Open access peer-reviewed chapter

Primordial Germ Cell Reprogramming

Written By

Maria P. De Miguel, Yago Alcaina and Diego Sainz de la Maza

Submitted: 31 October 2016 Reviewed: 31 May 2017 Published: 20 December 2017

DOI: 10.5772/intechopen.69965

From the Edited Volume

Germ Cell

Edited by Ahmed RG

Chapter metrics overview

1,719 Chapter Downloads

View Full Metrics

Abstract

Primordial germ cells (PGCs) are the embryonic precursors of the gametes. Thus, they are unipotent cells. However, PGCs share some common features with pluripotent stem cells. Among them, PGCs show alkaline phosphatase activity and express stage-specific embryonic antigens and pluripotency factors Lin28, Oct4, Sox2, and Nanog. Under specific conditions, they undergo spontaneous reprogramming in vivo. Moreover, they can be easily reprogrammed in vitro into pluripotent embryonic germ cells (EGCs) by culturing them in the presence of basic fibroblast growth factor or the epigenetic modulator trichostatin A. Previous work in our laboratory has also proven that hypoxia alone can reprogram PGCs into hypoxia-induced embryonic germ-like cells, which have a pluripotent phenotype but which do not show self-renewal capacity. Therefore, PGCs are an interesting model to further comprehensively understand the process of cell reprogramming. This chapter reviews various methods to achieve PGC reprogramming, as well as the molecular pathways involved. We focus on soluble factors and genetic strategies to obtain pluripotent cells from PGCs. Special emphasis will be given to factors implied in energetic metabolism, epigenetics, and cell signaling transduction, both in vitro and in vivo.

Keywords

  • cellular reprogramming
  • ROS
  • glycolysis
  • autophagy
  • primordial germ cells
  • pluripotency
  • metabolism
  • hypoxia
  • epigenetics

1. Introduction

During normal embryogenesis and throughout the life of an organism, cells maintain or restrain their developmental potential. This potential refers to the ability to give rise to various types of cells. After fertilization, the resulting zygote and the blastomeres are totipotent until the four-cell stage, meaning they can develop a complete organism alone, including extraembryonic tissues such as the placenta [1]. Pluripotent stem cells, like embryonic stem cells (ESCs), are defined by their potential to differentiate into cells of the three germ layers (endoderm, mesoderm, and ectoderm), but they do not give rise to extraembryonic structures. In a more differentiated state, we find multipotent cells capable of differentiating into several types of cells. This capability is common in progenitor cells in adults, which give rise to diverse tissue cells.

In some processes, differentiated cells revert to a less differentiated and higher potential state. These phenomena are called reprogramming and are shown both in vivo and in vitro. In vivo reprogramming can be demonstrated by the appearance of cancer stem cells, and in vitro reprogramming has been achieved by several methods, such as somatic cell nuclear transfer, cell fusion between somatic and pluripotent cells, and treatment with pluripotent cell extracts, among others [2].

The most important and well-studied method of in vitro reprogramming in the last decade has been the derivation of induced pluripotent stem cells (iPSCs) from somatic cells by transduction of specific transcription factors, Oct4, Sox2, Klf4, and c-Myc [3]. These cells represented a revolution in stem cell research because they eliminated the ethical concerns about the use of ESCs and allowed access to an endless and personalized source of pluripotent cells. The clinical potential of this discovery is enormous, with the possibility of generating patient-derived iPSCs with applications not only in autologous transplants, but also in disease modeling and regenerative therapy. The use of iPSCs is extensive, and new ways to improve their derivation are being studied to increase efficiency and to overcome the risks for their clinical use. Even with the progress that has been made, there is still much to understand about the mechanisms of reprogramming.

In the study of pluripotency induction, an important model could be the use of primordial germ cells (PGCs), which can give rise to pluripotent cells called embryonic germ cells (EGCs). This reprogramming is achieved relatively easily, without the need for gene transduction, thus avoiding the risks related to gene manipulation [4, 5, 6]. PGCs are the embryonic precursors of gametes, giving rise during normal development to either spermatozoids or oocytes. These cells have limited self-renewal ability and are unipotent, are incapable of forming pluripotent embryoid bodies or contributing to teratomas or chimeras [7]. However, PGCs are considered developmentally pluripotent, given they generate the whole totipotent zygote after fertilization. This dual identity of both differentiated and pluripotent stem cells make PGCs a unique model to study cell fate and flexibility.

PGCs suffer reprogramming both in vivo and in vitro. In vivo, PGCs can give rise to embryonal carcinoma cells (ECCs), which are the pluripotent stem cells of testicular tumors [7]. In vitro, PGCs are easily reprogrammed into pluripotent EGCs with a specific cocktail of growth factors [4].

The reprogramming ability of PGCs can be explained by their similarity to pluripotent cells and their latent totipotency. PGCs innately express several transcription factors related to pluripotency, such as Oct4, Sox2, Nanog, and Lin28 [7]. Some of these factors are retained during PGC development from the zygote, whereas others such as Sox2, Nanog, and Klf2 are reexpressed or upregulated [8].

Other markers, such as tissue nonspecific alkaline phosphatase (TNAP) and germ cell nuclear antigen, do not belong to the pluripotency network itself, but are also strongly expressed by PGCs and ESCs. Even typical germ line factors such as B lymphocyte-induced maturation protein-1 (Blimp1) and Stella are typically expressed by ESCs [9]. It has been described that derivation of ESCs from the inner cell mass (ICM) is preferably achieved from cells that express Blimp1 and other germ line markers (interferon-induced transmembrane protein 3, Lin28, Prdm14, Stella, & c-Kit) [10]. The similarities in gene expression between PGCs and ESCs lead to the idea that the closest in vivo equivalent to ESCs are germ cells, instead of the ICM or even the epiblast, because PGC precursors are specified within the epiblast around 6 days post coitum (dpc) in mice [11].

The proximity of the PGCs to pluripotent stem cells and the ease of their reprogramming agree with the results of experiments on iPSC derivation from differently developed cells. The more undifferentiated the cells, the more easily they give rise to iPSCs, in a more efficient manner and requiring the transduction of fewer transcription factors. Conversely, the more differentiated cells, the more difficult they are to reprogram [12]. PGCs, however, can be reprogrammed with the transduction of only one of any of the four traditional iPSC factors [13]. This result is especially interesting because PGCs already express Oct4 and Sox2, which means that a variation in the expression level of just one transcription factor can be sufficient for PGCs to become pluripotent.

Advertisement

2. PGC reprogramming in vivo

ECCs are the stem cells of testicular tumors, which can be maintained indefinitely in culture as pluripotent cells. ECCs were first established as cell lines from mouse teratocarcinomas 50 years ago. In humans, the first teratocarcinoma cell lines isolated in vitro were TERA1 and TERA2, but their identity as ECCs was not discovered until some years later [14].

These pluripotent stem cells share most of their characteristics with ESCs, such as self-renewal capacity, specific markers, and the ability for differentiation to any cell of the organism [15]. The main difference from other pluripotent cells is that they are usually aneuploid. The malignancy of these cells is highly dependent on the microenvironment, as has been observed in chimera formation experiments: ECCs injected into mouse blastocysts can contribute to the development of a normal chimera, or in some cases to tumors. Another difference from ESCs is the very low efficiency in colonizing the germ line, which makes them less suitable for establishing mutant rodent lines [2]. Some authors define these cells as multipotent rather than pluripotent due to limitations in the differentiation potential, which is even more limited in human lines that often show no differentiation potential at all [14]. This potential has not been consistent, changing from one ECC line to another, and varying with culture conditions, such as the F9 EC line, which was considered nullipotent until the discovery of the induction of differentiation by retinoic acid (RA) exposure [16]. Considering the definition of pluripotency as the ability to differentiate into cells of the three germ layers, most ECC lines are pluripotent, but could be considered incompletely or partially pluripotent cells.

The germ line origin of the ECCs was proven using transgenic Steel (Sl) mutant mice. These mice, carrying a homozygous mutation in the Sl locus are unable to express stem cell factor (Kit ligand), a growth factor required for PGC survival and proliferation [17]. When embryonic gonads of the teratogenic mouse strain 129/Sv are transplanted to the adult testis, teratoma formation occurs in wild-type mice but not in Sl mutants, suggesting the Kit ligand is implicated in ECCs.

