small rnas in spermatogenesis

35
Accepted Manuscript Small RNAs in spermatogenesis Ram Prakash Yadav, Noora Kotaja PII: S0303-7207(13)00160-3 DOI: http://dx.doi.org/10.1016/j.mce.2013.04.015 Reference: MCE 8498 To appear in: Molecular and Cellular Endocrinology Molecular and Cellular Endocrinology Please cite this article as: Yadav, R.P., Kotaja, N., Small RNAs in spermatogenesis, Molecular and Cellular Endocrinology Molecular and Cellular Endocrinology (2013), doi: http://dx.doi.org/10.1016/j.mce.2013.04.015 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Upload: noora

Post on 10-Dec-2016

215 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: Small RNAs in spermatogenesis

Accepted Manuscript

Small RNAs in spermatogenesis

Ram Prakash Yadav, Noora Kotaja

PII: S0303-7207(13)00160-3

DOI: http://dx.doi.org/10.1016/j.mce.2013.04.015

Reference: MCE 8498

To appear in: Molecular and Cellular Endocrinology Molecular

and Cellular Endocrinology

Please cite this article as: Yadav, R.P., Kotaja, N., Small RNAs in spermatogenesis, Molecular and Cellular

Endocrinology Molecular and Cellular Endocrinology (2013), doi: http://dx.doi.org/10.1016/j.mce.2013.04.015

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers

we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and

review of the resulting proof before it is published in its final form. Please note that during the production process

errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Page 2: Small RNAs in spermatogenesis

Small RNAs in spermatogenesis

Ram Prakash Yadava and Noora Kotajaa,*

a Department of Physiology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10,

FIN-20520 Turku, Finland, [email protected], [email protected]

*Corresponding author: Noora Kotaja

Department of Physiology

Institute of Biomedicine

University of Turku

Kiinamyllynkatu 10

FIN-20520 Turku, Finland

Tel: +358-2-3337283

E-mail: [email protected]

Page 3: Small RNAs in spermatogenesis

 

Abstract

Spermatogenesis is characterized by meiotic divisions and major morphological changes to

produce spermatozoa that are capable of independent movement and fertilization of an egg.

Male germ cell differentiation is governed by orchestrated, phase-specific gene expression

patterns that are tightly controlled at transcriptional and post-transcriptional level. Post-

transcriptional regulation of protein-coding mRNAs becomes prominent during the late steps

of spermatogenesis when the compacting sperm nucleus becomes transcriptionally inhibited.

Small non-coding RNAs are important regulators of gene expression that mainly function

post-transcriptionally to control the properties of their target mRNAs. Male germ cells express

several classes of small RNAs, including Dicer-dependent microRNAs (miRNAs) and

endogenous small interfering RNAs (endo-siRNAs), as well as Dicer-independent piwi-

interacting RNAs (piRNAs). Increasing evidence supports the essential role of small RNA-

mediated RNA regulation in normal spermatogenesis and male fertility.

Keywords Spermatogenesis; Germ cell; Fertility; Post-transcriptional; Non-coding RNA; Small RNA

Abbreviations PGC, primordial germ cell; CB, chromatoid body; miRNA, microRNA; siRNA, small interfering

RNA; endo-siRNA, endogenous small interfering RNAs; piRNA, PIWI-interacting RNA; RISC,

RNA-induced silencing complex; MSCI, meiotic sex chromosome inactivation; SSC,

spermatogonial stem cell; RNP, ribonucleoprotein; AGO, Argonaute protein; PND, post-natal

day; TE, transposable element;

Page 4: Small RNAs in spermatogenesis

 

1. Introduction to spermatogenesis Development of male gamete is a complex differentiation program that produces highly

specialized spermatozoa (Hess and Renato de Franca, 2008). Germ cells are unique in their

capability to generate new organisms, and therefore molecular events during

spermatogenesis have to be strictly regulated to enable correct transmission of genetic and

epigenetic information to subsequent generations. Spermatogenesis is an efficient process

and average healthy human male may produce as much as thousand spermatozoa each

second. Spermatogenesis is also prone to mistakes as demonstrated by the low percentage

of morphologically normal spermatozoa in an average human semen sample. Moreover,

alarming adverse trends in the semen quality in Western countries have been reported

(Sharpe, 2012). At the same time, incidence of testicular germ cell tumors and congenital

malformations of the reproductive organs is increasing, which highlights the importance of

the characterization of the environmental and genetic factors negatively affecting male

reproductive health.

Male germ cells are specified shortly after implantation from the pluripotent cells of epiblast.

During gastrulation, the nascent primordial germ cells (PGCs) start to migrate toward the

future gonads and rapidly proliferate (Matsui, 2010). After migrating to the gonadal ridge,

PGCs become gonocytes within cords that are formed by Sertoli precursors and surrounded

by peritubular cells. Gonocytes show a burst of mitotic activity, then arrest in the G0 phase of

cell cycle remaining mitotically quiescent until after birth, when they give rise to

spermatogonia (de Rooij and Russell, 2000). Embryonic germ cells undergo a genome wide

epigenetic reprogramming, which is initiated by near to complete erasure of somatic

epigenetic marks. Subsequently, novel sex-specific DNA methylation patterns, including

differential imprinting of the genes in the male and female germ cells, are established (Sasaki

and Matsui, 2008; Ewen and Koopman, 2010).

Spermatogenesis starts shortly after birth by mitotic proliferation of spermatogonia (Fig. 1).

After proliferation phase, spermatogonia enter the meiotic phase and become

spermatocytes. After the long-lasting meiotic prophase I that includes homologous

chromosome pairing, synapse and recombination, cells undergo a reduction division during

which the sister chromosomes are separated into two cells to generate secondary

spermatocytes. These cells very quickly divide again to give raise to haploid round

spermatids. All the dramatic morphological changes such as acrosome and flagellum

formation, nuclear shaping and chromatin condensation take place during the post-meiotic

spermatid differentiation (spermiogenesis). Chromatin is tightly compacted with the help of

Page 5: Small RNAs in spermatogenesis

 

sperm-specific protamines that replace the most of histones and enable more efficient

packing of DNA. As a result of spermiogenesis, the spermatozoa are equipped with all the

structures required for its prospective functions. Spermatozoa are released into a lumen of

seminiferous tubules, where they continue the journey to the epididymis. Transit of

spermatozoa through the long convoluted epididymal duct is important for their final

maturation and acquirement of the capacity for full motility.

Germ line stem cells located in the basal compartment of seminiferous epithelium provide a

source of undifferentiated cells that enable the production of spermatozoa throughout the

whole period of sexual maturity (de Rooij and Russell, 2000). Interestingly, recent studies

have succeeded to generate totipotent embryonic stem cells from male germ line stem cells

(Fagoonee et al., 2011). Therefore, male germ line stem cells isolated from the adult testis

may offer a powerful tool for regenerative medicine. The seminiferous epithelium is organized

in cyclic stages so that each cross-section of the tubule contains a defined grouping of germ

cell types at particular phases of development. In mice, there are twelve designated cell

associations or stages (I-XII) that are arranged in an ordered manner in the logical sequence

of developmental progression (Kotaja et al., 2004). This organization ensures the production

of a constant supply of mature spermatozoa. It also enables the coordinated regulation of

germ cells at different phases of differentiation by the surrounding Sertoli cells that embed

germ cells in the cytoplasmic pockets. Spermatogenesis is fully dependent on close

interactions with Sertoli cells that provide physical and nutritional support to germ cells (Rato

et al., 2012).

Male reproductive functions are controlled by the synchronized action of the hypothalamic-

pituitary-gonadal (HPG) axis. Gonadotropin-releasing hormone (GnRH) from hypothalamus

regulates the release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) from

anterior pituitary (Ruwanpura et al., 2010). FSH and LH are targeted to the testis where they

mostly act on somatic testicular cell types to stimulate testosterone production by interstitial

Leydig cells or to regulate Sertoli cells. Testosterone is absolutely required for maintaining

spermatogenesis, the main targets being meiosis and spermiogenesis (Ruwanpura et al.,

2010). In addition to the extrinsic hormonal and paracrine control, a wide variety of germline-

specific intrinsic delicate regulatory mechanisms are required. In this review, we concentrate

in describing the post-transcriptional gene control in male germ cells and summarizing the

results reporting the emerging importance of small non-coding RNAs in spermatogenesis and

male fertility.

2. Post-transcriptional gene regulation in spermatogenesis

Page 6: Small RNAs in spermatogenesis

 

The accurate, spatially and temporally regulated gene expression is of fundamental

importance for normal progression of spermatogenesis. Regulation takes place at

transcriptional and epigenetic levels, but in addition, post-transcriptional gene control has an

essential role especially at later steps of sperm differentiation when chromatin compaction

induces transcriptional silencing in elongating spermatids (Kimmins et al., 2004; Kimmins

and Sassone-Corsi, 2005). mRNAs for many spermiogenic proteins are synthesized already

in meiotic cells, and are temporarily stored and translationally regulated until needed during

later steps. As an example, mRNAs for protamines are kept inactive for several days until the

proteins are needed for histone-protamine transition, and premature translation leads to

spermatogenic defects (Lee et al., 1995). Fates of mRNAs are controlled mainly by RNA-

binding proteins that bind their targets either in a non-specific manner or through specific

motifs. There are a wide range of different RNA-binding proteins, also testis-specific ones,

expressed in meiotic and post-meiotic cells that participate in mRNA regulation by

recognizing target mRNAs and forming ribonucleoprotein (RNP) complexes (Paronetto and

Sette, 2010; Idler and Yan, 2012).

Cytoplasmic RNP granules provide important platforms for the post-transcriptional RNA

control (Meikar et al., 2011). For example processing bodies (P-bodies, PBs or GW/P bodies)

in mammalian cultured cells are known to coordinate mRNA decay, storage, transport and

miRNA-mediated pathways (Kulkarni et al., 2010). Germ cells of various organisms are

characterized by special cytoplasmic RNP granules that are commonly called germ granules.

The biological functions of the germ granules in non-mammalian and mammalian organisms

appear to be distinct, but they still share many protein components and are all involved in the

RNA regulation (Meikar et al., 2011; Kotaja and Sassone-Corsi, 2007; Chuma et al., 2009).

