Emerging clues for regenerative medicine in infertility: an overview for cell sources and results with a sensitizing modifier PRMT5
Review Article

Emerging clues for regenerative medicine in infertility: an overview for cell sources and results with a sensitizing modifier PRMT5

Jiayi Duan1, Qi Li2, Ziwei Li1

1Department of Surgery, The 1st Hospital Affiliated to Kunming Medical School, Kunming 650032, China; 2Department of Dentistry, Renji Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200001, China

Contributions: (I) Conception and design: All authors; (II) Administrative support: Z Li; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Ziwei Li. Department of Surgery, The 1st Hospital Affiliated to Kunming Medical School, Kunming 650032, China. Email: cocolzw@yeah.net.

Abstract: Germ cells are the most important cell type for generation continuation in all sexually reproductive species. Pluripotent stem cells (PSCs) are important cell resources for disease modeling and tissue-specific differentiation in regenerative medicine. Ground-state naïve PSCs share similar transcriptional and epigenetic features with primordial germ cells (PGCs) and PGC like cells can be derived from PSCs in mice. As a critical regulator in PSCs and PGCs, PRMT5 has a versatile role in regulating cell function both in pluripotency and early germline. This review discusses current progress from mammalian PSCs to germ cell differentiation and PRMT5’s important role in PSCs and PGCs, which will help future research and clinical investigation.

Keywords: Protein arginine methyltransferase 5 (PRMT5); pluripotent stem cells (PSCs); germ cells


Received: 04 April 2017; Accepted: 12 May 2017; Published: 07 June 2017.

doi: 10.21037/amj.2017.05.16


Our ancestors and our children are linked by a single, special cell lineage called the germline, “oocytes and sperm”, which are terminally differentiated and haploid cells necessary to fertilize, in order to form offspring. Germ cells are tasked with the role of accurately passing DNA from one generation to the next. In today’s society infertility is an important health concern as it is estimated to affect 10% of the reproductive age population (1). In many cases infertility can be traced to abnormal germ cell development. Therefore, study of the mechanisms within germ cell development is of utmost importance to guide clinical treatment of infertility (2-5).

Due to material limitations, the study of the very early events of human germline formation are inaccessible since germ cells start to form even before pregnancy is realized. Also, the fetal materials for study can only be obtained from elected terminations with approved IRB protocols. Therefore, the majority of studies about early mammalian germline formation are performed using the mouse model, given it is genetically malleable through transgenics or embryonic stem cells (ESCs) and these genetic changes can be passed through the germline (6-10). Very recently, early germline development has been scrutinized using Cynomolgus monkeys (Macaca fascicularis), which shows primate PGCs originate from the nascent amnion (11).

However, in vivo studies of newly specified mouse germ cells are challenging due to the fact that the onset of germline happens at the time of implantation [embryonic day (E) 5.5–E6.25] in mouse embryos (12-20) and approximately 40 germ cells are first specified at E7.5 (13,21-23). Germ cell number does not significantly increase until after the germ cells have entered the gonad at around E11.5 (24-27). In Sasaki et al., Cynomolgus germ cells are characterized from E36–E55 [Carnegie stage (CS) 17–23 and early fetal corresponding to E9.5–E12.5 in mice) (11). As early as E11 in monkeys, fewer than 10 SOX17+/TFAP2C+ cells were found in 3/5 embryos. Given this small number, large-scale biochemical studies that require a significant number of cells are impossible. The murine germ cells during this period from specification at E7.5 to complete colonization of the genital ridges and developmental arrest at E13.5 are referred to as primordial germ cells (PGCs).

In order to overcome this challenge, there are many studies investigating methods of generating PGC like cells or gamete like cells from a pluripotent cell type: ESCs and induced pluripotent stem cells (iPSCs) (28-38):

The efforts for generating functional gametes from ESCs started as early as 2003 by independent investigators from Dr. Hans Scholer’s group (30) and Dr. George Daley’s group (39). Hubner et al. from Scholer group identified a 2-D differentiation method using attached cell cultures with a gcOct4-GFP reporter ESC line, resulting in oocyte-like structure as early as Day (D) 26 and blastocyst-like structure at D 43 (30). At around the same time, Geijsen et al. from Dr. Daley’s group identified a method of differentiating germ cell like cells by formation of embryoid bodies (EBs) using the hanging drop method (39). 30 µL droplets containing 200 cells were plated on the inverted petri-dish lid as hanging drops and collected into non-attachment plates at D 3. After 3–4 more days of differentiation, SSEA1+ cells were sorted from EBs and further differentiated to form haploid male cells, which are able to fertilize oocytes following microinjections, with 50% of injected oocytes progressing to 2-cell stage and 20% progressing to blastocyst stage. These studies indicate that in vitro cell types sharing the feature of gametes could be achieved.

Following these studies, a majority of experiments have been performed in many groups around the world to form germ-cell like cells, either using the 2-D differentiation method, or using the hanging drop method to form EBs (28,29,31-38). Firstly, in order to understand germline development in vitro, investigators studied the timing of when known germline-specific genes become expressed in vivo. Some typical germline markers for gonadal stage differentiation included Tex14, Piwil2, Dazl and Vasa. In Geijsen et al., gonadal germline genes such as Tex14, Piwil2 and Dazl were evaluated in SSEA1+ cells derived from EBs (39). However in Hubner et al., a pregonadal germline marker ckit and a gonadal marker Vasa were both used for sorted gcOct4-GFP+/cKit+ populations, and oocyte markers ZP 1-3 were used for the oocyte-like structures from 2-D differentiation (30). More recently, Wei et al. examined germline specification genes, such as Blimp1 (expression starts at E6.25) and Stella/Dppa3 (expression starts at E7.25), in Stella-GFP+ cells from both adherent differentiation and EB differentiation (38). In conclusion, absent in these studies is a thorough characterization of germline markers which are able to distinguish PGCs by specific stages of development, such as specification, migration and gonadal colonization, to investigate the molecular events within each particular developmental window.

Secondly, most of these studies require ESC lines with a transgenic reporter that is often hard to obtain and create, such as gcOct4-GFP (30), Oct4ΔPE-GFP (29,32), Dazl-GFP(36), Mvh-RFP (31) and Stella-GFP (35,38), or rely on a single surface marker such as SSEA1 (39), which is not exclusively expressed in the germline (40). The limitations of using the transporter lines are: 1-Changing the genetic background of a transporter line to another is difficult, involving derivation of new ESC lines under different genetic backgrounds, which usually takes five generations of mouse breeding. 2-PGC like cells isolating from a particular transporter line are referring to a developmental stage expressing the promoter driving the GFP or RFP, therefore overlooking other stages of PGCs. 3-More than one reporter is often required to stage PGC like cells to endogenous equivalents. Therefore, a differentiation method without the use of transgenic mice, but with more than one marker to pinpoint the correct stage of in vitro PGCs is highly favorable. Significant progress has been made recently by inducing murine (m) ESCs/iPSCs into PGC like cells (mPGCLCs) passing through an intermediate epiblast-like cell (EpiLC) stage. These PGCLCs could contribute to spermatogenesis and oogenesis with an in vivo maturation step and are able to fertilize into live young (4,41). Moreover, human (h) ESCs/iPSCs have been induced into hPGCLCs using similar methods and media (42,43), predicting that human gametes would one day be produced in a dish to cure infertility. However, the first step is to bypass the ethical concerns.