Most ECC lines are derived from early mouse PGCs (8.5–10.5 dpc), and spontaneous teratomas have been described to start around 12.5 dpc. PGCs lose their ability to reprogram in vivo after 12.5 dpc, which is coincident with the time the PGCs are able to give rise to EGCs in vitro. When 129/Sv 12.5 dpc gonads were transplanted into adult testes, teratomas appear at an 80% incidence, whereas when 13.5 dpc gonads were grafted, the tumor incidence decreased to 8%. The induction of these tumors is also related to the strain used, and is far less efficient in strains other than 129/Sv [14, 15].

In vivo reprogramming of PGCs to ECCs depends on a variety of genetic factors. The best known mutation that affects the development of mice teratomas is Teratoma (Ter) [18] in the RNA-modifying gene DND microRNA-mediated repression inhibitor 1 (Dnd1). In homozygous 129/Sv-Ter mice, teratoma incidence increases up to 75%. This reprogramming appears to be linked with proliferation, because the PGCs of these mice continue proliferating after 13.5 dpc, whereas unmutated PGCs enter mitotic arrest. The Ter mutation appears to be caused by the surrounding somatic cells instead of the PGCs themselves. In a similar manner, doublesex-related transcription factor 1 (Dmrt1) mutants also develop teratomas at a high rate in 129/SV mice, but unlike the Ter mutation, this effect is achieved by the loss of Dmrt1 in PGCs [19, 20].

Also related to PGC transformation is the phosphoinositide-3 kinase (PI3K)/protein kinase B (Akt) pathway, whose upregulation could lead to the appearance of ECCs. This outcome can be observed in the effect of the specific inactivation in PGCs of the tumor suppressor phosphatase and tensin homologue (PTEN), which leads to an activation of PI3K/Akt signaling, and, therefore, testicular teratoma formation [21]. Among the Akt targets is the tumor suppressor Trp53, whose deletion increases the incidence of testicular teratomas in mice [22].

Testicular cancers in humans are classified into three categories: (1) teratomas and yolk sac tumors that develop in fetuses and infants and are classified as nonseminomas, (2) adult testicular cancer, both seminomas and nonseminomas, which appear in men aged between 20 and 40 years, and (3) spermatocytic cancer, which affects elderly men [20]. The first two types have characteristics in common with mouse carcinomas. Teratomas and yolk sac tumors that arise early in human life are the most similar to the teratocarcinomas described in mice, even if normal karyotypes are present, and probably also originate from PGCs. With respect to adult human testicular tumors, the cellular origin of these tumors is carcinoma in situ, which is considered to develop early from the germ line due to the similarity to PGCs and gonocytes. Between these similarities, there is the expression of specific membrane markers, such as Kit or TNAP; stem and early germ cell genes, such as Nanog or Vasa; and genomic imprinting [23]. These cells can remain nonpathological until adult life and later develop into teratomas. Unlike the previous type, these cancer cells show chromosomal abnormalities, such as isochromosome of the short arm of the 12th chromosome (iso-12p) [24].

As in the mouse, the PI3K/Akt pathway is related to teratoma formation in humans. AKT1 overexpression, Trp53 deficiency and PTEN inactivation, or KRAS and NRAS mutations, which lead to activation of this pathway, correlate with testicular cancer formation. The Kit signaling pathway can also be implicated in the origin of human ECCs. Both KIT and Kit ligand (stem cell factor) mutations are related to human teratomas [19].

In addition, typical reprogramming factors such as Oct4 and Sox2 [3] are related to in vivo germ cell reprogramming. The Oct4 expression level appears to affect germ cell reprogramming in vivo, given its reported overexpression in both teratomas and adult testicular cancers [25]. On the other hand, Sox2 is only expressed in ECC, but not in PGCs or other testicular cancer cells [26].

The appearance of human testicular cancer has also been shown to be related to disturbances in the environment of the germ cells in the embryo, and in diseases such as cryptorchidism, gonadal dysgenesis, or estrogen exposure during pregnancy [27, 28, 29]. Estrogen upregulates c-Kit in the genital ridges, which leads to an increased proliferation of PGCs and reprogramming [30, 31].

Advertisement

3. PGC reprogramming in vitro

EGCs are derived in vitro from PGCs when cultured with a specific cocktail of growth factors: stem cell factor (SCF), leukemia inhibitory factor (LIF), and basic fibroblast growth factor (bFGF) [4, 5, 32]. EGCs are complete pluripotent cells, which are, like ESCs, able to give rise to all cell types in the organism and to fully contribute to blastocyst complementation, including contributions to the germline transmission [5, 32]. EGCs also share specific markers, such as stage-specific embryonic antigens (SSEAs), Oct4, Nanog, and TNAP, with other pluripotent stem cells [33].

EGCs were first derived from mice [4, 5] and then from various other animals [34]. EGCs have also been derived from human PGCs of around 5–10 weeks of gestation, providing a source of pluripotent stem cells and a good model of reprogramming [32].

Contrary to ECCs, EGCs are euploid and the primary difference between them and ESCs is the epigenetic state [7]. At the time of reprogramming, PGCs find themselves in different phases of the erasure and reestablishment of genomic imprints, and these epigenetic features are transmitted to the resulting EGCs. The epigenetic state of the PGCs is related both to the maintenance of the latent totipotency and to the inhibition of the stemness [35]. The manipulation of PGC epigenetics has proven the capability of reprogramming to EGCs (as we discuss later), showing how close these cells are to pluripotency.

Pluripotent stem cells can be found in various development states, with mouse ESCs representing the most undifferentiated, or naïve. This state is characterized by small, compact colonies; better survival when passaged as single cells; higher efficiency in chimera formation; shorter doubling time; and different culture condition requirements [36]. Human ESCs and mouse epiblast stem cells represent the most differentiated or primed state, characterized by larger flat colonies [37]. Human EGCs share some of their features with naïve stem cells, such as colony shape and culture requirements, and other features related to a more differentiated state, such as the lack of teratoma formation when injected into mice and low efficiency when derived from subculture [38]. This outcome shows how close PGCs are to totipotency and how useful they can be as a study model. The only major problem for this model is the availability of PGCs, which are scarce at the time when they are able to reprogram and are difficult to expand, because they only survive approximately a week in culture [33].

Advertisement

4. Classical PGC reprogramming mechanism

As we previously explained, PGCs can be cultured with LIF and SCF, maintaining their phenotype and promoting their survival for approximately a week [11]. When basic fibroblast growth factor (bFGF) is added to the media, reprogramming of the PGCs is induced. The mechanisms involved in this process are still not fully understood. Several signaling pathways and genes have been shown to be implicated, such as Blimp1 downregulation, PTEN inactivation, Klf4, and c-Myc upregulation, PI3K/Akt signaling activation, and transforming growth factor (TGF)β signaling, among others [21, 39, 40, 41]. Most of these mechanisms are also involved in iPSC derivation, indicating a common regulatory network between the various reprogramming processes [19].

The dynamics of the transition from a unipotent germ cell to a pluripotent stem cell have been studied [42]. Three phases have been described, which are similar to those in iPSC derivation [43], based on the loss of germ cell characteristics and the expression of pluripotency genes: induction, preparation, and maintenance. This change in gene expression is a gradual process that takes approximately a week to complete. In the induction phase, along with the upregulation of Klf4 and embryonic stem cell-expressed ras (Eras), some germ cell markers, such as Dnd1 and Ddx4, start to be downregulated. Surprisingly, a large amount of PGCs that begin the reprogramming die in this first step. Some pluripotent factors, such as Klf4 and Eras, begin to be upregulated in the early phase, but they do not reach their higher expression until later phases, when most pluripotent factors, such as c-Myc and Nanog, are upregulated. In the preparation phase, some pluripotent markers, such as Klf4, Nanog, and Zfp42, reach their highest expression level, whereas others, such as Klf9 and Sox11, continue their gradual upregulation. It is also interesting to notice the high expression in this phase of the Meis family of transcription factors, a family related to the maintenance of hematopoietic stem cells.