Germ granules exist in the cytoplasm of germ cells during the whole course of

spermatogenesis. The most prominent germ granules are intermitochondrial cement (IMC) in

spermatocytes that form between mitochondrial clusters, and the chromatoid body (CB) in

haploid round spermatids. These two types of granules share many protein components that

are mainly RNA-binding proteins, RNA helicases, Tudor domain proteins, and other proteins

involved in RNA processing. The germ granules seem to be central structures in the

processes involving PIWI-interacting RNA (piRNA) pathways (Meikar et al., 2011). The CB is

the biggest of all the known RNP granules. In mouse, the CB appears in pachytene

spermatocytes as a fibrous–granular structure and condenses into one single finely

filamentous, lobulated, perinuclear granule in post-meiotic round spermatids (Meikar et al.,

2011; Kotaja and Sassone-Corsi, 2007). The important role of the CB in male reproduction is

Page 7: Small RNAs in spermatogenesis

 

highlighted by the infertile phenotype of many knockout mice lacking various CB components

(Kotaja and Sassone-Corsi, 2007).

Recent evidence demonstrates that most of the human genome is transcribed but only a

small fraction of the transcribed RNA encodes for proteins (Rinn and Chang, 2012; Guttman

and Rinn, 2012). Therefore, most of the RNA transcripts are non-coding, i.e. they are not

used as a template for protein synthesis. It has become clear that these non-coding RNAs

are not just “junk” as it was originally thought, but they serve important cellular functions. In

addition to ribosomal RNA, transfer RNA, small nucleolar RNA (snoRNA), small nuclear RNA

(snRNA) and other well characterized functional RNAs, cells transcribe a legion of other

kinds of non-coding RNAs with different sizes and different functions. Non-coding RNAs have

been shown to have critical role in the control of gene expression, functioning both at

transcriptional level as components of chromatin remodeling complexes or post-

transcriptionally (Rinn and Chang, 2012). Male germ cells, especially meiotic cells, are

transcriptionally very active. Male germ cells express also an outstanding amount of different

non-coding RNAs, including both yet poorly characterized long non-coding RNAs and small

RNAs that are known to be essential for male germ cell-specific processes.

3. Control of gene expression by small RNAs Small non-coding RNAs are probably the best characterized class of non-coding RNAs in

terms of their functions. These RNAs do not code for proteins, however, they function as

cellular modulators of gene expression (Ghildiyal and Zamore, 2009; Ketting, 2011; Carthew

and Sontheimer, 2009). Discovery of microRNAs (miRNAs) started in Victor Ambros’

laboratory in 1993 while working on lin4 and how it controls developmental timing in the C.

elegans (Lee et al., 1993). Today after 20 years of their discovery, miRNAs are recognized as

vital regulatory factors that control the expression of a broad range of protein coding genes.

miRNAs function mainly post-transcriptionally by affecting the stability of their target mRNAs

or in some cases the translation of mRNAs (Ghildiyal and Zamore, 2009; Ketting, 2011;

Carthew and Sontheimer, 2009). In addition, transcriptional control of gene expression is

well-established in the plants (Khraiwesh et al., 2010) and also recently observed in human

granulopoiesis (Zardo et al., 2012).

miRNAs are synthesized as hairpin loop precursors and processed first by the nuclear

enzyme Drosha (Fig. 2) (Siomi and Siomi, 2010; Krol et al., 2010; Winter et al., 2009). After

the transport to the cytoplasm, they are further processed by the endonuclease Dicer into 21-

25 nucleotide double-stranded RNAs (dsRNAs). The produced dsRNA is unwound and one

Page 8: Small RNAs in spermatogenesis

 

of the strands is used as template strand which in association with accessory proteins and

Argonaute (AGO) proteins form the RNA-induced silencing complex (RISC) (Pratt and

MacRae, 2009; Hutvagner and Simard, 2008). miRNAs mediate the sequence-specific

recognition of their target mRNAs by the RISC complex. Depending upon sequence

complementary, the target mRNA is either degraded or translationally silenced. The same

pathway including Dicer and RISC-directed cleavage is used in antiviral defense to

specifically target and inactivate invading nucleic acids. This RNA silencing mechanism

couple the destruction of viral RNA with the use of the resulting small interfering RNAs

(siRNAs) to target other nucleic acid molecules that contain the complementary sequence

(Mlotshwa et al., 2008).

Several miRNAs can target one mRNA. On the other hand, one miRNA can down-regulate

the expression level of hundreds of genes and act as a regulatory factor to control various

physiological processes and diseases (Sood et al., 2006). Mammalian genomes comprise

roughly a thousand miRNA genes. Tissue-specific expression patterns of miRNAs are

observed (Lagos-Quintana et al., 2002), and misregulation of miRNA expressions seems to

be a common finding in cancer cells. miRNA profiling is used for many cancer sub-

classifications and identifications of cancer tissue origin (Lu et al., 2005; Rosenfeld et al.,

2008). Due to the high requirements for gene regulation, it is not a surprise that small non-

coding RNAs are also involved in the control of sperm production. Male germ cells express

several classes of small RNAs, including Dicer-dependent miRNAs and endogenous siRNAs

(endo-siRNAs), as well as Dicer-independent PIWI-interacting RNAs (piRNAs) (Meikar et al.,

2011). These small RNAs and their roles in spermatogenesis will be discussed in the

following chapters.

4. Dicer-dependent small RNAs in male germ cells

4.1. miRNAs and endo-siRNAs

Post-transcriptional mRNA regulation is active during spermatogenesis, and miRNA-

mediated mechanisms are clearly involved in the orchestrated and stage-specific control of

gene expression during the whole course of male germ cell differentiation. Several studies

using miRNA microarrays, RT-PCR or small RNA sequencing have identified miRNAs that

are highly, exclusively or preferentially expressed in the testis and at the specific phases of

male germ cell differentiation (Papaioannou and Nef, 2010; McIver et al., 2012a). These

studies have demonstrated for example differentially expressed miRNAs between immature

and mature mouse testes (Yan et al., 2007), specific miRNA profiles in highly enriched

Page 9: Small RNAs in spermatogenesis

 

populations of gonocytes and spermatogonia (McIver et al., 2012b), and miRNA profiles

during the first wave of spermatogenesis by using prepubertal testes collected on post-natal

days (PND) 7, 10 and 14 (Buchold et al., 2010). miRNA profiles were identified also from

enriched populations of stem cells, pre-meiotic and meiotic cells (Smorag et al., 2012) or

spermatogonia, spermatocytes and spermatids (Marcon et al., 2008). These cell types seem

to express several common miRNAs, but also some miRNAs that are specific for the certain

cell type.

Intriguingly, densities of miRNA genes are significantly higher on the mammalian X

chromosome than on autosomes as a consequence of the organization of X-chromosomal

miRNA genes in clusters that consist of paralogous copies resulting from miRNA gene

duplication (Meunier et al., 2013). Sequencing of spermatocyte and spermatid RNA in

comparison to somatic cells revealed that the multimember family miRNAs on the X have

higher expression levels than all other miRNA categories in male germ cells (Meunier et al.,

2013). Sex chromosomal genes are normally silenced by the process of meiotic sex

chromosome inactivation (MSCI) during meiotic prophase I. However, miRNAs and

especially the miRNA families that expanded by gene duplication, seems to escape silencing

by MSCI, suggesting that the miRNA gene duplications on the X was selectively favored

during evolution to allow their expression in spermatocytes and spermatids in spite of sex

chromosome inactivation (Meunier et al., 2013; Song et al., 2009).

The identification of functions and target genes for specific miRNAs during spermatogenesis

has already begun. Several studies support the role of miRNA-mediated gene control in the

maintenance of undifferentiated state of spermatogonia and the induction of differentiation.

miR-146 that is highly expressed in undifferentiated spermatogonia is involved in the control

of retinoid acid-induced spermatogonial differentiation in the mouse (Huszar and Payne,

2013). This is at least partially mediated by the regulation of the expression of the mediator

complex subunit 1 (Med1), a coregulator of retinoid receptors. Other miRNAs involved in the

regulation of the undifferentiated vs. differentiated state of spermatogonia are miR-221 and

miR-222. The impaired function of these X chromosome-clustered miRNAs in mouse induces

spermatogonial differentiation (Yang et al., 2013). Mir-17-92 and its paralog Mir-106b-25

clusters are also expressed in undifferentiated spermatogonia and down-regulated during

differentiation. Deletion of Mir-17-92 cluster in mouse results in small testes and lower

number of epididymal sperm and increase in Mir-106b-25 expression which suggests a

functional co-operation of these two clusters (Tong et al., 2012). High-throughput sequencing

identified miR-21, along with miR-34c, -182, -183, and -146a, as being preferentially

expressed spermatogonial stem cell (SSC)-enriched population. The transient inhibition of

Page 10: Small RNAs in spermatogenesis

 

miR-21 in SSC-enriched germ cell cultures increased the number of germ cells undergoing

apoptosis and reduced the SSC potency, indicating that miR-21 is important for maintaining

the SSC population (Niu et al., 2011).

miR-34c is highly expressed in spermatocytes and round spermatids (Romero et al., 2011;

Bouhallier et al., 2010; Liang et al., 2012). Inhibition of miR-34c in primary spermatocytes

seems to prevent germ cell from testosterone deprivation-induced apoptosis, and

overexpression of miR-34c in cultured germ cells trigger apoptosis. This is at least partially

mediated by targeting ATF1 via direct binding to ATF1 3’UTR (Liang et al., 2012). When

overexpressed in HeLa cells, miR-34c induces a shift in the transcriptome towards the germ

lineage transcriptome (Bouhallier et al., 2010). Therefore, miR-34c was suggested to play a

role in enhancing the germinal phenotype of cells already committed to this lineage.

Interestingly, miR-34c is also found in mature sperm and the zygotes, and the sperm-borne

miR-34c appears to be important for the first zygotic cell division via modulation of Bcl-2

expression (Liu et al., 2012b). miR-34c is not an only example of sperm-derived miRNAs.

Despite the transcriptionally inactive state of mature spermatozoa, its nucleus contains a

complex population of both mRNAs and miRNAs that may have a role in early embryo

development, and that can be potentially exploited in the investigation and diagnostics of

male infertility (McIver et al., 2012a; Hamatani, 2012; Abu-Halima et al., 2013).