The in vitro model provides large amounts of cell number to enable molecular and biochemical studies that are impossible using the endogenous germline. After the initial identification of crucial targets by the in vitro model, we could then validate roles of key germline modifiers in vivo using transgenic mice. Combining both in vivo and in vitro data together, we are able to unveil the role of essential germline genes in a complete and thorough way, which will provide guidance and reference for future studies to generate functional gametes in vitro, ultimately leading to cure of infertility. The first step of achieving this goal is to explore the knowledge of mammalian germline which provides guidance for induced PGC (iPGC) differentiation in a dish.


Study of the mammalian germ line using mouse models

The study of human germline is limited due to human samples being rare, especially for the earliest stages of PGC development prior to 5 weeks post fertilization. Although non-human primates could be utilized as a new model, studies are extremely rare and difficult to perform due to difficulty in sample preparation and collection besides ethical and spending issues. Therefore, mouse models have served as a favorable tool to study mammalian germline, which allows different genetic manipulation and shares similar traits as human germline.

From the mouse model, we understand that PGC specification is initiated early in embryogenesis at around E6.0 by the expression of the germline master regulator Blimp1/Prdm1 in the proximal epiblast induced by paracrine signals from adjacent extraembryonic ectoderm (ExE) and visceral endoderm (VE), such as Bone Morphogenic Protein (BMP) 2, BMP4 and BMP8b (12,15-20,44,45). By E7.25, PGCs start to express Stella/Dppa3. Together with Blimp1, these are the earliest known markers of founder PGCs (13,46). After specification, PGCs undergo a migratory phase starting at around E8.0 from the allantois through the developing hindgut to finally reach the genital ridge at around E10.5 (23,47). During this migratory phase, epigenetic reprogramming also happens in the germline. After PGCs enter and reside in the gonads, PGCs undergo sex determination as early as E12.5. Later, female germ cells will undergo meiotic arrest at Prophase I, while male germ cells will undergo mitotic arrest.

Germ cell specification

The inductive signals for germ cell specification have been identified using knock out studies (48) and more recently culture experiments with cytokines (3). Shown by genetic knock out studies using the mouse model, BMP signals play essential roles in the generation of tissue-nonspecific alkaline phosphate (TNAP) positive PGCs from the epiblast. BMP4 is expressed in the inner cell mass (ICM), ExE from E5.5, and extraembryonic mesoderm (ExM) during gastrulation. Bmp4-/- embryos completely lacked PGCs and Bmp4+/- embryos had reduced PGC number. Loss of BMP4 in the ExM caused aberrant PGC localization and impaired survival (18,49,50). BMP8b is expressed in ExE from E5.5 and the phenotype with loss of BMP8b resembled loss of BMP4 (20,51). BMP2 is expressed in VE at E6.0–E6.75, with stronger expression in the boundary between ExE and epiblast. Loss of BMP2 resulted in significantly reduced PGC number in both heterozygotes and homozygotes (52,53). Since the downstream part of BMP signaling is mediated and amplified through the mothers against decapentaplegic (SMAD) proteins, similar phenotypes were observed in certain SMAD mutants such as Smad1 (12,17,54), Smad4 (55,56) and Smad5 (15,16,57). Smad1 and Smad5 are ubiquitously expressed in the epiblast and Smad4 is ubiquitously expressed during gastrulation. Smad1-/- and Smad5-/- embryos completely lacked PGCs. Smad4 mutants exhibits severely reduced PGC number. Therefore, the BMP induced cell signaling pathway mediated by the SMADs proteins are of absolute importance to the initiation of the mammalian germline.

In addition to the presence of BMP signaling from neighboring tissue, the proximal epiblast needs to be competent to receive the BMP signaling. Through the more recent embryo culture experiments (3), it is shown that wingless-related MMTV integration site (WNT) 3 is required in the epiblast to become BMP4 responsive and germ cell fate is a direct consequence of BMP4 induction from ExE. BMP4 signal is antagonized by signaling from the anterior visceral endoderm (AVE), which development is restricted by BMP8b at E5.5. With the correct gradient of BMP signaling, WNT signaling and inhibitory signaling, the founder germline is initiated in the proximal epiblast by expressing Blimp1.

To understand PGC specification at a single cell level, Saitou et al. [2002] (46) performed single cell gene expression profiling on individual TNAP positive cells and the neighboring cells in E7.5 embryos. An interferon inducible transmembrane protein, Fragilis (also known as interferon-induced transmembrane protein 3-Ifitm3), was found to mark the onset of germ cell competence induced by BMP4 signaling. From this cluster of cells highly expressing Fragilis, Stella was identified to express by E7.2 and therefore restricting these cells into germ cells. From this screen, the PGC specific gene B-lymphocyte-induced maturation protein-1 (Blimp1) also known as Prdm1 (PRDI-BF1 and RIZ) domain-containing 1, was identified to enable the visualization of germline as early as E6.25 and is required for germline development before E7.5 (13). Blimp1-/- embryos produced a cluster of TNAP positive PGC-like cells with high expression of Homeobox genes HoxB1 and HoxA1, in contrast to control Stella positive PGCs that repressed the somatic Hox genes. Moreover, PGCs were completely absent at E8.5 resulting in infertility of the animals. Another PR domain containing transcriptional regulator, PRDM14 starts to express as early as E6.75, which is downstream of BLIMP1. Loss of PRDM14 leads to reduced PGC number as early as E7.25 and Prdm14-/- male/female animals were both sterile (58). More recently, a transcriptional factor Tcfap2c (also known as AP2, gamma), is shown as another down-stream target of BLIMP1 and starts to express in PGCs from E7.25 to E12.5 (59). In Tcfap2c mutants, PGCs were specified but lost around E8.0. In vitro EB formation using Tcfap2c deficient ESCs indicated that Tcfap2c is required for the expression of germ cell markers such as Nanos3 and Dazl, as well as repression of the somatic genes HoxB1 and HoxA1. Given that Blimp1, Prdm14 and Tcfap2c are among the earliest genes enriched in the germline and loss of either results in a loss of germline in early embryogenesis, these three genes are considered the top three master regulators of the mammalian germline. This is further proved by a recent study that overexpressing the three factors in a cell type derived from PSCs results in generation of functional germline.

Germ cell epigenetics

A dramatic drop of genome-wide DNA methylation (with the DNA methylation at imprinted loci not affected) and remodeling of histone modifications happen in the window between E8.0 to approximately E10.5. At around the same time, PGCs decrease histone H3 lysine 9 dimethylation (H3K9me2) and gain levels of histone H3 lysine 27 tri-methylation (H3K27me3) (25,27). These pre-gonadal epigenetic changes in concomitant with PGC migration are termed Reprogramming Phase I (25,60-64). During the early stages of reprogramming phase I, there is a G2 pause in PGCs from E8.0–E9.0, which is also associated with a transcriptional pause (27). Notably, methylation is maintained at the imprinting control centers (ICCs) of imprinted genes, single copy genes and intracisternal A particle (IAP) elements, despite global loss of methylated cytosine (5mC) by immunofluorescence.

