C. Li et al. 2009; Malone et al. 2009). The piRNAs in both cell types are loaded onto Piwi to form the Piwi-piRNA complex, which subsequently translocates into the nucleus for transcriptional silencing of transposons.

Source of piRNAs—piRNA clusters and transcription of piRNA precursors

Generally, piRNA clusters can be classified on the basis of transcription of precursors. Most piRNA clusters that participate in piRNA biogenesis in ovarian somatic cells are transcribed in one direction, and thus called uni-strand clusters. By contrast, active clusters in germ cells are transcribed in a convergent manner from both directions, and thus called dual-strand clusters (reviewed by Hirakata and Siomi 2016).

Active piRNA clusters in germ cells and generation of piRNA precursors:

Most clusters active in germ cells are located in pericentric or subtelomeric regions, which are heterochromatic in nature, and yet they are readily transcribed and processed into piRNAs (Klattenhoff et al. 2009; Malone et al. 2009; Mohn et al. 2014; Z. Zhang et al. 2014). Although piRNA clusters are transcribed by RNA Pol II, majority of piRNA precursors are reported as nonpolyadenylated and lack capping at the 5′-end (Mohn et al. 2014; Z. Zhang et al. 2014; Chen et al. 2016) (Figure 3). Notably, piRNA precursor transcription depends on H3K9me3, and loss of a histone methyltransferase encoded by setDB1/egg causes severe reduction in cluster transcript levels from both uni- and bidirectional clusters (Rangan et al. 2011). A recent study showed that loss of rpp30, which encodes a subunit of RNase P for tRNA processing, leads to reduction of H3K9me3 at piRNA clusters and reduction in levels of cluster transcripts, supporting the importance of H3K9me3 for cluster transcription (Molla-Herman et al. 2015).

Transcription of bidirectional clusters in the germline. The H3K9me3 marks are deposited on the piRNA clusters by the histone methyl transferase, Egg, and also by Piwi at selected clusters. Rhino recognizes the H3K9me3 marks and prompts binding of the Rhi-Del-Cuff complex (RDC) to the piRNA cluster. The RDC licenses noncanonical transcription from piRNA clusters by recruiting RNA-pol II for transcription of piRNA clusters. Longer transcripts in the antisense direction to transposons are transcribed. Cuff represses Cleavage and Polyadenylation Specificity Factor (CPSF), preventing the termination of cluster transcription, as well as its splicing. In addition, Cuff recruits components of Transcription Export (TREX) complex to stabilize and accumulate cluster transcripts. The Rhi partner, UAP56, binds to the cluster transcripts in order to export them to nuage.
Figure 3

Transcription of bidirectional clusters in the germline. The H3K9me3 marks are deposited on the piRNA clusters by the histone methyl transferase, Egg, and also by Piwi at selected clusters. Rhino recognizes the H3K9me3 marks and prompts binding of the Rhi-Del-Cuff complex (RDC) to the piRNA cluster. The RDC licenses noncanonical transcription from piRNA clusters by recruiting RNA-pol II for transcription of piRNA clusters. Longer transcripts in the antisense direction to transposons are transcribed. Cuff represses Cleavage and Polyadenylation Specificity Factor (CPSF), preventing the termination of cluster transcription, as well as its splicing. In addition, Cuff recruits components of Transcription Export (TREX) complex to stabilize and accumulate cluster transcripts. The Rhi partner, UAP56, binds to the cluster transcripts in order to export them to nuage.

Transcription from piRNA clusters requires specialized complexes because many such clusters are present in gene-poor regions and lack canonical promoters, as well as canonical intron-exon boundaries. In the germline, the RDC (Rhino-Deadlock-Cutoff) protein complex licenses transcription from piRNA clusters (Mohn et al. 2014; Z. Zhang et al. 2014) (Figure 3). RDC is composed of Rhino (Rhi), a homolog of Heterochromatin Protein 1a (HP1a), Deadlock (Del) without any conserved domain, and Cutoff (Cuff), an Rai1/Dom3Z-family protein (Chen et al. 2007; Klattenhoff et al. 2009; Mohn et al. 2014; Z. Zhang et al. 2014). The chromodomain on Rhino recognizes H3K9me3 marks at clusters for the binding of RDC complex (Mohn et al. 2014; Z. Zhang et al. 2014; B. Yu et al. 2015). However, the detailed mechanism underlying the establishment of H3K9me3 at clusters is not known. The RDC complex binds to all dual-strand piRNA clusters, which are active in the germline, but not to the clusters active in somatic cells. The RDC complex prevents transcription termination of adjacent genes to allow for transcription of the clusters. The RDC complex also licenses transcription from noncanonical promoters in piRNA clusters (Le Thomas et al. 2014; Mohn et al. 2014; Z. Zhang et al. 2014). Loss of RDC complex leads to an increase in splicing of cluster transcripts, which could potentially destabilize these cluster transcripts (Mohn et al. 2014; Z. Zhang et al. 2014; Chen et al. 2016). Indeed, it was shown that RDC tethering to a transgene leads to intron stabilization and transcription beyond the polyA site. Aravin and colleagues suggest that Cuff in the RDC complex is necessary to prevent binding of Cleavage and Polyadenylation Specific Factor (CPSF) for a continuous transcription throughout the piRNA cluster (Chen et al. 2016). Cuff is also required for stabilizing the noncapped cluster transcripts, by antagonizing cluster transcript destabilization by 5′-3′ exonuclease dRat1 (Chen et al. 2016). The RDC complex is important not only for transcription from the piRNA clusters, but also for transcription and piRNA production from transgenes (Z. Zhang et al. 2014). In addition, RDC complex also participates in channeling the cluster transcripts to the piRNA processing site for piRNA generation.

Recently, it was reported that Tho5 and other THO subunits of the Transcription/Export (TREX) complex are recruited to piRNA clusters by Cuff, and are loaded onto cluster transcripts (Hur et al. 2016) (Figure 3). Tho proteins are required for accumulation of nascent cluster transcripts in a splicing-independent manner in nucleus. Another protein, UAP56, a component of the nuclear pore complex, binds to cluster transcripts in a Rhi-dependent manner. Through interaction with a nuage component, Vasa, an RNA helicase, UAP56 likely functions to export cluster transcripts to nuage, the site of piRNA processing (Zhang et al. 2012) (see below).

Transcription of piRNA clusters active in the ovarian somatic cells:

Somatic piRNA clusters do not require the RDC complex for their transcription. Details about their transcription remain limited. In contrast to germline cluster transcripts, somatic piRNA cluster transcripts are polyadenylated. A piRNA cluster, flamenco, located near the pericentric region of X-chromosome, is particularly active in the ovarian follicle cells. The flamenco locus has fragmented copies of transposons expressed in somatic cells, such as ZAM and gypsy (Prud’homme et al. 1995; Sarot et al. 2004; Desset et al. 2008). The flamenco-derived transcripts are in antisense orientation to the active transposons. Transcription from the flamenco locus is reported to be dependent on the transcription factor Cubitus interruptus (Ci). The flamenco-derived transcripts are alternatively spliced, probably for diversity (Goriaux et al. 2014).

piRNA biogenesis in somatic gonadal cells via primary processing

The somatic piRNA cluster transcripts are processed into piRNAs in the cytoplasm (Haase et al. 2010; Saito et al. 2010). Mechanisms of transport of somatic piRNA precursors from the nucleus to cytoplasmic processing sites remain elusive. Most primary piRNA processing components are localized to mitochondria. Yb-body and Zucchini (Zuc)-associated proteins have been shown to coordinate piRNA processing and loading of piRNAs to Piwi (Haase et al. 2010; Saito et al. 2010; Handler et al. 2011; Qi et al. 2011). The 5′-end of piRNAs is generated by an exonuclease, Zucchini (Zuc), which localizes to the mitochondrial surface (Ipsaro et al. 2012; Nishimasu et al. 2012). Zuc generates 5′-ends of piRNAs, the resultant ends of which are not enriched with U (Uracil). However, the 5′-ends of mature piRNAs are remarkably biased for U. In vitro study of the silkworm Piwi homolog Siwi suggested that this bias for U at the 5′-end could be introduced during piRNA loading to Siwi (Kawaoka et al. 2011). The 3′-ends of piRNAs are speculated to be generated while piRNAs are being loaded to PIWI proteins. Zuc is thought to function in 3′-end formation of piRNAs, although the resultant products have additional nucleotides at the 3′-end. These extra nucleotides are proposed to be trimmed by another exonuclease (Kawaoka et al. 2011). A Tudor-domain protein, PAPI, has also been suggested to be important for 3′-end trimming of piRNAs, although PAPI does not have any nuclease domain (Honda et al. 2013). The piRNAs are loaded to Piwi at Yb-bodies present at the outer mitochondrial surface close to Zuc (Olivieri et al. 2010; Saito et al. 2010). Piwi forms a complex with Yb-body proteins, such as Vretreno (Vret), Shutdown (Shu), and Armitage (Armi). Severe reduction of piRNAs in vret, shu, and armi mutants leads to cytoplasmic accumulation of Piwi, which likely results from the failure of piRNA loading on Piwi (Haase et al. 2010; Olivieri et al. 2010, 2012; Saito et al. 2010; Handler et al. 2011; Qi et al. 2011). In addition, Shu functions for piRNA loading onto Piwi with the help of Hsp 83 (Handler et al. 2011).

The Yb-body, where many proteins required for primary piRNA processing are localized, is named after the Yb protein containing both an RNA-helicase and a Tudor domain (King et al. 2001; Szakmary et al. 2009). Yb proteins bind to the flamenco-derived piRNA precursors through their DEAD box domain, channel them to the Yb-body, and stabilize the piRNA processing apparatus at Yb-bodies (Murota et al. 2014). Although all Yb-body components are required for piRNA biogenesis, their exact function remains unknown.

piRNA biogenesis in germ cells

In addition to primary piRNA processing, germ cells have an additional piRNA processing machinery called secondary amplification or the ping-pong cycle. This piRNA biogenesis mechanism allows more robust piRNA production against transposons and provides more flexibility to adapt for newer transposon threats.

Primary piRNA biogenesis in germ cells:

The mechanistic details of primary piRNA biogenesis in germ cells remain unknown. However, many components required for primary piRNA processing in ovarian somatic cells, such as Zuc, Armi, Gasz, Shu, Mino, and HSP90, are expressed in germ cells and suggested to function for primary piRNA processing in germ cells.

Secondary piRNA processing in germ cells; ping-pong amplification:

Secondary piRNA processing is a feed-forward amplification loop involving two PIWI-family proteins, Aub and Ago3, and it takes place at the nuage in germ cells where the key components of this secondary processing are localized. The Aub-bound antisense piRNAs target the transposon transcripts, and piRNA-loaded Aub harboring slicer activity cleaves the transposon transcript, generating the 5′-end of transposon-derived sense piRNA. The 3′-end of the piRNAs is generated either by slicer activity or by Zucchini. Ultimately the 3′ end generated by both the mechanisms requires trimming by an exonuclease Nibbler, generating mature Ago3-bound sense piRNAs of correct size. (Nishimasu et al. 2012; Hayashi et al. 2016). In turn, piRNA-bound Ago3 targets and cleaves cluster transcripts to generate more antisense piRNAs loaded onto Aub or Piwi, and the processing cycle amplifies piRNAs in a feed-forward loop. This processing leads to a significant 10-nt overlap between Aub and Ago3-bound piRNA sequences, with a U at position 1 of Aub-bound piRNAs and an adenine (A) at position 10 of Ago3-bound piRNAs (Brennecke et al. 2007; Gunawardane et al. 2007). Computational analysis suggested that Aub preferentially binds to mRNA targets with an A at the position opposing the first base of their piRNA partner. Upon target slicing and subsequent piRNA maturation, the A of target mRNA comes to the 10th position in a ping-pong-derived piRNA (Wang et al. 2014).

The secondary piRNAs also trigger production of Zuc-dependent, 3′-directed phased piRNAs. Phasing is triggered by so-called responder piRNAs, which result from Ago3-piRISC activity in the ping-pong cycle. The piRNAs downstream of responder piRNAs are associated with Piwi. These piRNAs are designated as trailer piRNAs. The responder piRNAs and trailer piRNAs show marked phasing with a ∼27 nt interval and a striking bias for U at the 5′-end. Production of these trailer piRNAs depends on Zuc. These findings also suggest that piRNA 3′-ends are defined by Zuc endonucleolytic activity (Han et al. 2015; Mohn et al. 2015) (Figure 4). Biogenesis of such phased piRNAs spreads piRNA production beyond the target cleavage sites of Ago3 and Aub, thus allowing sequence diversification in the piRNA pool, which could target transposon threat in an adaptive manner by both TGS (Transcriptional Gene Silencing) and PTGS (Post-Transcriptional Gene Silencing) mechanisms (Han et al. 2015; Homolka et al. 2015; Mohn et al. 2015; Sato et al. 2015; Senti et al. 2015; Wang et al. 2015; Webster et al. 2015).

piRNA biogenesis mechanisms in germ cells. (A) The piRNA production mechanisms at nuage. (i) Ping-pong amplification: Aub-bound antisense piRNA leads to the cleavage of the transposon transcript. The 5′-end of piRNAs is produced by Zucchini, while the 3′-end is either generated by Zucchini or by a splicer complex. The resulting 3′ end is trimmed by an exoribonuclease Nibbler, and subsequently, is methylated at 2′-O. The piRNA-Ago3 complex cleaves the anti-sense piRNA precursor transcript. Trimming of the resultant piRNAs occurs as described above. This creates a feed-forward amplification loop, called the ping-pong cycle. (ii) Phasing: The Ago3 cleaved transcript at 5′ end, bound by Aub, apart from going to ping-pong amplification, enters phasing. Piwi is proposed to bind downstream of the Aub cleavage site on a transposon transcript. Zuc cleaves at 5′- and 3′-ends of the transcript bound by Piwi. The Piwi-bound piRNAs, thus resulting from Zuc mediated cleavage, have their 5′-end 27 nucleotides apart from each other, leading to production of piRNAs in a phased manner. This phasing produces piRNAs beyond the Aub and Ago3 cleavage sites and broadens the piRNA repertoire to target transposon threats. (B) Tudor domain proteins, Krimp and Qin, ensure that the ping-pong cycle occurs at the nuage. Vas receives the transcripts of piRNA clusters at nuage. Krimp anchors both Aub and Ago3 at nuage, while Qin inhibits the loading of sense piRNAs on Aub, thus enforcing heterotypic ping-pong.
Figure 4

piRNA biogenesis mechanisms in germ cells. (A) The piRNA production mechanisms at nuage. (i) Ping-pong amplification: Aub-bound antisense piRNA leads to the cleavage of the transposon transcript. The 5′-end of piRNAs is produced by Zucchini, while the 3′-end is either generated by Zucchini or by a splicer complex. The resulting 3′ end is trimmed by an exoribonuclease Nibbler, and subsequently, is methylated at 2′-O. The piRNA-Ago3 complex cleaves the anti-sense piRNA precursor transcript. Trimming of the resultant piRNAs occurs as described above. This creates a feed-forward amplification loop, called the ping-pong cycle. (ii) Phasing: The Ago3 cleaved transcript at 5′ end, bound by Aub, apart from going to ping-pong amplification, enters phasing. Piwi is proposed to bind downstream of the Aub cleavage site on a transposon transcript. Zuc cleaves at 5′- and 3′-ends of the transcript bound by Piwi. The Piwi-bound piRNAs, thus resulting from Zuc mediated cleavage, have their 5′-end 27 nucleotides apart from each other, leading to production of piRNAs in a phased manner. This phasing produces piRNAs beyond the Aub and Ago3 cleavage sites and broadens the piRNA repertoire to target transposon threats. (B) Tudor domain proteins, Krimp and Qin, ensure that the ping-pong cycle occurs at the nuage. Vas receives the transcripts of piRNA clusters at nuage. Krimp anchors both Aub and Ago3 at nuage, while Qin inhibits the loading of sense piRNAs on Aub, thus enforcing heterotypic ping-pong.

Nuage as a site for ping-pong cycle in germ cells:

Both PIWI-family proteins, Aub and Ago3, involved in ping-pong amplification are found at the perinuclear foci in the cytoplasmic face called nuage (Brennecke et al. 2007). Nuage is an amorphous, electro-dense structure present at the cytoplasmic face of nuclear membrane (reviewed in Eddy 1975). Nuage has been widely recognized, albeit occasionally by different names, as a hallmark of germ cells in animals, and yet its function remained unknown for a long time.

Studies have shown that different kinds of proteins localize to nuage and participate in piRNA processing, including RNA helicase Vasa and Spindle-E (SpnE). Most nuage components are Tudor domain proteins, such as Tudor (Tud), Qin/Kumo, Tejas (Tej), Tapas (Tap), Krimp (Krimp), and SpnE. Other proteins include HMG box protein Mael and nucleases, such as Zuc and Squash (Squ) (Lim and Kai 2007; Pane et al. 2007; Malone et al. 2009; Patil and Kai 2010; Zhang et al. 2011; Anand and Kai 2012; Sienski et al. 2012; Patil et al. 2014). Tudor domains preferably bind to symmetrical demethylation of Arginine (sDMA) sites on PIWI-family proteins (Nishida et al. 2009). Tud binds to Aub and Ago3 in an sDMA-dependent manner and ensures proper binding of piRNAs to Aub and Ago3 (Nishida et al. 2009). However, the interaction between Tudor domains and PIWI-family proteins may not always be dependent on sDMA (Patil and Kai 2010).

The details of function of many nuage proteins have been revealed by a series of recent studies. For example, Vasa interacts with UAP56 and likely functions for transport of piRNA precursors to the nuage (Zhang et al. 2012). The Tudor domain protein Krimp maintains Aub and Ago3 on the nuage for proper ping-pong amplification. Krimp interacts with Ago3, which does not require piRNA loading on Ago3 or Arginine methylation. However, the binding of Krimp to Aub requires arginine methylation of Aub. Krimp promotes arginine methylation on Ago3, and prevents the loading of antisense piRNA on it (Sato et al. 2015; Webster et al. 2015) (Figure 4). The nuage components Tej and Tap function synergistically for piRNA production, and are required for maintenance of all other piRNA pathway components at the nuage (Patil and Kai 2010; Patil et al. 2014). Another Tudor domain protein, Qin/Kumo, is also required for proper maintenance of Aub and Ago3 at nuage, and it prevents the loading of sense piRNAs to Piwi and Aub, thereby enforcing heterotypic Aub: Ago3 ping-pong (Zhang et al. 2011; Anand and Kai 2012; Wang et al. 2015). The function of piRNA pathway proteins explains, to some extent, the observed mutual dependence for their localization to nuage based on a genetic hierarchical relationship (Lim and Kai 2007; Anand and Kai 2012; Patil et al. 2014). For example, Vasa, which is genetically farther upstream, also becomes functionally upstream. Similarly, Krimp, Qin/Kumo, Tej, and SpnE support ping-pong, and, therefore, also support Aub and Ago3 on nuage. The piRNA biogenesis requires a multistep mechanism, and, as such, a number of proteins assist Aub and Ago3 for piRNA generation and piRNA-mediated degradation of transposon transcripts.

piRNA-mediated transcriptional silencing of transposons

Transposon transcripts are post-transcriptionally silenced by piRNAs at the nuage, possibly at cytoplasmic processing bodies where piRNA pathway components are localized in later stages of oogenesis (Lim et al. 2009). In addition, piRNAs transcriptionally repress transposons, and piRNA loss leads to concomitant loss of repressive histone marks at transposons in the Drosophila female germline and somatic cells (Klenov et al. 2007, 2014; Sienski et al. 2012; Le Thomas et al. 2013; Rozhkov et al. 2013).

Piwi is a key mediator of transcriptional silencing:

The piRNA loading on Piwi is important for its entry in to the nucleus. Piwi-piRISC enters the nucleus to transcriptionally silence the transposons in both somatic and germ cells (Le Thomas et al. 2013; Rozhkov et al. 2013). Piwi loss does, indeed, cause an increase in RNA polymerase II occupancy at promoter regions of transposons, as well as reduction of H3K9me3 levels (Sienski et al. 2012; Czech et al. 2013; Huang et al. 2013; Le Thomas et al. 2013; Rozhkov et al. 2013; Klenov et al. 2014). Although the enrichment of Piwi at transposon loci has still not been established, predominant loading of antisense piRNAs to Piwi led to the speculation that Piwi-piRISC scans for nascent transposon transcripts to enforce transcriptional repression (Han et al. 2015; Homolka et al. 2015; Mohn et al. 2015; Sato et al. 2015; Senti et al. 2015; Wang et al. 2015; Webster et al. 2015). Notably, although Piwi is equipped with a slicer domain, this domain is dispensable for piRNA production and transposon silencing (Darricarrere et al. 2013).