Other miRNAs involved in the regulation of meiotic and post-meiotic gene expression have

been reported. miR-449 cluster miRNAs are drastically up-regulated upon meiotic initiation

during first wave of spermatogenesis, and their expression is under the regulation of

transcription factors CREMtau and SOX5 (Bao et al., 2012). miR-449 cluster and miR-34b/c

were shown to share some target genes that belong to the E2F transcription factor-

retinoblasoma regulatory network (Bao et al., 2012). The correct timing of the expression of

transition proteins (TP) and Protamines (Prm) is critical for successful histone-protamine

transition during late spermatogenesis. TPs and Prms are subjected to extensive

translational control, including miRNA-mediated mechanisms. TP2 and Prm2 mRNAs are

targeted by the testis-specific miR-469 that represses TP2 and Prm2 protein expression in

pachytene spermatocytes and round spermatids at the translation level with minor effect on

mRNA degradation (Dai et al., 2011). miR-122a that is enriched in late-stage male germ cells

can also directly control TP2 expression by inducing mRNA cleavage (Yu et al., 2005). miR-

18 that belongs to the Oncomir-1 or miR-17-92 cluster can directly target heat shock factor 2

(HSF2), a transcription factor that influences a wide range of developmental processes

including spermatogenesis (Bjork et al., 2010). The expressions of HSF2 and miR-18 exhibit

an inverse correlation during spermatogenesis and inhibition of miR-18 in intact seminiferous

Page 11: Small RNAs in spermatogenesis

10 

 

tubules leads to increased HSF2 protein levels and altered expression of HSF2 target genes

(Bjork et al., 2010).

Recently, endogenous siRNAs (endo-siRNAs) which are processed by Dicer were reported

to be highly expressed in mouse male germ cells (Song et al., 2011). Endo-siRNAs do not

need the microprocessor complex (Drosha-DGCR8) for their biogenesis because they are

processed from long dsRNA precursors; nuclear Drosha activity is required only for the

processing of the stem-loop structure-containing miRNA precursors (Krol et al., 2010). Endo-

siRNAs were initially identified in yeasts, plants, C. elegans and Drosophila (Czech et al.,

2008; Okamura et al., 2008; Chapman and Carrington, 2007). In the mouse, they have been

reported in embryonic stem cells (Babiarz et al., 2008) and oocytes (Watanabe et al., 2008;

Tam et al., 2008) in addition to the testis. A total of 73 endo-siRNAs were identified by

sequencing the testicular RNA. The majority of them were mapped to several (even

hundreds) of different sites on multiple chromosomes in contrast to miRNAs that usually

derive from a unique locus or very few loci (Song et al., 2011). Predicted targets for the

testicular endo-siRNAs were mainly mRNAs (~92%) but also transcripts of pseudogenes

(~3%), retrotransposons (~1%), and non-coding RNAs (~4%). In mouse oocytes, endo-

siRNAs seem to regulate both retrotransposons and protein-coding transcripts (Watanabe et

al., 2008; Tam et al., 2008). Testicular endo-siRNAs are probably involved in post-

transcriptional RNA regulation since they effectively induced target mRNA degradation in

vitro. Since they display numerous hits on multiple chromosomes, it is tempting to speculate

that in addition to their role in post-transcriptional RNA control, they could also have nuclear

effects on chromatin modifications similar to what has been shown for endo-siRNAs in other

organisms (Song et al., 2011).

4.2. Male germ cell-specific Dicer and Drosha knockout mouse models

miRNA and siRNA processing is globally dependent on the two endonucleases, the

microprocessor complex (Drosha/DGCR8) and Dicer. Both Dicer and DGCR8 have been

demonstrated to be essential genes in mouse since their deletion leads to an embryonic

lethal phenotype (Bernstein et al., 2003; Wang et al., 2007). Tissue and cell type–specific

knockout mouse models have enabled the assessment of the functions of these pathways in

certain tissues and in adult organism. By using this approach, the role of miRNA/siRNA

pathways in spermatogenesis has started to be revealed (Fig. 1, Table 1). Dicer has been

demonstrated to be required for the functions of both Sertoli cells that are the supporting

somatic cells of the seminiferous epithelium (Papaioannou et al., 2009; Papaioannou et al.,

2011) and germ cells that will be discussed in more detail. Removal of Dicer1 gene in

Page 12: Small RNAs in spermatogenesis

11 

 

primordial germ cells after embryonic day 10 (E10) by using the tissue-nonspecific alkaline

phosphatase (Tnap) promoter-driven Cre expression demonstrated for the first time the

importance of Dicer for male fertility (Hayashi et al., 2008; Maatouk et al., 2008). Upon

removal of functional Dicer, the PGCs showed defects in proliferation and post-natal

spermatogenesis was also affected (Hayashi et al., 2008; Maatouk et al., 2008). This mouse

model could not be used to address the exact role of Dicer in adult spermatogenesis due to

affected embryonic germ line development, and the low penetrance of TnapCre transgene

which interferes with the analysis.

Conditional deletion of Dicer1 in the male germ line just before birth by using the Ddx4

promoter-driven Cre expression resulted in severe cumulative defects in meiotic and post-

meiotic germ cells (Romero et al., 2011; Liu et al., 2012a). The spermatocytes were delayed

in the progression of meiotic prophase I and apoptosis was increased, resulting in reduced

number of round spermatids. The remaining round spermatids failed to undergo normal

differentiation and morphologically abnormal non-functional spermatozoa were generated

(Romero et al., 2011). The expression of transposable elements of the SINE (short

interspersed nuclear element) family was up-regulated in Dicer1 knockout spermatocytes

(Romero et al., 2011). Similar up-regulation of transposon expression (MT and SINE families)

was observed in Dicer1 deficient mouse oocytes (Murchison et al., 2007). Dicer function has

also been linked to transposon silencing in retinal pigmented epithelium where it is involved

in retrotransposon transcript degradation by a miRNA-independent mechanism (Kaneko et

al., 2011).

Interestingly, less severe phenotypes were observed when Dicer1 was deleted after birth in

post-natal spermatogonia in Neurogenin3 (Ngn3)-Cre and Stra8Cre knockout models

(Korhonen et al., 2011; Greenlee et al., 2012; Wu et al., 2012). Selective removal of Dicer1 in

type A spermatogonia by Ngn3Cre transgene did not have clear effects on meiotic

progression and the first visible impairments were found in haploid male germ cells

(Korhonen et al., 2011). Elongation phase of spermiogenesis was severely affected with

problems in chromatin organization and shaping and condensation of sperm head (Korhonen

et al., 2011). The number of epididymal spermatozoa was dramatically reduced and all

spermatozoa were morphologically abnormal. Interestingly, transposon expression was

unaffected in the knockout testis. The difference in transposon expression between the

Ddx4Cre and Ngn3Cre–induced Dicer1 knockout models indicate that Dicer may be involved

in transposon control in embryonic and/or very early post-natal phases before the onset

Ngn3 expression. On the other hand, major satellite expression was induced in the testis of

Ngn3Cre-Dicer1 knockout mice similarly to what is observed in mouse embryonic stem cells

Page 13: Small RNAs in spermatogenesis

12 

 

lacking functional Dicer (Korhonen et al., 2011; Murchison et al., 2005; Kanellopoulou et al.,

2005). Altogether the reported defects in repeat sequence expressions (transposon and

satellite repeats) in different Dicer knockout models suggest that Dicer is implicated in the

control of repetitive elements in addition to its essential role in miRNA biogenesis.

The induction of Dicer1 deletion in early spermatogonia by Stra8Cre transgene resulted in

the comparable phenotype to Ngn3Cre driven Dicer1 deletion (Greenlee et al., 2012; Wu et

al., 2012). The meiotic progression of Stra8Cre-Dicer1 knockout spermatocytes was delayed

and haploid differentiation was greatly affected. The microarray analysis of PND18 testes of

the control and knockout mice revealed that ~40% of the genes were differentially expressed

in the knockout testis (Greenlee et al., 2012). Interestingly, disproportionately high

percentage of X- and Y-linked genes was overexpressed in the knockout testis. However, it is

still unclear if the overexpression of the genes that should be silenced during MSCI is

induced by direct defects in MSCI or if it originates from the increased transcript stability or

by the enrichment of pre-MSCI spermatocytes in the analysed knockout testis samples

(Greenlee et al., 2012). The ablation of Dicer or Drosha in spermatogonia did not appear to

affect significantly the sex body formation in pachytene spermatocytes (Wu et al., 2012). It is

evident that the mechanisms of Dicer-mediated sex chromosomal gene silencing require

further investigations.

Male germ cell–specific Dicer1 knockout mouse has also been generated by using protamine

1 (Prm1)-Cre transgene, that is activated much later in spermatogenesis in haploid cells

(Chang et al., 2012). The spermatogenesis of these mice was not so severely affected as the

spermatogenesis in Dicer1 knockout models in which the deletion takes place already in

spermatogonia (Korhonen et al., 2011; Greenlee et al., 2012; Wu et al., 2012). In

consequence, a higher number of spermatozoa were also found in the epididymides of

Prm1Cre-Dicer1 knockout males. However, post-meiotic differentiation was disrupted and

elongating spermatids had abnormal head morphology and compromised chromatin integrity

(Chang et al., 2012). Dicer1 inactivation in round spermatids impaired translational activation

of germ cell transcripts including Prm1, possibly resulting from the transcript sequestration

into translationally inert RNP complexes. All these different Dicer1 knockout models jointly

suggest that Dicer activities are required throughout the male germ cell differentiation, and

the earlier the deletion takes place, the more defects accumulate and the more compromised

the spermatogenesis is.

Since Dicer is required for the processing of both miRNAs and endo-siRNAs, it is important

to dissect these pathways and clarify the involvement of specific RNA species in the

Page 14: Small RNAs in spermatogenesis

13 

 

regulation of spermatogenic processes. The functional difference between miRNA and endo-

siRNA pathways in spermatogenesis has been investigated by comparing the testicular

phenotypes of Dicer1 and Drosha knockout mice (deletion induced in spermatogonia by

Stra8Cre transgene) (Wu et al., 2012). Drosha knockout testis that has defective miRNA

pathway but intact endo-siRNA pathways displayed more severe spermatogenic disruptions,

thus highlighting the essential role of miRNA pathways in normal post-natal

spermatogenesis. This is in contrast to female germ cells that appear to be more dependent

on endo-siRNA pathways; mRNA profiles of wild-type and Dgcr8 null oocytes were identical,

whereas Dicer null oocytes depicted many misregulated transcripts, therefore suggesting

that endo-siRNAs are important during oocyte maturation and preimplantation development

(Suh et al., 2010). The transcriptome analysis revealed that mRNA profiles of Drosha- or

Dicer-null pachytene spermatocytes and round spermatids were altered (Wu et al., 2012).