Compared with PGC reprogramming, the traditional iPSC derivation process is far longer, taking approximately 3 weeks to complete. One primary difference between the EGC and iPSC derivation processes is that in the latter, a mesenchymal-to-epithelial transition takes place in an early phase. The lack of that event in PGC reprogramming could be due to the lack of the inverse process (epithelial-to-mesenchymal transition) during PGC specification, which is contrary to most somatic cells [8]. Moreover, the activation of genes already expressed in PGCs, such as SSEAs or TNAP, takes place in this first phase. The preparation phase is characterized by the upregulation of Nanog (as in EGC derivation), Sall4, and Esrrb. Other genes reactivated after these are Rex1, Lin28, and finally, Stella, Dppa4, or Pecam, among others. Between the transduced factors, endogenous Oct4 is typical of the preparation phase, whereas endogenous Sox2 is necessary for the maintenance phase [8].

Inhibition of Blimp could be the primary mechanism of bFGF-mediated EGC derivation. Blimp is the key germ cell specification gene [44], which has a potent repressive function. Among its targets are c-Myc and Klf-4, two of the primary pluripotency transcription factors; thus, Blimp inhibition leads to their upregulation [39]. These factors are particularly important in the acquisition of pluripotency, and they are the factors not expressed naturally in PGCs. It has recently been reported that deletion of Blimp1 in PGCs is sufficient to cause the derivation of EGCs in culture without bFGF [45].

Advertisement

5. Non-classical PGC reprogramming

It has been shown that the PI3K/AKT signaling pathway is involved in PGC reprogramming in vitro. PI3K is activated not only by bFGF but also by LIF and SCF, the three factors needed for EGC derivation. One of its primary downstream effectors is Akt, also known as protein kinase B, which has been observed to improve EGC derivation efficiency when activated in PGCs, even allowing the reprogramming in late PGCs up to 14.5 dpc. [40]. The specific in vivo inactivation of the tumor suppressor PTEN in PGCs, which leads to PI3K/Akt signaling activation, and enhances both EGC derivation and testicular teratoma formation [21]. Among Akt targets is the tumor suppressor Trp53. Akt inhibits its transcriptional activity by preventing its phosphorylation and nuclear accumulation. Deletion of Trp53 increases the incidence of testicular teratomas in mice and is enough to cause PGC reprogramming in culture in the absence of bFGF [40]. Trp53 deletion has similar effects on iPSCs, enhancing their induction [46].

The cell proliferation rate also appears to be as important in PGC reprogramming as it is in iPSC derivation [47]. The time when PGCs have the highest potential to give rise to EGCs coincides with the moment of the highest proliferation in vivo [5]. The three growth factors typically used for EGC derivation are mitogens that can alone promote proliferation [48]. Also, bFGF can be replaced by other known mitogens, such as RA or forskolin [11, 49], which activates protein kinase A by increasing the intracellular cyclic AMP. These pro-mitogenic effects could be triggered both by MAPK signaling [50] and the PI3K/Akt pathway, which enhances proliferation through several downstream effectors. One is by the already mentioned inhibition of Trp53, but there are others, such as the activation of cyclin D and inhibition of cyclin-dependent protein kinase inhibitors (CDKIs) [51]. The effects of CDKI inactivation have been observed in the mutation of the CDKI inhibitor of CDK4 (INK4), which enhances teratoma formation in mice, along with Trp53 inhibition [22].

PGC reprogramming can also be achieved by inhibition of MAPK/extracellular signal-regulated kinase (ERK), and glycogen synthase kinase-3 (GSK3) signaling. The two inhibitors that have been used for this purpose have been PD0325901 (PD) and CHIR99021, respectively (named 2i), which in combination with LIF can replace both SCF and bFGF and give rise to EGCs [52]. The same inhibitors have been used to enhance iPSC derivation, thus obtaining a more undifferentiated phenotype [53]. The mechanism followed by 2i or by bFGF reprogramming differs, because bFGF activates MAPK signaling [54]. It has been proposed that MAPK/ERK inhibition can lead to pluripotency by promoting long-term self-renewal and inhibiting differentiation through downregulation of Lef1. In ESCs, Lef1 promotes differentiation by inducing lineage specific genes and suppressing pluripotency gene expression. On the other hand, GSK3 inhibits the Wnt/β-catenin pathway, being responsible for the Tcf3-mediated repression of other pluripotency-related genes, such as Sox2, Oct4, Nanog, and Esrrb [55, 56]. The difference between the mechanism triggered by bFGF and 2i is also revealed by the timing of the required compounds, whereas bFGF is only needed during the first 24 h [57, 58], continuous culture with 2i is required [52].

Recently, another glycogen synthase kinase 3 (GSK3) inhibitor has been found to achieve EGC derivation [41]: kenpaullone, which inhibits not only GSK3, but a wide spectrum of kinases such as CDKIs, and is sufficient for late (13.5–14.5 dpc) PGC reprogramming. Also, TGFβR inhibition by SB431542 can promote reprogramming in 11.5 dpc PGCs. The fact that these treatments have no effect on early PGCs, contrary to bFGF and 2i treatments, underlines the differences between the mechanisms that can trigger PGC reprogramming. TGFβ inhibition could induce reprogramming through promotion of proliferation [59], reducing MAPK activity [60], and directing induction of the pluripotency network. TGFβ inhibition can replace Sox2 transduction in iPSC generation by inducing Nanog expression [61]. On the other hand, the effect of various treatments can be combined to enhance reprogramming efficiency, such as TGFβR inhibition with 2i, with an efficiency of 12% [42]; TGF-β inhibition + ERK inhibitor (PD) [62], or bFGF + 2i + RA + forskolin, with an efficiency of approximately 20% [48]. This outcome shows that various mechanisms can synergize their effects in the reprogramming of PGCs. It has also been reported that mutations in genes involved in PGC development, such as Dnd1, Pten, and Pgct1, improve EGC derivation efficiency [19].

Advertisement

6. PGCs and hypoxia reprogramming

Recently, we have shown the reprogramming of PGCs cultured in hypoxic conditions without bFGF. The EGCs obtained had proven to be pluripotent, even if they were not completely reprogrammed, as shown by their limited proliferation [6].

The positive effects of hypoxia in enhancing reprogramming have been reported in iPSCs [63]. These hypoxia effects could be related to the idea of cancer stem cells arising in vivo from differentiated cells [64], due to environmental causes.

Hypoxia induces a change in the cell’s energetic metabolism, from oxidative phosphorylation to glycolysis. This switch in the metabolism has been shown to be required for reprogramming of somatic cells to iPSCs. It has also been observed that the closest the somatic cell metabolism is to an ESC, the more efficient the reprogramming [65]. This metabolic change is an active process at the beginning of the reprogramming process; it has been shown that the expression of glycolytic genes, such as glucose transporter (Glut)1, Hxk2, Pfkm, and lactate dehydrogenase A (Ldha), is previous to pluripotency genes [66]. The relationship between stemness and glycolytic metabolism has been widely described in the Warburg effect, in which cancer cells in normoxia change their metabolism from oxidative phosphorylation to glycolysis [67]. It is still not clear whether the stem cell program triggers the metabolic change or whether it is the metabolic shift that activates the stem cell program; however, it has been demonstrated that these two processes are correlated. Our PGC data clearly supports the second hypothesis [6]; one described mechanism of this hypothesis is the positive feedback between glycolysis and the oncogene NF-κB [68].

The induction of PGC pluripotency by hypoxia has been demonstrated to be mediated by hypoxia-inducible factors (HIFs), given the inhibition of their degradation by dimethyloxaloylglycine mimics the effect of hypoxia in the EGC derivation. This agrees with the improved reprogramming efficiency observed in iPSC derivation when a prolonged expression of HIF1 is forced [69].

HIFs are transcription factors that regulate a large number of downstream effectors under hypoxic conditions. Among genes regulated by HIFs are pluripotency genes such as Oct4 and c-Myc, regulated by HIF2 or Notch, and ETS-1, regulated by HIF1 [70]. The metabolic switch can also be provoked by HIF activation. HIF regulates several metabolism-related genes, promoting the expression of glycolytic proteins such as GLUT1 & 3, LDHA, ENO1, aldolase A, phosphoglycerate kinase 1 (PGK1), glyceraldehyde 3-phosphate dehydrogenase (GAPDH), hexokinase 1 & 2, phosphofructokinase-2 (PFK2), and phosphofructokinase, liver type (PFKL)[65, 71].