From E10.5–E13.5, DNA methylation at ICCs, single copy genes and IAPs are further removed and the global DNA methylation is further reduced, nearly depleted from the PGC genome. These events are termed Reprogramming Phase II (60,61,63,65-68). Recent studies suggest that the ten-eleven translocation (TET) family proteins play critical roles in mediating active demethylation of the imprinted genes (68,69). Yamaguchi et al. [2012] (68) used paternal Tet1 knockout mice (paternalKO; progenies from Tet1-/- male X wild-type female) to analyze the effect of the paternal TET1 loss on the offspring (68), because Tet1-/- females had meiotic defects. In E9.5 paternalKO embryos, 11–46 out of 81 expressed imprinted genes were dysregulated and the Peg10 differentially methylated region (DMR) remained fully methylated compared to the controls. By performing reduced representative bisulfite sequencing (RRBS) on E13.5 male PGCs and sperm, 7 out of 12 commonly covered DMRs showed significantly enriched hypermethylation in paternalKO relative to control. Besides, Dawlaty et al. [2013] (69) found that Tet1/Tet2 deficiency partially compromised imprinting (69) because in some of the double knock out (DKO) embryos, the imprinting control regions (ICRs) showed aberrant hypermethylation and associated down-regulation of mRNA levels, such as Mest and Peg3. However, the defects of increased DNA methylation are dynamic among Tet1/Tet2 deficient embryos and one possible explanation is that TET3 is compensating for the loss of TET1 and 2. Therefore, demethylation events in Reprogramming Phase II greatly rely on TET family mediated active demethylation, although more detailed experiments need to be performed to investigate the compensation among TET family proteins or whether other mechanism is possible at the same time.


Different states of ESCs in culture

In recent years three PSCs states have been described: primed, naïve and ground state naïve. Primed PSCs are traditionally cultured with medium supplemented with basic fibroblast growth factor (bFGF) and knock out replacer on mouse embryonic fibroblast (MEF) feeder layers and are poised with low expression of lineage differentiation genes. Two examples are human embryonic stem cells (hESCs) and mouse epiblast stem cells (EpiSCs) (70-73) cultured in bFGF. Naïve PSCs refer to mESCs or iPSCs cultured in LIF with Fetal Bovine Serum (FBS) on MEFs. They have the potential to derive into all four germ layers and could generate chimera when injected into blastocysts. Ground state naïve PSCs often refer to mESCs cultured with MEK1 and GSK3 inhibitors (2i) in a chemically defined medium (74-76). ESCs in 2i culture can also differentiate very well both in vivo and in vitro, in terms of forming teratomas and living chimeras. However, the epigenetic features are quite different in 2i cultured ESCs compared to naïve ESCs including genome-wide DNA methylation levels (77-80). The hall marks for ground state pluripotency include driving Oct4 transcription by its distal enhancer, retaining a pre-inactivation X chromosome state in female lines, global reduction in DNA methylation and reduced deposition of H3K27me3 on developmental regulatory gene promoters. Although a lot about naïve and primed pluripotency have been known, the foundational mechanisms that govern ground state pluripotency remain to be fully established, especially when stem cell research is transitioning to the newly defined ground state pluripotency, due to its chemically defined nature and convenience to derivation of clinic related cell types.

Efforts have been spent on generating ground state naïve human ESCs by adding a few compounds into the media including a combination of 2i, FGF and hLIF (81,82). In these studies, naïve-like hESCs could resemble transcriptional profile as human preimplantation embryo (82) and from interspecies chimeras after injection into mouse morula (81). There are other studies reporting that supplementing 2i and FGF could contribute to naïve-like hESC in the presence (83) or absence of hLIF (84). Theunissen et al. compared the previous four reports and identified 2i and FGF are necessary in all media and optimized the media with three more inhibitors (BRAF, ROCK and SRC inhibitors) to facilitate cell expansion plus activin-A (85). Following the naïve-like hESCs, Cynomolgus monkey naïve-like ESCs/iPSCs have been converted to facilitate neural differentiation (86).


ESC as a tool for iPGC in vitro model

Currently Murine ESCs (mESCs) are cultured in two different states, naïve (serum + LIF) and ground state naive (2i+LIF). mESCs are in vitro derived pluripotent cells from the ICM of E3.5 blastocysts isolated from timed pregnant female mice and are able to differentiate into all four germ layers that compose the entire body: ectoderm, mesoderm, endoderm, and the germline (87,88). Previous studies have shown that ESCs cultured in naïve state could differentiate into germ cell like cells expressing germline genes by the method of 2-D differentiation (30,38) or forming EBs (28,29,31,33-39). By comparing these two methods, Wei et al. suggested that Stella-GFP+ cells from the EB method showed closer resemblance to the in vivo PGCs with regard to methylation levels of Peg3 and Igf2r DMRs and germline gene expression levels (38), indicating that 3-D structure is more in favor of in vitro germ cell like cell differentiation. Moreover, Wei et al. was the first to characterize differentiated in vitro PGCs focusing on genes expressed in specification and pre-gonadal stage. Although this study provided many preliminary data to understand in vitro PGCs in terms of specification, Wei et al. seeded ESCs in low-attachment plates to form variable sizes of EBs, utilized a single transgenic reporter line to isolate the PGC like cells and overlooked detailed analysis of global DNA methylation level and epigenetics relative to endogenous pre-gonadal germline. Therefore, more defined studies need to be performed to really characterize an in vitro PGC population that is correspondent to endogenous pre-gonadal germline. To build upon the present research for deriving pre-gonadal PGCs from ESCs, Vincent et al. devised a 3-D differentiation method based on forming EBs in hanging drops (defined starting cell numbers and differentiation period), that is correspondent to E9.5–E10.5 pre-gonadal PGCs in vivo (63,89).

Hayashi et al. invented a method to derive PGC-like cells (PGCLCs) starting from 2i + LIF cultured ground state pluripotent ESCs (4,41,90). To generate PGCLCs, this method first involved a 2-D differentiation to epiblast-like cells (EpiLCs) in the presence of activin A and bFGF, on fibronectin coated 12-well plates starting with 100,000 cells/well. After 2 days of EpiLC induction, EpiLCs were disassociated and plated as floating aggregates in low-cell-binding 96-well plates (2,000 cells/well) in the presence of a cocktail of growth factors and cytokines, BMP4 (500 ng/mL), LIF (1,000 /mL), Stem Cell Factor (SCF, 100 ng/mL), BMP8b (500 ng/mL) and Epidermal Growth Factor (EGF, 50 ng/mL) (90). Initially Hayashi et al. utilized a transgenic reporter ESC line with Blimp1-mVenus and Stella-ECFP (BVSC) and isolated BVSC positive PGCLCs from D 4–6 of PGCLC induction (41). At the end of this paper, the authors identified surface markers CD61 (also named integrin 3) and SSEA1 to isolate the same PGCLC population. Both BVSC+ and CD61+/SSEA1+ PGCLCs are able to further differentiate in reconstituted gonads into haploid gametes (spermatozoa and oocytes), which could be fertilized and generate live young (4,41). This is the first study showing the creation of functional gametes from a pluripotent cell source able to produce live young.

Based on the mPGCLC induction, there are two groups reporting the induction of hPGCLCs from primed hPSCs (believed to mimic an epiblast-like status) on feeders (43) or feeder-free using MEF-conditioned media (91). To explore hPGCLC induction in a chemically defined environment, Sasaki et al. demonstrated robust hPGCLC induction with the cells expressing gonadal transcriptome similar to Cynomolgus monkeys and human gonadal PGCs (42).