Even though some mRNAs showed similar changes in both mouse lines, many changes in

the mRNA expressions were unique to either of the two genotypes. This indicates that

miRNAs and other Dicer-dependent pathways (e.g. endo-siRNAs) have divergent roles in the

regulation of protein-coding mRNAs during post-natal spermatogenesis (Wu et al., 2012).

The roles of small RNA pathways can be also studied by inactivating the effector proteins

that mediate miRNA/siRNA functions. AGO proteins (AGO1-AGO4) associate with miRNAs

and siRNAs and mediate their effects through formation of RISC complexes. AGO proteins

are ubiquitously expressed, but AGO4 is highly expressed in testis and interestingly, it

localizes to the transcriptionally silenced sex body during the meiotic prophase I

(Modzelewski et al., 2012). Ago4 knockout spermatogonia initiate meiosis early and manifest

incorrect sex body assembly, which leads to disrupted MSCI. Interestingly, AGO4 deletion is

accompanied by the decreased expression of X-chromosomal miRNAs known to escape

MSCI (Song et al., 2009; Modzelewski et al., 2012). These findings clearly demonstrate the

role of AGO4 in the nucleus of pachytene spermatocytes in regulating MSCI, and implicates

that small RNA pathways are involved in this process. Some miRNAs localize in the nucleus

of pachytene spermatocytes and in the sex body further supporting the role of miRNA

pathway in MSCI (Marcon et al., 2008). Sex chromosome regulation is clearly a central target

for Dicer and small RNAs, but the exact relationship between Dicer, miRNAs, AGO4 and

MSCI remains to be explored.

5. PIWI proteins and piRNAs

piRNAs form a big and complex group of small RNAs that were discovered in 2006 by high-

throughput sequencing of either total testis RNA or RNA species interacting with PIWI

Page 15: Small RNAs in spermatogenesis

14 

 

proteins (Aravin et al., 2006; Girard et al., 2006; Grivna et al., 2006; Watanabe et al., 2006).

piRNAs are 24-30 nucleotides, single-stranded RNAs that are synthesized in very high

numbers by Dicer-independent biogenesis mechanism and that are expressed predominantly

in the male germ line (Chuma and Nakano, 2013). piRNA encoding genes are mostly found

in genomic clusters ranging from a few to hundreds of kilobases in length. piRNAs in the

clusters map typically to only one genome strand of each cluster. This kind of asymmetric

organization suggests that long single-stranded precursor piRNA transcripts are transcribed

form the piRNA clusters. piRNA biogenesis from long transcripts requires at least two

nucleolytic steps to process both the monophosphorylated 5´end and the 3´end. The details

of the primary piRNA biogenesis are still obscure, but recently a protein

PLD6/MITOPLD/Zucchini was suggested to act in primary piRNA biogenesis as single-

strand-specific nuclease (Nishimasu et al., 2012; Ipsaro et al., 2012). To support this function,

primary piRNA generation was demonstrated to be defective in Pld6 knockout mice

(Watanabe et al., 2011; Huang et al., 2011). In mammals, piRNAs are classified mainly into

three groups on the basis of their stage-specific expression patterns: fetal piRNAs and post-

natal pre-pachytene and pachytene piRNAs (Fig. 1) (Chuma and Nakano, 2013). Sequence

comparison shows that these piRNA populations expressed at different developmental

stages are quite distinct and they originate from chromosomal clusters showing little overlap.

piRNA sequences are not conserved between the species but the locations of piRNA clusters

seems to be conserved. This suggests the involvement of chromatin-based regulatory

mechanisms in the control of piRNA cluster expression, even though nothing is known so far

about the transcriptional or developmental regulation of the piRNA clusters.

5.1. piRNAs and genome defense

To date, piRNAs in mammals and lower organisms have been demonstrated to be involved

in male germ line development, epigenetic regulation and mobile element repression (Chuma

and Nakano, 2013). Especially the role of piRNA machinery in genome defense and

transposable element (TE) control has been convincingly demonstrated in fruit fly, worm and

fetal testis of the mouse (Aravin et al., 2008; Kuramochi-Miyagawa et al., 2008; Siomi et al.,

2011; Bao and Yan, 2012). TEs are mobile DNA segment that can move and replicate within

the host genome and if aberrantly expressed, can result in potentially harmful insertional

mutagenesis and genomic rearrangements. TE expression is controlled at transcriptional

level by DNA methylation and heterochromatin formation, and post-transcriptionally by small

RNAs. Male germ cells are at high risk of transposon attack during the time of epigenetic

reprogramming in the fetal testis which in turn leads to temporary relaxation of TEs from

epigenetic control. Therefore, an adaptive strategy has evolved in germ cells to silence TEs

Page 16: Small RNAs in spermatogenesis

15 

 

and to maintain genomic integrity. piRNA machinery seems to contribute to this genome

defense in a significant way. piRNA-mediated transposon silencing takes place at the post-

transcriptional level to cleave transposon transcripts, but a connection between piRNA action

and DNA methylation of transposon genes has also been demonstrated (Aravin et al., 2008;

Kuramochi-Miyagawa et al., 2008). A specific mechanism, so-called ping-pong amplification

cycle, is utilized to amplify piRNAs during TE control (Fig. 3). In this pathway, primary piRNAs

loaded to PIWI proteins recognize complementary target RNAs that are cleaved to produce

secondary piRNAs that in turn continues the cycle to amplify piRNA response.

piRNA functions are mediated through binding to PIWI proteins. There are three PIWI

subfamily members in the mouse, PIWIL1/MIWI, PIWIL2/MILI and PIWIL4/MIWI2 that are

expressed solely in the germ line with distinct expression patterns (Deng and Lin, 2002;

Kuramochi-Miyagawa et al., 2004; Carmell et al., 2007). MIWI2 has a very narrow expression

window from embryonic prospermatogonia until the very early post-natal spermatogonia,

corresponding to the time of epigenetic reprogramming and de novo DNA methylation

(Carmell et al., 2007; Aravin et al., 2009). In these cells, MIWI2 localizes in the nucleus and

in cytoplasmic granules (piP-bodies) that are also positive for MEAL, DDX4, TDRD9 and

canonical Processing body (P-body) proteins (Aravin et al., 2009). MILI co-exists with MIWI2

in prospermatogonia but it localizes in another kind of germ granules, pi-bodies, together with

TDRD1 and DDX4. These two types of germ granules are in close connection and participate

in piRNA biogenesis, de novo DNA methylation and transposon silencing (Aravin et al.,

2009). Knockout mouse models for Miwi2 or Mili genes generate a testicular phenotype with

spermatogenesis arrested at early meiotic stage (Kuramochi-Miyagawa et al., 2004; Carmell

et al., 2007) possibly resulting from the genomic instability due to an aberrant TE

expressions.

5.2. piRNAs in post-natal spermatogenesis

Two of three PIWI proteins are expressed during post-natal spermatogenesis where the

functions of piRNAs are much less understood than in prospermatogonia. MILI is found in the

cytoplasm of spermatogonia, pachytene spermatocytes and round spermatids, whereas

MIWI expression starts in meiotic cells and continues during the whole course of round

spermatid differentiation (Chuma and Nakano, 2013). In meiotic and post-meiotic cells, MIWI

and MILI predominantly localize in germ granules such as IMC in spermatocytes and CBs in

spermatids (Meikar et al., 2011; Deng and Lin, 2002; Kuramochi-Miyagawa et al., 2004;

Beyret and Lin, 2011). piRNA expression is massively induced in pachytene spermatocytes

and this specific subset of piRNAs is generally called the pachytene piRNAs (Aravin et al.,

Page 17: Small RNAs in spermatogenesis

16 

 

2006; Grivna et al., 2006). Pachytene piRNAs are different from fetal piRNAs or pre-

pachytene piRNAs that bind MILI and are expressed before the pachytene stage of the first

meiotic prophase (Chuma and Nakano, 2013). The production of pachytene piRNAs has

been demonstrated to be independent of the ping-pong amplification mechanism (Fig. 3).

Instead, pachytene piRNAs are more likely to be produced by primary processing pathway

(Beyret et al., 2012).

Deletion of Miwi gene in mouse results in arrest of spermatogenesis at round spermatid

stage with no obvious defects in meiotic progression and complete lack of elongating

spermatids (Deng and Lin, 2002). The expression of pachytene piRNAs is significantly

reduced in the Miwi knockout testis (Grivna et al., 2006) suggesting that their expression is

dependent on MIWI function. Members of the Argonaute family including PIWI proteins are

small RNA-guided nucleases that can cleave target nucleic acids (Ender and Meister, 2010).

Interestingly, a mutant knock-in mouse line expression catalytically inactive form of MIWI

devoid of RNA slicing activity manifest almost identical spermatogenic phenotype than the

Miwi full knockout mouse, which indicates that a substantial portion of the biology of MIWI is

implemented via its small RNA-guided slicer activity. This activity includes the post-

transcriptional cleavage of LINE1 (long interspersed nuclear elements) retrotransposon

transcripts (Reuter et al., 2011). The primary piRNA biogenesis was unaffected in the testes

defective in MIWI slicer activity. Therefore, pachytene piRNA production seems to require

MIWI but not the MIWI slicer activity. The mechanism for pachytene piRNA production

remains unclear, however recent studies have started to reveal critical components, such as

Zucchini/MitoPLD/PLD6 as discussed above, and an RNA helicase MOV10L1 that has been

demonstrated to act upstream of PIWI proteins in the primary processing of pachytene

piRNAs by knockout mouse models (Zheng and Wang, 2012; Zheng et al., 2010; Frost et al.,

2010). Primary piRNA biogenesis has been also suggested to involve 3´ to 5´ exonucleolytic

processing for maturation of piRNA 3´end, after the precursor transcript with processed

5´end has been loaded to PIWI proteins (Vourekas et al., 2012).