Distinct components act together with Piwi:

Piwi-piRISC interacts with proteins to enforce transposon silencing. Recent work has identified several downstream components of Piwi-piRISC for transposon transcriptional silencing in both the germline and somatic cells. Many proteins have been shown to act downstream of Piwi-piRISC for H3K9me3 enrichment at transposon loci.

The double CHHC zinc finger protein, gametocyte-specific factor 1 (GTSF1), has been shown as a downstream Piwi-piRISC partner in both germline and somatic cells in the ovary (Dönertas et al. 2013; Ohtani et al. 2013). GTSF1 interacts with Piwi to establish H3K9me3 at the transposon loci for repression of transposons (Dönertas et al. 2013; Ohtani et al. 2013). The downstream nature of GTSF1 is suggested by unchanged piRNA levels upon GTSF1 loss, but transposon derepression profile and loss of H3K9me3 at transposon loci mimic what has been observed upon Piwi loss (Dönertas et al. 2013; Ohtani et al. 2013). However, the precise molecular mechanisms that explain how GTSF1 engages Piwi in TGS remain elusive. Another protein, Panoramix/Silencio (Panx), is vital for transposon repression through transcriptional silencing (TS) in germ cells (Sienski et al. 2015; Y. Yu et al. 2015). Panx interacts with Piwi and recruits the methyltransferase Egg to deposit H3K9me3 for heterochromatin formation at transposon loci. The loss of Panx leads to global transposon derepression without any reduction of piRNAs, suggesting its role as a downstream partner of Piwi-piRISC (Sienski et al. 2015; Y. Yu et al. 2015). In addition, Piwi-piRISC may silence transposons through the removal of H3K4me2 (Fadloun et al. 2013; Klenov et al. 2014). Depletion of the Lsd1 demethylase in Drosophila ovaries resulted in derepression of a subset of transposons, which is independent of Panx (Czech et al. 2013).

The function of Piwi-RISC for TGS of transposons is through not only histone modifications but also mediated by chromatin binding proteins. For example, Piwi loss leads to reduction of HP1a at few transposons in both germline and somatic cells in the ovary (Ohtani et al. 2013; Klenov et al. 2014). In germ cells, HP1a loss leads to derepression of transposons (Wang and Elgin 2011). However, in somatic cells, transposon derepression resulting from HP1a loss is not necessarily correlated with that caused by Piwi loss (Ohtani et al. 2013). This suggests that other chromatin regulators could repress some transposons independent of Piwi. Functions of these proteins also overlap, for example, HP1a likely enforces transposon silencing downstream of Piwi-piRISC by recruiting SetDB1 via interaction with Piwi-RISC tethered at the transposon loci (Brower-Toland et al. 2007; Sienski et al. 2015). Recently, histone H1 was also shown to be one of the downstream components of Piwi-piRISC for transposon repression in ovarian somatic cells, functioning in parallel with HP1a. However, unlike HP1a, H1 function is independent of H3K9Me3 marks (Iwasaki et al. 2016). Hence, it is possible that Piwi recruits different downstream factors, such as HP1a and H1, for more efficient transposon repression.

Current studies suggest that Piwi acts with different downstream factors to repress different sets of transposons. Although the underlying mechanism is not fully understood, the transposon location, type and evolutionary age are speculated to contribute to this. Indeed, the evolutionarily older transposons are shown to be enriched at pericentric regions and are targeted by fewer piRNAs (Kofler et al. 2012; Kelleher and Barbash 2013). It has been suggested that evolutionarily older transposons are more likely to be silenced transcriptionally, while the evolutionarily younger transposons depend more on the post-transcriptional silencing mechanism (Senti et al. 2015). In summary, the piRNA pathway incorporates a wide variety of partners to maintain piRNA clusters, transport, TGS and PTGS, for effective silencing of transposon globally and better sustainability of species in an evolutionary arms race with transposons.

Mechanisms that Regulate Meiotic Cell Maturation

The maturation of both male and female gametes is a step-wise developmental process that requires the coordinated control of the cell cycle, cellular morphology, and cellular positioning. The coordination of these processes are absolutely essential, and require exquisite transcriptional, as well as translational, regulation of a diverse set of genes.

Spermatocyte maturation

In the male germline, the transition from spermatogonia to spermatocytes is accompanied by a series of transcriptional, epigenetic, and morphological changes. After transit-amplification, germ cells undergo the last S phase followed by an extended G2 phase that initiates the spermatocyte stage. Spermatocytes grow 25 times in volume and turn on a robust transcription program to activate genes required for spermatocyte maturation, as well as genes needed for meiotic divisions and terminal differentiation (White-Cooper et al. 1998).

Transcriptional regulators:

Many genes required for meiotic divisions and terminal differentiation are under translational repression until a later time when their encoded proteins are required (Schafer et al. 1995). The G2/M transition in meiosis I requires Cyclin B, Boule (a RNA-binding protein) and Twine (Cdc25 homolog), all transcribed in spermatocytes (Alphey et al. 1992; Courtot et al. 1992; White-Cooper et al. 1998). Boule translocates from the nucleus to the cytoplasm to trigger the G2/M transition in meiosis I by allowing translation of Twine (Maines and Wasserman 1999). At this point in time, Cyclin B also escapes from translational repression and accumulates Cyc B protein in the cytoplasm of spermatocytes (White-Cooper et al. 1998). In both boule and twine mutant testes, spermatid differentiation occurs in a manner independent of meiotic cell cycle progression, suggesting that these two processes can be uncoupled (Alphey et al. 1992; Eberhart et al. 1996). However, the discovery of two classes of genes expressed in early spermatocytes reveals a high degree of coordination between meiotic divisions and spermatid differentiation (Lin et al. 1996). Mutations in any of these genes arrest meiosis and block spermatid differentiation, leading to testes filled with immature spermatocytes. These genes are named “meiotic arrest” genes, which are further classified into “aly-class” and “can-class” based on morphological differences of the chromosomal structure in the mutant spermatocytes (Lin et al. 1996; White-Cooper et al. 1998) and their distinct target genes (Lin et al. 1996; White-Cooper et al. 1998, 2000; Hiller et al. 2001, 2004; Ayyar et al. 2003; Jiang and White-Cooper 2003; Perezgasga et al. 2004; Beall et al. 2007; Jiang et al. 2007; Chen et al. 2011). For example, transcription of meiotic cell cycle genes, such as Cyclin B, boule, and twine, rely on aly-class, but not can-class, genes (White-Cooper et al. 1998). However, Boule protein accumulation requires the can-class genes (Chen et al. 2005). Since meiotic arrest genes regulate transcription or translation of meiotic cell cycle genes, the meiotic cell cycle cannot proceed until terminal differentiation genes are robustly transcribed (Lin et al. 1996; White-Cooper et al. 1998).

The six known aly-class genes are always early (aly), cookie monster (comr), matotopetli (topi), tombola (tomb), achintya/vismay (achi/vis), and Caf1 (Beall et al. 2007). All of the aly-class genes, except achi/vis, are expressed exclusively in primary spermatocytes (Ayyar et al. 2003; Jiang and White-Cooper 2003; Wang and Mann 2003; Perezgasga et al. 2004; Jiang et al. 2007; White-Cooper 2009). Four aly-class proteins have putative DNA-binding domains, including Comr, which contains a winged helix; Topi, which contains multiple Zn-finger motifs; Tomb, which has a CXC domain; and Achi/Vis, products from a gene duplication, which have homeodomains. Thus, it is thought that these proteins regulate the transcription of target genes by directly binding to DNA sequences, even though their direct target genes have not been identified. Immunoaffinity purification studies have revealed that Aly and Tomb proteins are copurified with Mip40 (Myb interacting protein, 40 kDa) to form the testis meiotic arrest complex tMAC, which also contains Topi, Comr, and CAF1 (Beall et al. 2007). A second form of tMAC contains Aly, Comr, and Achi/Vis (Wang and Mann 2003). The tMAC resembles the MIP/dREAM complex in mammals and the SynMuv complexes in C. elegans (White-Cooper et al. 1998, 2000; Ayyar et al. 2003; Jiang and White-Cooper 2003; Perezgasga et al. 2004; Beall et al. 2007; Jiang et al. 2007). Studies using the DamID method profiled ∼300 direct target genes of Comr in testis, most of which have decreased expression in the comr mutant, suggesting that it functions mainly as a transcriptional activator (Laktionov et al. 2014). This is consistent with earlier results demonstrating that expression of Achi/Vis fused with a strong transactivation domain, VP16, rescued the achi/vis mutant phenotype, while the fusion of Achi/Vis with a repression domain, EnR, failed to rescue (Wang et al. 2008). Consistent with these findings, all tMAC subunits have been found to colocalize with euchromatin in primary spermatocytes (White-Cooper et al. 2000; Jiang and White-Cooper 2003; Wang and Mann 2003; Jiang et al. 2007).

The can-class genes encode testis-specific homologs of ubiquitously expressed subunits of the general transcription factor II D (TFIID). TFIID is one of the general transcription factors that constitute the RNA Pol II preinitiation complex composed of TATA-binding protein (TBP) and 13–14 TBP-associated factors (TAFs) (Tora 2002; Matangkasombut et al. 2004; Cler et al. 2009). TFIID coordinates the interaction between RNA Pol II and gene promoter regions. The characterized can-class genes include cannonball (can, TAF5L), meiosis I arrest (mia, TAF6L), no hitter (nht, TAF4L), ryan express (rye, TAF12L), and spermatocyte arrest (sa, TAF8L). Among the five TAF homologs, four, including Mia, Nht, Rye, and Sa, share similar structural domains called histone folding motifs for protein–protein interaction, while Can is a WD40-repeat-containing protein (Hiller et al. 2001). Indeed, Nht and Rye form a heterodimer in vitro (Hiller et al. 2004). These testis-specific TAFs (tTAFs) are thought to form a testis-specific complex required for transcriptional activation of the terminal differentiation genes (Hiller et al. 2001, 2004). Such predicted functions of tTAFs suggest that they localize at the euchromatin in spermatocyte nuclei. However, while a proportion of the total protein of each tTAF associates with chromosomes in spermatocytes, most tTAF protein is localized to a subcompartment within nucleolus (Chen et al. 2005; Metcalf and Wassarman 2007). Interestingly, Polycomb and other components of PRC1 are colocalized to the same nucleolar subcompartment with tTAFs in spermatocytes. Furthermore, localization of PRC1 components to the spermatocyte nucleolus is coincident with tTAF expression and dependent on wild-type tTAF function (Chen et al. 2005). These results suggest that tTAFs act as derepressors by sequestering PRC1 to the spermatocyte nucleolus to counteract PcG-induced repression. However, removing PcG activity is not sufficient to turn on terminal differentiation genes in the absence of tTAFs (Chen et al. 2011), suggesting that chromatin-associated tTAFs are required to activate terminal differentiation genes. Consistent with these observations, tTAFs were reported to turn on transcription of >1000 genes, many of which are required for spermatid differentiation (White-Cooper et al. 1998; Chen et al. 2011). Among the tTAF-dependent genes, three are shown to be direct target genes of tTAF by ChIP assay: fuzzy onions (fzo), which encodes a protein required for mitochondrial fusion in early spermatids (Hales and Fuller 1997); mst87F, which encodes a component of the sperm tail (Schafer et al. 1993) and don juan (dj), which encodes a sperm-specific DNA-binding protein that also localizes to mitochondria (Santel et al. 1998). ChIP analysis at the promoter regions of these three genes directly targeted by tTAF showed that levels of the repressive H3K27me3 mark and paused Pol II are high, while levels of the active H3K4me3 mark are low in can and aly mutant testes (Chen et al. 2011). These data suggest that tTAFs and tMAC might recruit TrxG, whose activities antagonize PcG, to methylate H3K4 at promoters of terminal differentiation genes and activate robust transcription (Chen et al. 2005).

Although the mode of interaction between tMAC components (aly-class) and tTAFs (can-class) is not fully understood, the transcription coactivator Mediator likely acts to coordinate tMAC and tTAFs. The tMAC recruits Mediator components to spermatocyte chromatin, and Mediator subsequently helps proper tTAF localization. Together, tMAC, tTAFs and Mediator coregulate a cohort of spermatid differentiation gene expression (Lu and Fuller 2015). It was also reported that the function of aly is required for the binding of TAF8L to target gene promoters. Aly is also required for the proper nucleolar localization of several tTAFs and Polycomb in spermatocytes, suggesting that tMAC acts upstream of tTAFs (Chen et al. 2011). This is consistent with assays using Northern blot, in situ hybridization, and microarray analysis (White-Cooper et al. 1998; Hiller et al. 2001; Chen et al. 2011). In addition, while Mip40 is coimmunoprecipitated with tMAC components, loss of mip40 results in spermatocytes with condensed chromosomes, a phenotype similar to mutants of can-class genes (Beall et al. 2007), suggesting that Mip40 might mediate the interaction between tMAC and tTAFs. Both tMAC and tTAFs have their canonical counterparts that act generally in other tissues, as well as in spermatocytes, probably by regulating target genes distinct from the testis-specific forms. Similarly, the canonical chromatin remodeler NURF has a germline-specific function in regulating meiotic divisions and spermatocyte differentiation (Kwon et al. 2009), most likely through using an alternatively spliced isoform.

Five other meiotic arrest genes, which cannot be classified as either aly-class or can-class, were identified and characterized. Wake-up-call (Wuc) was identified by its physical interaction with Aly in a yeast-two-hybrid screen (Jiang et al. 2007). In spermatocytes, the Wuc protein is highly expressed and associated with chromatin, similar to other tMAC components. However, unlike tMAC or tTAF mutants, loss of wuc does not abolish expression of either meiotic cell cycle genes or spermatid differentiation genes (Doggett et al. 2011). Another study showed that disruption of a component of the THO complex, THOC5, led to the meiotic arrest phenotype. The THO complex is known to export mRNAs from nucleus to cytoplasm. However, no mRNA export defects were detectable in the thoc5 mutant. Moreover, neither meiotic cell cycle genes nor spermatid differentiation genes have decreased transcription in the thoc5 mutant, even though a more comprehensive study is needed. THOC5 is localized to a perinucleolar region, and loss of thoc5 function leads to disrupted nucleolar structure and the localization of tTAFs, which could contribute to its mutant phenotype (Moon et al. 2011). A more recent study identified Ntx1, another mRNA export machinery component, as required for accumulation of many spermatogenesis-specific mRNAs. However, the dependence of these transcripts on Ntx1 has a distinct mode compared to tMAC- or tTAF-dependent genes, which is regulated by the primary transcript structure (Caporilli et al. 2013). Moreover, through characterization of a meiotic arrest mutant magellan (magn), the Ubi-p63E gene encoding polyubiquitin has been shown to regulate proper spermatocyte chromatin structure, meiotic cell cycle progression, and spermiogenesis. However, the different phenotypes caused by loss-of-function of proteasome subunits suggest that Ubi-p63E acts in a protein degradation-independent manner in spermatocytes (Lu et al. 2013). Finally, a very recent study identified a novel meiotic arrest gene kumgang (kmg), which encodes a zinc finger-containing protein. The kmg gene is specifically turned on in early spermatocytes, independent of either tMAC or tTAF. Interestingly, Kmg is required to maintain germline identity by suppressing the expression of hundreds of somatic genes. Genetic, genomic, and biochemical analyses reveal that Kmg acts with the chromatin remodeler dMi-2 to restrict the tMAC component Aly from helping to fire transcription from cryptic promoters of a cohort of somatic genes, which are normally turned on in somatic tissues, such as gut and brain (Kim et al. 2017). Both identification of wuc, thoc5, Ntx1, magn, and kmg mutants, and detailed characterization of their phenotypes and mechanisms demonstrate the existence of meiotic arrest genes other than aly- and can-class. Further understanding of their molecular and cellular mechanisms will lead to new information about spermatocyte maturation.

MicroRNAs:

In males, bam mRNA is detectable, but Bam protein is undetectable in the meiotic spermatocytes. It has been shown that a specific miRNA, miR-275, represses Bam protein accumulation through bam 3′UTR in spermatocytes. If this repression of Bam protein accumulation in spermatocytes is misregulated, spermiogenesis cannot proceed properly, and this will lead to decreased male fertility (Eun et al. 2013). Therefore, although Bam is an important differentiation factor to initiate GSC differentiation, its downregulation is also critical for proper spermatid terminal differentiation. Furthermore, this post-transcriptional regulation of Bam protein accumulation does not occur in the female germline, again suggesting sex-specific modes in the regulation of meiotic germ cell maturation.

Oocyte specification and maturation

Oocyte development begins with oocyte specification in the germarium; once specified, the oocyte migrates to the posterior region of the cyst, which will be enclosed by follicle cells and buds off as an egg chamber. While the egg chamber is growing, the polarity of the oocyte is established. These processes involve both signal transduction pathways, as well as the cytoskeletal machinery. Furthermore, the piRNA pathway has also been reported to have a significant role in establishing oocyte polarity and proper oocyte development.

Oocyte specification and polarity establishment in the germarium:

As an extremely specialized cell type, oocyte formation requires a series of developmentally regulated processes to break the symmetry and give rise to this highly polarized, gigantic cell (Roth and Lynch 2009). Polarity formation begins in the germarium with the specification of oocytes, and this polarity is already established as early as the first division of the CB (de Cuevas and Spradling 1998). In regions 2a and 2b (Figure 1A), mRNA transport allows accumulation of specific markers in the developing oocyte (Suter and Steward 1991; Lantz et al. 1994; Mach and Lehmann 1997). After completing the transit-amplification stage, the microtubule organizing center (MTOC) appears in one or two cells inside the 16-cell cyst, which has four ring canals connected to other cells in the cyst. These two cells, called pro-oocytes, start meiosis, which is characterized by the appearance of double-strand breaks (DBSs) and the formation of synaptonemal complex (SC). Later, one of them will be committed to an oocyte. Upon the specification of oocyte, DBSs, which have been seen in both pro-oocytes, become restricted to one oocyte in region 2a/2b, and are repaired thereafter (Jang et al. 2003). In piRNA pathway mutants, such as armitage (armi) and aub, DSBs persist longer and are accumulated in the oocyte during later stages of development (Klattenhoff et al. 2007). The upregulation of transposons, and their increased transpositions in the oocyte, are believed to cause accumulation of DSBs in the oocyte, although no study has explicitly proven this. At region 2b/3 (Figure 1A), oocyte determination is evident in that the SC remains in only one germ cell determined to be an oocyte (Huynh and St Johnston 2000; Page and Hawley 2001). In addition, MTOC appearance is more pronounced, and gurken (grk) mRNA is localized posteriorly (Neuman-Silberberg and Schupbach 1993, 1996). Egg chambers (Stage 1–14) are assembled posterior to the germarium, which contains the nurturing nurse cells and the developing oocyte ensheathed by follicle cells derived from FSCs.

Oocyte determination and polarity formation during oocyte maturation:

As oogenesis proceeds toward region 3, the oocyte meiotic chromosomes form a compact spherical structure called the karyosome, which is mainly transcriptionally silent (Parfenov et al. 1989; Bastock and St Johnston 2008; Lancaster et al. 2010). At this stage, the oocyte development is also determined by the coordinated activity of both cell cycle genes and polarity genes (Lilly and Spradling 1996; Mach and Lehmann 1997; Mata et al. 2000; Huynh et al. 2001a; Hong et al. 2003).