PRMT5 is an important protein arginine methyltransferase (PRMT) involved in critical biological processes

Protein arginine methyltransferase 5 (PRMT5) is a type II PRMT that modifies symmetrical di-methylated arginines (SDMA) in glycine and arginine-rich motifs of proteins involved in cancer biology, neurogenesis and reprogramming (92). There are three type II PRMTs that have been described: PRMT5, PRMT7 (93,94) and PRMT10, out of which PRMT5 is the mostly well characterized. PRMT5 modifies a large number of protein substrates, including both cytoplasmic and nuclear proteins (2,78,80,95-105). The cytoplasmic proteins include Sm-class proteins composing the Sm core in spliceosomes (106,107). The nuclear proteins include arginines on tails of multiple histones H2A (2), H3 (96,104) and H4 (2,99) as well as germline RNA binding proteins such as VASA (103), PIWI (101), MILI and MIWI (102,108). Therefore, the molecular function of PRMT5 is implicated in regulating splicing, transcription, RNA biogenesis and transposon repression.

Most interestingly, PRMT5 is required for governing the pluripotency in naïve ESCs cultured in serum + LIF; PRMT5 is required for the Drosophila germline and is essential for mammalian germline. Detailed discussion is as follows:

PRMT5 in Drosophila germline

The Drosophila homolog of Prmt5, dart5, is a grand-childless gene (95). Loss of functional allele of dart5 results in sex-dependent germline phenotypes in flies. In males, homozygous dart5-1 (a mutant allele with piggyBac transposon inserted in exon 2 of dart5) flies are infertile due to the lack of mature spermatocytes although homozygous dart5-1 females are fertile with slightly reduced fecundity. However, when mated with wild-type male flies, the fertilized embryos are devoid of pole cells, thus are completely agametic, due to failure of nuage formation in the pre-fertilized oocyte and pole plasm assembly in fertilized embryo. Also, DART5 [also known as dPRMT5 and Capsuleen (Csul)] was shown to be responsible for SDMA modification of PIWI proteins required for piRNA (also known as rasiRNAs-repeat associated small interfering RNAs) biogenesis in Drosophila (101). Loss of DART5 activity resulted in reduction of piRNA levels and accumulation of retrotransposons in the ovary (101). The mechanism is that DART5 is required for the association between Tud and Aub/AGO3 and loss of DART5 resulted in lower roo piRNA (a subtype of piRNA) loading onto Aub (109). In summary, these data suggests that PRMT5 is involved in the initiation of the germline as well as germline development when piRNA biogenesis is required in Drosophila.

Although mammalian germline forms by signal induction from BMPs, other than the pre-formation in Drosophila, PRMT5 is also indicated to be important in mammalian germline by functioning together with the mammalian germline master regulator BLIMP1 (2).

BLIMP1 and PRMT5 in mammalian germline

BLIMP1 is a transcriptional repressor containing a SET domain and Kruppel-type zinc fingers, which enable its DNA-binding activity (27,110). BLIMP1 is known as a master regulator of terminal B cell differentiation into plasma cells through repression of the mature B-cell program (89,111-116). Global deletion of Blimp1 in mouse results in embryonic lethality by E10.5 (2,117,118) and reduction of Alkaline Phosphatase (AP) positive PGCs at E9.5 in a dose-dependent manner (13,41,117,118). AP positive Blimp1 null PGC-like cells fail to repress somatic lineage genes such as the homeobox genes Hoxa1 and Hoxb1 by single cell PCR (13,26,118). Another study has shown that compared to the somatic neighboring cells, Blimp1 null PGC-like cells fail to repress genes of multiple somatic lineages that are normally repressed in wild-type PGCs, such as genes involved in pattern specification, morphogenesis and DNA methylation, indicating that BLIMP1 functions as a repressor of somatic gene programs to facilitate PGC fate (41,119).

At the same time, Ancelin et al. showed that BLIMP1 physically interacts with PRMT5 (Protein arginine methyltransferase 5) by co-immunoprecipitation (co-IP) in 293T cells. PRMT5 was enriched at a BLIMP1 consensus motif within the Dhx38 locus as a result from chromatin immunoprecipitation (ChIP) followed by PCR using E10.5 mouse genital ridges, although the epigenetic marks directed by PRMT5: symmetrical dimethylation of arginine 3 on histone H2A/H4 (H2A/H4R3sme2), were not enriched in the same locus (2,26). The authors concluded that failure to identify the H2A/H4R3sme2 in the Dhx38 BLIMP1-PRMT5 binding sites was due to low number of PGCs (300 PGCs per embryo at E10.5) in the genital ridges that precludes efficient ChIP analysis. Therefore, the authors turned to an in vitro cell line to address this, over-expression of Myc-Blimp1 in P19 cells (an embryonic carcinoma cell line) down regulated Dhx38, and an increased enrichment of H2A/H4R3sme2 on BLIMP1 targeted consensus sequence was seen in Dhx38 locus (2,120). Taken together, these results led to a model that the interaction of BLIMP1 with PRMT5 results in recruitment to key Blimp1-consensus sequences, resulting in the deposition of H2A/H4R3sme2 at these sites, to repress somatic gene expression and promote PGC development. However, lack of cellular material (around 300 cells/E10.5 genital ridge from data of our lab) restricts efforts to clarify PRMT5’s role in germline regulation. Whether BLIMP1 and PRMT5 physically interact in a pure PGC population remain to be proved. Li et al. has utilized a conditional Cre, Blimp1-Cre to knock out Prmt5 specifically in the germline as early as E6.25, resulting in reduction of germ cell numbers as early as E10.5 and a complete “wipe-out” of germ cells at E13.5 in both male and female mouse embryos (121). The defected germ cells exit cell cycle, undergo apoptosis and arrest in G2/M.

PRMT5 in splicing

PRMT5 is able to modify SmB/B’, SmD1/D3 proteins in Drosophila, mouse cells and human cells (72,97,122). In flies, DART5 (Drosophila Prmt5) and DART7 (Drosophila Prmt7) were both required for methylating SmB and SmD1/D3 proteins (97). However, snRNP assembly in either dart5 or dart7 mutants was unaffected, due to the fact that methylation of Sm proteins is not required for Sm-core assembly.

Different from Drosophila, methylation of the Sm proteins is indeed required for efficient association with the SMN (survival of the motor neurons) complex in both human HeLa cells (122) and murine Neural Progenitor Cells (NPCs) (72). In HeLa cells, both PRMT5 and PRMT7 are required for methylation of the Sm proteins and snRNP assembly. However, whether splicing is affected in Prmt5 or Prmt7 Knock down (KD) HeLa cells remains to be determined. In Bezzi et al. (72) loss of PRMT5 resulted in increased apoptosis and affected homeostasis of NPCs. Prmt5 null NPCs showed differentially spliced events mainly in the categories of retained introns (RI). Among the 300 genes with affected splicing events, the authors focused on alternatively spliced Mdm4 (a P53 inhibitor), because MDM4 was down-regulated after inhibiting splicing machinery (72,123). In Prmt5 null NPCs, Mdm4 transcripts comprised a short unstable form relative to the control, leading to the reduction of full length MDM4 protein. As a result, the repression of P53 pathway by MDM4 was released, leading to increased cell death. However, the phenotype by loss of PRMT5 was only partially rescued with a Trp53-/- background, indicating that a P53 independent pathway for regulating cell survival is present.