The functions of the pachytene piRNAs and the reason of their overwhelmingly high

expression are still under active investigation. As discussed above, piRNAs can guide MIWI-

mediated slicing of TEs (Reuter et al., 2011). However, pachytene piRNA population is

diverse and only around 20% of piRNAs map to TE genes. The majority of pachytene

piRNAs cluster in repeat-devoid intergenic regions and a fraction maps also to protein-coding

genes (RefSeq exons). High-throughput sequencing after cross-linking and

immunoprecipitation (HITS-CLIP) of MILI and MIWI bound RNAs coupled with RNA-

sequencing revealed that the majority of the intergenic and uniquely mapped RefSeq-exon

Page 18: Small RNAs in spermatogenesis

17 

 

piRNAs had no complementary larger RNA targets (Vourekas et al., 2012). This suggests

that MILI and MIWI proteins do not use piRNAs as guides to target non-repeat RNAs. It is

currently unknown for what purpose these piRNAs without complementary targets are

produced, and one possibility is that they are the end products of a degradation mechanism

acting on e.g. meiotic RNA transcripts that are no longer required in haploid cells (Vourekas

et al., 2012). The functional characterization of pachytene piRNAs is challenging due to the

complexity of spermatogenic system and the lack of in vitro culture model for meiotic and

post-meiotic differentiation. Moreover, proteins that function in piRNA pathways seem to have

other piRNA-independent functions as well. For example, MIWI has been demonstrated to

bind, protect and translationally repress spermiogenic mRNAs without using piRNAs as

guides (Vourekas et al., 2012; Nishibu et al., 2012).

5.3. Epigenetic inheritance and non-coding RNAs

Transgenerational effects have wide-ranging implications for human health, biological

adaptation and evolution, and germ line cells play a key role in this process. In addition to

genetic information, the epigenetic information can be transmitted from parent to offspring via

the gametes, which enables transgenerational epigenetic inheritance. Because the

environmentally induced changes in the epigenome of the germline become permanently

programmed, the altered epigenome and phenotype can be transmitted to subsequent

progeny. Environmental epigenetics has certainly a critical role in disease etiology (Skinner,

2011). DNA methylation has always been considered to be a likely candidate for mediating

transgenerational epigenetic inheritance because of vast changes of DNA methylation

patterns during early mammalian development (Lange and Schneider, 2010). However,

recent evidence points towards diffusible factors, in particular RNA, as a possible mechanism

(Daxinger and Whitelaw, 2012). Non-coding RNAs have the ability to modify epigenetic

marks, and therefore could mediate epigenetic inheritance through homology-dependent

regulation. Mature sperm contain different RNA populations, and sperm-borne RNA has been

detected in the zygote, and it is easy to envisage that RNAs transmitted from sperm to early

embryo at fertilization, could act as mobile signals that influence the epigenetic status of

certain elements (Daxinger and Whitelaw, 2012).

piRNAs that have a role in the silencing of mobile genetic elements are also considered as

potential mediators of epigenetic transgenerational inheritance (Aravin et al., 2008;

Kuramochi-Miyagawa et al., 2008; Daxinger and Whitelaw, 2012). Recent reports show that

in C. elegans germ line, piRNAs can initiate a multigenerational epigenetic memory of RNA

that is recognizes as “nonself” (Shirayama et al., 2012; Ashe et al., 2012; Lee et al., 2012).

Page 19: Small RNAs in spermatogenesis

18 

 

By using the RNA-based recognition mechanism, foreign sequences could be detected not

by any molecular signature, but by comparing the foreign sequence to a memory of previous

gene expression. On the other hand, endogenous germline-expressed genes are actively

protected from piRNA-induced silencing (Lee et al., 2012). This raises an intriguing possibility

that mammalian small RNAs could function in epigenetic programming and generation of an

epigenetic memory.

6. Future perspectives

The ability of germ cells to transmit their genetic and epigenetic information to following

generations highlights the importance of the correct establishment and maintenance of

epigenetic marks, as well as the protection of information to ensure the quality and to prevent

the transmission of faulty information to offspring. Extensive and accurate transcriptional and

post-transcriptional regulation is required to support the highly orchestrated expression of

specific genes required for each step of spermatogenesis. In addition, spermatogenesis is

accompanied by major chromosomal rearrangement during meiosis and haploid cell

differentiation. The combined actions of miRNAs, endo-siRNAs and piRNAs, and possibly

several other classes of yet unidentified small RNAs have major contribution to the control of

gene expression and chromatin in male germ cells. The functions of small RNAs, especially

those of piRNAs are diverse and require still extensive long-term investigation before we are

able to obtain the full picture. In the future it is also essential to identify the regulatory

network governing the expression of small RNAs, as well as to clarify the importance of the

spatial control of RNA regulation in specific subcellular compartments.

People in Europe and other developed countries have been lately challenged by increasing

problems in fertility. Both genetic factors and environmental interference contribute to the

etiology of subfertility and infertility. Male infertility is often reflecting faults in

spermatogenesis, and these faults are usually overcome by assisted reproductive

technologies. We know that the epigenetic reprogramming taking place during the whole

course of spermatogenesis is important for correct development of spermatozoa. However,

we do not understand the possible far-reaching transgenerational consequences resulted

from the bypassing of the critical differentiation steps. Since small RNAs act as versatile

regulators of gene expression and epigenetic events during spermatogenesis, they may

provide a potent tool for the diagnosis of infertility and the assessment of the suitable

methods for infertility treatments.

Page 20: Small RNAs in spermatogenesis

19 

 

Acknowledgements We want to thank all the lab members for their comments and critical reading of the

manuscript. The authors are funded by the Academy of Finland, Emil Aaltonen Foundation,

Novo Nordisk Foundation and Sigrid Jusélius Foundation.

References

Abu-Halima, M., Hammadeh, M., Schmitt, J., Leidinger, P., Keller, A., Meese, E., Backes, C., 2013. Altered microRNA expression profiles of human spermatozoa in patients with different spermatogenic impairments. Fertil. Steril. 99, 1249-1255.

Aravin, A., Gaidatzis, D., Pfeffer, S., Lagos-Quintana, M., Landgraf, P., Iovino, N., Morris, P., Brownstein, M.J., Kuramochi-Miyagawa, S., Nakano, T., Chien, M., Russo, J.J., Ju, J., Sheridan, R., Sander, C., Zavolan, M., Tuschl, T., 2006. A novel class of small RNAs bind to MILI protein in mouse testes. Nature 442, 203-207.

Aravin, A.A., Sachidanandam, R., Bourc'his, D., Schaefer, C., Pezic, D., Toth, K.F., Bestor, T., Hannon, G.J., 2008. A piRNA pathway primed by individual transposons is linked to de novo DNA methylation in mice. Mol. Cell 31, 785-799.

Aravin, A.A., van der Heijden, G.W., Castaneda, J., Vagin, V.V., Hannon, G.J., Bortvin, A., 2009. Cytoplasmic compartmentalization of the fetal piRNA pathway in mice. PLoS Genet. 5, e1000764.

Ashe, A., Sapetschnig, A., Weick, E.M., Mitchell, J., Bagijn, M.P., Cording, A.C., Doebley, A.L., Goldstein, L.D., Lehrbach, N.J., Le Pen, J., Pintacuda, G., Sakaguchi, A., Sarkies, P., Ahmed, S., Miska, E.A., 2012. piRNAs can trigger a multigenerational epigenetic memory in the germline of C. elegans. Cell 150, 88-99.

Babiarz, J.E., Ruby, J.G., Wang, Y., Bartel, D.P., Blelloch, R., 2008. Mouse ES cells express endogenous shRNAs, siRNAs, and other Microprocessor-independent, Dicer-dependent small RNAs. Genes Dev. 22, 2773-2785.

Bao, J., Li, D., Wang, L., Wu, J., Hu, Y., Wang, Z., Chen, Y., Cao, X., Jiang, C., Yan, W., Xu, C., 2012. MicroRNA-449 and microRNA-34b/c function redundantly in murine testes by targeting E2F transcription factor-retinoblastoma protein (E2F-pRb) pathway. J. Biol. Chem. 287, 21686-21698.

Bao, J., Yan, W., 2012. Male germline control of transposable elements. Biol. Reprod. 86, 162, 1-14.

Bernstein, E., Kim, S.Y., Carmell, M.A., Murchison, E.P., Alcorn, H., Li, M.Z., Mills, A.A., Elledge, S.J., Anderson, K.V., Hannon, G.J., 2003. Dicer is essential for mouse development. Nat. Genet. 35, 215-217.

Beyret, E., Lin, H., 2011. Pinpointing the expression of piRNAs and function of the PIWI protein subfamily during spermatogenesis in the mouse. Dev. Biol. 355, 215-226.

Page 21: Small RNAs in spermatogenesis

20 

 

Beyret, E., Liu, N., Lin, H., 2012. piRNA biogenesis during adult spermatogenesis in mice is independent of the ping-pong mechanism. Cell Res. 22, 1429-1439.

Bjork, J.K., Sandqvist, A., Elsing, A.N., Kotaja, N., Sistonen, L., 2010. miR-18, a member of Oncomir-1, targets heat shock transcription factor 2 in spermatogenesis. Development 137, 3177-3184.

Bouhallier, F., Allioli, N., Lavial, F., Chalmel, F., Perrard, M.H., Durand, P., Samarut, J., Pain, B., Rouault, J.P., 2010. Role of miR-34c microRNA in the late steps of spermatogenesis. RNA 16, 720-731.

Buchold, G.M., Coarfa, C., Kim, J., Milosavljevic, A., Gunaratne, P.H., Matzuk, M.M., 2010. Analysis of microRNA expression in the prepubertal testis. PLoS One 5, e15317.

Carmell, M.A., Girard, A., van de Kant, H.J., Bourc'his, D., Bestor, T.H., de Rooij, D.G., Hannon, G.J., 2007. MIWI2 is essential for spermatogenesis and repression of transposons in the mouse male germline. Dev. Cell. 12, 503-514.

Carthew, R.W., Sontheimer, E.J., 2009. Origins and Mechanisms of miRNAs and siRNAs. Cell 136, 642-655.

Chang, Y.F., Lee-Chang, J.S., Imam, J.S., Buddavarapu, K.C., Subaran, S.S., Sinha-Hikim, A.P., Gorospe, M., Rao, M.K., 2012. Interaction between microRNAs and actin-associated protein Arpc5 regulates translational suppression during male germ cell differentiation. Proc. Natl. Acad. Sci. U. S. A. 109, 5750-5755.

Chapman, E.J., Carrington, J.C., 2007. Specialization and evolution of endogenous small RNA pathways. Nat. Rev. Genet. 8, 884-896.

Chuma, S., Hosokawa, M., Tanaka, T., Nakatsuji, N., 2009. Ultrastructural characterization of spermatogenesis and its evolutionary conservation in the germline: germinal granules in mammals. Mol. Cell. Endocrinol. 306, 17-23.