Microtubule networks, as organized by spectrosome/fusome structure (Grieder et al. 2000), play important roles in the formation and maintenance of oocyte polarity. The MTOCs shift from the anterior to the posterior side of the oocyte (Theurkauf et al. 1992, 1993; Huynh et al. 2001b; Vaccari and Ephrussi 2002). The reorganized microtubule network is important for proper localization of maternal components, such as bicoid and oskar (osk), as polarity determinants, and define the anterior and posterior sides of the developing oocyte, respectively (Ephrussi et al. 1991; Kim-Ha et al. 1991; Brendza et al. 2000; Januschke et al. 2002). This polarity within the oocyte also defines the embryonic anterior−posterior axis. Live imaging of Osk particle movement during oogenesis showed that the mRNA is actively transported along microtubules in all directions, with a slight bias toward the posterior (Zimyanin et al. 2008). Meanwhile, grk mRNA is found at the posterior of oocyte in the germarium, while in the later stages, grk mRNA is repositioned at the dorsal anterior corner of the oocyte (Neuman-Silberberg and Schupbach 1996; Van Buskirk and Schupbach 1999; St Johnston 2005). Grk accumulation defines the dorsal−ventral axis of the oocyte, as well as the embryos (Schupbach 1987; Neuman-Silberberg and Schupbach 1993; Nilson and Schupbach 1999; Moussian and Roth 2005). Grk localization and oocyte nucleus migration are mediated by microtubules (reviewed by Roth and Lynch 2009). Although the exact mechanism for grk mRNA localization is not known, microtubules from MTOC and dynein are important for Grk localization in the oocyte and mediate communication with follicle cells (Brendza et al. 2000, 2002; Duncan and Warrior 2002; Januschke et al. 2002). Together, polarized localization of bicoid, osk, and grk mRNAs defines both the AP and DV axes of the oocyte and embryo. However, imaging data revealed that the overall microtubule network is actually much less polarized than previously expected (MacDougall et al. 2003; Zimyanin et al. 2008), provoking more studies using new techniques such as live cell imaging.

By midoogenesis, a specialized cytoplasm, termed as pole plasm, assembles at the posterior end of oocyte (Hay et al. 1988; Lasko and Ashburner 1988; Ephrussi et al. 1991; Golumbeski et al. 1991; Ephrussi and Lehmann 1992; Harris and Macdonald 2001;). The pole plasm contains granules of ribonucleoproteins (RNPs) enriched with RNAs and proteins, which are required for formation of primordial germ cells (PGCs; Santos and Lehmann 2004). The pole plasm is maternally transmitted to the embryo and retained at the posterior side, where the nuclei are first cellularized to form the primordial germline cells (Starz-Gaiano and Lehmann 2001; Santos and Lehmann 2004; Laver et al. 2015). As previously noted, piRNAs are also deposited maternally to the embryos, conferring defense against transposons in the next generation (Brennecke et al. 2008). In addition to piRNAs, several piRNA pathway components, such as Piwi, Vasa, Tud, and Aub, are found in the pole plasm and required for pole plasm formation (Hay et al. 1988; Lasko and Ashburner 1988; Ephrussi et al. 1991; Golumbeski et al. 1991; Ephrussi and Lehmann 1992; Harris and Macdonald 2001; Megosh et al. 2006).

Function of PcG in oocyte specification:

While the determined oocyte will initiate extraordinary cell growth and meiotic cell cycle, the 15 nurse cells in the Drosophila ovary will enter the endocycle and become polyploid cells to provide RNAs and proteins to the developing oocyte. Transdetermination from oocyte to nurse-like cells was observed when PRC2 components E(z) and Su(z)12 were knocked down in the Drosophila female germline. This cell fate change results from derepression of Cyclin E and cyclin-dependent kinase inhibitor Dacapo upon loss of the repressive H3K27me3 mark (Iovino et al. 2013). However, such cell fate switch does not occur in the male germline. In males, all 16 spermatogonial cells enter meiosis simultaneously after mitosis and differentiate into mature sperm synchronously. This phenomenon suggests that differences between the female and male germline differentiation pathways require distinct epigenetic regulators.

piRNA pathway components for polarity formation:

In addition to cytoskeletal machinery and RNA-binding proteins, loss of many piRNA pathway proteins results in discernible defects in polarity formation. For example, in armi, spnE, zuc, mael, and krimp mutants, Grk and Osk proteins fail to localize to the dorsal–anterior region and to the posterior region, respectively (Findley et al. 2003; Cook et al. 2004; Chen et al. 2007; Klattenhoff et al. 2007; Lim and Kai 2007; Pane et al. 2007). Failure of microtubule network polarization in piRNA pathway mutants is believed to cause mislocalization of these components. In addition, Osk is precociously translated in some of the piRNA pathway mutants (Cook et al. 2004; Lim and Kai 2007; Pane et al. 2007). The piRNA pathway component Mael interacts with MTOC components, including centrosomin, mini spindles, and γ−tubulin (Sato et al. 2011). The interaction of Mael with cytoskeletal structure further strengthens the role of piRNA pathway components in oocyte polarity formation. Interestingly, upregulation of I-element transposon is known to perturb the localization of grk and bcd mRNAs (Van De Bor et al. 2005). However, some piRNA pathway mutants, such as tej, and qin/kumo, do not show defects in the localization of grk or polarity formation of oocyte, despite the severe depression of transposons including I-element (Patil and Kai 2010; Anand and Kai 2012; Patil et al. 2014), suggesting that derepression of transposons alone is not sufficient to cause polarity defects in oocytes.

Consensus has still not formed around the role of piRNA pathway proteins in DNA damage response or polarity determination. In some piRNA pathway mutants, ablation of DNA damage checkpoint components, such as mei41 and chk2, could suppress oocyte polarity defects (Klattenhoff et al. 2007), suggesting crosstalk between some microtubule polarization components and the DNA damage response pathway. In contrast, Grk mislocalization in mael, squ, and zuc mutants could not be restored by ablating mei41 function (Pane et al. 2007; Sato et al. 2011). Therefore, either DNA damage response in those mutants is initiated by a different downstream component or a different DNA damage-sensing pathway is involved. Except for Mael, no other piRNA pathway protein is known to directly interact with cytoskeleton components (Sato et al. 2011). Under these circumstances, the effect on polarity formation is a direct or indirect effect of loss of these proteins.

Mechanisms in Regulating Intergenerational and Transgenerational Epigenetic Inheritance

Traditionally, heritability is a characteristic feature of the genetic material of an organism, notably its DNA. Nonetheless, many phenomena and mechanisms of non-DNA sequence-based inheritance of vastly different phenotypes have been described from one generation to the next in multiple organisms ranging from plants to vertebrates (Youngson and Whitelaw 2008; Heard and Martienssen 2014). This inheritance of information beyond the primary DNA sequence is known as epigenetic. Direct epigenetic inheritance from parent to offspring is termed intergenerational epigenetic inheritance (IEI), and it is distinguished from transgenerational epigenetic inheritance (TEI), which is observed in generations that were not exposed to the initial signal or environment that triggered the acquired change.

One of the earliest reports of TEI in Drosophila was uncovered studying the Fab-7 chromosomal boundary element (Cavalli and Paro 1998). The Fab-7 boundary element, also a member of Polycomb Response Elements, is derived from the bithorax complex (BX-C) and is required to prevent crosstalk between adjacent regulatory regions, iab-6 and iab-7, which control the spatial expression of the Abd-B gene (Hagstrom et al. 1996). To determine the function of the defined Fab-7 element, transgenic reporter strains were engineered to carry the Fab-7 element upstream of a GAL4 UAS-inducible lacZ reporter and a mini-white gene. The Fab-7 element was found to act as a strong silencer, repressing expression of both lacZ and the distantly located mini-white gene (Zink and Paro 1995). Increased GAL4 expression could stably activate both lacZ reporter and mini-white gene. Furthermore, a short, single pulse of GAL4 expression, regulated by a heat-shock promoter (hs-GAL4), during embryogenesis was sufficient to induce activation of the mini-white gene throughout development, resulting in adult flies with red eyes (Cavalli and Paro 1998). This continued expression of the reporter suggests a loss of silencing that is mitotically inheritable over many cell divisions and not dependent on the duration of the GAL4 protein. Surprisingly, GAL4-independent transmission of the active mini-white gene could be propagated through the female gametes for four generations. Inheritance of the expression pattern was not observed in the male germline. This was tested under conditions where offspring did not inherit the hs-GAL4, demonstrating that a short pulse of GAL4 induced during early embryogenesis alters the epigenetic landscape of a gene in a way that is stably inherited during both mitosis and meiosis. The molecular carrier for the maintenance of these patterns of expression through meiosis remains to be determined. Despite the fact that the mechanistic basis underlying both IEI and TEI is largely unknown and under intense investigation, three epigenetic information carriers have, in fact, been identified, including DNA methylation, chromatin structure, and RNAs.

DNA methylation in intergenerational and transgenerational epigenetic inheritance

DNA methylation may function as a molecular carrier during IEI in Drosophila. Inheritance of 5mC DNA methylation has been well documented in both mammalian and plant models of epigenetic inheritance (Heard and Martienssen 2014). Although the full extent to which DNA methylation participates in IEI in Drosophila remains elusive, investigations of sister chromatid inheritance, as well as tumor susceptibility, have highlighted two separate cases of intergenerational epigenetic effects in genetically compromised backgrounds.

During ACD of male GSCs, sister chromatids of the X and Y chromosome are distinguished and segregated in a nonrandom manner (Yadlapalli and Yamashita 2013). Loss of DNMT2 leads to randomized sister chromatid segregation of both X and Y chromosomes, suggesting that DNMT2 confers epigenetic information to the X and Y chromosomes that leads to distinct sister chromatid segregation. Furthermore, systematic crosses between homozygotic and heterozygotic DNMT2 parents revealed that parental DNMT2 function is necessary for proper segregation of X and Y sister chromatids in the next generation in a parent-dependent manner, very much like the imprinting phenomenon. These data suggest that parental DNMT2 functions during gametogenesis in both males and females to transmit heritable information on the X and Y chromosomes, and such information is maintained during early embryogenesis (Yadlapalli and Yamashita 2013).

Little is known about epigenetic reprogramming of DNA methylation during gametogenesis and early embryogenesis in Drosophila. Studies of an oncogenic JAK kinase encoded by hopscotchtum-1 (hoptum-1) demonstrated that this temperature-sensitive hypermorphic allele is able to antagonize a cellular program that erases DNA methylation of parental origin, allowing the epigenetic alterations to be maintained in the absence of the original mutation (Xing et al. 2007). Tumorous-lethal (Tum-1) is a dominant temperature-sensitive mutation in the hop locus that leads to overproliferation of hemocytes and formation of melonotic tumors, which are black masses of hemocytes correlated with lethality. In a genetic approach to identify genes important for hoptum-1-induced tumorigenesis, 37 modifiers that either enhanced or suppressed hoptum-1 tumorigenesis were identified (Shi et al. 2006). Interestingly, many of the identified mutations exhibited paternal-effect modification of hoptum-1 tumor susceptibility. For example, one of the modifiers, Kruppel (Kr), enhances hoptum-1 tumorigenicity. When hoptum-1 heterozygotic females were mated with male heterozygotes for Kr, tumorigenesis associated with hoptum-1 was enhanced in the F1 generation, irrespective of the inheritance of the modifier mutation itself. This enhancement persisted into the F2 generation, but diminished by the F3 generation. Further studies indicate that the Kr mutation establishes DNA methylation at promoters during early embryogenesis. Furthermore, the epigenetic alterations induced by Kr are normally erased in the next generation. However, in the presence of the hoptum-1 allele, the increased DNA methylation induced by Kr was transmitted to the next generation.

Chromatin structure in intergenerational and transgenerational epigenetic inheritance

Histones and histone variants have become primary candidates for mediating germline epigenetic inheritance. Histone modifications and variants are capable of transmitting epigenetic information through mitosis and meiosis to the next generation (Gaydos et al. 2014). A major barrier to IEI or TEI is the epigenetic reprogramming during gametogenesis and early embryo development, during which global changes in histone modifications and variants occur (Harrison and Eisen 2015). An extreme example of histone replacement is the transition from nucleosome-based to protamine-based chromatin structure during Drosophila spermatogenesis (Rathke et al. 2007). Upon fertilization, chromatin undergoes dramatic remodeling again when the paternal genome is remodeled, replacing protamines with the histone variant H3.3 (Loppin et al. 2005). The Drosophila sesame mutant exhibits lesions at the HIRA gene encoding H3.3/H4 replication-independent nucleosome assembly chaperone. Characterization of sesame revealed that protamines are replaced by maternal H3.3 prior to the first S phase during embryogenesis. Upon fertilization, H3.3 is used to remodel paternal chromatin. Despite these sweeping changes, recent proteomic analysis of whole sperm mass spectrometry has revealed that all four canonical histones, as well as histone variants, are retained in mature sperm (Dorus et al. 2006). This retention in mature sperm raises the possibility that they retain epigenetic information for transmission across generations.

The centromere-specific histone variant Centromere identifier (Cid) is also present in mature sperm (Raychaudhuri et al. 2012). In Drosophila, nucleosomes with Cid, instead of other histone H3 variants, are stably incorporated exclusively at the centromeric region. Analysis of centromere identity has indicated that the centromere is specified epigenetically (Black and Cleveland 2011). Cid is retained in mature sperm and during the protamine-to-histone transition after fertilization. This paternally inherited Cid is required for the maintenance of paternal chromosomes in the next generation. In the absence of paternally inherited Cid, paternal chromosomes fail to recruit the maternally provided Cid and cannot generate functional kinetochores during the first mitosis.

Paternal genome stability in the embryo also relies on a heterochromatin-associated protein 1 (HP1) paralog, HP1E. The Drosophila genome encodes five HP1 paralogs, HP1A–E. The genes that encode HP1A, HP1B, and HP1C are expressed in all tissues and localize primarily to heterochromatin, with the exception of HP1C, which localizes exclusively to euchromatin (Smothers and Henikoff 2001). HP1D and HP1E have special roles in the female and male germline, respectively (Volpe et al. 2001; Vermaak and Malik 2009). HP1D is required for transposon silencing in the female germline, and loss of HP1D results in female sterility (Volpe et al. 2001; Klattenhoff et al. 2009). Expression of HP1E is developmentally restricted within the male germline where it localizes to the developing spermatids and functions in heterochromatin integrity. Specifically, HP1E localizes to developing spermatids subsequent to the completion of Meiosis II, but it is not detectable in mature sperm. In the absence of HP1E in males, embryos have defects in paternal chromatin condensation and fail to separate chromosomes during mitosis, resulting in “chromatin bridges” and lethality (Levine et al. 2015). Unlike Cid, which is inherited from the previous generation, HP1E is not inherited from sperm; instead, HP1E primes paternal chromosomes during spermatogenesis to ensure proper segregation in the next generation.

Role of piRNA pathway in maternal deposition of transcripts and their clearance

The oocyte provides transcripts and proteins to embryos for their early development. Transcripts of many genes, including those required for early development, are maternally deposited through the ring canals from nurse cells to the oocyte during oogenesis (reviewed by Laver et al. 2015). Transcription from zygotic genome starts at 2 hr postfertilization, and concurrently, a subset of maternally deposited materials is eliminated. This process is referred to as Maternal to Zygotic Transition (MZT; reviewed by Laver et al. 2015). During MZT, the transition of gene expression is tightly regulated in several different ways, including clearance of the maternally deposited transcripts. Recent high-throughput analyses identified that a significant number of transcripts, from 7000 to 10,000, are maternally transmitted to embryos (Lecuyer et al. 2007; Thomsen et al. 2010). Approximately two-thirds of them are either degraded, or significantly reduced, within 3 hr postfertilization (Thomsen et al. 2010; Laver et al. 2015). An RNA-binding protein, Smaug, triggers degradation of those RNAs by deadenylation through the CCR4/POP2/NOT4 deadenylase complex. Several studies revealed that piRNA pathway proteins and piRNAs promote the decay of a subset of posteriorly localizing maternally deposited RNAs in the bulk of embryo, possibly via the deadenylation complex, leading to the enrichment of germline determinants at pole plasm (Rouget et al. 2010; Barckmann et al. 2015; Vourekas et al. 2016).

Enrichment of nos transcript at pole plasm:

Simonelig and colleagues first reported that piRNA pathway proteins and piRNAs promote the deadenylation and decay of maternally deposited nanos (nos) transcripts (Rouget et al. 2010). Maternally deposited nos mRNA is present throughout embryos at very early stages, but it is translationally repressed in the somatic part and degraded in a deadenylation-dependent manner involving Smaug (Dahanukar and Wharton 1996). Osk, the key component of pole plasm formation, prevents deadenylation of nos transcript, and promotes its translation at the posterior pole, which helps to form Nos gradient at the posterior region (Santos and Lehmann 2004; Zaessinger et al. 2006). In addition to Osk, the piRNA pathway components aub, ago3, spnE, and piwi were shown to be required for deadenylation and decay of the maternal nos transcript (Rouget et al. 2010). Aub and Ago3 are present throughout embryos and likely trigger deadenylation of nos mRNA by recruiting the deadenylation complex. The piRNAs arising from 412 and roo transposons target nos 3′UTR for nos mRNA decay. Those piRNAs bound to Aub and Ago3 likely recruit Smaug and the CCR4-NOT adenylation complex to nos transcript and degrade it in the bulk embryo, but not at the pole, forming the Nos gradient (Rouget et al. 2010).

Decay and anchoring of maternal transcripts en masse:

Recent genome-wide analyses of Aub-bound RNAs using CLIP (crosslinking and immunoprecipitation) experiments by two groups further revealed the functions of piRNAs in anchoring and enriching maternally deposited transcripts at the posterior pole (Konig et al. 2010; Barckmann et al. 2015; Vourekas et al. 2016). While both small RNAs and long RNAs are found in the Aub-CLIP libraries, the small RNAs, mostly comprised of piRNAs, are more abundant than long ones. Almost all Aub-bound long RNAs bind to Aub in a piRNA-dependent manner, and do not contain transposon sequences, suggesting that RNAs bound to Aub are unrelated to piRNA biogenesis (Barckmann et al. 2015).

Both studies also reported that Aub-bound transcripts are derived from genes involved in diverse functions (Barckmann et al. 2015; Vourekas et al. 2016). Posterior localization of Aub does not seem to be necessary for binding with these transcripts, except for some localized at the posterior pole (Barckmann et al. 2015; Vourekas et al. 2016). A large number of posteriorly localizing transcripts, including osk, germ cell-less (gcl), polar granule component (pgc), hsp83, and nos, depend on Aub for their degradation in the bulk of embryos (Barckmann et al. 2015; Vourekas et al. 2016). Many of them are degraded by the deadenylation-dependent pathway because they are stabilized in the embryos with mutations of deadenylation complex components (Rouget et al. 2010; Barckmann et al. 2015). In addition, Simonelig and colleagues found very few secondary piRNAs pairing with Aub-bound transcripts, suggesting that the ping-pong mechanism contribute to the clearance of fewer maternal transcripts (Barckmann et al. 2015). Hence, Aub is likely acting for MZT by its endonucleolytic activity and through the deadenylation-dependent pathway. It is possible that Smaug and the downstream components are involved in degradation of these transcripts, possibly triggered by piRNAs.

Extensive computational analysis by Zissimos and colleagues further elucidated the importance of piRNAs in anchoring transcripts to Aub in embryos. Aub-bound piRNAs exhibit rather weak complementarity to mRNA in a manner reminiscent of miRNA–mRNA interaction (Vourekas et al. 2016). The transcripts localized at the posterior pole are enriched with such piRNA binding sites, suggesting that the Aub-piRNA complex in the pole granules may serve as nucleation sites for the proper localization of these transcripts. It is speculated that the same Aub-piRNA complex could also scan the transposon transcripts to maintain the fitness of species in the next generation.

Overall, piRNAs are important for maintaining mRNAs in Aub-RNA complexes. Aub functions to degrade transcripts of germline determinants in the bulk of cytoplasm, possibly by either endonucleolytic activity of Aub itself or triggering deadenylation, in turn leading to their enrichment in the pole plasm. In addition, Aub-bound piRNAs play an important role in anchoring the transcripts involved in posterior localization and germline development. Further studies are needed to elucidate the molecular mechanism that underlies the spatiotemporal regulation of Aub-piRNA complex/mRNAs interaction.

Perspectives

In summary, to understand both oogenesis and spermatogenesis, developmental genetics and cell biology approaches can take advantage of the reliable developmental processes and distinct morphologies for each stage of germ cell differentiation. Formation of both male and female gametes requires the interaction between the germline and the somatic gonadal cells. During this process, the germline identity is protected, while GSC self-renewal, differentiation, and meiotic cell cycle genes are tightly regulated by the sequential changes of the chromatin structure in germ cells (Figure 5). On the other hand, mature gametes carry both genetic and epigenetic information from one generation to the next.