PRMT5 in pluripotency

In Tee et al., PRMT5 was shown to safeguard naïve pluripotency in mouse ESCs by modifying R3 of pre-deposited histone H2A (H2AR3sme2) in the cytoplasm (26). Knock down of Prmt5 by shRNA in mESCs cultured in serum showed precocious differentiation, i.e., down regulation of Oct4 and Sox2 and up regulation of somatic genes such as FoxA2, Gata4 and HoxD9. Over expression of a mutated H2A that cannot be methylated at R3 lead to partial resemblance to Prmt5 knock down phenotype (26). In this paper, symmetrical di-methylation of arginine 3 on histone H4 (H4R3sme2) was not reduced in Prmt5 KD naïve ESCs. However, this is possibly due to the non-specificity of the antibodies recognizing unmodified H4, because the same antibody from the same catalog number (Abcam, #ab5823) still detects a band corresponding to H4 in Prmt5 null MEFs (in collaboration with Dr. Mark Bedford). Using a new antibody from Active Motif, a reduction of H4R3sme2 is observed in Prmt5 null MEFs (Dr. Mark Bedford unpublished) and Prmt5 null ESCs cultured in 2i+LIF. Therefore, H4R3sme2 and H2AR3sme2 are possibly both required for repression of somatic genes.

In primed PSCs such as hESCs cultured with bFGF, PRMT5 is not required for pluripotency, but instead for cell proliferation (120). KD of Prmt5 in hESCs resulted in no change of OCT4 and NANOG protein levels. Instead, affected cell proliferation was seen with loss of PRMT5 using competition assays. This was due to induction of P57, resulting in cell cycle arrest in G1/G0 phase. Given that PRMT5 is expressed in all states of pluripotency and the role of PRMT5 in naïve and primed pluripotency is known, it is important to test the function of PRMT5 in ground state pluripotency, therefore the 2i + LIF cultures, to understand the different mechanisms mediated by a common factor in different states of pluripotency. Tee et al. showed that Prmt5-/- blastocysts cannot give rise to ESC outgrowths in serum culture (26).

In Li et al., ground state naïve mESCs lacking PRMT5 also exhibited many defects in pools of RNA transcripts including go terms “RNA processing”, “Cell cycle”, “DNA damage response” and “Epigenetic modification” etc., with compromised Mdm4 splicing acting as a known downstream indicator of aberrant splicing. Moreover, the SDMAs on SmB, SmD1/D3 were missing without PRMT5 (121).


Summary

This review summarizes current trends in progress about making early mammalian germ cells in vitro starting from a PSC type either in mouse or human. The studies within the field have greatly inspired the investigation for gametogenesis in a dish to cure infertility, which is an exacerbating health problem worldwide. PRMT5 is a critical modifier important for and exhibits functional difference in multi-species pluripotent states. And PRMT5 has different functions in fly and mouse germline probably due to evolution. Therefore, it is interesting to investigate PRMT5’s role systematically in mouse and human PSCs under primed, naïve and ground state naive pluripotent states. It is also interesting to investigate PRMT5’s role in the germline from early non-human primate embryos. Given that PSCs and PGCs share intrinsic genetic and epigenetic characteristics, exploration about PRMT5 in core regulation network will not only provide clues for regenerative and reproductive medicine, but also hint the research for cancer stem cell maintenance and clinical treatment for potential type of cancers.


Acknowledgements

Funding: This work was supported by the NNSFC (National Natural Science Foundation of China), grant No. 31501187.