Chuma, S., Nakano, T., 2013. piRNA and spermatogenesis in mice. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 368, 20110338.

Czech, B., Malone, C.D., Zhou, R., Stark, A., Schlingeheyde, C., Dus, M., Perrimon, N., Kellis, M., Wohlschlegel, J.A., Sachidanandam, R., Hannon, G.J., Brennecke, J., 2008. An endogenous small interfering RNA pathway in Drosophila. Nature 453, 798-802.

Dai, L., Tsai-Morris, C.H., Sato, H., Villar, J., Kang, J.H., Zhang, J., Dufau, M.L., 2011. Testis-specific miRNA-469 up-regulated in gonadotropin-regulated testicular RNA helicase (GRTH/DDX25)-null mice silences transition protein 2 and protamine 2 messages at sites within coding region: implications of its role in germ cell development. J. Biol. Chem. 286, 44306-44318.

Daxinger, L., Whitelaw, E., 2012. Understanding transgenerational epigenetic inheritance via the gametes in mammals. Nat. Rev. Genet. 13, 153-162.

de Rooij, D.G., Russell, L.D., 2000. All you wanted to know about spermatogonia but were afraid to ask. J. Androl. 21, 776-798.

Page 22: Small RNAs in spermatogenesis

21 

 

Deng, W., Lin, H., 2002. Miwi, a Murine Homolog of Piwi, Encodes a Cytoplasmic Protein Essential for Spermatogenesis. Dev. Cell. 2, 819-830.

Ender, C., Meister, G., 2010. Argonaute proteins at a glance. J. Cell. Sci. 123, 1819-1823.

Ewen, K.A., Koopman, P., 2010. Mouse germ cell development: from specification to sex determination. Mol. Cell. Endocrinol. 323, 76-93.

Fagoonee, S., Pellicano, R., Silengo, L., Altruda, F., 2011. Potential applications of germline cell-derived pluripotent stem cells in organ regeneration. Organogenesis 7, 116-122.

Frost, R.J., Hamra, F.K., Richardson, J.A., Qi, X., Bassel-Duby, R., Olson, E.N., 2010. MOV10L1 is necessary for protection of spermatocytes against retrotransposons by Piwi-interacting RNAs. Proc. Natl. Acad. Sci. U. S. A. 107, 11847-11852.

Ghildiyal, M., Zamore, P.D., 2009. Small silencing RNAs: an expanding universe. Nat. Rev. Genet. 10, 94-108.

Girard, A., Sachidanandam, R., Hannon, G.J., Carmell, M.A., 2006. A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature 442, 199-202.

Greenlee, A.R., Shiao, M.S., Snyder, E., Buaas, F.W., Gu, T., Stearns, T.M., Sharma, M., Murchison, E.P., Puente, G.C., Braun, R.E., 2012. Deregulated sex chromosome gene expression with male germ cell-specific loss of Dicer1. PLoS One 7, e46359.

Grivna, S.T., Beyret, E., Wang, Z., Lin, H., 2006. A novel class of small RNAs in mouse spermatogenic cells. Genes Dev. 20, 1709-1714.

Guttman, M., Rinn, J.L., 2012. Modular regulatory principles of large non-coding RNAs. Nature 482, 339-346.

Hamatani, T., 2012. Human spermatozoal RNAs. Fertil. Steril. 97, 275-281.

Hayashi, K., Chuva de Sousa Lopes, S.M., Kaneda, M., Tang, F., Hajkova, P., Lao, K., O'Carroll, D., Das, P.P., Tarakhovsky, A., Miska, E.A., Surani, M.A., 2008. MicroRNA biogenesis is required for mouse primordial germ cell development and spermatogenesis. PLoS One 3, e1738.

Hess, R.A., Renato de Franca, L., 2008. Spermatogenesis and cycle of the seminiferous epithelium. Adv. Exp. Med. Biol. 636, 1-15.

Huang, H., Gao, Q., Peng, X., Choi, S.Y., Sarma, K., Ren, H., Morris, A.J., Frohman, M.A., 2011. piRNA-associated germline nuage formation and spermatogenesis require MitoPLD profusogenic mitochondrial-surface lipid signaling. Dev. Cell. 20, 376-387.

Huszar, J.M., Payne, C.J., 2013. MicroRNA 146 (Mir146) modulates spermatogonial differentiation by retinoic acid in mice. Biol. Reprod. 88, 15.

Hutvagner, G., Simard, M.J., 2008. Argonaute proteins: key players in RNA silencing. Nat. Rev. Mol. Cell Biol. 9, 22-32.

Idler, R.K., Yan, W., 2012. Control of messenger RNA fate by RNA-binding proteins: an emphasis on mammalian spermatogenesis. J. Androl. 33, 309-337.

Page 23: Small RNAs in spermatogenesis

22 

 

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.

Kaneko, H., Dridi, S., Tarallo, V., Gelfand, B.D., Fowler, B.J., Cho, W.G., Kleinman, M.E., Ponicsan, S.L., Hauswirth, W.W., Chiodo, V.A., Kariko, K., Yoo, J.W., Lee, D.K., Hadziahmetovic, M., Song, Y., Misra, S., Chaudhuri, G., Buaas, F.W., Braun, R.E., Hinton, D.R., Zhang, Q., Grossniklaus, H.E., Provis, J.M., Madigan, M.C., Milam, A.H., Justice, N.L., Albuquerque, R.J., Blandford, A.D., Bogdanovich, S., Hirano, Y., Witta, J., Fuchs, E., Littman, D.R., Ambati, B.K., Rudin, C.M., Chong, M.M., Provost, P., Kugel, J.F., Goodrich, J.A., Dunaief, J.L., Baffi, J.Z., Ambati, J., 2011. DICER1 deficit induces Alu RNA toxicity in age-related macular degeneration. Nature 471, 325-330.

Kanellopoulou, C., Muljo, S.A., Kung, A.L., Ganesan, S., Drapkin, R., Jenuwein, T., Livingston, D.M., Rajewsky, K., 2005. Dicer-deficient mouse embryonic stem cells are defective in differentiation and centromeric silencing. Genes Dev. 19, 489-501.

Ketting, R.F., 2011. The many faces of RNAi. Dev. Cell. 20, 148-161.

Khraiwesh, B., Arif, M.A., Seumel, G.I., Ossowski, S., Weigel, D., Reski, R., Frank, W., 2010. Transcriptional control of gene expression by microRNAs. Cell 140, 111-122.

Kimmins, S., Kotaja, N., Davidson, I., Sassone-Corsi, P., 2004. Testis-specific transcription mechanisms promoting male germ-cell differentiation. Reproduction 128, 5-12.

Kimmins, S., Sassone-Corsi, P., 2005. Chromatin remodelling and epigenetic features of germ cells. Nature 434, 583-589.

Korhonen, H.M., Meikar, O., Yadav, R.P., Papaioannou, M.D., Romero, Y., Da Ros, M., Herrera, P.L., Toppari, J., Nef, S., Kotaja, N., 2011. Dicer is required for haploid male germ cell differentiation in mice. PLoS One 6, e24821.

Kotaja, N., Kimmins, S., Brancorsini, S., Hentsch, D., Vonesch, J.L., Davidson, I., Parvinen, M., Sassone-Corsi, P., 2004. Preparation, isolation and characterization of stage-specific spermatogenic cells for cellular and molecular analysis. Nat. Methods 1, 249-254.

Kotaja, N., Sassone-Corsi, P., 2007. The chromatoid body: a germ-cell-specific RNA-processing centre. Nat. Rev. Mol. Cell Biol. 8, 85-90.

Krol, J., Loedige, I., Filipowicz, W., 2010. The widespread regulation of microRNA biogenesis, function and decay. Nat. Rev. Genet. 11, 597-610.

Kulkarni, M., Ozgur, S., Stoecklin, G., 2010. On track with P-bodies. Biochem. Soc. Trans. 38, 242-251.

Kuramochi-Miyagawa, S., Kimura, T., Ijiri, T.W., Isobe, T., Asada, N., Fujita, Y., Ikawa, M., Iwai, N., Okabe, M., Deng, W., Lin, H., Matsuda, Y., Nakano, T., 2004. Mili, a mammalian member of piwi family gene, is essential for spermatogenesis. Development 131, 839-849.

Kuramochi-Miyagawa, S., Watanabe, T., Gotoh, K., Totoki, Y., Toyoda, A., Ikawa, M., Asada, N., Kojima, K., Yamaguchi, Y., Ijiri, T.W., Hata, K., Li, E., Matsuda, Y., Kimura, T., Okabe, M., Sakaki, Y., Sasaki, H., Nakano, T., 2008. DNA methylation of retrotransposon genes is

Page 24: Small RNAs in spermatogenesis

23 

 

regulated by Piwi family members MILI and MIWI2 in murine fetal testes. Genes Dev. 22, 908-917.

Lagos-Quintana, M., Rauhut, R., Yalcin, A., Meyer, J., Lendeckel, W., Tuschl, T., 2002. Identification of tissue-specific microRNAs from mouse. Curr. Biol. 12, 735-739.

Lange, U.C., Schneider, R., 2010. What an epigenome remembers. Bioessays 32, 659-668.

Lee, H.C., Gu, W., Shirayama, M., Youngman, E., Conte, D.,Jr, Mello, C.C., 2012. C. elegans piRNAs mediate the genome-wide surveillance of germline transcripts. Cell 150, 78-87.

Lee, K., Haugen, H.S., Clegg, C.H., Braun, R.E., 1995. Premature translation of protamine 1 mRNA causes precocious nuclear condensation and arrests spermatid differentiation in mice. Proc. Natl. Acad. Sci. U. S. A. 92, 12451-12455.

Lee, R.C., Feinbaum, R.L., Ambros, V., 1993. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75, 843-854.

Liang, X., Zhou, D., Wei, C., Luo, H., Liu, J., Fu, R., Cui, S., 2012. MicroRNA-34c enhances murine male germ cell apoptosis through targeting ATF1. PLoS One 7, e33861.

Liu, D., Li, L., Fu, H., Li, S., Li, J., 2012a. Inactivation of Dicer1 has a severe cumulative impact on the formation of mature germ cells in mouse testes. Biochem. Biophys. Res. Commun. 422, 114-120.