Epigenetic regulation of germ cell development in Drosophila. Male spermatogenesis (Blue arrow) and female oogenesis (Pink arrow) are highlighted at distinct differentiation stages (circles). Chromatin regulators, histone modifications, histone variants, and histone modifying enzymes are placed along the developmental timeline at specific stages in which previous studies have uncovered a critical developmental role. Schematic drawings of the zygote and early embryo include female (Venus symbol) and male (Mars symbol) pronuclei (gray) marked in the zygote, as well as PGCs, marked in the early embryo. Detailed information of these developmental mechanisms are discussed throughout the text.
Figure 5

Epigenetic regulation of germ cell development in Drosophila. Male spermatogenesis (Blue arrow) and female oogenesis (Pink arrow) are highlighted at distinct differentiation stages (circles). Chromatin regulators, histone modifications, histone variants, and histone modifying enzymes are placed along the developmental timeline at specific stages in which previous studies have uncovered a critical developmental role. Schematic drawings of the zygote and early embryo include female (Venus symbol) and male (Mars symbol) pronuclei (gray) marked in the zygote, as well as PGCs, marked in the early embryo. Detailed information of these developmental mechanisms are discussed throughout the text.

As described in this review, the germline genome must be protected against transposable elements (TE), or transposons, which are DNA sequences that can alter the genetic identity of a cell by changing their position in the genome. Charged with this task is the piRNA pathway, which is directly involved with the silencing of transposons. Both the steady state of TE repression, and the dynamics of the piRNA pathway during germline development are better understood today. Newly introduced transposons initially escape from repression via the piRNA pathway, but germ cells quickly acquire adaptation to new invasion of transposons by producing piRNAs in a single generation (Khurana et al. 2011). Repression of evolutionarily older transposons needs fewer piRNAs, while recent transposon insertions attract a higher number of piRNAs (Kelleher and Barbash 2013). Certainly, the study of piRNA pathway function from a population-wide perspective might shed light on the evolutionary nature and adaptive events in the piRNA population over generations. While studies have provided a glimpse of piRNA precursor transcription and processing, a proper understanding of events prior to piRNA processing remain enigmatic.

In the future, we can expect a better understanding of the unique features of germ cells, which will greatly facilitate applying them for the treatment of diseases and regenerative medicine. For example, piRNA pathway proteins are reported for transposon repression of embryonic stem cells in mammals (Darricarrere et al. 2013; Marchetto et al. 2013; Peng et al. 2016). Recently, the piRNA pathway proteins have also been shown to support survival and proliferation of cancer cells from flies to human (Janic et al. 2010; Fagegaltier et al. 2016; Ng et al. 2016; Sumiyoshi et al. 2016). In higher vertebrates, PIWIs are implicated in somatic stem cell functions and/or regeneration of the tissues (Rizzo et al. 2014). Based on the conserved nature of the piRNA pathway across species and during development and disease, understanding the relationship between piRNAs and transposons during early development may provide insight into the development of tumors, highlighting the importance of studying noncoding RNA regulation, and in turn, leading to the identification of new therapeutic targets.

However, it remains challenging to study epigenetic regulation at the individual gene in germ cells at particular stages, such as PGCs in embryos and GSCs in adults. A major technical hurdle involves obtaining a sufficient number of homogeneous cells to investigate their chromatin structure. However, technological advances have significantly reduced the required cell number for such studies, thus providing unprecedented opportunities to understand germ cell identity and activity. This step will be invaluable for treating diseases associated with defects in germ cell differentiation, such as infertility and germ cell tumors, as well as applying germ cells in regenerative medicine. Also, new imaging techniques, such as live cell imaging and superresolution imaging, in combination with genomic engineering, will allow us to trace distinct molecules, such as mRNAs and proteins, as well as organelles and subcellular structures in order to gain new insights into germ cell differentiation at individual developmental stages (Cheng et al. 2008, 2011; Sheng et al. 2009; Morris and Spradling 2011; Spradling 2011; Lenhart and DiNardo 2015; Shalaby and Buszczak 2017). Furthermore, new advances in genomic analyses, including RNA-seq, ChIP-seq, and Hi-C, are beginning to reveal chromatin structure during germline development in a sequence-specific manner. We anticipate rapid progress in the near future to resolve dynamic epigenetic regulation of germ cell differentiation at single-cell resolution, in real-time, and at both genomic and specific gene loci in Drosophila.

Acknowledgments

This work has been supported by National Institutes of Health (NIH) F32GM119347-02 (R.J.G.); Japan Society for the Pormotion of Science (JSPS) Grants-in-aid for Scientific Research (KAKENHI) 26291048 and the Naito Foundation (T.K.); NIH RO1GM112008, R21HD084959, the David and Lucile Packard Foundation, Faculty Scholar from Howard Hughes Medical Institute, Bill and Melinda Gates Foundation, and the Simons Foundation, as well as Johns Hopkins University start-up (X.C.).

Footnotes

Communicating editor: A. Spradling

Literature Cited

Abad
J P
,
De Pablos
B
,
Osoegawa
K
,
De Jong
P J
,
Martin-Gallardo
A
et al. ,
2004
Genomic analysis of Drosophila melanogaster telomeres: full-length copies of HeT-A and TART elements at telomeres.
Mol. Biol. Evol.
21
:
1613
1619
.

Ables
E T
,
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
.

Akiyama
T
,
2002
Mutations of stonewall disrupt the maintenance of female germline stem cells in Drosophila melanogaster.
Dev. Growth Differ.
44
:
97
102
.

Alphey
L
,
Jimenez
J
,
White-Cooper
H
,
Dawson
I
,
Nurse
P
et al. ,
1992
Twine, a cdc25 homolog that functions in the male and female germline of Drosophila.
Cell
69
:
977
988
.

Anand
A
,
Kai
T
,
2012
The tudor domain protein kumo is required to assemble the nuage and to generate germline piRNAs in Drosophila.
EMBO J.
31
:
870
882
.

Arama
E
,
Agapite
J
,
Steller
H
,
2003
Caspase activity and a specific cytochrome C are required for sperm differentiation in Drosophila.
Dev. Cell
4
:
687
697
.

Aravin
A A
,
Naumova
N M
,
Tulin
A V
,
Vagin
V V
,
Rozovsky
Y M
et al. ,
2001
Double-stranded RNA-mediated silencing of genomic tandem repeats and transposable elements in the D. melanogaster germline.
Curr. Biol.
11
:
1017
1027
.

Aravin
A A
,
Lagos-Quintana
M
,
Yalcin
A
,
Zavolan
M
,
Marks
D
et al. ,
2003
The small RNA profile during Drosophila melanogaster development.
Dev. Cell
5
:
337
350
.

Aravin
A A
,
Klenov
M S
,
Vagin
V V
,
Bantignies
F
,
Cavalli
G
et al. ,
2004
Dissection of a natural RNA silencing process in the Drosophila melanogaster germ line.
Mol. Cell. Biol.
24
:
6742
6750
.

Ayyar
S
,
Jiang
J
,
Collu
A
,
White-Cooper
H
,
White
R A
,
2003
Drosophila TGIF is essential for developmentally regulated transcription in spermatogenesis.
Development
130
:
2841
2852
.

Badenhorst
P
,
Voas
M
,
Rebay
I
,
Wu
C
,
2002
Biological functions of the ISWI chromatin remodeling complex NURF.
Genes Dev.
16
:
3186
3198
.

Balakireva
M D
,
Shevelyov
Y
,
Nurminsky
D I
,
Livak
K J
,
Gvozdev
V A
,
1992
Structural organization and diversification of Y-linked sequences comprising Su(Ste) genes in Drosophila melanogaster.
Nucleic Acids Res.
20
:
3731
3736
.

Baldi
S
,
Becker
P B
,
2013
The variant histone H2A.V of Drosophila–three roles, two guises.
Chromosoma
122
:
245
258
.

Barckmann
B
,
Pierson
S
,
Dufourt
J
,
Papin
C
,
Armenise
C
et al. ,
2015
Aubergine iCLIP reveals piRNA-dependent decay of mRNAs involved in germ cell development in the early embryo.
Cell Rep.
12
:
1205
1216
.

Bastock
R
,
St Johnston
D
,
2008
Drosophila oogenesis.
Curr. Biol.
18
:
R1082
R1087
.

Beall
E L
,
Lewis
P W
,
Bell
M
,
Rocha
M
,
Jones
D L
et al. ,
2007
Discovery of tMAC: a Drosophila testis-specific meiotic arrest complex paralogous to Myb-Muv B.
Genes Dev.
21
:
904
919
.

Beisel
C
,
Imhof
A
,
Greene
J
,
Kremmer
E
,
Sauer
F
,
2002
Histone methylation by the Drosophila epigenetic transcriptional regulator Ash1.
Nature
419
:
857
862
.

Bernstein
B E
,
Mikkelsen
T S
,
Xie
X
,
Kamal
M
,
Huebert
D J
et al. ,
2006
A bivalent chromatin structure marks key developmental genes in embryonic stem cells.
Cell
125
:
315
326
.

Black
B E
,
Cleveland
D W
,
2011
Epigenetic centromere propagation and the nature of CENP-a nucleosomes.
Cell
144
:
471
479
.

Borner
K
,
Becker
P B
,
2016
Splice variants of the SWR1-type nucleosome remodeling factor Domino have distinct functions during Drosophila melanogaster oogenesis.
Development
143
:
3154
3167
.

Boudreault
A A
,
Cronier
D
,
Selleck
W
,
Lacoste
N
,
Utley
R T
et al. ,
2003
Yeast enhancer of polycomb defines global Esa1-dependent acetylation of chromatin.
Genes Dev.
17
:
1415
1428
.

Brasset
E
,
Taddei
A R
,
Arnaud
F
,
Faye
B
,
Fausto
A M
et al. ,
2006
Viral particles of the endogenous retrovirus ZAM from Drosophila melanogaster use a pre-existing endosome/exosome pathway for transfer to the oocyte.
Retrovirology
3
:
25
.

Brawley
C
,
Matunis
E
,
2004
Regeneration of male germline stem cells by spermatogonial dedifferentiation in vivo.
Science
304
:
1331
1334
.

Brendza
R P
,
Serbus
L R
,
Duffy
J B
,
Saxton
W M
,
2000
A function for kinesin I in the posterior transport of oskar mRNA and Staufen protein.
Science
289
:
2120
2122
.

Brendza
R P
,
Serbus
L R
,
Saxton
W M
,
Duffy
J B
,
2002
Posterior localization of dynein and dorsal-ventral axis formation depend on kinesin in Drosophila oocytes.
Curr. Biol.
12
:
1541
1545
.

Brennecke
J
,
Aravin
A A
,
Stark
A
,
Dus
M
,
Kellis
M
et al. ,
2007
Discrete small RNA-generating loci as master regulators of transposon activity in Drosophila.
Cell
128
:
1089
1103
.

Brennecke
J
,
Malone
C D
,
Aravin
A A
,
Sachidanandam
R
,
Stark
A
et al. ,
2008
An epigenetic role for maternally inherited piRNAs in transposon silencing.
Science
322
:
1387
1392
.

Brizuela
B J
,
Elfring
L
,
Ballard
J
,
Tamkun
J W
,
Kennison
J A
,
1994
Genetic analysis of the brahma gene of Drosophila melanogaster and polytene chromosome subdivisions 72AB.
Genetics
137
:
803
813
.

Brower-Toland
B
,
Findley
S D
,
Jiang
L
,
Liu
L
,
Yin
H
et al. ,
2007
Drosophila PIWI associates with chromatin and interacts directly with HP1a.
Genes Dev.
21
:
2300
2311
.

Buszczak
M
,
Spradling
A C
,
2006
Searching chromatin for stem cell identity.
Cell
125
:
233
236
.

Buszczak
M
,
Freeman
M R
,
Carlson
J R
,
Bender
M
,
Cooley
L
et al. ,
1999
Ecdysone response genes govern egg chamber development during mid-oogenesis in Drosophila.
Development
126
:
4581
4589
.

Buszczak
M
,
Paterno
S
,
Spradling
A C
,
2009
Drosophila stem cells share a common requirement for the histone H2B ubiquitin protease scrawny.
Science
323
:
248
251
.

Byrd
K N
,
Shearn
A
,
2003
ASH1, a Drosophila trithorax group protein, is required for methylation of lysine 4 residues on histone H3.
Proc. Natl. Acad. Sci. USA
100
:
11535
11540
.

Cabrera
G R
,
Godt
D
,
Fang
P Y
,
Couderc
J L
,
Laski
F A
,
2002
Expression pattern of Gal4 enhancer trap insertions into the bric a brac locus generated by P element replacement.
Genesis
34
:
62
65
.

Cairns
J
,
1975
Mutation selection and the natural history of cancer.
Nature
255
:
197
200
.

Cao
R
,
Wang
L
,
Wang
H
,
Xia
L
,
Erdjument-Bromage
H
et al. ,
2002
Role of histone H3 lysine 27 methylation in Polycomb-group silencing.
Science
298
:
1039
1043
.

Caporilli
S
,
Yu
Y
,
Jiang
J
,
White-Cooper
H
,
2013
The RNA export factor, Nxt1, is required for tissue specific transcriptional regulation.
PLoS Genet.
9
:
e1003526
.

Capuano
F
,
Mulleder
M
,
Kok
R
,
Blom
H J
,
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
.

Carreira-Rosario
A
,
Bhargava
V
,
Hillebrand
J
,
Kollipara
R K
,
Ramaswami
M
et al. ,
2016
Repression of pumilio protein expression by Rbfox1 promotes germ cell differentiation.
Dev. Cell
36
:
562
571
.

Casper
A L
,
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
.

Cavalli
G
,
Paro
R
,
1998
The Drosophila Fab-7 chromosomal element conveys epigenetic inheritance during mitosis and meiosis.
Cell
93
:
505
518
.

Chambeyron
S
,
Popkova
A
,
Payen-Groschene
G
,
Brun
C
,
Laouini
D
et al. ,
2008
piRNA-mediated nuclear accumulation of retrotransposon transcripts in the Drosophila female germline.
Proc. Natl. Acad. Sci. USA
105
:
14964
14969
.

Chaturvedi
D
,
Inaba
M
,
Scoggin
S
,
Buszczak
M
,
2016
Drosophila CG2469 encodes a homolog of human CTR9 and is essential for development.
G3
6
:
3849
3857
.

Chen
D
,
McKearin
D
,
2003
a
Dpp signaling silences bam transcription directly to establish asymmetric divisions of germline stem cells.
Curr. Biol.
13
:
1786
1791
.

Chen
D
,
McKearin
D
,
2005
Gene circuitry controlling a stem cell niche.
Curr. Biol.
15
:
179
184
.

Chen
D
,
McKearin
D M
,
2003
b
A discrete transcriptional silencer in the bam gene determines asymmetric division of the Drosophila germline stem cell.
Development
130
:
1159
1170
.

Chen
D
,
Wu
C
,
Zhao
S
,
Geng
Q
,
Gao
Y
et al. ,
2014
Three RNA binding proteins form a complex to promote differentiation of germline stem cell lineage in Drosophila.
PLoS Genet.
10
:
e1004797
.

Chen
H
,
Chen
X
,
Zheng
Y
,
2013
The nuclear lamina regulates germline stem cell niche organization via modulation of EGFR signaling.
Cell Stem Cell
13
:
73
86
.

Chen
X
,
Hiller
M
,
Sancak
Y
,
Fuller
M T
,
2005
Tissue-specific TAFs counteract Polycomb to turn on terminal differentiation.
Science
310
:
869
872
.

Chen
X
,
Lu
C
,
Prado
J R
,
Eun
S H
,
Fuller
M T
,
2011
Sequential changes at differentiation gene promoters as they become active in a stem cell lineage.
Development
138
:
2441
2450
.

Chen
Y
,
Pane
A
,
Schupbach
T
,
2007
Cutoff and aubergine mutations result in retrotransposon upregulation and checkpoint activation in Drosophila.
Curr. Biol.
17
:
637
642
.

Chen
Y C
,
Stuwe
E
,
Luo
Y
,
Ninova
M
,
Le Thomas
A
et al. ,
2016
Cutoff suppresses RNA polymerase II termination to ensure expression of piRNA precursors.
Mol. Cell
63
:
97
109
.

Cheng
J
,
Turkel
N
,
Hemati
N
,
Fuller
M T
,
Hunt
A J
et al. ,
2008
Centrosome misorientation reduces stem cell division during ageing.
Nature
456
:
599
604
.

Cheng
J
,
Tiyaboonchai
A
,
Yamashita
Y M
,
Hunt
A J
,
2011
Asymmetric division of cyst stem cells in Drosophila testis is ensured by anaphase spindle repositioning.
Development
138
:
831
837
.

Cherry
C M
,
Matunis
E L
,
2010
Epigenetic regulation of stem cell maintenance in the Drosophila testis via the nucleosome-remodeling factor NURF.
Cell Stem Cell
6
:
557
567
.

Chittuluru
J R
,
Chaban
Y
,
Monnet-Saksouk
J
,
Carrozza
M J
,
Sapountzi
V
et al. ,
2011
Structure and nucleosome interaction of the yeast NuA4 and Piccolo-NuA4 histone acetyltransferase complexes.
Nat. Struct. Mol. Biol.
18
:
1196
1203
.

Cinalli
R M
,
Rangan
P
,
Lehmann
R
,
2008
Germ cells are forever.
Cell
132
:
559
562
.

Clarke
M F
,
Fuller
M
,
2006
Stem cells and cancer: two faces of eve.
Cell
124
:
1111
1115
.

Cler
E
,
Papai
G
,
Schultz
P
,
Davidson
I
,
2009
Recent advances in understanding the structure and function of general transcription factor TFIID.
Cell. Mol. Life Sci.
66
:
2123
2134
.

Clough
E
,
Moon
W
,
Wang
S
,
Smith
K
,
Hazelrigg
T
,
2007
Histone methylation is required for oogenesis in Drosophila.
Development
134
:
157
165
.

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
.

Cook
H A
,
Koppetsch
B S
,
Wu
J
,
Theurkauf
W E
,
2004
The Drosophila SDE3 homolog armitage is required for oskar mRNA silencing and embryonic axis specification.
Cell
116
:
817
829
.

Corona
D F
,
Clapier
C R
,
Becker
P B
,
Tamkun
J W
,
2002
Modulation of ISWI function by site-specific histone acetylation.
EMBO Rep.
3
:
242
247
.

Courtot
C
,
Fankhauser
C
,
Simanis
V
,
Lehner
C F
,
1992
The Drosophila cdc25 homolog twine is required for meiosis.
Development
116
:
405
416
.

Creyghton
M P
,
Markoulaki
S
,
Levine
S S
,
Hanna
J
,
Lodato
M A
et al. ,
2008
H2AZ is enriched at polycomb complex target genes in ES cells and is necessary for lineage commitment.
Cell
135
:
649
661
.

Czech
B
,
Preall
J B
,
McGinn
J
,
Hannon
G J
,
2013
A transcriptome-wide RNAi screen in the Drosophila ovary reveals factors of the germline piRNA pathway.
Mol. Cell
50
:
749
761
.

Czermin
B
,
Melfi
R
,
McCabe
D
,
Seitz
V
,
Imhof
A
et al. ,
2002
Drosophila enhancer of zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Polycomb sites.
Cell
111
:
185
196
.

Dahanukar
A
,
Wharton
R P
,
1996
The nanos gradient in Drosophila embryos is generated by translational regulation.
Genes Dev.
10
:
2610
2620
.

Darricarrere
N
,
Liu
N
,
Watanabe
T
,
Lin
H
,
2013
Function of Piwi, a nuclear Piwi/Argonaute protein, is independent of its slicer activity.
Proc. Natl. Acad. Sci. USA
110
:
1297
1302
.

Davies
E L
,
Lim
J G
,
Joo
W J
,
Tam
C H
,
Fuller
M T
,
2013
The transcriptional regulator lola is required for stem cell maintenance and germ cell differentiation in the Drosophila testis.
Dev. Biol.
373
:
310
321
.