Footnote

Conflicts of Interest: The authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/amj.2017.05.16). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Chandra A, Copen CE, Stephen EH. Infertility and impaired fecundity in the United States, 1982-2010: data from the National Survey of Family Growth. Natl Health Stat Report 2013:1-18, 1 p following 9.
  2. Ancelin K, Lange UC, Hajkova P, et al. Blimp1 associates with Prmt5 and directs histone arginine methylation in mouse germ cells. Nat Cell Biol 2006;8:623-30. [Crossref] [PubMed]
  3. Ohinata Y, Ohta H, Shigeta M, et al. A signaling principle for the specification of the germ cell lineage in mice. Cell 2009;137:571-84. [Crossref] [PubMed]
  4. Hayashi K, Ogushi S, Kurimoto K, et al. Offspring from oocytes derived from in vitro primordial germ cell-like cells in mice. Science 2012;338:971-5. [Crossref] [PubMed]
  5. Reijo R, Seligman J, Dinulos MB, et al. Mouse autosomal homolog of DAZ, a candidate male sterility gene in humans, is expressed in male germ cells before and after puberty. Genomics 1996;35:346-52. [Crossref] [PubMed]
  6. Hackett JA, Zylicz JJ, Surani MA. Parallel mechanisms of epigenetic reprogramming in the germline. Trends Genet 2012;28:164-74. [Crossref] [PubMed]
  7. Skarnes WC, Rosen B, West AP, et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 2011;474:337-42. [Crossref] [PubMed]
  8. Leitch HG, Tang WW, Surani MA. Primordial germ-cell development and epigenetic reprogramming in mammals. Curr Top Dev Biol 2013;104:149-87. [Crossref] [PubMed]
  9. Seisenberger S, Peat JR, Reik W. Conceptual links between DNA methylation reprogramming in the early embryo and primordial germ cells. Curr Opin Cell Biol 2013;25:281-8. [Crossref] [PubMed]
  10. Saitou M, Yamaji M. Primordial germ cells in mice. Cold Spring Harb Perspect Biol 2012;4. [PubMed]
  11. Sasaki K, Nakamura T, Okamoto I, et al. The Germ Cell Fate of Cynomolgus Monkeys Is Specified in the Nascent Amnion. Dev Cell 2016;39:169-85. [Crossref] [PubMed]
  12. Tremblay KD, Dunn NR, Robertson EJ. Mouse embryos lacking Smad1 signals display defects in extra-embryonic tissues and germ cell formation. Development 2001;128:3609-21. [PubMed]
  13. Ohinata Y, Payer B, O'Carroll D, et al. Blimp1 is a critical determinant of the germ cell lineage in mice. Nature 2005;436:207-13. [Crossref] [PubMed]
  14. Sadaie M, Salama R, Carroll T, et al. Redistribution of the Lamin B1 genomic binding profile affects rearrangement of heterochromatic domains and SAHF formation during senescence. Genes Dev 2013;27:1800-8. [Crossref] [PubMed]
  15. Arnold SJ, Maretto S, Islam A, et al. Dose-dependent Smad1, Smad5 and Smad8 signaling in the early mouse embryo. Dev Biol 2006;296:104-18. [Crossref] [PubMed]
  16. Chang H, Matzuk MM. Smad5 is required for mouse primordial germ cell development. Mech Dev 2001;104:61-7. [Crossref] [PubMed]
  17. Hayashi K, Kobayashi T, Umino T, et al. SMAD1 signaling is critical for initial commitment of germ cell lineage from mouse epiblast. Mech Dev 2002;118:99-109. [Crossref] [PubMed]
  18. Lawson KA, Dunn NR, Roelen BA, et al. Bmp4 is required for the generation of primordial germ cells in the mouse embryo. Genes Dev 1999;13:424-36. [Crossref] [PubMed]
  19. Ying Y, Qi X, Zhao GQ. Induction of primordial germ cells from murine epiblasts by synergistic action of BMP4 and BMP8B signaling pathways. Proc Natl Acad Sci U S A 2001;98:7858-62. [Crossref] [PubMed]
  20. Ying Y, Liu XM, Marble A, et al. Requirement of Bmp8b for the generation of primordial germ cells in the mouse. Mol Endocrinol 2000;14:1053-63. [Crossref] [PubMed]
  21. Mah LJ, El-Osta A, Karagiannis TC. gammaH2AX: a sensitive molecular marker of DNA damage and repair. Leukemia 2010;24:679-86. [Crossref] [PubMed]
  22. Ginsburg M, Snow MH, McLaren A. Primordial germ cells in the mouse embryo during gastrulation. Development 1990;110:521-8. [PubMed]
  23. Saitou M, Kagiwada S, Kurimoto K. Epigenetic reprogramming in mouse pre-implantation development and primordial germ cells. Development 2012;139:15-31. [Crossref] [PubMed]
  24. Tam PP, Snow MH. Proliferation and migration of primordial germ cells during compensatory growth in mouse embryos. J Embryol Exp Morphol 1981;64:133-47. [PubMed]
  25. Seki Y, Hayashi K, Itoh K, et al. Extensive and orderly reprogramming of genome-wide chromatin modifications associated with specification and early development of germ cells in mice. Dev Biol 2005;278:440-58. [Crossref] [PubMed]
  26. Tee WW, Pardo M, Theunissen TW, et al. Prmt5 is essential for early mouse development and acts in the cytoplasm to maintain ES cell pluripotency. Genes Dev 2010;24:2772-7. [Crossref] [PubMed]
  27. Seki Y, Yamaji M, Yabuta Y, et al. Cellular dynamics associated with the genome-wide epigenetic reprogramming in migrating primordial germ cells in mice. Development 2007;134:2627-38. [Crossref] [PubMed]
  28. Toyooka Y, Tsunekawa N, Akasu R, et al. Embryonic stem cells can form germ cells in vitro. Proc Natl Acad Sci U S A 2003;100:11457-62. [Crossref] [PubMed]
  29. Haston KM, Tung JY, Reijo Pera RA. Dazl functions in maintenance of pluripotency and genetic and epigenetic programs of differentiation in mouse primordial germ cells in vivo and in vitro. PLoS One 2009;4:e5654. [Crossref] [PubMed]
  30. Hübner K, Fuhrmann G, Christenson LK, et al. Derivation of oocytes from mouse embryonic stem cells. Science 2003;300:1251-6. [Crossref] [PubMed]
  31. Imamura M, Aoi T, Tokumasu A, et al. Induction of primordial germ cells from mouse induced pluripotent stem cells derived from adult hepatocytes. Mol Reprod Dev 2010;77:802-11. [Crossref] [PubMed]
  32. Sabour D, Arauzo-Bravo MJ, Hubner K, et al. Identification of genes specific to mouse primordial germ cells through dynamic global gene expression. Hum Mol Genet 2011;20:115-25. [Crossref] [PubMed]
  33. Young JC, Dias VL, Loveland KL. Defining the window of germline genesis in vitro from murine embryonic stem cells. Biol Reprod 2010;82:390-401. [Crossref] [PubMed]
  34. Payer B, Chuva de Sousa Lopes SM, Barton SC, et al. Generation of stella-GFP transgenic mice: a novel tool to study germ cell development. Genesis 2006;44:75-83. [Crossref] [PubMed]
  35. West JA, Viswanathan SR, Yabuuchi A, et al. A role for Lin28 in primordial germ-cell development and germ-cell malignancy. Nature 2009;460:909-13. [PubMed]
  36. Nicholas CR, Xu EY, Banani SF, et al. Characterization of a Dazl-GFP germ cell-specific reporter. Genesis 2009;47:74-84. [Crossref] [PubMed]
  37. Mizuno K, Tokumasu A, Nakamura A, et al. Genes associated with the formation of germ cells from embryonic stem cells in cultures containing different glucose concentrations. Mol Reprod Dev 2006;73:437-45. [Crossref] [PubMed]
  38. Wei W, Qing T, Ye X, et al. Primordial germ cell specification from embryonic stem cells. PLoS One 2008;3:e4013. [Crossref] [PubMed]
  39. Geijsen N, Horoschak M, Kim K, et al. Derivation of embryonic germ cells and male gametes from embryonic stem cells. Nature 2004;427:148-54. [Crossref] [PubMed]
  40. Anjos-Afonso F, Bonnet D. Nonhematopoietic/endothelial SSEA-1+ cells define the most primitive progenitors in the adult murine bone marrow mesenchymal compartment. Blood 2007;109:1298-306. [Crossref] [PubMed]
  41. Hayashi K, Ohta H, Kurimoto K, et al. Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 2011;146:519-32. [Crossref] [PubMed]
  42. Sasaki K, Yokobayashi S, Nakamura T, et al. Robust In Vitro Induction of Human Germ Cell Fate from Pluripotent Stem Cells. Cell Stem Cell 2015;17:178-94. [Crossref] [PubMed]