These effects in the metabolism coincide with those provoked by Akt signaling, which is consistent with the mechanisms proposed for bFGF-mediated reprogramming of PGCs. Akt promotes glycolysis by the inhibition of FoxO transcription factors [51]. In somatic cell reprogramming, FoxO1 Akt-dependent phosphorylation enhances both glycolysis and iPSC derivation [72].

The mechanism by which HIFs provoke dedifferentiation in PGCs is partially known. It has been shown that the deregulation of Oct4 mediated by HIFs could be directly responsible for hypoxia-induced reprogramming [6]. Oct4 is one of the primary components of the pluripotency network; small changes in its expression levels result in great effects on stem cell development, promoting both undifferentiated and differentiated states, depending on the context [73]. Thus, in addition to the effect of its transduction on iPSC derivation, high levels of Oct4 can lead to differentiation and low levels can lead to pluripotency entry, provoking a dose-dependent induction of differentiation between mesoderm and trophectoderm in ESCs.

However, pluripotent hypoxia-derived EGCs cannot be passaged long-term, probably due to a lack of upregulation of c-Myc and Klf4. Gene expression analysis of these cells suggests that they have not reached the stabilization phase of cell reprogramming. Comparing the gene expression of these phases with those of hypoxia-derived EGCs has shown that the genes typical from initiation and maturation are upregulated; however, those belonging to the stabilization phase, such as Dppa3, Dppa4, Utf1, Eras, Lin28, Sox2, and Dnmt3l, are not [6].

As in 2i-mediated derivation of EGCs, and unlike bFGF, hypoxia is needed continuously to provoke PGC reprogramming. This need suggests a mechanism closer to that triggered by 2i. The relationship between hypoxia and GSK3 inhibition has been demonstrated by the fact that HIF1α stabilization depends on an inactive GSK3β pathway [74]. Under long hypoxia exposure, HIF1 is downregulated though activation of GSK3β. It has also been reported that Akt, which inhibits GSK3β, can upregulate HIFs through mammalian target of rapamycin (mTOR) activation [51]. This correlation supports the hypothesis of these two methods of PGC reprogramming, hypoxia and 2i, being connected.

Advertisement

7. Primordial germ cell reprogramming and energetic metabolism

Energetic metabolism has deep implications in germ cell development. As evidence, human PGCs show lipid droplets and glycogen accumulations on their cytoplasm in order to obtain energy [75].

Gene expression analysis comparing PGCs and EGCs showed no relevant differences in expression of pluripotency factors, whereas glycolytic enzymes displayed elevated levels in EGCs. This link between metabolism and PGC reprogramming has also been reported in PGCs cultured in hypoxia. This process leads to the induction of pluripotency, which is dependent on HIF1α stabilization and in turn provokes metabolic reprogramming and Oct4 deregulation [6]. Other studies have also observed that low Oct4 expression favors a robust pluripotent state in embryonic stem cells or acquisition of pluripotency, whereas high Oct4 levels relate to differentiation processes [76, 77]. In addition, Oct4, which remains active in PGCs, participates in metabolism regulation. In embryonic stem cells, Oct4 can induce hexokinase, pyruvate kinase, and pyruvate dehydrogenase (PDH) kinase expression, and the overexpression of these enzymes can prevent cell differentiation [78]. This has also been observed in PGCs [6]. In fact, when glycolysis is favored at low-oxygen concentrations, an increase in iPSC efficiency, and an enhancement of the expression of pluripotency factors via HIF expression are observed. Furthermore, iPSC derivation can be achieved in hypoxic conditions using only Oct4 and Klf4 [63, 79]. As mentioned for the comparison between PGCs and their in vitro pluripotent counterparts, EGCs, metabolism is also involved in the malignant transformation of PGCs into their pluripotent partners in vivo, ECCs. In particular, miRNA-regulated expression of enzymes involved in glycolytic metabolism contributes to the growth of germ cell tumors [80].

Oxygen levels are closely related to metabolism and potentiality. PGCs cultured in hypoxia are reprogrammed toward pluripotent cells and cause an increase in glycolytic genes, while they downregulate genes involved in oxidative phosphorylation (OXPHOS) [6]. Abundant evidence relates hypoxia to inhibition of oxidative metabolism. Among its effects, hypoxia inhibits cytochrome c oxidase and complex II of the electron transport chain. In fact, usage of the OXPHOS inhibitor carbonyl cyanide 3-chlorophenylhydrazone (CCCP) induces an upregulation of Oct4, Nanog, and Sox2 in embryonic stem cells [81]. On the other hand, culture of these cells in normoxia upregulates genes involved in differentiation [82, 83].

As previously mentioned, PGCs cultured in hypoxia give rise to pluripotent cells, and this process takes place in parallel with a metabolic shift toward glycolysis, which is governed by HIF1. Consequently, HIF1 inhibition disrupts PGC reprogramming, and HIF stabilization induces reprogramming [6]. HIF1 acts as a link between oxygen levels and metabolic phenotype inducing the expression of several genes related to glycolysis, such as glucose transporters, hexokinase, pyruvate dehydrogenase kinase 1, and lactate dehydrogenase. In fact, PGCs reprogrammed through culture in hypoxia show pyruvate dehydrogenase inhibition and mitochondrial inactivation [6]. Recent work from our laboratory has shown that PDH needs to be inhibited to achieve PGC reprogramming. Once the glycolytic profile is established under hypoxia, an increase in glycolytic flux through PKM2 activation renders in a synergetic effect with hypoxia (Sainz de la Maza et al. [129]).

Hypoxia can also alter the metabolic profile through mitochondrial mass modification. Mitochondria in pre-migratory PGCs are globular and localized around the nucleus. During migration, mitochondria increase in number and, when they undergo differentiation, they increase their number and size even further, and they acquire a more ovoid shape, further developing their cristae [84].

HIF1 inhibits PGC1β and induces mitophagy through Bnip3 upregulation [85]. In fact, autophagy is required in the early steps of cell reprogramming [86]. A shift has been reported in iPSC derivation from complex, active mitochondrial networks with developed cristae in fibroblasts to small, spherical, perinuclear, inactive mitochondria without cristae in pluripotent stem cells [66]. The results from our laboratory have shown that hypoxia-induced reprogramming causes a marked increase in Bnip3 expression, an essential gene involved in mitophagy. Autophagy takes places during reprogramming and is required for pluripotency acquisition (Sainz de la Maza et al. [129]).

Additionally, the Lin28/let7 pathway is crucial in glucose metabolism. In particular, Lin28 increases glucose uptake and metabolism through PI3K/mTOR activation and is involved in the translation of genes related to glycolysis, glucose metabolism, cellular carbohydrate metabolism, oxidative metabolism, and mitochondria in human embryonic stem cells [87, 88, 89].

Lin28 is also closely related to pluripotency, since it contributes to cell reprogramming and is present in the reprogramming cocktail, giving rise to iPSCs [90], and is also capable of activating the translation of Oct4 at the post-transcriptional level in human embryonic stem cells [91].

Primordial germ cells express Lin28 from 7.5 dpc in mouse PGCs and play a key role in Blimp1 expression. Lin28 is an RNA-binding protein that is able to bind the let7 precursor, impairing let7 processing. Therefore, miRNA let7 is not synthesized and is not able to inhibit Blimp1 translation; Lin28 indirectly stabilizes Blimp1 so PGCs can fulfill their germ cell specification [44, 92].

Advertisement

8. PGC reprogramming and epigenetics

PGCs undergo profound epigenetic reprogramming during their development [42]. Once specified, PGC express Blimp1, which is the master regulator of PGCs responsible for somatic program repression and germ cell identity [44]. In order to maintain Blimp1 expression and establish germ cell fate, Lin28 acts a negative regulator of let7 [92]. In human PGCs, Blimp1 is the effector that represses the somatic program, and Sox17 is the determinant transcription factor that establishes the germinal fate [93]. A recent study has shown a stable and elevated expression of Sox15 in early PGCs, which infers a possible role for this molecule in human PGCs as a master regulator, rather than Sox17 [94].