Liu, W.M., Pang, R.T., Chiu, P.C., Wong, B.P., Lao, K., Lee, K.F., Yeung, W.S., 2012b. Sperm-borne microRNA-34c is required for the first cleavage division in mouse. Proc. Natl. Acad. Sci. U. S. A. 109, 490-494.

Lu, J., Getz, G., Miska, E.A., Alvarez-Saavedra, E., Lamb, J., Peck, D., Sweet-Cordero, A., Ebert, B.L., Mak, R.H., Ferrando, A.A., Downing, J.R., Jacks, T., Horvitz, H.R., Golub, T.R., 2005. MicroRNA expression profiles classify human cancers. Nature 435, 834-838.

Maatouk, D.M., Loveland, K.L., McManus, M.T., Moore, K., Harfe, B.D., 2008. Dicer1 is required for differentiation of the mouse male germline. Biol. Reprod. 79, 696-703.

Marcon, E., Babak, T., Chua, G., Hughes, T., Moens, P.B., 2008. miRNA and piRNA localization in the male mammalian meiotic nucleus. Chromosome Res. 16, 243-260.

Matsui, Y., 2010. The molecular mechanisms regulating germ cell development and potential. J. Androl. 31, 61-65.

McIver, S.C., Roman, S.D., Nixon, B., McLaughlin, E.A., 2012a. miRNA and mammalian male germ cells. Hum. Reprod. Update 18, 44-59.

McIver, S.C., Stanger, S.J., Santarelli, D.M., Roman, S.D., Nixon, B., McLaughlin, E.A., 2012b. A unique combination of male germ cell miRNAs coordinates gonocyte differentiation. PLoS One 7, e35553.

Meikar, O., Da Ros, M., Korhonen, H., Kotaja, N., 2011. Chromatoid body and small RNAs in male germ cells. Reproduction 142, 195-209.

Page 25: Small RNAs in spermatogenesis

24 

 

Meunier, J., Lemoine, F., Soumillon, M., Liechti, A., Weier, M., Guschanski, K., Hu, H., Khaitovich, P., Kaessmann, H., 2013. Birth and expression evolution of mammalian microRNA genes. Genome Res. 23, 34-45.

Mlotshwa, S., Pruss, G.J., Vance, V., 2008. Small RNAs in viral infection and host defense. Trends Plant Sci. 13, 375-382.

Modzelewski, A.J., Holmes, R.J., Hilz, S., Grimson, A., Cohen, P.E., 2012. AGO4 regulates entry into meiosis and influences silencing of sex chromosomes in the male mouse germline. Dev. Cell. 23, 251-264.

Murchison, E.P., Partridge, J.F., Tam, O.H., Cheloufi, S., Hannon, G.J., 2005. Characterization of Dicer-deficient murine embryonic stem cells. Proc. Natl. Acad. Sci. U. S. A. 102, 12135-12140.

Murchison, E.P., Stein, P., Xuan, Z., Pan, H., Zhang, M.Q., Schultz, R.M., Hannon, G.J., 2007. Critical roles for Dicer in the female germline. Genes Dev. 21, 682-693.

Nishibu, T., Hayashida, Y., Tani, S., Kurono, S., Kojima-Kita, K., Ukekawa, R., Kurokawa, T., Kuramochi-Miyagawa, S., Nakano, T., Inoue, K., Honda, S., 2012. Identification of MIWI-associated Poly(A) RNAs by immunoprecipitation with an anti-MIWI monoclonal antibody. Biosci. Trends 6, 248-261.

Nishimasu, H., Ishizu, H., Saito, K., Fukuhara, S., Kamatani, M.K., Bonnefond, L., Matsumoto, N., Nishizawa, T., Nakanaga, K., Aoki, J., Ishitani, R., Siomi, H., Siomi, M.C., Nureki, O., 2012. Structure and function of Zucchini endoribonuclease in piRNA biogenesis. Nature 491, 284-287.

Niu, Z., Goodyear, S.M., Rao, S., Wu, X., Tobias, J.W., Avarbock, M.R., Brinster, R.L., 2011. MicroRNA-21 regulates the self-renewal of mouse spermatogonial stem cells. Proc. Natl. Acad. Sci. U. S. A. 108, 12740-12745.

Okamura, K., Chung, W.J., Ruby, J.G., Guo, H., Bartel, D.P., Lai, E.C., 2008. The Drosophila hairpin RNA pathway generates endogenous short interfering RNAs. Nature 453, 803-806.

Papaioannou, M.D., Lagarrigue, M., Vejnar, C.E., Rolland, A.D., Kuhne, F., Aubry, F., Schaad, O., Fort, A., Descombes, P., Neerman-Arbez, M., Guillou, F., Zdobnov, E.M., Pineau, C., Nef, S., 2011. Loss of Dicer in Sertoli cells has a major impact on the testicular proteome of mice. Mol. Cell. Proteomics 10, M900587MCP200.

Papaioannou, M.D., Nef, S., 2010. microRNAs in the testis: building up male fertility. J. Androl. 31, 26-33.

Papaioannou, M.D., Pitetti, J.L., Ro, S., Park, C., Aubry, F., Schaad, O., Vejnar, C.E., Kuhne, F., Descombes, P., Zdobnov, E.M., McManus, M.T., Guillou, F., Harfe, B.D., Yan, W., Jegou, B., Nef, S., 2009. Sertoli cell Dicer is essential for spermatogenesis in mice. Dev. Biol. 326, 250-259.

Paronetto, M.P., Sette, C., 2010. Role of RNA-binding proteins in mammalian spermatogenesis. Int. J. Androl. 33, 2-12.

Pratt, A.J., MacRae, I.J., 2009. The RNA-induced silencing complex: a versatile gene-silencing machine. J. Biol. Chem. 284, 17897-17901.

Page 26: Small RNAs in spermatogenesis

25 

 

Rato, L., Alves, M.G., Socorro, S., Duarte, A.I., Cavaco, J.E., Oliveira, P.F., 2012. Metabolic regulation is important for spermatogenesis. Nat. Rev. Urol. 9, 330-338.

Reuter, M., Berninger, P., Chuma, S., Shah, H., Hosokawa, M., Funaya, C., Antony, C., Sachidanandam, R., Pillai, R.S., 2011. Miwi catalysis is required for piRNA amplification-independent LINE1 transposon silencing. Nature 480, 264-267.

Rinn, J.L., Chang, H.Y., 2012. Genome regulation by long noncoding RNAs. Annu. Rev. Biochem. 81, 145-166.

Romero, Y., Meikar, O., Papaioannou, M.D., Conne, B., Grey, C., Weier, M., Pralong, F., De Massy, B., Kaessmann, H., Vassalli, J.D., Kotaja, N., Nef, S., 2011. Dicer1 depletion in male germ cells leads to infertility due to cumulative meiotic and spermiogenic defects. PLoS One 6, e25241.

Rosenfeld, N., Aharonov, R., Meiri, E., Rosenwald, S., Spector, Y., Zepeniuk, M., Benjamin, H., Shabes, N., Tabak, S., Levy, A., Lebanony, D., Goren, Y., Silberschein, E., Targan, N., Ben-Ari, A., Gilad, S., Sion-Vardy, N., Tobar, A., Feinmesser, M., Kharenko, O., Nativ, O., Nass, D., Perelman, M., Yosepovich, A., Shalmon, B., Polak-Charcon, S., Fridman, E., Avniel, A., Bentwich, I., Bentwich, Z., Cohen, D., Chajut, A., Barshack, I., 2008. MicroRNAs accurately identify cancer tissue origin. Nat. Biotechnol. 26, 462-469.

Ruwanpura, S.M., McLachlan, R.I., Meachem, S.J., 2010. Hormonal regulation of male germ cell development. J. Endocrinol. 205, 117-131.

Sasaki, H., Matsui, Y., 2008. Epigenetic events in mammalian germ-cell development: reprogramming and beyond. Nat. Rev. Genet. 9, 129-140.

Sharpe, R.M., 2012. Sperm counts and fertility in men: a rocky road ahead. Science & Society Series on Sex and Science. EMBO Rep. 13, 398-403.

Shirayama, M., Seth, M., Lee, H.C., Gu, W., Ishidate, T., Conte, D.,Jr, Mello, C.C., 2012. piRNAs initiate an epigenetic memory of nonself RNA in the C. elegans germline. Cell 150, 65-77.

Siomi, H., Siomi, M.C., 2010. Posttranscriptional regulation of microRNA biogenesis in animals. Mol. Cell 38, 323-332.

Siomi, M.C., Sato, K., Pezic, D., Aravin, A.A., 2011. PIWI-interacting small RNAs: the vanguard of genome defence. Nat. Rev. Mol. Cell Biol. 12, 246-258.

Skinner, M.K., 2011. Role of epigenetics in developmental biology and transgenerational inheritance. Birth Defects Res. C. Embryo. Today 93, 51-55.

Smorag, L., Zheng, Y., Nolte, J., Zechner, U., Engel, W., Pantakani, D.V., 2012. MicroRNA signature in various cell types of mouse spermatogenesis: evidence for stage-specifically expressed miRNA-221, -203 and -34b-5p mediated spermatogenesis regulation. Biol. Cell. 104, 677-692.

Song, R., Hennig, G.W., Wu, Q., Jose, C., Zheng, H., Yan, W., 2011. Male germ cells express abundant endogenous siRNAs. Proc. Natl. Acad. Sci. U. S. A. 108, 13159-13164.

Page 27: Small RNAs in spermatogenesis

26 

 

Song, R., Ro, S., Michaels, J.D., Park, C., McCarrey, J.R., Yan, W., 2009. Many X-linked microRNAs escape meiotic sex chromosome inactivation. Nat. Genet. 41, 488-493.

Sood, P., Krek, A., Zavolan, M., Macino, G., Rajewsky, N., 2006. Cell-type-specific signatures of microRNAs on target mRNA expression. Proc. Natl. Acad. Sci. U. S. A. 103, 2746-2751.

Suh, N., Baehner, L., Moltzahn, F., Melton, C., Shenoy, A., Chen, J., Blelloch, R., 2010. MicroRNA function is globally suppressed in mouse oocytes and early embryos. Curr. Biol. 20, 271-277.

Tam, O.H., Aravin, A.A., Stein, P., Girard, A., Murchison, E.P., Cheloufi, S., Hodges, E., Anger, M., Sachidanandam, R., Schultz, R.M., Hannon, G.J., 2008. Pseudogene-derived small interfering RNAs regulate gene expression in mouse oocytes. Nature 453, 534-538.