Davring
L
,
Sunner
M
,
1973
Female meiosis and embryonic mitosis in Drosophila melanogaster. I. Meiosis and fertilization.
Hereditas
73
:
51
64
.

de Cuevas
M
,
Spradling
A C
,
1998
Morphogenesis of the Drosophila fusome and its implications for oocyte specification.
Development
125
:
2781
2789
.

Desset
S
,
Buchon
N
,
Meignin
C
,
Coiffet
M
,
Vaury
C
,
2008
In Drosophila melanogaster the COM locus directs the somatic silencing of two retrotransposons through both Piwi-dependent and -independent pathways.
PLoS One
3
:
e1526
.

Deuring
R
,
Fanti
L
,
Armstrong
J A
,
Sarte
M
,
Papoulas
O
et al. ,
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
.

Di Stefano
L
,
Ji
J Y
,
Moon
N S
,
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
.

Doggett
K
,
Jiang
J
,
Aleti
G
,
White-Cooper
H
,
2011
Wake-up-call, a lin-52 paralogue, and always early, a lin-9 homologue physically interact, but have opposing functions in regulating testis-specific gene expression.
Dev. Biol.
355
:
381
393
.

Dönertas
D
,
Sienski
G
,
Brennecke
J
,
2013
Drosophila Gtsf1 is an essential component of the Piwi-mediated transcriptional silencing complex.
Genes Dev.
27
:
1693
1705
.

Dorus
S
,
Busby
S A
,
Gerike
U
,
Shabanowitz
J
,
Hunt
D F
et al. ,
2006
Genomic and functional evolution of the Drosophila melanogaster sperm proteome.
Nat. Genet.
38
:
1440
1445
.

Duncan
J E
,
Warrior
R
,
2002
The cytoplasmic dynein and kinesin motors have interdependent roles in patterning the Drosophila oocyte.
Curr. Biol.
12
:
1982
1991
.

Eberhart
C G
,
Maines
J Z
,
Wasserman
S A
,
1996
Meiotic cell cycle requirement for a fly homologue of human deleted in Azoospermia.
Nature
381
:
783
785
.

Ebert
A
,
Schotta
G
,
Lein
S
,
Kubicek
S
,
Krauss
V
et al. ,
2004
Su(var) genes regulate the balance between euchromatin and heterochromatin in Drosophila.
Genes Dev.
18
:
2973
2983
.

Ebert
A
,
Lein
S
,
Schotta
G
,
Reuter
G
,
2006
Histone modification and the control of heterochromatic gene silencing in Drosophila.
Chromosome Res.
14
:
377
392
.

Eddy
E M
,
1975
Germ plasm and the differentiation of the germ cell line.
Int. Rev. Cytol.
43
:
229
280
.

Eisen
J A
,
Sweder
K S
,
Hanawalt
P C
,
1995
Evolution of the SNF2 family of proteins: subfamilies with distinct sequences and functions.
Nucleic Acids Res.
23
:
2715
2723
.

Eissenberg
J C
,
Wong
M
,
Chrivia
J C
,
2005
Human SRCAP and Drosophila melanogaster DOM are homologs that function in the notch signaling pathway.
Mol. Cell. Biol.
25
:
6559
6569
.

Elfring
L K
,
Daniel
C
,
Papoulas
O
,
Deuring
R
,
Sarte
M
et al. ,
1998
Genetic analysis of brahma: the Drosophila homolog of the yeast chromatin remodeling factor SWI2/SNF2.
Genetics
148
:
251
265
.

Eliazer
S
,
Shalaby
N A
,
Buszczak
M
,
2011
Loss of lysine-specific demethylase 1 nonautonomously causes stem cell tumors in the Drosophila ovary.
Proc. Natl. Acad. Sci. USA
108
:
7064
7069
.

Eliazer
S
,
Palacios
V
,
Wang
Z
,
Kollipara
R K
,
Kittler
R
et al. ,
2014
Lsd1 restricts the number of germline stem cells by regulating multiple targets in escort cells.
PLoS Genet.
10
:
e1004200
.

Ephrussi
A
,
Lehmann
R
,
1992
Induction of germ cell formation by oskar.
Nature
358
:
387
392
.

Ephrussi
A
,
Dickinson
L K
,
Lehmann
R
,
1991
Oskar organizes the germ plasm and directs localization of the posterior determinant nanos.
Cell
66
:
37
50
.

Eun
S H
,
Stoiber
P M
,
Wright
H J
,
McMurdie
K E
,
Choi
C H
et al. ,
2013
MicroRNAs downregulate bag of marbles to ensure proper terminal differentiation in the Drosophila male germline.
Development
140
:
23
30
.

Eun
S H
,
Shi
Z
,
Cui
K
,
Zhao
K
,
Chen
X
,
2014
A non-cell autonomous role of E(z) to prevent germ cells from turning on a somatic cell marker.
Science
343
:
1513
1516
.

Fadloun
A
,
Le Gras
S
,
Jost
B
,
Ziegler-Birling
C
,
Takahashi
H
et al. ,
2013
Chromatin signatures and retrotransposon profiling in mouse embryos reveal regulation of LINE-1 by RNA.
Nat. Struct. Mol. Biol.
20
:
332
338
.

Fagegaltier
D
,
Falciatori
I
,
Czech
B
,
Castel
S
,
Perrimon
N
et al. ,
2016
Oncogenic transformation of Drosophila somatic cells induces a functional piRNA pathway.
Genes Dev.
30
:
1623
1635
.

Fanti
L
,
Giovinazzo
G
,
Berloco
M
,
Pimpinelli
S
,
1998
The heterochromatin protein 1 prevents telomere fusions in Drosophila.
Mol. Cell
2
:
527
538
.

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
.

Filipowicz
W
,
Bhattacharyya
S N
,
Sonenberg
N
,
2008
Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight?
Nat. Rev. Genet.
9
:
102
114
.

Findley
S D
,
Tamanaha
M
,
Clegg
N J
,
Ruohola-Baker
H
,
2003
Maelstrom, a Drosophila spindle-class gene, encodes a protein that colocalizes with Vasa and RDE1/AGO1 homolog, aubergine, in nuage.
Development
130
:
859
871
.

Fischle
W
,
Wang
Y
,
Jacobs
S A
,
Kim
Y
,
Allis
C D
et al. ,
2003
Molecular basis for the discrimination of repressive methyl-lysine marks in histone H3 by Polycomb and HP1 chromodomains.
Genes Dev.
17
:
1870
1881
.

Foley
K
,
Cooley
L
,
1998
Apoptosis in late stage Drosophila nurse cells does not require genes within the H99 deficiency.
Development
125
:
1075
1082
.

Forstemann
K
,
Tomari
Y
,
Du
T
,
Vagin
V V
,
Denli
A M
et al. ,
2005
Normal microRNA maturation and germ-line stem cell maintenance requires loquacious, a double-stranded RNA-binding domain protein.
PLoS Biol.
3
:
e236
.

Frydrychova
R C
,
Mason
J M
,
Archer
T K
,
2008
HP1 is distributed within distinct chromatin domains at Drosophila telomeres.
Genetics
180
:
121
131
.

Fuller
M T
,
1993
Spermatogenesis
, in
The Development of Drosophila melanogaster
,
Vol. I
, edited by
Bate
M
,
Martinez Arias
A
.
Cold Spring Harbor Press
,
Cold Spring Harbor, NY
.

Galarneau
L
,
Nourani
A
,
Boudreault
A A
,
Zhang
Y
,
Heliot
L
et al. ,
2000
Multiple links between the NuA4 histone acetyltransferase complex and epigenetic control of transcription.
Mol. Cell
5
:
927
937
.

Gan
Q
,
Chepelev
I
,
Wei
G
,
Tarayrah
L
,
Cui
K
et al. ,
2010
a
Dynamic regulation of alternative splicing and chromatin structure in Drosophila gonads revealed by RNA-seq.
Cell Res.
20
:
763
783
.

Gan
Q
,
Schones
D E
,
Ho Eun
S
,
Wei
G
,
Cui
K
et al. ,
2010
b
Monovalent and unpoised status of most genes in undifferentiated cell-enriched Drosophila testis.
Genome Biol.
11
:
R42
.

Gaydos
L J
,
Wang
W
,
Strome
S
,
2014
Gene repression. H3K27me and PRC2 transmit a memory of repression across generations and during development.
Science
345
:
1515
1518
.

Goll
M G
,
Kirpekar
F
,
Maggert
K A
,
Yoder
J A
,
Hsieh
C L
et al. ,
2006
Methylation of tRNAAsp by the DNA methyltransferase homolog Dnmt2.
Science
311
:
395
398
.

Golumbeski
G S
,
Bardsley
A
,
Tax
F
,
Boswell
R E
,
1991
Tudor, a posterior-group gene of Drosophila melanogaster, encodes a novel protein and an mRNA localized during mid-oogenesis.
Genes Dev.
5
:
2060
2070
.

Gonczy
P
,
Matunis
E
,
DiNardo
S
,
1997
Bag-of-marbles and benign gonial cell neoplasm act in the germline to restrict proliferation during Drosophila spermatogenesis.
Development
124
:
4361
4371
.

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
.

Goriaux
C
,
Desset
S
,
Renaud
Y
,
Vaury
C
,
Brasset
E
,
2014
Transcriptional properties and splicing of the flamenco piRNA cluster.
EMBO Rep.
15
:
411
418
.

Gowher
H
,
Leismann
O
,
Jeltsch
A
,
2000
DNA of Drosophila melanogaster contains 5-methylcytosine.
EMBO J.
19
:
6918
6923
.

Grieder
N C
,
de Cuevas
M
,
Spradling
A C
,
2000
The fusome organizes the microtubule network during oocyte differentiation in Drosophila.
Development
127
:
4253
4264
.

Gu
S
,
Jin
L
,
Zhang
F
,
Sarnow
P
,
Kay
M A
,
2009
Biological basis for restriction of microRNA targets to the 3′ untranslated region in mammalian mRNAs.
Nat. Struct. Mol. Biol.
16
:
144
150
.

Guenther
M G
,
Levine
S S
,
Boyer
L A
,
Jaenisch
R
,
Young
R A
,
2007
A chromatin landmark and transcription initiation at most promoters in human cells.
Cell
130
:
77
88
.

Gunawardane
L S
,
Saito
K
,
Nishida
K M
,
Miyoshi
K
,
Kawamura
Y
et al. ,
2007
A slicer-mediated mechanism for repeat-associated siRNA 5′ end formation in Drosophila.
Science
315
:
1587
1590
.

Haase
A D
,
Fenoglio
S
,
Muerdter
F
,
Guzzardo
P M
,
Czech
B
et al. ,
2010
Probing the initiation and effector phases of the somatic piRNA pathway in Drosophila.
Genes Dev.
24
:
2499
2504
.

Hagstrom
K
,
Muller
M
,
Schedl
P
,
1996
Fab-7 functions as a chromatin domain boundary to ensure proper segment specification by the Drosophila bithorax complex.
Genes Dev.
10
:
3202
3215
.

Hales
K G
,
Fuller
M T
,
1997
Developmentally regulated mitochondrial fusion mediated by a conserved, novel, predicted GTPase.
Cell
90
:
121
129
.

Hallson
G
,
Hollebakken
R E
,
Li
T
,
Syrzycka
M
,
Kim
I
et al. ,
2012
dSet1 is the main H3K4 di- and tri-methyltransferase throughout Drosophila development.
Genetics
190
:
91
100
.

Han
B W
,
Wang
W
,
Li
C
,
Weng
Z
,
Zamore
P D
,
2015
Noncoding RNA. piRNA-guided transposon cleavage initiates Zucchini-dependent, phased piRNA production.
Science
348
:
817
821
.

Handler
D
,
Olivieri
D
,
Novatchkova
M
,
Gruber
F S
,
Meixner
K
et al. ,
2011
A systematic analysis of Drosophila TUDOR domain-containing proteins identifies Vreteno and the Tdrd12 family as essential primary piRNA pathway factors.
EMBO J.
30
:
3977
3993
.

Handler
D
,
Meixner
K
,
Pizka
M
,
Lauss
K
,
Schmied
C
et al. ,
2013
The genetic makeup of the Drosophila piRNA pathway.
Mol. Cell
50
:
762
777
.

Hardy
R W
,
Tokuyasu
K T
,
Lindsley
D L
,
Garavito
M
,
1979
The germinal proliferation center in the testis of Drosophila melanogaster.
J. Ultrastruct. Res.
69
:
180
190
.

Harris
A N
,
Macdonald
P M
,
2001
Aubergine encodes a Drosophila polar granule component required for pole cell formation and related to eIF2C.
Development
128
:
2823
2832
.

Harrison
M M
,
Eisen
M B
,
2015
Transcriptional activation of the zygotic genome in Drosophila.
Curr. Top. Dev. Biol.
113
:
85
112
.

Hartman
T R
,
Zinshteyn
D
,
Schofield
H K
,
Nicolas
E
,
Okada
A
et al. ,
2010
Drosophila Boi limits Hedgehog levels to suppress follicle stem cell proliferation.
J. Cell Biol.
191
:
943
952
.

Hay
B
,
Jan
L Y
,
Jan
Y N
,
1988
A protein component of Drosophila polar granules is encoded by vasa and has extensive sequence similarity to ATP-dependent helicases.
Cell
55
:
577
587
.

Hayashi
R
,
Schnabl
J
,
Handler
D
,
Mohn
F
,
Ameres
S L
et al. ,
2016
Genetic and mechanistic diversity of piRNA 3′-end formation.
Nature
539
:
588
592
.

Hayashi
S
,
Ito
K
,
Sado
Y
,
Taniguchi
M
,
Akimoto
A
et al. ,
2002
GETDB, a database compiling expression patterns and molecular locations of a collection of Gal4 enhancer traps.
Genesis
34
:
58
61
.

He
J
,
Xuan
T
,
Xin
T
,
An
H
,
Wang
J
et al. ,
2014
Evidence for chromatin-remodeling complex PBAP-controlled maintenance of the Drosophila ovarian germline stem cells.
PLoS One
9
:
e103473
.

Heard
E
,
Martienssen
R A
,
2014
Transgenerational epigenetic inheritance: myths and mechanisms.
Cell
157
:
95
109
.

Henikoff
S
,
Furuyama
T
,
Ahmad
K
,
2004
Histone variants, nucleosome assembly and epigenetic inheritance.
Trends Genet.
20
:
320
326
.

Hennig
W
,
Weyrich
A
,
2013
Histone modifications in the male germ line of Drosophila.
BMC Dev. Biol.
13
:
7
.

Hermann
A
,
Schmitt
S
,
Jeltsch
A
,
2003
The human Dnmt2 has residual DNA-(cytosine-C5) methyltransferase activity.
J. Biol. Chem.
278
:
31717
31721
.

Hermant
C
,
Boivin
A
,
Teysset
L
,
Delmarre
V
,
Asif-Laidin
A
et al. ,
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
.

Herz
H M
,
Madden
L D
,
Chen
Z
,
Bolduc
C
,
Buff
E
et al. ,
2010
The H3K27me3 demethylase dUTX is a suppressor of Notch- and Rb-dependent tumors in Drosophila.
Mol. Cell. Biol.
30
:
2485
2497
.

Hiller
M
,
Chen
X
,
Pringle
M J
,
Suchorolski
M
,
Sancak
Y
et al. ,
2004
Testis-specific TAF homologs collaborate to control a tissue-specific transcription program.
Development
131
:
5297
5308
.

Hiller
M A
,
Lin
T Y
,
Wood
C
,
Fuller
M T
,
2001
Developmental regulation of transcription by a tissue-specific TAF homolog.
Genes Dev.
15
:
1021
1030
.

Hirakata
S
,
Siomi
M C
,
2016
piRNA biogenesis in the germline: from transcription of piRNA genomic sources to piRNA maturation.
Biochim. Biophys. Acta
1859
:
82
92
.

Holtzman
S
,
Miller
D
,
Eisman
R
,
Kuwayama
H
,
Niimi
T
et al. ,
2010
Transgenic tools for members of the genus Drosophila with sequenced genomes.
Fly (Austin)
4
:
349
362
.

Homolka
D
,
Pandey
R R
,
Goriaux
C
,
Brasset
E
,
Vaury
C
et al. ,
2015
PIWI slicing and RNA elements in precursors instruct directional primary piRNA biogenesis.
Cell Rep.
12
:
418
428
.

Honda
S
,
Kirino
Y
,
Maragkakis
M
,
Alexiou
P
,
Ohtaki
A
et al. ,
2013
Mitochondrial protein BmPAPI modulates the length of mature piRNAs.
RNA
19
:
1405
1418
.

Hong
A
,
Lee-Kong
S
,
Iida
T
,
Sugimura
I
,
Lilly
M A
,
2003
The p27cip/kip ortholog dacapo maintains the Drosophila oocyte in prophase of meiosis I.
Development
130
:
1235
1242
.

Hota
S K
,
Bruneau
B G
,
2016
ATP-dependent chromatin remodeling during mammalian development.
Development
143
:
2882
2897
.

Houwing
S
,
Kamminga
L M
,
Berezikov
E
,
Cronembold
D
,
Girard
A
et al. ,
2007
A role for Piwi and piRNAs in germ cell maintenance and transposon silencing in zebrafish.
Cell
129
:
69
82
.

Houwing
S
,
Berezikov
E
,
Ketting
R F
,
2008
Zili is required for germ cell differentiation and meiosis in zebrafish.
EMBO J.
27
:
2702
2711
.

Huang
C R
,
Burns
K H
,
Boeke
J D
,
2012
Active transposition in genomes.
Annu. Rev. Genet.
46
:
651
675
.

Huang
F
,
Paulson
A
,
Dutta
A
,
Venkatesh
S
,
Smolle
M
et al. ,
2014
Histone acetyltransferase Enok regulates oocyte polarization by promoting expression of the actin nucleation factor spire.
Genes Dev.
28
:
2750
2763
.

Huang
X A
,
Yin
H
,
Sweeney
S
,
Raha
D
,
Snyder
M
et al. ,
2013
A major epigenetic programming mechanism guided by piRNAs.
Dev. Cell
24
:
502
516
.

Hur
J K
,
Luo
Y
,
Moon
S
,
Ninova
M
,
Marinov
G K
et al. ,
2016
Splicing-independent loading of TREX on nascent RNA is required for efficient expression of dual-strand piRNA clusters in Drosophila.
Genes Dev.
30
:
840
855
.

Huynh
J R
,
St Johnston
D
,
2000
The role of BicD, Egl, Orb and the microtubules in the restriction of meiosis to the Drosophila oocyte.
Development
127
:
2785
2794
.

Huynh
J R
,
Petronczki
M
,
Knoblich
J A
,
St Johnston
D
,
2001
a
Bazooka and PAR-6 are required with PAR-1 for the maintenance of oocyte fate in Drosophila.
Curr. Biol.
11
:
901
906
.

Huynh
J R
,
Shulman
J M
,
Benton
R
,
St Johnston
D
,
2001
b
PAR-1 is required for the maintenance of oocyte fate in Drosophila.
Development
128
:
1201
1209
.

Inaba
M
,
Yuan
H
,
Salzmann
V
,
Fuller
M T
,
Yamashita
Y M
,
2010
E-cadherin is required for centrosome and spindle orientation in Drosophila male germline stem cells.
PLoS One
5
:
e12473
.

Inaba
M
,
Venkei
Z G
,
Yamashita
Y M
,
2015
The polarity protein Baz forms a platform for the centrosome orientation during asymmetric stem cell division in the Drosophila male germline.
Elife
4:
04960.

Insco
M L
,
Leon
A
,
Tam
C H
,
McKearin
D M
,
Fuller
M T
,
2009
Accumulation of a differentiation regulator specifies transit amplifying division number in an adult stem cell lineage.
Proc. Natl. Acad. Sci. USA
106
:
22311
22316
.

Insco
M L
,
Bailey
A S
,
Kim
J
,
Olivares
G H
,
Wapinski
O L
et al. ,
2012
A self-limiting switch based on translational control regulates the transition from proliferation to differentiation in an adult stem cell lineage.
Cell Stem Cell
11
:
689
700
.

Iovino
N
,
Pane
A
,
Gaul
U
,
2009
miR-184 has multiple roles in Drosophila female germline development.
Dev. Cell
17
:
123
133
.