At the onset of specification, PGCs show several epigenetic marks, such as H3K4me2, H3K4me3, H3K9ac, H3K9me1, H3K9me2, H3K9me3, H3K27me2, and H3K27me3. However, these marks are shared at this time by neighboring, future somatic cells [95]. Once PGC migration begins, PGCs reduce the repressive mark H3K9me2 due to downregulation of the enzymes Ehmt1 and Ehmt2. PGCs also reduce DNA methylation by downregulation of the DNA methyltransferases Dnmt1, Dnmt3a, and Dnmt3b [8, 96]. Prdm14, an essential partner of Blimp1 in mouse germ cell development, and Tcfap2c intervene in this demethylation process downregulating Ehmt1, Ehmt2, Dnmt3a, and Dnmt3b. This partnership is also involved in the reexpression of transcription factors related to pluripotency, such as Sox2 and Klf2. However, Prdm14 is not necessary for human PGC development [97, 98]. PGCs progressively increase the repressive mark H3K27me3 via polycomb repressive complex 2 (PRC2) [95].

Later, once PGCs have reached the future gonads, they remove imprinting tags, increase H4R3me2 mark, and reactivate the inactive X chromosome in female PGCs [95, 99]. H4R3me2 modification, which takes place on arginine residues, is catalyzed by Prmt5, an epigenetic modulator that binds Blimp1, and exerts its epigenetic modifications until it is translocated back to the cytoplasm after E11.5 [100]. Another important event that occurs at this stage is major global DNA demethylation. It has been proposed that the vast extent of this DNA modification infers an active process and expression of the hydroxylase Tet1, the cytidine deaminase AID, and genes involved in base excision repair response (BER) have been detected. Nucleotide excision repair (NER) does not appear to be involved in PGC demethylation because no upregulation in molecules that take part in NER has been detected [101]. Finally, PGCs lose histone H1, increase nuclear size, lose chromocenters, reduce the epigenetic marks H3K9me3, H3K9me2, H3K27me2, H3K27me3, H3K9ac, and H4R3me2, and also lose nucleosomal, noncanonical histone H2a.Z [102, 103]. Blimp1 also induces the expression of Jmjd3 [104], responsible for removing the repressive epigenetic marks H3K9me2 and H3K9me3. In B cells, Blimp1 can also interact with chromatin modifier enzymes such as Kdm1a, a histone lysine transferase that catalyzes removal of methyl groups from lysines 4 and 9 of histone 3 [105]. In contrast to Blimp1, which appears to be essential in the repression of the somatic program, its effector Prdm14 contributes to establishing potential pluripotency through Sox2 upregulation in mouse PGCs, given that Sox2 is absent in human PGCs [97, 106]. Kdm6a is the histone demethylase that catalyzes the removal of the mark H3K27me3 during this second wave of reprogramming. The disruption of this process leads to an aberrant epigenetic reprogramming and loss of pluripotency markers Nanog, Oct4, and SSEA1 [107].

Sex determination begins at approximately E12.5. At this moment, DNA methylation is very low both in male and female PGCs [108, 109]. However, settlement of the epigenetic signature through DNA methylation occurs differently in male and female germlines. In males, de novo DNA methylation starts at around E13.5 and is accomplished before birth. Once methylation has been completed, gonocytes undergo vast proliferation and then they enter meiosis at the onset of gametogenesis. A disruption of methylation in male germ cells renders infertility [110]. In females, de novo methylation starts after birth and is not fulfilled until approximately P21 [111]. Oocytes undergo cell cycle arrest in meiotic prophase I, and it is not until ovulation that they re-enter meiosis. This coincides with the inability of PGCs to become reprogrammed.

Few studies have been performed on human PGCs, and the majority has focused on later stages of epigenetic reprogramming. Among the data available, early gonadal PGCs in humans (6–8 weeks of gestation) show low H3K9me2 and high H3K27me3 epigenetic markers [112]. Studies on pigs have also reported that changes in H3K9me2 and H3K27me3 markers are previous to DNA methylation, as observed in mouse PGC development [113]. Human PGCs also display active H3K4me1 and H3K4me3 marks and a peak in H3K9ac from 10 to 13 weeks of gestation. DNA showed a hypomethylated status and a loss of imprinting marks from gestation week 10 onwards. It is noteworthy that Blimp1 is restricted to the nucleus from week 7 to week 12, whereas Prmt5 is located in the cytoplasm, showing that there is no Blimp1/Prmt5 association in human PGCs [112].

Demethylated human PGCs later recover a methylated status, from week 13 of gestation to birth [114]. In addition, human postnatal gonocytes show a different epigenetic pattern from E13.5 mouse PGCs. Low levels of H3K9me2 are shared with mouse PGCs, but H3K27me3 shows low levels in post-migratory PGCs [112]. Also in contrast to mouse PGCs, the H3K9me3 repressive mark is observed in human gonocytes, as well as increased levels of H3K9ac active marks [115, 116]. Mouse PGCs show the lowest methylation at E13.5, when less than 10% of cytosines located at CpG islands are methylated [109]. Analyses of global methylation status from early stages of development have led to the observation that the ICM maintains its methylation status and, consequently, to the hypothesis that the germline is responsible for most of the DNA demethylation that takes place during development [117]. Specifically, the pattern appears to be a global demethylation during PGC migration, in which specific methylation marks in CpG islands (CGIs) of PGC-specific genes, CGIs on the X chromosome and differentially methylated regions in imprinted genes are conserved. Secondly, a new demethylation wave occurs when PGCs reach the genital ridges, affecting the previously mentioned sequences with epigenetic memory. This process involves both active and passive demethylation pathways [109]. Specifically, mouse PGCs fall from a 78% of global methylation at the epiblast stage to 5% at E11.5 (Figure 1). In human PGCs, demethylation takes place during the first 12 weeks of development, falling to 7% of methylation [118].

Figure 1.

Modifications in DNA methylation during mouse PGC and gamete development. Both male and female germ cells share a first phase of epigenetic modifications, in which several processes lead to DNA demethylation and histone modifications. This phase corresponds to PGC migration and colonization of genital ridges in the first phase and development of gonads in the second. After sex determination, differential methylation is observed among male and female germ cells. Whereas male germ cells undergo de novo methylation at E13.5, female germ cells maintain hypomethylated status until birth.

EGCs and ESCs can be fused to B-cells in order to originate a tetraploid hybrid. Methylation analysis has shown that the generated hybrids from EGCs showed lower methylation levels than those generated from ESCs, probably showing some sort of epigenetic memory in which EGCs resemble the low methylation status of their precedent PGCs. These hybrids eliminate the imprints that were present in ESC cells, suggesting that epigenetic reprogramming in EGCs is dominant over ESCs [119]. As previously mentioned, PGCs eliminate their imprinting marks at approximately E11-5. EGCs obtained from E11-5 or later PGCs also show demethylated imprinting marks, which cause serious alterations in developing chimeras [120, 121]. However, EGCs originated from previous PGCs show a less profound imprinting erasure and a more hypomethylated status regulated by Prdm14 when EGCs are cultured in the presence of GSK3β and MEK (2i) inhibitors [122, 123]. In fact, EGCs eliminate imprinting marks in the genes Igf2, Igf2rr, Dlk1, and H19, among others, which are established shortly after PGC specification [124, 125]. EGCs and ESCs also share as a common feature the activation of both X chromosomes in female cells [126].

PGCs can be reprogrammed into EGCs when cultured in the presence of trichostatin A (TSA), an inhibitor of histone deacetylase, in substitution for bFGF, stem cell factor and LIF [39]. In the case of bFGF, this growth factor needs to be added in the first 24 h of culture. Substitution of bFGF by TSA accelerates the kinetics of EGC derivation, with a quicker downregulation of Blimp1 [39]. As stated in the classical PGC reprogramming section, bFGF-induced reprogramming of PGCs causes a downregulation of Blimp1, which provokes an upregulation of Klf4, c-Myc, and Dhx38. This downregulation does not take place if bFGF is added later than 24 h of culture [39, 45, 127]. In EGC derivation by TSA, Blimp1 is absent in EGCs, whereas expression of Klf4, c-Myc, and Eras is detected. In bFGF-induced PGC reprogramming, Blimp1 disappears from PGC after 48 h [39].