  43. Irie N, Weinberger L, Tang WW, et al. SOX17 is a critical specifier of human primordial germ cell fate. Cell 2015;160:253-68. [Crossref] [PubMed]
  44. Chapman JR, Barral P, Vannier JB, et al. RIF1 is essential for 53BP1-dependent nonhomologous end joining and suppression of DNA double-strand break resection. Mol Cell 2013;49:858-71. [Crossref] [PubMed]
  45. Buonomo SB, Wu Y, Ferguson D, et al. Mammalian Rif1 contributes to replication stress survival and homology-directed repair. J Cell Biol 2009;187:385-98. [Crossref] [PubMed]
  46. Saitou M, Barton SC, Surani MA. A molecular programme for the specification of germ cell fate in mice. Nature 2002;418:293-300. [Crossref] [PubMed]
  47. Saitou M. Germ cell specification in mice. Curr Opin Genet Dev 2009;19:386-95. [Crossref] [PubMed]
  48. Saitou M. Specification of the germ cell lineage in mice. Front Biosci (Landmark Ed) 2009;14:1068-87. [Crossref] [PubMed]
  49. Winnier G, Blessing M, Labosky PA, et al. Bone morphogenetic protein-4 is required for mesoderm formation and patterning in the mouse. Genes Dev 1995;9:2105-16. [Crossref] [PubMed]
  50. Fujiwara T, Dunn NR, Hogan BL. Bone morphogenetic protein 4 in the extraembryonic mesoderm is required for allantois development and the localization and survival of primordial germ cells in the mouse. Proc Natl Acad Sci U S A 2001;98:13739-44. [Crossref] [PubMed]
  51. Zhao GQ, Deng K, Labosky PA, et al. The gene encoding bone morphogenetic protein 8B is required for the initiation and maintenance of spermatogenesis in the mouse. Genes Dev 1996;10:1657-69. [Crossref] [PubMed]
  52. Ying Y, Zhao GQ. Cooperation of endoderm-derived BMP2 and extraembryonic ectoderm-derived BMP4 in primordial germ cell generation in the mouse. Dev Biol 2001;232:484-92. [Crossref] [PubMed]
  53. Zhang H, Bradley A. Mice deficient for BMP2 are nonviable and have defects in amnion/chorion and cardiac development. Development 1996;122:2977-86. [PubMed]
  54. Aubin J, Davy A, Soriano P. In vivo convergence of BMP and MAPK signaling pathways: impact of differential Smad1 phosphorylation on development and homeostasis. Genes Dev 2004;18:1482-94. [Crossref] [PubMed]
  55. Yang X, Li C, Xu X, et al. The tumor suppressor SMAD4/DPC4 is essential for epiblast proliferation and mesoderm induction in mice. Proc Natl Acad Sci U S A 1998;95:3667-72. [Crossref] [PubMed]
  56. Sirard C, de la Pompa JL, Elia A, et al. The tumor suppressor gene Smad4/Dpc4 is required for gastrulation and later for anterior development of the mouse embryo. Genes Dev 1998;12:107-19. [Crossref] [PubMed]
  57. Chang H, Huylebroeck D, Verschueren K, et al. Smad5 knockout mice die at mid-gestation due to multiple embryonic and extraembryonic defects. Development 1999;126:1631-42. [PubMed]
  58. Yamaji M, Seki Y, Kurimoto K, et al. Critical function of Prdm14 for the establishment of the germ cell lineage in mice. Nat Genet 2008;40:1016-22. [Crossref] [PubMed]
  59. Weber S, Eckert D, Nettersheim D, et al. Critical function of AP-2 gamma/TCFAP2C in mouse embryonic germ cell maintenance. Biol Reprod 2010;82:214-23. [Crossref] [PubMed]
  60. Guibert S, Forne T, Weber M. Global profiling of DNA methylation erasure in mouse primordial germ cells. Genome Res 2012;22:633-41. [Crossref] [PubMed]
  61. Hajkova P, Erhardt S, Lane N, et al. Epigenetic reprogramming in mouse primordial germ cells. Mech Dev 2002;117:15-23. [Crossref] [PubMed]
  62. Lane N, Dean W, Erhardt S, et al. Resistance of IAPs to methylation reprogramming may provide a mechanism for epigenetic inheritance in the mouse. Genesis 2003;35:88-93. [Crossref] [PubMed]
  63. Vincent JJ, Huang Y, Chen PY, et al. Stage-specific roles for tet1 and tet2 in DNA demethylation in primordial germ cells. Cell Stem Cell 2013;12:470-8. [Crossref] [PubMed]
  64. Lees-Murdock DJ, De Felici M, Walsh CP. Methylation dynamics of repetitive DNA elements in the mouse germ cell lineage. Genomics 2003;82:230-7. [Crossref] [PubMed]
  65. Hajkova P, Ancelin K, Waldmann T, et al. Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature 2008;452:877-81. [Crossref] [PubMed]
  66. Hajkova P, Jeffries SJ, Lee C, et al. Genome-wide reprogramming in the mouse germ line entails the base excision repair pathway. Science 2010;329:78-82. [Crossref] [PubMed]
  67. Popp C, Dean W, Feng S, et al. Genome-wide erasure of DNA methylation in mouse primordial germ cells is affected by AID deficiency. Nature 2010;463:1101-5. [Crossref] [PubMed]
  68. Yamaguchi S, Hong K, Liu R, et al. Tet1 controls meiosis by regulating meiotic gene expression. Nature 2012;492:443-7. [Crossref] [PubMed]
  69. Dawlaty MM, Breiling A, Le T, et al. Combined deficiency of Tet1 and Tet2 causes epigenetic abnormalities but is compatible with postnatal development. Dev Cell 2013;24:310-23. [Crossref] [PubMed]
  70. Tesar PJ, Chenoweth JG, Brook FA, et al. New cell lines from mouse epiblast share defining features with human embryonic stem cells. Nature 2007;448:196-9. [Crossref] [PubMed]
  71. Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145-7. [Crossref] [PubMed]
  72. Bezzi M, Teo SX, Muller J, et al. Regulation of constitutive and alternative splicing by PRMT5 reveals a role for Mdm4 pre-mRNA in sensing defects in the spliceosomal machinery. Genes Dev 2013;27:1903-16. [Crossref] [PubMed]
  73. Brons IG, Smithers LE, Trotter MW, et al. Derivation of pluripotent epiblast stem cells from mammalian embryos. Nature 2007;448:191-5. [Crossref] [PubMed]
  74. Ying QL, Wray J, Nichols J, et al. The ground state of embryonic stem cell self-renewal. Nature 2008;453:519-23. [Crossref] [PubMed]
  75. Legerski RJ. The Pso4 complex splices into the DNA damage response. Cell Cycle 2009;8:3448-9. [Crossref] [PubMed]
  76. Chen PH, Lee CI, Weng YT, et al. BCAS2 is essential for Drosophila viability and functions in pre-mRNA splicing. RNA 2013;19:208-18. [Crossref] [PubMed]
  77. Marks H, Kalkan T, Menafra R, et al. The transcriptional and epigenomic foundations of ground state pluripotency. Cell 2012;149:590-604. [Crossref] [PubMed]
  78. Ficz G, Hore TA, Santos F, et al. FGF signaling inhibition in ESCs drives rapid genome-wide demethylation to the epigenetic ground state of pluripotency. Cell Stem Cell 2013;13:351-9. [Crossref] [PubMed]
  79. Wan L, Huang J. The PSO4 protein complex associates with replication protein A (RPA) and modulates the activation of ataxia telangiectasia-mutated and Rad3-related (ATR). J Biol Chem 2014;289:6619-26. [Crossref] [PubMed]
  80. Leitch HG, McEwen KR, Turp A, et al. Naive pluripotency is associated with global DNA hypomethylation. Nat Struct Mol Biol 2013;20:311-6. [Crossref] [PubMed]
  81. Gafni O, Weinberger L, Mansour AA, et al. Derivation of novel human ground state naive pluripotent stem cells. Nature 2013;504:282-6. [Crossref] [PubMed]
  82. Chan YS, Goke J, Ng JH, et al. Induction of a human pluripotent state with distinct regulatory circuitry that resembles preimplantation epiblast. Cell Stem Cell 2013;13:663-75. [Crossref] [PubMed]
  83. Valamehr B, Robinson M, Abujarour R, et al. Platform for induction and maintenance of transgene-free hiPSCs resembling ground state pluripotent stem cells. Stem Cell Reports 2014;2:366-81. [Crossref] [PubMed]
  84. Ware CB, Nelson AM, Mecham B, et al. Derivation of naive human embryonic stem cells. Proc Natl Acad Sci U S A 2014;111:4484-9. [Crossref] [PubMed]
  85. Theunissen TW, Powell BE, Wang H, et al. Systematic identification of culture conditions for induction and maintenance of naive human pluripotency. Cell Stem Cell 2014;15:471-87. [Crossref] [PubMed]