Iovino
N
,
Ciabrelli
F
,
Cavalli
G
,
2013
PRC2 controls Drosophila oocyte cell fate by repressing cell cycle genes.
Dev. Cell
26
:
431
439
.

Ipsaro
J J
,
Haase
A D
,
Knott
S R
,
Joshua-Tor
L
,
Hannon
G J
,
2012
The structural biochemistry of Zucchini implicates it as a nuclease in piRNA biogenesis.
Nature
491
:
279
283
.

Issigonis
M
,
Tulina
N
,
de Cuevas
M
,
Brawley
C
,
Sandler
L
et al. ,
2009
JAK-STAT signal inhibition regulates competition in the Drosophila testis stem cell niche.
Science
326
:
153
156
.

Iwasaki
Y W
,
Siomi
M C
,
Siomi
H
,
2015
PIWI-interacting RNA: its biogenesis and functions.
Annu. Rev. Biochem.
84
:
405
433
.

Iwasaki
Y W
,
Murano
K
,
Ishizu
H
,
Shibuya
A
,
Iyoda
Y
et al. ,
2016
Piwi modulates chromatin accessibility by regulating multiple factors including histone H1 to repress transposons.
Mol. Cell
.
63
:
408
419
.

Jang
J K
,
Sherizen
D E
,
Bhagat
R
,
Manheim
E A
,
McKim
K S
,
2003
Relationship of DNA double-strand breaks to synapsis in Drosophila.
J. Cell Sci.
116
:
3069
3077
.

Janic
A
,
Mendizabal
L
,
Llamazares
S
,
Rossell
D
,
Gonzalez
C
,
2010
Ectopic expression of germline genes drives malignant brain tumor growth in Drosophila.
Science
330
:
1824
1827
.

Januschke
J
,
Gervais
L
,
Dass
S
,
Kaltschmidt
J A
,
Lopez-Schier
H
et al. ,
2002
Polar transport in the Drosophila oocyte requires dynein and kinesin I cooperation.
Curr. Biol.
12
:
1971
1981
.

Ji
S
,
Li
C
,
Hu
L
,
Liu
K
,
Mei
J
et al. ,
2017
Bam-dependent deubiquitinase complex can disrupt germ-line stem cell maintenance by targeting cyclin A.
Proc. Natl. Acad. Sci. USA
114
:
6316
6321
.

Jiang
J
,
White-Cooper
H
,
2003
Transcriptional activation in Drosophila spermatogenesis involves the mutually dependent function of aly and a novel meiotic arrest gene cookie monster.
Development
130
:
563
573
.

Jiang
J
,
Benson
E
,
Bausek
N
,
Doggett
K
,
White-Cooper
H
,
2007
Tombola, a tesmin/TSO1-family protein, regulates transcriptional activation in the Drosophila male germline and physically interacts with always early.
Development
134
:
1549
1559
.

Jin
Z
,
Xie
T
,
2007
Dcr-1 maintains Drosophila ovarian stem cells.
Curr. Biol.
17
:
539
544
.

Kai
T
,
Spradling
A
,
2003
An empty Drosophila stem cell niche reactivates the proliferation of ectopic cells.
Proc. Natl. Acad. Sci. USA
100
:
4633
4638
.

Kai
T
,
Spradling
A
,
2004
Differentiating germ cells can revert into functional stem cells in Drosophila melanogaster ovaries.
Nature
428
:
564
569
.

Kaminker
J S
,
Bergman
C M
,
Kronmiller
B
,
Carlson
J
,
Svirskas
R
et al. ,
2002
The transposable elements of the Drosophila melanogaster euchromatin: a genomics perspective.
Genome Biol.
3
:
RESEARCH0084
.

Kawaoka
S
,
Izumi
N
,
Katsuma
S
,
Tomari
Y
,
2011
3′ end formation of PIWI-interacting RNAs in vitro.
Mol. Cell
43
:
1015
1022
.

Kawase
E
,
Wong
M D
,
Ding
B C
,
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
.

Kelleher
E S
,
Barbash
D A
,
2013
Analysis of piRNA-mediated silencing of active TEs in Drosophila melanogaster suggests limits on the evolution of host genome defense.
Mol. Biol. Evol.
30
:
1816
1829
.

Khurana
J S
,
Wang
J
,
Xu
J
,
Koppetsch
B S
,
Thomson
T C
et al. ,
2011
Adaptation to P element transposon invasion in Drosophila melanogaster.
Cell
147
:
1551
1563
.

Kiger
A A
,
White-Cooper
H
,
Fuller
M T
,
2000
Somatic support cells restrict germline stem cell self-renewal and promote differentiation.
Nature
407
:
750
754
.

Kiger
A A
,
Jones
D L
,
Schulz
C
,
Rogers
M B
,
Fuller
M T
,
2001
Stem cell self-renewal specified by JAK-STAT activation in response to a support cell cue.
Science
294
:
2542
2545
.

Kim
J
,
Lu
C
,
Srinivasan
S
,
Awe
S
,
Brehm
A
et al. ,
2017
Blocking promiscuous activation at cryptic promoters directs cell type-specific gene expression.
Science
356
:
717
721
.

Kim-Ha
J
,
Smith
J L
,
Macdonald
P M
,
1991
Oskar mRNA is localized to the posterior pole of the Drosophila oocyte.
Cell
66
:
23
35
.

King
F J
,
Szakmary
A
,
Cox
D N
,
Lin
H
,
2001
Yb modulates the divisions of both germline and somatic stem cells through piwi- and hh-mediated mechanisms in the Drosophila ovary.
Mol. Cell
7
:
497
508
.

King
R C
,
1970
The meiotic behavior of the Drosophila oocyte.
Int. Rev. Cytol.
28
:
125
168
.

Klar
A J
,
2007
Lessons learned from studies of fission yeast mating-type switching and silencing.
Annu. Rev. Genet.
41
:
213
236
.

Klattenhoff
C
,
Bratu
D P
,
McGinnis-Schultz
N
,
Koppetsch
B S
,
Cook
H A
et al. ,
2007
Drosophila rasiRNA pathway mutations disrupt embryonic axis specification through activation of an ATR/Chk2 DNA damage response.
Dev. Cell
12
:
45
55
.

Klattenhoff
C
,
Xi
H
,
Li
C
,
Lee
S
,
Xu
J
et al. ,
2009
The Drosophila HP1 homolog Rhino is required for transposon silencing and piRNA production by dual-strand clusters.
Cell
138
:
1137
1149
.

Klenov
M S
,
Lavrov
S A
,
Stolyarenko
A D
,
Ryazansky
S S
,
Aravin
A A
et al. ,
2007
Repeat-associated siRNAs cause chromatin silencing of retrotransposons in the Drosophila melanogaster germline.
Nucleic Acids Res.
35
:
5430
5438
.

Klenov
M S
,
Sokolova
O A
,
Yakushev
E Y
,
Stolyarenko
A D
,
Mikhaleva
E A
et al. ,
2011
Separation of stem cell maintenance and transposon silencing functions of Piwi protein.
Proc. Natl. Acad. Sci. USA
108
:
18760
18765
.

Klenov
M S
,
Lavrov
S A
,
Korbut
A P
,
Stolyarenko
A D
,
Yakushev
E Y
et al. ,
2014
Impact of nuclear Piwi elimination on chromatin state in Drosophila melanogaster ovaries.
Nucleic Acids Res.
42
:
6208
6218
.

Klymenko
T
,
Muller
J
,
2004
The histone methyltransferases Trithorax and Ash1 prevent transcriptional silencing by Polycomb group proteins.
EMBO Rep.
5
:
373
377
.

Kofler
R
,
Betancourt
A J
,
Schlotterer
C
,
2012
Sequencing of pooled DNA samples (Pool-Seq) uncovers complex dynamics of transposable element insertions in Drosophila melanogaster.
PLoS Genet.
8
:
e1002487
.

Konig
J
,
Zarnack
K
,
Rot
G
,
Curk
T
,
Kayikci
M
et al. ,
2010
iCLIP reveals the function of hnRNP particles in splicing at individual nucleotide resolution.
Nat. Struct. Mol. Biol.
17
:
909
915
.

Krauss
V
,
Reuter
G
,
2011
DNA methylation in Drosophila–a critical evaluation.
Prog. Mol. Biol. Transl. Sci.
101
:
177
191
.

Kusch
T
,
Florens
L
,
Macdonald
W H
,
Swanson
S K
,
Glaser
R L
et al. ,
2004
Acetylation by Tip60 is required for selective histone variant exchange at DNA lesions.
Science
306
:
2084
2087
.

Kuzmichev
A
,
Nishioka
K
,
Erdjument-Bromage
H
,
Tempst
P
,
Reinberg
D
,
2002
Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein.
Genes Dev.
16
:
2893
2905
.

Kwon
S Y
,
Xiao
H
,
Wu
C
,
Badenhorst
P
,
2009
Alternative splicing of NURF301 generates distinct NURF chromatin remodeling complexes with altered modified histone binding specificities.
PLoS Genet.
5
:
e1000574
.

Laktionov
P P
,
White-Cooper
H
,
Maksimov
D A
,
Beliakin
S N
,
2014
[Transcription factor comr acts as a direct activator in the genetic program controlling spermatogenesis in D. melanogaster].
Mol. Biol. (Mosk.)
48
:
153
165
.

Lancaster
O M
,
Breuer
M
,
Cullen
C F
,
Ito
T
,
Ohkura
H
,
2010
The meiotic recombination checkpoint suppresses NHK-1 kinase to prevent reorganisation of the oocyte nucleus in Drosophila.
PLoS Genet.
6
:
e1001179
.

Landais
S
,
D’Alterio
C
,
Jones
D L
,
2014
Persistent replicative stress alters polycomb phenotypes and tissue homeostasis in Drosophila melanogaster.
Cell Rep.
7
:
859
870
.

Lander
E S
,
Linton
L M
,
Birren
B
,
Nusbaum
C
,
Zody
M C
et al. ,
2001
Initial sequencing and analysis of the human genome.
Nature
409
:
860
921
.

Lansdorp
P M
,
2007
Immortal strands? Give me a break.
Cell
129
:
1244
1247
.

Lantz
V
,
Chang
J S
,
Horabin
J I
,
Bopp
D
,
Schedl
P
,
1994
The Drosophila orb RNA-binding protein is required for the formation of the egg chamber and establishment of polarity.
Genes Dev.
8
:
598
613
.

Lasko
P F
,
Ashburner
M
,
1988
The product of the Drosophila gene vasa is very similar to eukaryotic initiation factor-4A.
Nature
335
:
611
617
.

Laver
J D
,
Marsolais
A J
,
Smibert
C A
,
Lipshitz
H D
,
2015
Regulation and function of maternal gene products during the maternal-to-zygotic transition in Drosophila.
Curr. Top. Dev. Biol.
113
:
43
84
.

Leatherman
J L
,
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
.

Leatherman
J L
,
Dinardo
S
,
2010
Germline self-renewal requires cyst stem cells and stat regulates niche adhesion in Drosophila testes.
Nat. Cell Biol.
12
:
806
811
.

Leblanc
P
,
Desset
S
,
Giorgi
F
,
Taddei
A R
,
Fausto
A M
et al. ,
2000
Life cycle of an endogenous retrovirus, ZAM, in Drosophila melanogaster.
J. Virol.
74
:
10658
10669
.

Lecuyer
E
,
Yoshida
H
,
Parthasarathy
N
,
Alm
C
,
Babak
T
et al. ,
2007
Global analysis of mRNA localization reveals a prominent role in organizing cellular architecture and function.
Cell
131
:
174
187
.

Lee
T
,
Luo
L
,
1999
Mosaic analysis with a repressible cell marker for studies of gene function in neuronal morphogenesis.
Neuron
22
:
451
461
.

Lenhart
K F
,
DiNardo
S
,
2015
Somatic cell encystment promotes abscission in germline stem cells following a regulated block in cytokinesis.
Dev. Cell
34
:
192
205
.

Le Thomas
A
,
Rogers
A K
,
Webster
A
,
Marinov
G K
,
Liao
S E
et al. ,
2013
Piwi induces piRNA-guided transcriptional silencing and establishment of a repressive chromatin state.
Genes Dev.
27
:
390
399
.

Le Thomas
A
,
Stuwe
E
,
Li
S
,
Du
J
,
Marinov
G
et al. ,
2014
Transgenerationally inherited piRNAs trigger piRNA biogenesis by changing the chromatin of piRNA clusters and inducing precursor processing.
Genes Dev.
28
:
1667
1680
.

Levine
M T
,
Vander Wende
H M
,
Malik
H S
,
2015
Mitotic fidelity requires transgenerational action of a testis-restricted HP1.
Elife
4
:
e07378
.

Li
C
,
Vagin
V V
,
Lee
S
,
Xu
J
,
Ma
S
et al. ,
2009
Collapse of germline piRNAs in the absence of Argonaute3 reveals somatic piRNAs in flies.
Cell
137
:
509
521
.

Li
X
,
Han
Y
,
Xi
R
,
2010
Polycomb group genes Psc and Su(z)2 restrict follicle stem cell self-renewal and extrusion by controlling canonical and noncanonical Wnt signaling.
Genes Dev.
24
:
933
946
.

Li
Y
,
Minor
N T
,
Park
J K
,
McKearin
D M
,
Maines
J Z
,
2009
Bam and Bgcn antagonize Nanos-dependent germ-line stem cell maintenance.
Proc. Natl. Acad. Sci. USA
106
:
9304
9309
.

Li
Y
,
Maines
J Z
,
Tastan
O Y
,
McKearin
D M
,
Buszczak
M
,
2012
Mei-P26 regulates the maintenance of ovarian germline stem cells by promoting BMP signaling.
Development
139
:
1547
1556
.

Li
Y
,
Zhang
Q
,
Carreira-Rosario
A
,
Maines
J Z
,
McKearin
D M
et al. ,
2013
Mei-p26 cooperates with Bam, Bgcn and Sxl to promote early germline development in the Drosophila ovary.
PLoS One
8
:
e58301
.

Li
Y
,
Ma
Q
,
Cherry
C M
,
Matunis
E L
,
2014
Steroid signaling promotes stem cell maintenance in the Drosophila testis.
Dev. Biol.
394
:
129
141
.

Lilly
M A
,
Spradling
A C
,
1996
The Drosophila endocycle is controlled by Cyclin E and lacks a checkpoint ensuring S-phase completion.
Genes Dev.
10
:
2514
2526
.

Lim
A K
,
Kai
T
,
2007
Unique germ-line organelle, nuage, functions to repress selfish genetic elements in Drosophila melanogaster.
Proc. Natl. Acad. Sci. USA
104
:
6714
6719
.

Lim
A K
,
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
.

Lim
C
,
Gandhi
S
,
Biniossek
M L
,
Feng
L
,
Schilling
O
et al. ,
2015
An aminopeptidase in the Drosophila testicular niche acts in germline stem cell maintenance and spermatogonial dedifferentiation.
Cell Rep.
13
:
315
325
.

Lim
R S
,
Kai
T
,
2015
A piece of the pi(e): the diverse roles of animal piRNAs and their PIWI partners.
Semin. Cell Dev. Biol.
47–48
:
17
31
.

Lin
T Y
,
Viswanathan
S
,
Wood
C
,
Wilson
P G
,
Wolf
N
et al. ,
1996
Coordinate developmental control of the meiotic cell cycle and spermatid differentiation in Drosophila males.
Development
122
:
1331
1341
.

Lingel
A
,
Simon
B
,
Izaurralde
E
,
Sattler
M
,
2003
Structure and nucleic-acid binding of the Drosophila Argonaute 2 PAZ domain.
Nature
426
:
465
469
.

Loppin
B
,
Bonnefoy
E
,
Anselme
C
,
Laurencon
A
,
Karr
T L
et al. ,
2005
The histone H3.3 chaperone HIRA is essential for chromatin assembly in the male pronucleus.
Nature
437
:
1386
1390
.

Lu
C
,
Fuller
M T
,
2015
Recruitment of mediator complex by cell type and stage-specific factors required for tissue-specific TAF dependent gene activation in an adult stem cell lineage.
PLoS Genet.
11
:
e1005701
.

Lu
C
,
Kim
J
,
Fuller
M T
,
2013
The polyubiquitin gene Ubi-p63E is essential for male meiotic cell cycle progression and germ cell differentiation in Drosophila.
Development
140
:
3522
3531
.

Luo
G Z
,
Blanco
M A
,
Greer
E L
,
He
C
,
Shi
Y
,
2015
DNA N(6)-methyladenine: a new epigenetic mark in eukaryotes?
Nat. Rev. Mol. Cell Biol.
16
:
705
710
.

Lyko
F
,
Ramsahoye
B H
,
Jaenisch
R
,
2000
a
DNA methylation in Drosophila melanogaster.
Nature
408
:
538
540
.

Lyko
F
,
Whittaker
A J
,
Orr-Weaver
T L
,
Jaenisch
R
,
2000
b
The putative Drosophila methyltransferase gene dDnmt2 is contained in a transposon-like element and is expressed specifically in ovaries.
Mech. Dev.
95
:
215
217
.

Ma
X
,
Wang
S
,
Do
T
,
Song
X
,
Inaba
M
et al. ,
2014
Piwi is required in multiple cell types to control germline stem cell lineage development in the Drosophila ovary.
PLoS One
9
:
e90267
.

MacDougall
N
,
Clark
A
,
MacDougall
E
,
Davis
I
,
2003
Drosophila gurken (TGFalpha) mRNA localizes as particles that move within the oocyte in two dynein-dependent steps.
Dev. Cell
4
:
307
319
.

Mach
J M
,
Lehmann
R
,
1997
An Egalitarian-BicaudalD complex is essential for oocyte specification and axis determination in Drosophila.
Genes Dev.
11
:
423
435
.

Maines
J Z
,
Wasserman
S A
,
1999
Post-transcriptional regulation of the meiotic Cdc25 protein Twine by the Dazl orthologue Boule.
Nat. Cell Biol.
1
:
171
174
.

Maines
J Z
,
Park
J K
,
Williams
M
,
McKearin
D M
,
2007
Stonewalling Drosophila stem cell differentiation by epigenetic controls.
Development
134
:
1471
1479
.

Malone
C D
,
Brennecke
J
,
Dus
M
,
Stark
A
,
McCombie
W R
et al. ,
2009
Specialized piRNA pathways act in germline and somatic tissues of the Drosophila ovary.
Cell
137
:
522
535
.

Marchetto
M C
,
Narvaiza
I
,
Denli
A M
,
Benner
C
,
Lazzarini
T A
et al. ,
2013
Differential L1 regulation in pluripotent stem cells of humans and apes.
Nature
503
:
525
529
.

Mata
J
,
Curado
S
,
Ephrussi
A
,
Rorth
P
,
2000
Tribbles coordinates mitosis and morphogenesis in Drosophila by regulating string/CDC25 proteolysis.
Cell
101
:
511
522
.

Matangkasombut
O
,
Auty
R
,
Buratowski
S
,
2004
Structure and function of the TFIID complex.
Adv. Protein Chem.
67
:
67
92
.

McKearin
D M
,
Spradling
A C
,
1990
Bag-of-marbles: a Drosophila gene required to initiate both male and female gametogenesis.
Genes Dev.
4
:
2242
2251
.

Megosh
H B
,
Cox
D N
,
Campbell
C
,
Lin
H
,
2006
The role of PIWI and the miRNA machinery in Drosophila germline determination.
Curr. Biol.
16
:
1884
1894
.

Mejlumian
L
,
Pelisson
A
,
Bucheton
A
,
Terzian
C
,
2002
Comparative and functional studies of Drosophila species invasion by the gypsy endogenous retrovirus.
Genetics
160
:
201
209
.

Metcalf
C E
,
Wassarman
D A
,
2007
Nucleolar colocalization of TAF1 and testis-specific TAFs during Drosophila spermatogenesis.
Dev. Dyn.
236
:
2836
2843
.

Min
J
,
Zhang
Y
,
Xu
R M
,
2003
Structural basis for specific binding of Polycomb chromodomain to histone H3 methylated at Lys 27.
Genes Dev.
17
:
1823
1828
.

Mis
J
,
Ner
S S
,
Grigliatti
T A
,
2006
Identification of three histone methyltransferases in Drosophila: dG9a is a suppressor of PEV and is required for gene silencing.
Mol. Genet. Genomics
275
:
513
526
.