As far as Prmt5 is concerned, this epigenetic modifier stays in the nucleus up to 7 days of culture, when it translocates from the nucleus to the cytoplasm in EGC colonies. Since Blimp1 binds Prmt5 to repress gene expression through H2A/H4R3me2s, contributing to maintaining the germ cell phenotype, absence of Blimp1 again appears to be a crucial event in PGC reprogramming. Direct targets of this mentioned repression, such as Dhx38 or c-Myc, are detected a few days after a bFGF-induced reprogramming procedure. These events were also observed in TSA-induced reprogramming of PGCs. [39]. Prmt5 has also been linked to pluripotency in ESCs. Its loss disrupts pluripotency and causes differentiation of these cells, where it is located in their cytoplasm [128]. Prdm14 is also essential for PGC derivation into EGCs. As previously mentioned, Prdm14 is an effector of Blimp1 and is involved in the downregulation of methylation enzymes during the first demethylation wave and in the reexpression of pluripotency factors. PGCs deficient in Prdm14 are unable to reprogram into EGCs because they cannot downregulate the repressive marker H3K9me2 and upregulate the epigenetic marker H3K27me3 via PRC2 [97]. Recent studies from our laboratory have shown that PRC2 is not involved in hypoxia-induced PGC reprogramming, whereas addition of histone deacetylase inhibitor valproic acid (VPA) is capable of inducing PGC reprogramming (Sainz de la Maza et al. [129]).

It is not surprising that epigenetic modifications relate to potency. Pluripotent stem cells show an open conformation of chromatin and active chromatin markers, such as H3K4me and H3K9ac [130, 131]. On the other hand, differentiated cells display repressed chromatin markers, such as H3K27me. Partially differentiated cells show a bivalent chromatin, with both active and repressing markers. Akt, one of the primary factors related to the reprogramming of PGCs, as noted before, also promotes a more active chromatin, mainly by inhibition of Mbd3, a component of the nucleosome remodeling deacetylase complex. Mbd3 is important in heterochromatin formation, and its inhibition promotes reprogramming in both EGC and iPSC derivation [19].

Epigenetics is also involved in the derivation of iPSCs. Fibroblast reprogramming can be achieved exclusively using soluble factors. In the reprogramming cocktail used by Hou et al [131], some epigenetic modifiers were included, such as the histone deacetylase inhibitors (HDACi) sodium butyrate and VPA, and the Kdm1a inhibitor tranylcypromine [132]. HDACi are strongly related to pluripotency acquisition. VPA has been proven to enhance iPSC generation, and usage of HDACi can turn pluripotent colonies with fuzzy edges into typical, compact pluripotent colonies [133, 134, 135]. VPA can also eliminate the imprinting marks located at the Dlk1-Dio3 gene cluster [136]. Inhibition of the polycomb complex, responsible for gene repression through DNA and histone methylation, results in lower iPSC derivation efficiency, probably because this complex is essential to the repression of the somatic program [137]. However, inhibition of histone methyltransferases by using BIX-01294 or inhibition of DNA methyltransferases using 5-azacytidine has been reported to improve iPSC derivation. The usage of BIX-01294 on fibroblasts with induced expression of only Oct4 and Klf4 rendered a comparable efficiency to that of using the four factors [138, 139]. A deeper understanding of the impact of epigenetics in reprogramming is required in order to elucidate the role of chromatin and histone modifications in the acquisition of pluripotency. Further research should be performed regarding the link between pluripotency and the germline program. For example, the induction of the expression of Prdm14, which is a Blimp1 effector, enhances iPSC derivation from mouse and human fibroblasts [140].

Advertisement

9. Concluding remarks and future directions

Various methods and soluble factors can be used for PGC reprogramming, including the classical bFGF [4, 5, 7, 141], the deletion of Trp53 [40], the addition of mitogens such as RA [11] or forskolin, [49], the inhibition of MAPK/ERK kinase and GSK3 signaling [52], and the addition of epigenetic modifiers such as TSA [39] and VPA (Sainz de la Maza et al. [129]). In addition, inducers of glycolysis such as hypoxia [6] and manipulation of cell metabolism are able to induce pluripotency (Table 1) and partial reprogramming, implicating many cytoplasmic and nuclear proteins (Figure 2).

Method Mechanism Age (dpc) Efficiency Laboratory/year
bFGF MAPK and PI3K/Akt activation 8.5 10/80 Hogan [5] and McLaren-Surani [58]
Forskolin cAMP agonist 11.5 14.5 ± 3.9/(1/2 genital ridge) Nakatsuji [49]
Retinoic acid Mitogen 11.5 12.5 ± 5.2/(1/2 genital ridge) Nakatsuji 1996 [49]
Trichostatin A HDAC inhibitor 8.5 25/80 McLaren-Surani [58]
Valproic acid HDAC inhibitor 8.5 ND De Miguel, submitted
2i PD0325901 MAPK/ERK inhibitor 8.5 9.4% Smith [52]
CHIR99021 GSK3β inhibitor
SB431542 TGFβR inhibition 11.5 2.08/104 Nakano [41]
Kenpaullone GSK3β and CDKs inhibitor 13.5 2.27/104 Nakano [41]
Hypoxia HIFs activation 8.5 6.9% De Miguel [6]

Table 1.

Comparison of different methods of PGC to EGC reprogramming.

Figure 2.

Schematic representation of the external factors (in green), signal transduction molecules (in red or white), and processes (in yellow) implicated in PGC reprogramming. Directly inhibited molecules in red. Arrows indicate induction and broken lines indicate repression of the pathway. PD: PD0325901; SB: SB431542; CHIR: CHIR99021; RA: retinoic acid; VPA: valproic acid; TSA: trichostatin A.

All these data highlight the inherent potency of germ cells, allow for further and detailed characterization of the reprogramming process and are useful tools for the identification of genes involved in germ cell malignant transformation and the development of testicular tumors.

Advertisement

Acknowledgments

The authors thank financing from Roche Farma SA and Juliette Siegfried and her team at Servingmed.com for the English language editing.

Abbreviations

2itwo inhibitors (PD0325901 and CHIR99021)
BERbase excision repair response
bFGFbasic fibroblast growth factor
Blimp1B lymphocyte-induced maturation protein-1
Bnip3BCL2/adenovirus E1B 19 kDa protein-interacting protein 3
CCCPcyanide 3-chlorophenylhydrazone
CDK4cyclin-dependent kinase 4
CDKIcyclin-dependent protein kinase inhibitor
CpGcytosine phosphate guanine
Ddx4DEAD-Box Helicase 4
Dlk1delta like non-canonical notch ligand 1
Dmrt1doublesex-related transcription factor 1
Dnd1DND microRNA-mediated repression inhibitor 1
DnmtDNA methyltransferase
dpcdays post coitum
Dppadevelopmental pluripotency associated
ECCembryonal carcinoma cells
EGCembryonic germ cells
Ehmteuchromatin histone methyltransferase
ENO1enolase 1
Erasembryonic stem cell-expressed Ras
ESCembryonic stem cells
Esrrbestrogen-related receptor beta
FoxOforkhead box class O
GAPDHglyceraldehyde 3-phosphate dehydrogenase
Glutglucose transporter
Hand1heart and neural crest derivatives expressed 1
HDAChistone deacetylase
HIFhypoxia-inducible factor
Hxk2hexokinase 2
ICMinner cell mass of the blastocyst
iPSCinduced pluripotent stem cell
JmjdJumonji domain containing
Kdmlysine demethylase
Ldhalactate dehydrogenase A
Lef1lymphoid enhancer binding factor 1
Mbd3methyl-CpG binding domain protein 3
MEFmouse embryo fibroblast
Meismyeloid ecotropic viral integration site
mTORmammalian target of rapamycin
NERnucleotide excision repair
Octoctamer-binding transcription factor
PARPpoly(ADP-ribose) polymerase
PDHpyruvate dehydrogenase
Pecamplatelet endothelial cell adhesion molecule
PFKphosphofructokinase
PGCprimordial germ cell
PGK1phosphoglycerate Kinase 1
PKMpyruvate kinase muscle
PRC2polycomb repressive complex 2
Prmtprotein arginine methyltransferases
RAretinoic acid
SCFstem cell factor
SSEAstage-specific embryonic antigen
Tbx3T-box protein 3
Terteratoma
TGFβtransforming growth factor β
TNAPtissue non-specific alkaline phosphatase
TSAtrichostatin A
Utfundifferentiated embryonic cell transcription factor
VPAvalproic acid
Zfp42zinc finger protein