  86. Honda A, Kawano Y, Izu H, et al. Discrimination of Stem Cell Status after Subjecting Cynomolgus Monkey Pluripotent Stem Cells to Naive Conversion. Sci Rep 2017;7:45285. [Crossref] [PubMed]
  87. Bradley A, Evans M, Kaufman MH, et al. Formation of germ-line chimaeras from embryo-derived teratocarcinoma cell lines. Nature 1984;309:255-6. [Crossref] [PubMed]
  88. Baribault H, Kemler R. Embryonic stem cell culture and gene targeting in transgenic mice. Mol Biol Med 1989;6:481-92. [PubMed]
  89. Vincent JJ, Li Z, Lee SA, et al. Single cell analysis facilitates staging of Blimp1-dependent primordial germ cells derived from mouse embryonic stem cells. PLoS One 2011;6:e28960. [Crossref] [PubMed]
  90. Hayashi K, Saitou M. Generation of eggs from mouse embryonic stem cells and induced pluripotent stem cells. Nat Protoc 2013;8:1513-24. [Crossref] [PubMed]
  91. Sugawa F, Arauzo-Bravo MJ, Yoon J, et al. Human primordial germ cell commitment in vitro associates with a unique PRDM14 expression profile. EMBO J 2015;34:1009-24. [Crossref] [PubMed]
  92. Karkhanis V, Hu YJ, Baiocchi RA, et al. Versatility of PRMT5-induced methylation in growth control and development. Trends Biochem Sci 2011;36:633-41. [Crossref] [PubMed]
  93. Jelinic P, Stehle JC, Shaw P. The testis-specific factor CTCFL cooperates with the protein methyltransferase PRMT7 in H19 imprinting control region methylation. PLoS Biol 2006;4:e355. [Crossref] [PubMed]
  94. Lee JH, Cook JR, Yang ZH, et al. PRMT7, a new protein arginine methyltransferase that synthesizes symmetric dimethylarginine. J Biol Chem 2005;280:3656-64. [Crossref] [PubMed]
  95. Gonsalvez GB, Rajendra TK, Tian L, et al. The Sm-protein methyltransferase, dart5, is essential for germ-cell specification and maintenance. Curr Biol 2006;16:1077-89. [Crossref] [PubMed]
  96. Pal S, Vishwanath SN, Erdjument-Bromage H, et al. Human SWI/SNF-associated PRMT5 methylates histone H3 arginine 8 and negatively regulates expression of ST7 and NM23 tumor suppressor genes. Mol Cell Biol 2004;24:9630-45. [Crossref] [PubMed]
  97. Gonsalvez GB, Praveen K, Hicks AJ, et al. Sm protein methylation is dispensable for snRNP assembly in Drosophila melanogaster. RNA 2008;14:878-87. [Crossref] [PubMed]
  98. Jansson M, Durant ST, Cho EC, et al. Arginine methylation regulates the p53 response. Nat Cell Biol 2008;10:1431-9. [Crossref] [PubMed]
  99. Zhao Q, Rank G, Tan YT, et al. PRMT5-mediated methylation of histone H4R3 recruits DNMT3A, coupling histone and DNA methylation in gene silencing. Nat Struct Mol Biol 2009;16:304-11. [Crossref] [PubMed]
  100. Anne J, Ollo R, Ephrussi A, et al. Arginine methyltransferase Capsuleen is essential for methylation of spliceosomal Sm proteins and germ cell formation in Drosophila. Development 2007;134:137-46. [Crossref] [PubMed]
  101. Kirino Y, Kim N, de Planell-Saguer M, et al. Arginine methylation of Piwi proteins catalysed by dPRMT5 is required for Ago3 and Aub stability. Nat Cell Biol 2009;11:652-8. [Crossref] [PubMed]
  102. Vagin VV, Wohlschlegel J, Qu J, et al. Proteomic analysis of murine Piwi proteins reveals a role for arginine methylation in specifying interaction with Tudor family members. Genes Dev 2009;23:1749-62. [Crossref] [PubMed]
  103. Kirino Y, Vourekas A, Kim N, et al. Arginine methylation of vasa protein is conserved across phyla. J Biol Chem 2010;285:8148-54. [Crossref] [PubMed]
  104. Migliori V, Muller J, Phalke S, et al. Symmetric dimethylation of H3R2 is a newly identified histone mark that supports euchromatin maintenance. Nat Struct Mol Biol 2012;19:136-44. [Crossref] [PubMed]
  105. Cho EC, Zheng S, Munro S, et al. Arginine methylation controls growth regulation by E2F-1. EMBO J 2012;31:1785-97. [Crossref] [PubMed]
  106. Rho J, Choi S, Seong YR, et al. Prmt5, which forms distinct homo-oligomers, is a member of the protein-arginine methyltransferase family. J Biol Chem 2001;276:11393-401. [Crossref] [PubMed]
  107. Branscombe TL, Frankel A, Lee JH, et al. PRMT5 (Janus kinase-binding protein 1) catalyzes the formation of symmetric dimethylarginine residues in proteins. J Biol Chem 2001;276:32971-6. [Crossref] [PubMed]
  108. Chen C, Jin J, James DA, et al. Mouse Piwi interactome identifies binding mechanism of Tdrkh Tudor domain to arginine methylated Miwi. Proc Natl Acad Sci U S A 2009;106:20336-41. [Crossref] [PubMed]
  109. Nishida KM, Okada TN, Kawamura T, et al. Functional involvement of Tudor and dPRMT5 in the piRNA processing pathway in Drosophila germlines. EMBO J 2009;28:3820-31. [Crossref] [PubMed]
  110. Keller AD, Maniatis T. Identification and characterization of a novel repressor of beta-interferon gene expression. Genes Dev 1991;5:868-79. [Crossref] [PubMed]
  111. Toyooka Y, Tsunekawa N, Takahashi Y, et al. Expression and intracellular localization of mouse Vasa-homologue protein during germ cell development. Mech Dev 2000;93:139-49. [Crossref] [PubMed]
  112. John SA, Clements JL, Russell LM, et al. Ets-1 regulates plasma cell differentiation by interfering with the activity of the transcription factor Blimp-1. J Biol Chem 2008;283:951-62. [Crossref] [PubMed]
  113. Kuo TC, Calame KL. B lymphocyte-induced maturation protein (Blimp)-1, IFN regulatory factor (IRF)-1, and IRF-2 can bind to the same regulatory sites. J Immunol 2004;173:5556-63. [Crossref] [PubMed]
  114. Shaffer AL, Lin KI, Kuo TC, et al. Blimp-1 orchestrates plasma cell differentiation by extinguishing the mature B cell gene expression program. Immunity 2002;17:51-62. [Crossref] [PubMed]
  115. Shapiro-Shelef M, Lin KI, McHeyzer-Williams LJ, et al. Blimp-1 is required for the formation of immunoglobulin secreting plasma cells and pre-plasma memory B cells. Immunity 2003;19:607-20. [Crossref] [PubMed]
  116. Piskurich JF, Lin KI, Lin Y, et al. BLIMP-I mediates extinction of major histocompatibility class II transactivator expression in plasma cells. Nat Immunol 2000;1:526-32. [Crossref] [PubMed]
  117. Robertson EJ, Charatsi I, Joyner CJ, et al. Blimp1 regulates development of the posterior forelimb, caudal pharyngeal arches, heart and sensory vibrissae in mice. Development 2007;134:4335-45. [Crossref] [PubMed]
  118. Vincent SD, Dunn NR, Sciammas R, et al. The zinc finger transcriptional repressor Blimp1/Prdm1 is dispensable for early axis formation but is required for specification of primordial germ cells in the mouse. Development 2005;132:1315-25. [Crossref] [PubMed]
  119. Kurimoto K, Yabuta Y, Ohinata Y, et al. Complex genome-wide transcription dynamics orchestrated by Blimp1 for the specification of the germ cell lineage in mice. Genes Dev 2008;22:1617-35. [Crossref] [PubMed]
  120. Gkountela S, Li Z, Chin CJ, et al. PRMT5 is required for human embryonic stem cell proliferation but not pluripotency. Stem Cell Rev 2014;10:230-9. [Crossref] [PubMed]
  121. Li Z, Yu J, Hosohama L, et al. The Sm protein methyltransferase PRMT5 is not required for primordial germ cell specification in mice. EMBO J 2015;34:748-58. [Crossref] [PubMed]
  122. Gonsalvez GB, Tian L, Ospina JK, et al. Two distinct arginine methyltransferases are required for biogenesis of Sm-class ribonucleoproteins. J Cell Biol 2007;178:733-40. [Crossref] [PubMed]
  123. Allende-Vega N, Dayal S, Agarwala U, et al. p53 is activated in response to disruption of the pre-mRNA splicing machinery. Oncogene 2013;32:1-14. [Crossref] [PubMed]
doi: 10.21037/amj.2017.05.16
Cite this article as: Duan J, Li Q, Li Z. Emerging clues for regenerative medicine in infertility: an overview for cell sources and results with a sensitizing modifier PRMT5. AME Med J 2017;2:70.

Download Citation