Mohn
F
,
Sienski
G
,
Handler
D
,
Brennecke
J
,
2014
The rhino-deadlock-cutoff complex licenses noncanonical transcription of dual-strand piRNA clusters in Drosophila.
Cell
157
:
1364
1379
.

Mohn
F
,
Handler
D
,
Brennecke
J
,
2015
Noncoding RNA. piRNA-guided slicing specifies transcripts for Zucchini-dependent, phased piRNA biogenesis.
Science
348
:
812
817
.

Mohrmann
L
,
Langenberg
K
,
Krijgsveld
J
,
Kal
A J
,
Heck
A J
et al. ,
2004
Differential targeting of two distinct SWI/SNF-related Drosophila chromatin-remodeling complexes.
Mol. Cell. Biol.
24
:
3077
3088
.

Molla-Herman
A
,
Valles
A M
,
Ganem-Elbaz
C
,
Antoniewski
C
,
Huynh
J R
,
2015
tRNA processing defects induce replication stress and Chk2-dependent disruption of piRNA transcription.
EMBO J.
34
:
3009
3027
.

Monk
A C
,
Siddall
N A
,
Volk
T
,
Fraser
B
,
Quinn
L M
et al. ,
2010
HOW is required for stem cell maintenance in the Drosophila testis and for the onset of transit-amplifying divisions.
Cell Stem Cell
6
:
348
360
.

Moon
S
,
Cho
B
,
Min
S H
,
Lee
D
,
Chung
Y D
,
2011
The THO complex is required for nucleolar integrity in Drosophila spermatocytes.
Development
138
:
3835
3845
.

Morillo Prado
J R
,
Chen
X
,
Fuller
M T
,
2012
Polycomb group genes Psc and Su(z)2 maintain somatic stem cell identity and activity in Drosophila.
PLoS One
7
:
e52892
.

Morillo Prado
J R
,
Srinivasan
S
,
Fuller
M T
,
2013
The histone variant His2Av is required for adult stem cell maintenance in the Drosophila testis.
PLoS Genet.
9
:
e1003903
.

Morris
L X
,
Spradling
A C
,
2011
Long-term live imaging provides new insight into stem cell regulation and germline-soma coordination in the Drosophila ovary.
Development
138
:
2207
2215
.

Moussian
B
,
Roth
S
,
2005
Dorsoventral axis formation in the Drosophila embryo–shaping and transducing a morphogen gradient.
Curr. Biol.
15
:
R887
R899
.

Muller
J
,
Hart
C M
,
Francis
N J
,
Vargas
M L
,
Sengupta
A
et al. ,
2002
Histone methyltransferase activity of a Drosophila Polycomb group repressor complex.
Cell
111
:
197
208
.

Murota
Y
,
Ishizu
H
,
Nakagawa
S
,
Iwasaki
Y W
,
Shibata
S
et al. ,
2014
Yb integrates piRNA intermediates and processing factors into perinuclear bodies to enhance piRISC assembly.
Cell Rep.
8
:
103
113
.

Narlikar
G J
,
Sundaramoorthy
R
,
Owen-Hughes
T
,
2013
Mechanisms and functions of ATP-dependent chromatin-remodeling enzymes.
Cell
154
:
490
503
.

Neuman-Silberberg
F S
,
Schupbach
T
,
1993
The Drosophila dorsoventral patterning gene gurken produces a dorsally localized RNA and encodes a TGF alpha-like protein.
Cell
75
:
165
174
.

Neuman-Silberberg
F S
,
Schupbach
T
,
1996
The Drosophila TGF-alpha-like protein gurken: expression and cellular localization during Drosophila oogenesis.
Mech. Dev.
59
:
105
113
.

Ng
K W
,
Anderson
C
,
Marshall
E A
,
Minatel
B C
,
Enfield
K S
et al. ,
2016
Piwi-interacting RNAs in cancer: emerging functions and clinical utility.
Mol. Cancer
15
:
5
.

Nilson
L A
,
Schupbach
T
,
1999
EGF receptor signaling in Drosophila oogenesis.
Curr. Top. Dev. Biol.
44
:
203
243
.

Nishida
K M
,
Okada
T N
,
Kawamura
T
,
Mituyama
T
,
Kawamura
Y
et al. ,
2009
Functional involvement of Tudor and dPRMT5 in the piRNA processing pathway in Drosophila germlines.
EMBO J.
28
:
3820
3831
.

Nishimasu
H
,
Ishizu
H
,
Saito
K
,
Fukuhara
S
,
Kamatani
M K
et al. ,
2012
Structure and function of Zucchini endoribonuclease in piRNA biogenesis.
Nature
491
:
284
287
.

Nystul
T
,
Spradling
A
,
2007
An epithelial niche in the Drosophila ovary undergoes long-range stem cell replacement.
Cell Stem Cell
1
:
277
285
.

Nystul
T
,
Spradling
A
,
2010
Regulation of epithelial stem cell replacement and follicle formation in the Drosophila ovary.
Genetics
184
:
503
515
.

Ohlstein
B
,
McKearin
D
,
1997
Ectopic expression of the Drosophila Bam protein eliminates oogenic germline stem cells.
Development
124
:
3651
3662
.

Ohtani
H
,
Iwasaki
Y W
,
Shibuya
A
,
Siomi
H
,
Siomi
M C
et al. ,
2013
DmGTSF1 is necessary for Piwi-piRISC-mediated transcriptional transposon silencing in the Drosophila ovary.
Genes Dev.
27
:
1656
1661
.

Olivieri
D
,
Sykora
M M
,
Sachidanandam
R
,
Mechtler
K
,
Brennecke
J
,
2010
An in vivo RNAi assay identifies major genetic and cellular requirements for primary piRNA biogenesis in Drosophila.
EMBO J.
29
:
3301
3317
.

Olivieri
D
,
Senti
K A
,
Subramanian
S
,
Sachidanandam
R
,
Brennecke
J
,
2012
The cochaperone shutdown defines a group of biogenesis factors essential for all piRNA populations in Drosophila.
Mol. Cell
47
:
954
969
.

Page
S L
,
Hawley
R S
,
2001
c(3)G encodes a Drosophila synaptonemal complex protein.
Genes Dev.
15
:
3130
3143
.

Pancratov
R
,
Peng
F
,
Smibert
P
,
Yang
S
Jr.
,
Olson
E R
et al. ,
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
.

Pane
A
,
Wehr
K
,
Schupbach
T
,
2007
Zucchini and squash encode two putative nucleases required for rasiRNA production in the Drosophila germline.
Dev. Cell
12
:
851
862
.

Pardue
M L
,
DeBaryshe
P G
,
2003
Retrotransposons provide an evolutionarily robust non-telomerase mechanism to maintain telomeres.
Annu. Rev. Genet.
37
:
485
511
.

Pardue
M L
,
DeBaryshe
P G
,
2011
Adapting to life at the end of the line: how Drosophila telomeric retrotransposons cope with their job.
Mob. Genet. Elements
1
:
128
134
.

Parfenov
V
,
Potchukalina
G
,
Dudina
L
,
Kostyuchek
D
,
Gruzova
M
,
1989
Human antral follicles: oocyte nucleus and the karyosphere formation (electron microscopic and autoradiographic data).
Gamete Res.
22
:
219
231
.

Park
J K
,
Liu
X
,
Strauss
T J
,
McKearin
D M
,
Liu
Q
,
2007
The miRNA pathway intrinsically controls self-renewal of Drosophila germline stem cells.
Curr. Biol.
17
:
533
538
.

Parrott
B B
,
Chiang
Y
,
Hudson
A
,
Sarkar
A
,
Guichet
A
et al. ,
2011
Nucleoporin98–96 function is required for transit amplification divisions in the germ line of Drosophila melanogaster.
PLoS One
6
:
e25087
.

Parrott
B B
,
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
.

Patil
V S
,
Kai
T
,
2010
Repression of retroelements in Drosophila germline via piRNA pathway by the Tudor domain protein Tejas.
Curr. Biol.
20
:
724
730
.

Patil
V S
,
Anand
A
,
Chakrabarti
A
,
Kai
T
,
2014
The Tudor domain protein Tapas, a homolog of the vertebrate Tdrd7, functions in the piRNA pathway to regulate retrotransposons in germline of Drosophila melanogaster.
BMC Biol.
12
:
61
.

Pek
J W
,
Lim
A K
,
Kai
T
,
2009
Drosophila maelstrom ensures proper germline stem cell lineage differentiation by repressing microRNA-7.
Dev. Cell
17
:
417
424
.

Peng
J C
,
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
.

Pengelly
A R
,
Copur
O
,
Jackle
H
,
Herzig
A
,
Muller
J
,
2013
A histone mutant reproduces the phenotype caused by loss of histone-modifying factor Polycomb.
Science
339
:
698
699
.

Perezgasga
L
,
Jiang
J
,
Bolival
B
Jr
,
Hiller
M
,
Benson
E
et al. ,
2004
Regulation of transcription of meiotic cell cycle and terminal differentiation genes by the testis-specific Zn-finger protein matotopetli.
Development
131
:
1691
1702
.

Perrini
B
,
Piacentini
L
,
Fanti
L
,
Altieri
F
,
Chichiarelli
S
et al. ,
2004
HP1 controls telomere capping, telomere elongation, and telomere silencing by two different mechanisms in Drosophila.
Mol. Cell
15
:
467
476
.

Petrella
L N
,
Smith-Leiker
T
,
Cooley
L
,
2007
The Ovhts polyprotein is cleaved to produce fusome and ring canal proteins required for Drosophila oogenesis.
Development
134
:
703
712
.

Phalke
S
,
Nickel
O
,
Walluscheck
D
,
Hortig
F
,
Onorati
M C
et al. ,
2009
Retrotransposon silencing and telomere integrity in somatic cells of Drosophila depends on the cytosine-5 methyltransferase DNMT2.
Nat. Genet.
41
:
696
702
.

Prud’homme
N
,
Gans
M
,
Masson
M
,
Terzian
C
,
Bucheton
A
,
1995
Flamenco, a gene controlling the gypsy retrovirus of Drosophila melanogaster.
Genetics
139
:
697
711
.

Qi
H
,
Watanabe
T
,
Ku
H Y
,
Liu
N
,
Zhong
M
et al. ,
2011
The Yb body, a major site for Piwi-associated RNA biogenesis and a gateway for Piwi expression and transport to the nucleus in somatic cells.
J. Biol. Chem.
286
:
3789
3797
.

Raddatz
G
,
Guzzardo
P M
,
Olova
N
,
Fantappie
M R
,
Rampp
M
et al. ,
2013
Dnmt2-dependent methylomes lack defined DNA methylation patterns.
Proc. Natl. Acad. Sci. USA
110
:
8627
8631
.

Rangan
P
,
Malone
C D
,
Navarro
C
,
Newbold
S P
,
Hayes
P S
et al. ,
2011
piRNA production requires heterochromatin formation in Drosophila.
Curr. Biol.
21
:
1373
1379
.

Rathke
C
,
Baarends
W M
,
Jayaramaiah-Raja
S
,
Bartkuhn
M
,
Renkawitz
R
et al. ,
2007
Transition from a nucleosome-based to a protamine-based chromatin configuration during spermiogenesis in Drosophila.
J. Cell Sci.
120
:
1689
1700
.

Raychaudhuri
N
,
Dubruille
R
,
Orsi
G A
,
Bagheri
H C
,
Loppin
B
et al. ,
2012
Transgenerational propagation and quantitative maintenance of paternal centromeres depends on Cid/Cenp-A presence in Drosophila sperm.
PLoS Biol.
10
:
e1001434
.

Riddiford
L M
,
Cherbas
P
,
Truman
J W
,
2000
Ecdysone receptors and their biological actions.
Vitam. Horm.
60
:
1
73
.

Ringrose
L
,
Paro
R
,
2004
Epigenetic regulation of cellular memory by the Polycomb and Trithorax group proteins.
Annu. Rev. Genet.
38
:
413
443
.

Rizzo
F
,
Hashim
A
,
Marchese
G
,
Ravo
M
,
Tarallo
R
et al. ,
2014
Timed regulation of P-element-induced wimpy testis-interacting RNA expression during rat liver regeneration.
Hepatology
60
:
798
806
.

Rorth
P
,
1998
Gal4 in the Drosophila female germline.
Mech. Dev.
78
:
113
118
.

Roth
S
,
Lynch
J A
,
2009
Symmetry breaking during Drosophila oogenesis.
Cold Spring Harb. Perspect. Biol.
1
:
a001891
.

Rouget
C
,
Papin
C
,
Boureux
A
,
Meunier
A C
,
Franco
B
et al. ,
2010
Maternal mRNA deadenylation and decay by the piRNA pathway in the early Drosophila embryo.
Nature
467
:
1128
1132
.

Rozhkov
N V
,
Hammell
M
,
Hannon
G J
,
2013
Multiple roles for Piwi in silencing Drosophila transposons.
Genes Dev.
27
:
400
412
.

Saito
K
,
Nishida
K M
,
Mori
T
,
Kawamura
Y
,
Miyoshi
K
et al. ,
2006
Specific association of Piwi with rasiRNAs derived from retrotransposon and heterochromatic regions in the Drosophila genome.
Genes Dev.
20
:
2214
2222
.

Saito
K
,
Ishizu
H
,
Komai
M
,
Kotani
H
,
Kawamura
Y
et al. ,
2010
Roles for the Yb body components Armitage and Yb in primary piRNA biogenesis in Drosophila.
Genes Dev.
24
:
2493
2498
.

Sanson
B
,
White
P
,
Vincent
J P
,
1996
Uncoupling cadherin-based adhesion from wingless signalling in Drosophila.
Nature
383
:
627
630
.

Santel
A
,
Blumer
N
,
Kampfer
M
,
Renkawitz-Pohl
R
,
1998
Flagellar mitochondrial association of the male-specific Don Juan protein in Drosophila spermatozoa.
J. Cell Sci.
111
:
3299
3309
.

Santos
A C
,
Lehmann
R
,
2004
Germ cell specification and migration in Drosophila and beyond.
Curr. Biol.
14
:
R578
R589
.

Sarkar
A
,
Parikh
N
,
Hearn
S A
,
Fuller
M T
,
Tazuke
S I
et al. ,
2007
Antagonistic roles of Rac and Rho in organizing the germ cell microenvironment.
Curr. Biol.
17
:
1253
1258
.

Sarmento
O F
,
Digilio
L C
,
Wang
Y
,
Perlin
J
,
Herr
J C
et al. ,
2004
Dynamic alterations of specific histone modifications during early murine development.
J. Cell Sci.
117
:
4449
4459
.

Sarot
E
,
Payen-Groschene
G
,
Bucheton
A
,
Pelisson
A
,
2004
Evidence for a piwi-dependent RNA silencing of the gypsy endogenous retrovirus by the Drosophila melanogaster flamenco gene.
Genetics
166
:
1313
1321
.

Sato
K
,
Nishida
K M
,
Shibuya
A
,
Siomi
M C
,
Siomi
H
,
2011
Maelstrom coordinates microtubule organization during Drosophila oogenesis through interaction with components of the MTOC.
Genes Dev.
25
:
2361
2373
.

Sato
K
,
Iwasaki
Y W
,
Shibuya
A
,
Carninci
P
,
Tsuchizawa
Y
et al. ,
2015
Krimper enforces an antisense bias on piRNA pools by binding AGO3 in the Drosophila germline.
Mol. Cell
59
:
553
563
.

Schaefer
M
,
Pollex
T
,
Hanna
K
,
Tuorto
F
,
Meusburger
M
et al. ,
2010
RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage.
Genes Dev.
24
:
1590
1595
.

Schafer
M
,
Borsch
D
,
Hulster
A
,
Schafer
U
,
1993
Expression of a gene duplication encoding conserved sperm tail proteins is translationally regulated in Drosophila melanogaster.
Mol. Cell. Biol.
13
:
1708
1718
.

Schafer
M
,
Nayernia
K
,
Engel
W
,
Schafer
U
,
1995
Translational control in spermatogenesis.
Dev. Biol.
172
:
344
352
.

Scheuermann
J C
,
de Ayala Alonso
A G
,
Oktaba
K
,
Ly-Hartig
N
,
McGinty
R K
et al. ,
2010
Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB.
Nature
465
:
243
247
.

Schulz
C
,
Wood
C G
,
Jones
D L
,
Tazuke
S I
,
Fuller
M T
,
2002
Signaling from germ cells mediated by the rhomboid homolog stet organizes encapsulation by somatic support cells.
Development
129
:
4523
4534
.

Schulz
C
,
Kiger
A A
,
Tazuke
S I
,
Yamashita
Y M
,
Pantalena-Filho
L C
et al. ,
2004
A misexpression screen reveals effects of bag-of-marbles and TGF beta class signaling on the Drosophila male germ-line stem cell lineage.
Genetics
167
:
707
723
.

Schupbach
T
,
1987
Germ line and soma cooperate during oogenesis to establish the dorsoventral pattern of egg shell and embryo in Drosophila melanogaster.
Cell
49
:
699
707
.

Schwartz
Y B
,
Pirrotta
V
,
2007
Polycomb silencing mechanisms and the management of genomic programmes.
Nat. Rev. Genet.
8
:
9
22
.

Scott
E K
,
Lee
T
,
Luo
L
,
2001
Enok encodes a Drosophila putative histone acetyltransferase required for mushroom body neuroblast proliferation.
Curr. Biol.
11
:
99
104
.

Sedkov
Y
,
Cho
E
,
Petruk
S
,
Cherbas
L
,
Smith
S T
et al. ,
2003
Methylation at lysine 4 of histone H3 in ecdysone-dependent development of Drosophila.
Nature
426
:
78
83
.

Seligson
D B
,
Horvath
S
,
Shi
T
,
Yu
H
,
Tze
S
et al. ,
2005
Global histone modification patterns predict risk of prostate cancer recurrence.
Nature
435
:
1262
1266
.

Senti
K A
,
Jurczak
D
,
Sachidanandam
R
,
Brennecke
J
,
2015
piRNA-guided slicing of transposon transcripts enforces their transcriptional silencing via specifying the nuclear piRNA repertoire.
Genes Dev.
29
:
1747
1762
.

Shalaby
N A
,
Buszczak
M
,
2017
Live-cell imaging of the adult Drosophila ovary using confocal microscopy.
Methods Mol. Biol.
1463
:
85
91
.

Shan
L
,
Wu
C
,
Chen
D
,
Hou
L
,
Li
X
et al. ,
2017
Regulators of alternative polyadenylation operate at the transition from mitosis to meiosis.
J. Genet. Genomics
44
:
95
106
.

Sheng
X R
,
Brawley
C M
,
Matunis
E L
,
2009
Dedifferentiating spermatogonia outcompete somatic stem cells for niche occupancy in the Drosophila testis.
Cell Stem Cell
5
:
191
203
.

Shi
S
,
Calhoun
H C
,
Xia
F
,
Li
J
,
Le
L
et al. ,
2006
JAK signaling globally counteracts heterochromatic gene silencing.
Nat. Genet.
38
:
1071
1076
.

Shivdasani
A A
,
Ingham
P W
,
2003
Regulation of stem cell maintenance and transit amplifying cell proliferation by tgf-beta signaling in Drosophila spermatogenesis.
Curr. Biol.
13
:
2065
2072
.

Siddall
N A
,
McLaughlin
E A
,
Marriner
N L
,
Hime
G R
,
2006
The RNA-binding protein Musashi is required intrinsically to maintain stem cell identity.
Proc. Natl. Acad. Sci. USA
103
:
8402
8407
.

Sienski
G
,
Donertas
D
,
Brennecke
J
,
2012
Transcriptional silencing of transposons by Piwi and maelstrom and its impact on chromatin state and gene expression.
Cell
151
:
964
980
.

Sienski
G
,
Batki
J
,
Senti
K A
,
Donertas
D
,
Tirian
L
et al. ,
2015
Silencio/CG9754 connects the Piwi-piRNA complex to the cellular heterochromatin machinery.
Genes Dev.
29
:
2258
2271
.

Smothers
J F
,
Henikoff
S
,
2001
The hinge and chromo shadow domain impart distinct targeting of HP1-like proteins.
Mol. Cell. Biol.
21
:
2555
2569
.

Song
J J
,
Smith
S K
,
Hannon
G J
,
Joshua-Tor
L
,
2004
Crystal structure of Argonaute and its implications for RISC slicer activity.
Science
305
:
1434
1437
.