Tong, M.H., Mitchell, D.A., McGowan, S.D., Evanoff, R., Griswold, M.D., 2012. Two miRNA clusters, Mir-17-92 (Mirc1) and Mir-106b-25 (Mirc3), are involved in the regulation of spermatogonial differentiation in mice. Biol. Reprod. 86, 72.

Vourekas, A., Zheng, Q., Alexiou, P., Maragkakis, M., Kirino, Y., Gregory, B.D., Mourelatos, Z., 2012. Mili and Miwi target RNA repertoire reveals piRNA biogenesis and function of Miwi in spermiogenesis. Nat. Struct. Mol. Biol. 19, 773-781.

Wang, Y., Medvid, R., Melton, C., Jaenisch, R., Blelloch, R., 2007. DGCR8 is essential for microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nat. Genet. 39, 380-385.

Watanabe, T., Chuma, S., Yamamoto, Y., Kuramochi-Miyagawa, S., Totoki, Y., Toyoda, A., Hoki, Y., Fujiyama, A., Shibata, T., Sado, T., Noce, T., Nakano, T., Nakatsuji, N., Lin, H., Sasaki, H., 2011. MITOPLD is a mitochondrial protein essential for nuage formation and piRNA biogenesis in the mouse germline. Dev. Cell. 20, 364-375.

Watanabe, T., Takeda, A., Tsukiyama, T., Mise, K., Okuno, T., Sasaki, H., Minami, N., Imai, H., 2006. Identification and characterization of two novel classes of small RNAs in the mouse germline: retrotransposon-derived siRNAs in oocytes and germline small RNAs in testes. Genes Dev. 20, 1732-1743.

Watanabe, T., Totoki, Y., Toyoda, A., Kaneda, M., Kuramochi-Miyagawa, S., Obata, Y., Chiba, H., Kohara, Y., Kono, T., Nakano, T., Surani, M.A., Sakaki, Y., Sasaki, H., 2008. Endogenous siRNAs from naturally formed dsRNAs regulate transcripts in mouse oocytes. Nature 453, 539-543.

Winter, J., Jung, S., Keller, S., Gregory, R.I., Diederichs, S., 2009. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat. Cell Biol. 11, 228-234.

Wu, Q., Song, R., Ortogero, N., Zheng, H., Evanoff, R., Small, C.L., Griswold, M.D., Namekawa, S.H., Royo, H., Turner, J.M., Yan, W., 2012. The RNase III enzyme DROSHA is essential for microRNA production and spermatogenesis. J. Biol. Chem. 287, 25173-25190.

Yan, N., Lu, Y., Sun, H., Tao, D., Zhang, S., Liu, W., Ma, Y., 2007. A microarray for microRNA profiling in mouse testis tissues. Reproduction 134, 73-79.

Page 28: Small RNAs in spermatogenesis

27 

 

Yang, Q.E., Racicot, K.E., Kaucher, A.V., Oatley, M.J., Oatley, J.M., 2013. MicroRNAs 221 and 222 regulate the undifferentiated state in mammalian male germ cells. Development 140, 280-290.

Yu, Z., Raabe, T., Hecht, N.B., 2005. MicroRNA Mirn122a reduces expression of the posttranscriptionally regulated germ cell transition protein 2 (Tnp2) messenger RNA (mRNA) by mRNA cleavage. Biol. Reprod. 73, 427-433.

Zardo, G., Ciolfi, A., Vian, L., Starnes, L.M., Billi, M., Racanicchi, S., Maresca, C., Fazi, F., Travaglini, L., Noguera, N., Mancini, M., Nanni, M., Cimino, G., Lo-Coco, F., Grignani, F., Nervi, C., 2012. Polycombs and microRNA-223 regulate human granulopoiesis by transcriptional control of target gene expression. Blood 119, 4034-4046.

Zheng, K., Wang, P.J., 2012. Blockade of pachytene piRNA biogenesis reveals a novel requirement for maintaining post-meiotic germline genome integrity. PLoS Genet. 8, e1003038.

Zheng, K., Xiol, J., Reuter, M., Eckardt, S., Leu, N.A., McLaughlin, K.J., Stark, A., Sachidanandam, R., Pillai, R.S., Wang, P.J., 2010. Mouse MOV10L1 associates with Piwi proteins and is an essential component of the Piwi-interacting RNA (piRNA) pathway. Proc. Natl. Acad. Sci. U. S. A. 107, 11841-11846.

Page 29: Small RNAs in spermatogenesis

28 

 

Figure captions:

Fig. 1. Male germ cell differentiation. Spermatogenesis is initiated after birth by mitotic

proliferation of spermatogonia that finally enter into the meiotic program and become

spermatocytes. After long meiotic prophase I that includes synaptonemal complex formation,

crossing over, and homologous recombination, spermatocytes undergo two fast divisions (MI

and MII) to produce haploid round spermatids (RS). Post-meiotic differentiation is

characterized by dramatic morphological changes including chromatin compaction by

histone-protamine transition, nuclear reshaping, and acrosome and flagellum formation. The

transcriptional activity is high until elongating spermatids (ES). After histone-protamine

transition, transcription in late ESs is largely silenced and protein production in these cells is

mostly dependent on mRNAs that are transcribed earlier and translationally regulated in RNP

complexes. The timing of Cre-mediated gene deletion in different germ cell-specific Dicer1

and Drosha knockout mouse models discussed in this review are shown by arrows. The

expressions of fetal, pre-pachytene and pachytene piRNAs are also indicated.

Fig. 2. Biosynthesis and functions of Dicer-dependent small RNAs. The nuclear

microprocessor complex that comprises endonuclease Drosha and DGCR8 processes the

nascent hairpin loop primary miRNA (pri-miRNA) transcripts into 70-nucleotide pre-miRNAs

that are transported to the cytoplasm. The pre-miRNAs are further processed by the

endoribonuclease Dicer into 21-25 nucleotide small dsRNAs that subsequently unwind and

associate with RNA-induced silencing complex (RISC). Endo-siRNAs are processed by long

double-stranded RNA precursors and their processing does not require Drosha activity. Small

RNAs mediate sequence-specific binding of RISC complex to their target mRNAs that are

directed either to cleavage or translational regulation. Small RNAs are also known to have

nuclear effects on gene expression at transcriptional level.

Fig. 3. piRNA pathways in murine male germ cells. piRNA precursors are transcribed from

piRNA clusters and transported to the cytoplasm for further processing in the cytoplasmic

germ granules. In fetal male germ cells, primary piRNAs produced by primary processing

pathway are loaded to MILI. The response is amplified by the secondary processing pathway

(ping-pong cycle) including cyclic production of MILI-bound primary piRNAs and MIWI2-

bound secondary piRNAs. This pathway is exploited in fetal prospermatogonia to silence

transposon expression. MIWI2 can also be imported to the nucleus where it controls the

methylation of transposon genes. In meiotic and post-meiotic cells, piRNAs are produced

Page 30: Small RNAs in spermatogenesis

29 

 

only by primary processing pathway and are loaded to MILI and MIWI. The targets and

mechanisms of these post-natal piRNAs are still under investigation.

Page 31: Small RNAs in spermatogenesis

30 

 

Birth

Stra8Cre

Ddx4Cre Ngn3Cre TnapCre

TRANSCRIPTIONAL ACTIVITY

HISTONES

pachytene piRNAs

Prm1Cre

ACTIVE TRANSLATIONAL REGULATIONfetal

piRNAs pre-pachytene piRNAs

PGCs Prospermatogonia Spermatogonia Spermatocyte M-I and M-II RS ES Spermatozoa

PROTAMINES

Page 32: Small RNAs in spermatogenesis

31 

 

Pri-miRNA Pre-miRNA

NUCLEUS

CYTOPLASM

DGCR8 Drosha

endo-siRNAs

Dicer

target mRNA

RISC AGO

RISC

AGO

Translational repressionmRNA cleavage

Pre-miRNA

Transcription

?

Small RNA gene

21-25 nt small RNA

Page 33: Small RNAs in spermatogenesis

32 

 

piRNA loci

NUCLEUS

CYTOPLASM

Primary processing

MIWI2

MOUSE GERM CELL

Transcription

Sense transcripts

MILI

MILI

 MILI MIWI2

 MIWI

 Post-natal

germ cells

Fetal prospermatogonia

Antisense transcripts

Primary piRNAs Secondary piRNAs

Secondary processing

ping-pong cycle

Page 34: Small RNAs in spermatogenesis

33 

 

Table 1. Male germ line-specific Dicer1 and Drosha knockout mouse models.

Mouse model Cre expression Spermatogenic phenotype References

TnapCre-Dicer1

Primordial germ cells (PGCs)

Defects in PGC proliferation and post-natal spermatogenesis.

(Hayashi et al., 2008; Maatouk et al., 2008)

Ddx4Cre-Dicer1

Prospermatogonia (E18)

Meiotic defects, reduced number of haploid cells, drastic abnormalities in head and tail morphology of the remaining spermatozoa.

(Liu et al., 2012a; Romero et al., 2011)

Ngn3Cre-Dicer1 Spermatogonia

No gross abnormalities in spermatogonia and spermatocytes. Major defects in haploid differentiation including chromatin organization, condensation and nuclear shaping of elongating spermatids.

(Korhonen et al., 2011)

Stra8Cre-Dicer1 Spermatogonia

Morphological disruptions of spermatocytes and round spermatids, impaired meiotic progression.

(Wu et al., 2012; Greenlee et al., 2012)

Stra8Cre-Drosha Spermatogonia

Severe morphological disruptions in spermatocytes and round spermatids. Stronger phenotype than for the Stra8Cre-driven deletion of Dicer1 gene.

(Wu et al., 2012)

Prm1Cre-Dicer1 Spermatids

Defects mainly in post-meiotic differentiation, abnormal head morphology.

(Chang et al., 2012)

 

Page 35: Small RNAs in spermatogenesis

34 

 

Small RNAs in spermatogenesis Ram Prakash Yadav and Noora Kotaja

Highlights

Post-transcriptional gene regulation has important role in spermatogenesis

Small RNAs are essential regulators of gene expression

Small RNAs function at post-transcriptional or epigenetic level

Male germ cells express many classes of small RNAs (miRNAs, endo-siRNAs,

piRNAs)

Knockout mouse models have revealed the importance of small RNAs in

spermatogenesis