References

  1. 1. Nicholas JS, Hall BV. Experiments on developing rats. II. The development of isolated blastomeres and fused eggs. Journal of Experimental Zoology. 1942;90(3):441-459
  2. 2. De Miguel MP, Fuentes-Julian S, Alcaina Y. Pluripotent stem cells: Origin, maintenance and induction. Stem Cell Reviews. 2010 Dec;6(4):633-649
  3. 3. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006 Aug 25;126(4):663-676
  4. 4. Resnick JL, Bixler LS, Cheng L, Donovan PJ. Long-term proliferation of mouse primordial germ cells in culture. Nature. 1992 Oct 08;359(6395):550-551
  5. 5. Matsui Y, Zsebo K, Hogan BL. Derivation of pluripotential embryonic stem cells from murine primordial germ cells in culture. Cell. 1992 Sep 4;70(5):841-847
  6. 6. Lopez-Iglesias P, Alcaina Y, Tapia N, Sabour D, Arauzo-Bravo MJ, Sainz de la Maza D, et al. Hypoxia induces pluripotency in primordial germ cells by HIF1alpha stabilization and Oct4 deregulation. Antioxidants & Redox Signaling. 2015 Jan 20;22(3):205-223
  7. 7. Donovan PJ, de Miguel MP. Turning germ cells into stem cells. Current Opinion in Genetics & Development. 2003 Oct;13(5):463-471
  8. 8. Kurimoto K, Yabuta Y, Ohinata Y, Shigeta M, Yamanaka K, Saitou M. Complex genome-wide transcription dynamics orchestrated by Blimp1 for the specification of the germ cell lineage in mice. Genes & Development. 2008 Jun 15;22(12):1617-1635
  9. 9. Saitou M, Barton SC, Surani MA. A molecular programme for the specification of germ cell fate in mice. Nature. 2002;418(6895):293-300
  10. 10. Chu L-F, Surani MA, Jaenisch R, Zwaka TP. Blimp1 expression predicts embryonic stem cell development in vitro. Current Biology. 2011;21(20):1759-1765
  11. 11. De Felici M, Farini D, Dolci S. In or out stemness: Comparing growth factor signalling in mouse embryonic stem cells and primordial germ cells. Current Stem Cell Research & Therapy. 2009;4(2):87-97
  12. 12. Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, et al. Epigenetic memory in induced pluripotent stem cells. Nature. 2010 Sep 16;467(7313):285-290
  13. 13. Nagamatsu G, Kosaka T, Saito S, Honda H, Takubo K, Kinoshita T, et al. Induction of pluripotent stem cells from primordial germ cells by single reprogramming factors. Stem Cells. 2013 Mar;31(3):479-487
  14. 14. Andrews PW. From teratocarcinomas to embryonic stem cells. Philosophical transactions of the Royal Society of London Series B, Biological Sciences. 2002 Apr 29;357(1420):405-417
  15. 15. Stevens LC. The development of transplantable teratocarcinomas from intratesticular grafts of pre- and postimplantation mouse embryos. Developmental Biology. 1970 Mar;21(3):364-382
  16. 16. Strickland S, Mahdavi V. The induction of differentiation in teratocarcinoma stem cells by retinoic acid. Cell. 1978 Oct;15(2):393-403
  17. 17. Donovan PJ, De Miguel MP. The role of the C-Kit/Kit ligand axis in mammalian gametogenesis. In: Matzuk MM, CW Brown, TR Kumar, editors. Contemporary Endocrinology: Transgenics in Endocrinology. Totowa, NJ: Humana Press Inc.; 2001. pp. 147-163
  18. 18. Noguchi T, Noguchi M. A recessive mutation (ter) causing germ cell deficiency and a high incidence of congenital testicular teratomas in 129/Sv-ter mice. Journal of the National Cancer Institute. 1985 Aug;75(2):385-392
  19. 19. Sekita Y, Nakamura T, Kimura T. Reprogramming of germ cells into pluripotency. World Journal of Stem Cells. 2016 Aug 26;8(8):251-259
  20. 20. Kimura T, Murayama K, Nakamura T, Watanabe S, Umehara H, Tomooka M, et al. Testicular teratomas: Back to pluripotent stem cells. Progress in Molecular and Subcellular Biology. 2005;40:133-150
  21. 21. Kimura T, Suzuki A, Fujita Y, Yomogida K, Lomeli H, Asada N, et al. Conditional loss of PTEN leads to testicular teratoma and enhances embryonic germ cell production. Development. 2003 Apr;130(8):1691-1700
  22. 22. Sotillo R, Dubus P, Martin J, de la Cueva E, Ortega S, Malumbres M, et al. Wide spectrum of tumors in knock-in mice carrying a Cdk4 protein insensitive to INK4 inhibitors. The EMBO Journal. 2001 Dec 03;20(23):6637-6647
  23. 23. Almstrup K, Hoei-Hansen CE, Nielsen JE, Wirkner U, Ansorge W, Skakkebaek NE, et al. Genome-wide gene expression profiling of testicular carcinoma in situ progression into overt tumours. British Journal of Cancer. 2005 May 23;92(10):1934-1941
  24. 24. von Eyben FE. Chromosomes, genes, and development of testicular germ cell tumors. Cancer Genetics and Cytogenetics. 2004 Jun;151(2):93-138
  25. 25. Rajpert-De Meyts E, Hanstein R, Jorgensen N, Graem N, Vogt PH, Skakkebaek NE. Developmental expression of POU5F1 (OCT-3/4) in normal and dysgenetic human gonads. Human Reproduction. 2004 Jun;19(6):1338-1344
  26. 26. de Jong J, Stoop H, Gillis AJ, van Gurp RJ, van de Geijn GJ, Boer M, et al. Differential expression of SOX17 and SOX2 in germ cells and stem cells has biological and clinical implications. The Journal of Pathology. 2008 May;215(1):21-30
  27. 27. Looijenga LH, Oosterhuis JW. Pathogenesis of testicular germ cell tumours. Reviews of Reproduction. 1999 May;4(2):90-100
  28. 28. Kristensen DM, Sonne SB, Ottesen AM, Perrett RM, Nielsen JE, Almstrup K, et al. Origin of pluripotent germ cell tumours: The role of microenvironment during embryonic development. Molecular and Cellular Endocrinology. 2008 Jun 25;288(1-2):111-118
  29. 29. Nistal M, Gonzalez-Peramato P, Regadera J, Serrano A, Tarin V, De Miguel MP. Primary testicular lesions are associated with testicular germ cell tumors of adult men. The American Journal of Surgical Pathology. 2006 Oct;30(10):1260-1268
  30. 30. Moe-Behrens GH, Klinger FG, Eskild W, Grotmol T, Haugen TB, De Felici M. Akt/PTEN signaling mediates estrogen-dependent proliferation of primordial germ cells in vitro. Molecular Endocrinology. 2003 Dec;17(12):2630-2638
  31. 31. Nistal M, Gonzalez-Peramato P, De Miguel MP. Sertoli cell dedifferentiation in human cryptorchidism and gender reassignment shows similarities between fetal environmental and adult medical treatment estrogen and antiandrogen exposure. Reproductive Toxicology. 2013 Dec;42:172-179
  32. 32. Shamblott MJ, Axelman J, Wang S, Bugg EM, Littlefield JW, Donovan PJ, et al. Derivation of pluripotent stem cells from cultured human primordial germ cells. Proceedings of the National Academy of Sciences of the United States of America. 1998 Nov 10;95(23):13726-13731
  33. 33. De Miguel MP, Donovan PJ. Isolation and culture of mouse germ cells. Methods in Molecular Biology. 2000;137:403-408
  34. 34. Miguel MPD, Kerr CL, López-Iglesias P, Alcaina Y. Techniques and conditions for embryonic germ cell derivation and culture. In: Kallos PM, editor. Embryonic Stem Cells - Basic Biology to Bioengineering. InTech; 2011;22:425-448, Michael S. Kallos (ed). Intech publishing, Rijeka, ISBN: 978-953-307-278-4. DOI: 10.5772/24632