Song
X
,
Wong
M D
,
Kawase
E
,
Xi
R
,
Ding
B C
et al. ,
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
.

Spradling
A C
,
1993
Developmental genetics of oogenesis
, pp.
1
70
in
The development of Drosophila melanogaster
,
Vol. I
, edited by
Bate
M
,
Martinez Arias
A
.
Cold Spring Harbor Press
,
Cold Spring Harbor, NY
.

Spradling
A C
,
2011
The living-tissue microscope: the importance of studying stem cells in their natural, undisturbed microenvironment.
J. Pathol.
225
:
161
162
.

St Johnston
D
,
2005
Moving messages: the intracellular localization of mRNAs.
Nat. Rev. Mol. Cell Biol.
6
:
363
375
.

Stabell
M
,
Bjorkmo
M
,
Aalen
R B
,
Lambertsson
A
,
2006
The Drosophila SET domain encoding gene dEset is essential for proper development.
Hereditas
143
:
177
188
.

Starz-Gaiano
M
,
Lehmann
R
,
2001
Moving towards the next generation.
Mech. Dev.
105
:
5
18
.

Stock
J K
,
Giadrossi
S
,
Casanova
M
,
Brookes
E
,
Vidal
M
et al. ,
2007
Ring1-mediated ubiquitination of H2A restrains poised RNA polymerase II at bivalent genes in mouse ES cells.
Nat. Cell Biol.
9
:
1428
1435
.

Sumiyoshi
T
,
Sato
K
,
Yamamoto
H
,
Iwasaki
Y W
,
Siomi
H
et al. ,
2016
Loss of l(3)mbt leads to acquisition of the ping-pong cycle in Drosophila ovarian somatic cells.
Genes Dev.
30
:
1617
1622
.

Surface
L E
,
Thornton
S R
,
Boyer
L A
,
2010
Polycomb group proteins set the stage for early lineage commitment.
Cell Stem Cell
7
:
288
298
.

Suter
B
,
Steward
R
,
1991
Requirement for phosphorylation and localization of the Bicaudal-D protein in Drosophila oocyte differentiation.
Cell
67
:
917
926
.

Suzuki
M M
,
Bird
A
,
2008
DNA methylation landscapes: provocative insights from epigenomics.
Nat. Rev. Genet.
9
:
465
476
.

Swaminathan
J
,
Baxter
E M
,
Corces
V G
,
2005
The role of histone H2Av variant replacement and histone H4 acetylation in the establishment of Drosophila heterochromatin.
Genes Dev.
19
:
65
76
.

Szakmary
A
,
Reedy
M
,
Qi
H
,
Lin
H
,
2009
The Yb protein defines a novel organelle and regulates male germline stem cell self-renewal in Drosophila melanogaster.
J. Cell Biol.
185
:
613
627
.

Takayama
S
,
Dhahbi
J
,
Roberts
A
,
Mao
G
,
Heo
S J
et al. ,
2014
Genome methylation in D. melanogaster is found at specific short motifs and is independent of DNMT2 activity.
Genome Res.
24
:
821
830
.

Talbert
P B
,
Henikoff
S
,
2010
Histone variants–ancient wrap artists of the epigenome.
Nat. Rev. Mol. Cell Biol.
11
:
264
275
.

Tang
Y
,
Geng
Q
,
Chen
D
,
Zhao
S
,
Liu
X
et al. ,
2017
Germline proliferation is regulated by somatic endocytic genes via JNK and BMP signaling in Drosophila.
Genetics
206
:
189
197
.

Tanimoto
H
,
Itoh
S
,
ten Dijke
P
,
Tabata
T
,
2000
Hedgehog creates a gradient of DPP activity in Drosophila wing imaginal discs.
Mol. Cell
5
:
59
71
.

Tarayrah
L
,
Herz
H M
,
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
.

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
.

Tastan
O Y
,
Maines
J Z
,
Li
Y
,
McKearin
D M
,
Buszczak
M
,
2010
Drosophila ataxin 2-binding protein 1 marks an intermediate step in the molecular differentiation of female germline cysts.
Development
137
:
3167
3176
.

Tazuke
S I
,
Schulz
C
,
Gilboa
L
,
Fogarty
M
,
Mahowald
A P
et al. ,
2002
A germline-specific gap junction protein required for survival of differentiating early germ cells.
Development
129
:
2529
2539
.

Terry
N A
,
Tulina
N
,
Matunis
E
,
DiNardo
S
,
2006
Novel regulators revealed by profiling Drosophila testis stem cells within their niche.
Dev. Biol.
294
:
246
257
.

Theurkauf
W E
,
Smiley
S
,
Wong
M L
,
Alberts
B M
,
1992
Reorganization of the cytoskeleton during Drosophila oogenesis: implications for axis specification and intercellular transport.
Development
115
:
923
936
.

Theurkauf
W E
,
Alberts
B M
,
Jan
Y N
,
Jongens
T A
,
1993
A central role for microtubules in the differentiation of Drosophila oocytes.
Development
118
:
1169
1180
.

Thomsen
S
,
Anders
S
,
Janga
S C
,
Huber
W
,
Alonso
C R
,
2010
Genome-wide analysis of mRNA decay patterns during early Drosophila development.
Genome Biol.
11
:
R93
.

Toledano
H
,
D’Alterio
C
,
Czech
B
,
Levine
E
,
Jones
D L
,
2012
The let-7-Imp axis regulates ageing of the Drosophila testis stem-cell niche.
Nature
485
:
605
610
.

Tora
L
,
2002
A unified nomenclature for TATA box binding protein (TBP)-associated factors (TAFs) involved in RNA polymerase II transcription.
Genes Dev.
16
:
673
675
.

Tran
J
,
Brenner
T J
,
DiNardo
S
,
2000
Somatic control over the germline stem cell lineage during Drosophila spermatogenesis.
Nature
407
:
754
757
.

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
.

Tschiersch
B
,
Hofmann
A
,
Krauss
V
,
Dorn
R
,
Korge
G
et al. ,
1994
The protein encoded by the Drosophila position-effect variegation suppressor gene Su(var)3–9 combines domains of antagonistic regulators of homeotic gene complexes.
EMBO J.
13
:
3822
3831
.

Tulina
N
,
Matunis
E
,
2001
Control of stem cell self-renewal in Drosophila spermatogenesis by JAK-STAT signaling.
Science
294
:
2546
2549
.

Unhavaithaya
Y
,
Hao
Y
,
Beyret
E
,
Yin
H
,
Kuramochi-Miyagawa
S
et al. ,
2009
MILI, a PIWI-interacting RNA-binding protein, is required for germ line stem cell self-renewal and appears to positively regulate translation.
J. Biol. Chem.
284
:
6507
6519
.

Vaccari
T
,
Ephrussi
A
,
2002
The fusome and microtubules enrich Par-1 in the oocyte, where it effects polarization in conjunction with Par-3, BicD, Egl, and dynein.
Curr. Biol.
12
:
1524
1528
.

Vagin
V V
,
Sigova
A
,
Li
C
,
Seitz
H
,
Gvozdev
V
et al. ,
2006
A distinct small RNA pathway silences selfish genetic elements in the germline.
Science
313
:
320
324
.

Van Buskirk
C
,
Schupbach
T
,
1999
Versatility in signalling: multiple responses to EGF receptor activation during Drosophila oogenesis.
Trends Cell Biol.
9
:
1
4
.

Van De Bor
V
,
Hartswood
E
,
Jones
C
,
Finnegan
D
,
Davis
I
,
2005
Gurken and the I factor retrotransposon RNAs share common localization signals and machinery.
Dev. Cell
9
:
51
62
.

Van Doren
M
,
Williamson
A L
,
Lehmann
R
,
1998
Regulation of zygotic gene expression in Drosophila primordial germ cells.
Curr. Biol.
8
:
243
246
.

Vasudevan
S
,
Tong
Y
,
Steitz
J A
,
2007
Switching from repression to activation: microRNAs can up-regulate translation.
Science
318
:
1931
1934
.

Venkatesh
S
,
Workman
J L
,
2015
Histone exchange, chromatin structure and the regulation of transcription.
Nat. Rev. Mol. Cell Biol.
16
:
178
189
.

Vermaak
D
,
Malik
H S
,
2009
Multiple roles for heterochromatin protein 1 genes in Drosophila.
Annu. Rev. Genet.
43
:
467
492
.

Volpe
A M
,
Horowitz
H
,
Grafer
C M
,
Jackson
S M
,
Berg
C A
,
2001
Drosophila rhino encodes a female-specific chromo-domain protein that affects chromosome structure and egg polarity.
Genetics
159
:
1117
1134
.

Vourekas
A
,
Alexiou
P
,
Vrettos
N
,
Maragkakis
M
,
Mourelatos
Z
,
2016
Sequence-dependent but not sequence-specific piRNA adhesion traps mRNAs to the germ plasm.
Nature
531
:
390
394
.

Wallenfang
M R
,
Nayak
R
,
DiNardo
S
,
2006
Dynamics of the male germline stem cell population during aging of Drosophila melanogaster.
Aging Cell
5
:
297
304
.

Wang
H
,
Wang
L
,
Erdjument-Bromage
H
,
Vidal
M
,
Tempst
P
et al. ,
2004
Role of histone H2A ubiquitination in Polycomb silencing.
Nature
431
:
873
878
.

Wang
S H
,
Elgin
S C
,
2011
Drosophila Piwi functions downstream of piRNA production mediating a chromatin-based transposon silencing mechanism in female germ line.
Proc. Natl. Acad. Sci. USA
108
:
21164
21169
.

Wang
W
,
Yoshikawa
M
,
Han
B W
,
Izumi
N
,
Tomari
Y
et al. ,
2014
The initial uridine of primary piRNAs does not create the tenth adenine that Is the hallmark of secondary piRNAs.
Mol. Cell
56
:
708
716
.

Wang
W
,
Han
B W
,
Tipping
C
,
Ge
D T
,
Zhang
Z
et al. ,
2015
Slicing and binding by Ago3 or Aub trigger Piwi-bound piRNA production by distinct mechanisms.
Mol. Cell
59
:
819
830
.

Wang
X
,
Pan
L
,
Wang
S
,
Zhou
J
,
McDowell
W
et al. ,
2011
Histone H3K9 trimethylase eggless controls germline stem cell maintenance and differentiation.
PLoS Genet.
7
:
e1002426
.

Wang
Y
,
Wang
L
,
Wang
Z
,
2008
Transgenic analyses of TGIF family proteins in Drosophila imply their role in cell growth.
J. Genet. Genomics
35
:
457
465
.

Wang
Z
,
Mann
R S
,
2003
Requirement for two nearly identical TGIF-related homeobox genes in Drosophila spermatogenesis.
Development
130
:
2853
2865
.

Ward
E J
,
Thaipisuttikul
I
,
Terayama
M
,
French
R L
,
Jackson
S M
et al. ,
2002
GAL4 enhancer trap patterns during Drosophila development.
Genesis
34
:
46
50
.

Webster
A
,
Li
S
,
Hur
J K
,
Wachsmuth
M
,
Bois
J S
et al. ,
2015
Aub and Ago3 are recruited to nuage through two mechanisms to form a ping-pong complex assembled by Krimper.
Mol. Cell
59
:
564
575
.

White-Cooper
H
,
2009
Molecular mechanisms of gene regulation during Drosophila spermatogenesis.
Reproduction
139
:
11
21
.

White-Cooper
H
,
Schafer
M A
,
Alphey
L S
,
Fuller
M T
,
1998
Transcriptional and post-transcriptional control mechanisms coordinate the onset of spermatid differentiation with meiosis I in Drosophila.
Development
125
:
125
134
.

White-Cooper
H
,
Leroy
D
,
MacQueen
A
,
Fuller
M T
,
2000
Transcription of meiotic cell cycle and terminal differentiation genes depends on a conserved chromatin associated protein, whose nuclear localisation is regulated.
Development
127
:
5463
5473
.

Wion
D
,
Casadesus
J
,
2006
N6-methyl-adenine: an epigenetic signal for DNA-protein interactions.
Nat. Rev. Microbiol.
4
:
183
192
.

Wolfstetter
G
,
Holz
A
,
2012
The role of LamininB2 (LanB2) during mesoderm differentiation in Drosophila.
Cell. Mol. Life Sci.
69
:
267
282
.

Wong
L H
,
Choo
K H
,
2004
Evolutionary dynamics of transposable elements at the centromere.
Trends Genet.
20
:
611
616
.

Wu
H
,
Zhang
Y
,
2014
Reversing DNA methylation: mechanisms, genomics, and biological functions.
Cell
156
:
45
68
.

Xi
R
,
Xie
T
,
2005
Stem cell self-renewal controlled by chromatin remodeling factors.
Science
310
:
1487
1489
.

Xia
L
,
Jia
S
,
Huang
S
,
Wang
H
,
Zhu
Y
et al. ,
2010
The fused/Smurf complex controls the fate of Drosophila germline stem cells by generating a gradient BMP response.
Cell
143
:
978
990
.

Xia
L
,
Zheng
X
,
Zheng
W
,
Zhang
G
,
Wang
H
et al. ,
2012
The niche-dependent feedback loop generates a BMP activity gradient to determine the germline stem cell fate.
Curr. Biol.
22
:
515
521
.

Xie
J
,
Wooten
M
,
Tran
V
,
Chen
B C
,
Pozmanter
C
et al. ,
2015
Histone H3 threonine phosphorylation regulates asymmetric histone inheritance in the Drosophila male germline.
Cell
163
:
920
933
.

Xin
T
,
Xuan
T
,
Tan
J
,
Li
M
,
Zhao
G
et al. ,
2013
The Drosophila putative histone acetyltransferase enok maintains female germline stem cells through regulating Bruno and the niche.
Dev. Biol.
384
:
1
12
.

Xing
Y
,
Shi
S
,
Le
L
,
Lee
C A
,
Silver-Morse
L
et al. ,
2007
Evidence for transgenerational transmission of epigenetic tumor susceptibility in Drosophila.
PLoS Genet.
3
:
1598
1606
.

Xu
T
,
Rubin
G M
,
1993
Analysis of genetic mosaics in developing and adult Drosophila tissues.
Development
117
:
1223
1237
.

Xuan
T
,
Xin
T
,
He
J
,
Tan
J
,
Gao
Y
et al. ,
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
.

Yadlapalli
S
,
Yamashita
Y M
,
2013
Chromosome-specific nonrandom sister chromatid segregation during stem-cell division.
Nature
498
:
251
254
.

Yadlapalli
S
,
Cheng
J
,
Yamashita
Y M
,
2011
Drosophila male germline stem cells do not asymmetrically segregate chromosome strands.
J. Cell Sci.
124
:
933
939
.

Yan
K S
,
Yan
S
,
Farooq
A
,
Han
A
,
Zeng
L
et al. ,
2003
Structure and conserved RNA binding of the PAZ domain.
Nature
426
:
468
474
.

Yang
L
,
Chen
D
,
Duan
R
,
Xia
L
,
Wang
J
et al. ,
2007
Argonaute 1 regulates the fate of germline stem cells in Drosophila.
Development
134
:
4265
4272
.

Yang
S Y
,
Baxter
E M
,
Van Doren
M
,
2012
Phf7 controls male sex determination in the Drosophila germline.
Dev. Cell
22
:
1041
1051
.

Yang
Y
,
Xu
S
,
Xia
L
,
Wang
J
,
Wen
S
et al. ,
2009
The bantam microRNA is associated with drosophila fragile X mental retardation protein and regulates the fate of germline stem cells.
PLoS Genet.
5
:
e1000444
.

Yi
X
,
de Vries
H I
,
Siudeja
K
,
Rana
A
,
Lemstra
W
et al. ,
2009
Stwl modifies chromatin compaction and is required to maintain DNA integrity in the presence of perturbed DNA replication.
Mol. Biol. Cell
20
:
983
994
.

Yin
H
,
Lin
H
,
2007
An epigenetic activation role of Piwi and a Piwi-associated piRNA in Drosophila melanogaster.
Nature
450
:
304
308
.

Yoon
J
,
Lee
K S
,
Park
J S
,
Yu
K
,
Paik
S G
et al. ,
2008
dSETDB1 and SU(VAR)3–9 sequentially function during germline-stem cell differentiation in Drosophila melanogaster.
PLoS One
3
:
e2234
.

Youngson
N A
,
Whitelaw
E
,
2008
Transgenerational epigenetic effects.
Annu. Rev. Genomics Hum. Genet.
9
:
233
257
.

Yu
B
,
Cassani
M
,
Wang
M
,
Liu
M
,
Ma
J
et al. ,
2015
Structural insights into Rhino-mediated germline piRNA cluster formation.
Cell Res.
25
:
525
528
.

Yu
Y
,
Gu
J
,
Jin
Y
,
Luo
Y
,
Preall
J B
et al. ,
2015
Panoramix enforces piRNA-dependent cotranscriptional silencing.
Science
350
:
339
342
.

Yuan
H
,
Chiang
C Y
,
Cheng
J
,
Salzmann
V
,
Yamashita
Y M
,
2012
Regulation of cyclin A localization downstream of Par-1 function is critical for the centrosome orientation checkpoint in Drosophila male germline stem cells.
Dev. Biol.
361
:
57
67
.

Zaessinger
S
,
Busseau
I
,
Simonelig
M
,
2006
Oskar allows nanos mRNA translation in Drosophila embryos by preventing its deadenylation by Smaug/CCR4.
Development
133
:
4573
4583
.

Zeidler
M P
,
Perrimon
N
,
Strutt
D I
,
1999
Polarity determination in the Drosophila eye: a novel role for unpaired and JAK/STAT signaling.
Genes Dev.
13
:
1342
1353
.

Zemach
A
,
McDaniel
I E
,
Silva
P
,
Zilberman
D
,
2010
Genome-wide evolutionary analysis of eukaryotic DNA methylation.
Science
328
:
916
919
.

Zhang
F
,
Wang
J
,
Xu
J
,
Zhang
Z
,
Koppetsch
B S
et al. ,
2012
UAP56 couples piRNA clusters to the perinuclear transposon silencing machinery.
Cell
151
:
871
884
.

Zhang
G
,
Huang
H
,
Liu
D
,
Cheng
Y
,
Liu
X
et al. ,
2015
N6-methyladenine DNA modification in Drosophila.
Cell
161
:
893
906
.

Zhang
Q
,
Shalaby
N A
,
Buszczak
M
,
2014
Changes in rRNA transcription influence proliferation and cell fate within a stem cell lineage.
Science
343
:
298
301
.

Zhang
Z
,
Xu
J
,
Koppetsch
B S
,
Wang
J
,
Tipping
C
et al. ,
2011
Heterotypic piRNA Ping-Pong requires qin, a protein with both E3 ligase and tudor domains.
Mol. Cell
44
:
572
584
.

Zhang
Z
,
Wang
J
,
Schultz
N
,
Zhang
F
,
Parhad
S S
et al. ,
2014
The HP1 homolog rhino anchors a nuclear complex that suppresses piRNA precursor splicing.
Cell
157
:
1353
1363
.

Zhao
S
,
Chen
D
,
Geng
Q
,
Wang
Z
,
2013
The highly conserved LAMMER/CLK2 protein kinases prevent germ cell overproliferation in Drosophila.
Dev. Biol.
376
:
163
170
.

Zhu
C H
,
Xie
T
,
2003
Clonal expansion of ovarian germline stem cells during niche formation in Drosophila.
Development
130
:
2579
2588
.

Zimyanin
V L
,
Belaya
K
,
Pecreaux
J
,
Gilchrist
M J
,
Clark
A
et al. ,
2008
In vivo imaging of oskar mRNA transport reveals the mechanism of posterior localization.
Cell
134
:
843
853
.

Zink
D
,
Paro
R
,
1995
Drosophila Polycomb-group regulated chromatin inhibits the accessibility of a trans-activator to its target DNA.
EMBO J.
14
:
5660
5671
.

Zraly
C B
,
Marenda
D R
,
Nanchal
R
,
Cavalli
G
,
Muchardt
C
et al. ,
2003
SNR1 is an essential subunit in a subset of Drosophila brm complexes, targeting specific functions during development.
Dev. Biol.
253
:
291
308
.

Author notes

1

These authors contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)