  35. 35. Surani MA, Hayashi K, Hajkova P. Genetic and epigenetic regulators of pluripotency. Cell. 2007 Feb 23;128(4):747-762
  36. 36. Nichols J, Smith A. Naive and primed pluripotent states. Cell Stem Cell. 2009 Jun 05;4(6):487-492
  37. 37. Tesar PJ, Chenoweth JG, Brook FA, Davies TJ, Evans EP, Mack DL, et al. New cell lines from mouse epiblast share defining features with human embryonic stem cells. Nature. 2007;448(7150):196-199
  38. 38. Shamblott MJ, Axelman J, Littlefield JW, Blumenthal PD, Huggins GR, Cui Y, et al. Human embryonic germ cell derivatives express a broad range of developmentally distinct markers and proliferate extensively in vitro. Proceedings of the National Academy of Sciences of the United States of America. 2001 Jan 02;98(1):113-118
  39. 39. Durcova-Hills G, Tang F, Doody G, Tooze R, Surani MA. Reprogramming primordial germ cells into pluripotent stem cells. PloS One. 2008;3(10):e3531
  40. 40. Kimura T, Tomooka M, Yamano N, Murayama K, Matoba S, Umehara H, et al. AKT signaling promotes derivation of embryonic germ cells from primordial germ cells. Development. 2008 Mar;135(5):869-879
  41. 41. Kimura T, Kaga Y, Sekita Y, Fujikawa K, Nakatani T, Odamoto M, et al. Pluripotent stem cells derived from mouse primordial germ cells by small molecule compounds. Stem Cells. 2015 Jan;33(1):45-55
  42. 42. Nagamatsu G, Kosaka T, Saito S, Takubo K, Akiyama H, Sudo T, et al. Tracing the conversion process from primordial germ cells to pluripotent stem cells in mice. Biology of Reproduction. 2012 Jun;86(6):182
  43. 43. Samavarchi-Tehrani P, Golipour A, David L, Sung HK, Beyer TA, Datti A, et al. Functional genomics reveals a BMP-driven mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming. Cell Stem Cell. 2010 Jul 2;7(1):64-77
  44. 44. Ohinata Y, Payer B, O’Carroll D, Ancelin K, Ono Y, Sano M, et al. Blimp1 is a critical determinant of the germ cell lineage in mice. Nature. 2005 Jul 14;436(7048):207-213
  45. 45. Nagamatsu G, Saito S, Takubo K, Suda T. Integrative analysis of the acquisition of pluripotency in PGCs reveals the mutually exclusive roles of blimp-1 and AKT signaling. Stem Cell Reports. 2015 Jul 14;5(1):111-124
  46. 46. Krizhanovsky V, Lowe SW. Stem cells: The promises and perils of p53. Nature. 2009 Aug 27;460(7259):1085-1086
  47. 47. Hanna J, Saha K, Pando B, van Zon J, Lengner CJ, Creyghton MP, et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature. 2009 Dec 03;462(7273):595-601
  48. 48. Leitch HGN, Nichols J, Humphreys P, Mulas C, Martello G, Lee C, Jones K, Surani MA, Smith A. Rebuilding pluripotency from primordial germ cells. Stem Cell Reports. 2013;1(1):66-78
  49. 49. Koshimizu U, Taga T, Watanabe M, Saito M, Shirayoshi Y, Kishimoto T, et al. Functional requirement of gp130-mediated signaling for growth and survival of mouse primordial germ cells in vitro and derivation of embryonic germ (EG) cells. Development. 1996;122(4):1235-1242
  50. 50. De Miguel MP, Cheng L, Holland EC, Federspiel MJ, Donovan PJ. Dissection of the c-Kit signaling pathway in mouse primordial germ cells by retroviral-mediated gene transfer. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(16):10458-10463
  51. 51. Manning BD, Cantley LC. AKT/PKB signaling: Navigating downstream. Cell. 2007 Jun 29;129(7):1261-1274
  52. 52. Leitch HG, Blair K, Mansfield W, Ayetey H, Humphreys P, Nichols J, et al. Embryonic germ cells from mice and rats exhibit properties consistent with a generic pluripotent ground state. Development. 2010;137(14):2279-2287
  53. 53. Silva J, Barrandon O, Nichols J, Kawaguchi J, Theunissen TW, Smith A. Promotion of reprogramming to ground state pluripotency by signal inhibition. PLoS biology. 2008 Oct 21;6(10):e253
  54. 54. Eiselleova L, Matulka K, Kriz V, Kunova M, Schmidtova Z, Neradil J, et al. A complex role for FGF-2 in self-renewal, survival, and adhesion of human embryonic stem cells. Stem Cells. 2009 Aug;27(8):1847-1857
  55. 55. Hackett JA, Surani MA. Regulatory principles of pluripotency: From the ground state up. Cell Stem Cell. 2014 Oct 02;15(4):416-430
  56. 56. Ye S, Zhang T, Tong C, Zhou X, He K, Ban Q, et al. Depletion of Tcf3 and Lef1 maintains mouse embryonic stem cell self-renewal. Biology Open. 2017 Apr 15;6(4):511-517
  57. 57. Donovan PJ, De Miguel MP, Hirano MP, Parsons MS, Lincoln AJ. Germ cell biology–from generation to generation. The International Journal of Developmental Biology. 2001;45(3):523-531
  58. 58. Durcova-Hills G, Adams IR, Barton SC, Surani MA, McLaren A. The role of exogenous fibroblast growth factor-2 on the reprogramming of primordial germ cells into pluripotent stem cells. Stem Cells. 2006;24(6):1441-1449
  59. 59. Godin I, Wylie CC. TGF beta 1 inhibits proliferation and has a chemotropic effect on mouse primordial germ cells in culture. Development. 1991 Dec;113(4):1451-1457
  60. 60. Tan F, Qian C, Tang K, Abd-Allah SM, Jing N. Inhibition of transforming growth factor beta (TGF-beta) signaling can substitute for Oct4 protein in reprogramming and maintain pluripotency. The Journal of Biological Chemistry. 2015 Feb 13;290(7):4500-4511
  61. 61. Ichida JK, Blanchard J, Lam K, Son EY, Chung JE, Egli D, et al. A small-molecule inhibitor of tgf-Beta signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell. 2009 Nov 6;5(5):491-503
  62. 62. Attari F, Sepehri H, Ansari H, Hassani SN, Esfandiari F, Asgari B, et al. Efficient induction of pluripotency in primordial germ cells by dual inhibition of TGF-beta and ERK signaling pathways. Stem Cells and Development. 2014 May 15;23(10):1050-1061
  63. 63. Yoshida Y, Takahashi K, Okita K, Ichisaka T, Yamanaka S. Hypoxia enhances the generation of induced pluripotent stem cells. Cell Stem Cell. 2009 Sep 4;5(3):237-241
  64. 64. Krivtsov AV, Twomey D, Feng Z, Stubbs MC, Wang Y, Faber J, et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 2006 Aug 17;442(7104):818-822
  65. 65. De Miguel MP, Alcaina Y, de la Maza DS, Lopez-Iglesias P. Cell metabolism under microenvironmental low oxygen tension levels in stemness, proliferation and pluripotency. Current Molecular Medicine. 2015;15(4):343-359
  66. 66. Folmes CD, Nelson TJ, Martinez-Fernandez A, Arrell DK, Lindor JZ, Dzeja PP, et al. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming. Cell Metabolism. 2011 Aug 3;14(2):264-271
  67. 67. Kondoh H, Lleonart ME, Nakashima Y, Yokode M, Tanaka M, Bernard D, et al. A high glycolytic flux supports the proliferative potential of murine embryonic stem cells. Antioxidants & Redox Signaling. 2007;9(3):293-299
  68. 68. Kawauchi K, Araki K, Tobiume K, Tanaka N. p53 regulates glucose metabolism through an IKK-NF-kappaB pathway and inhibits cell transformation. Nature Cell Biology. 2008 May;10(5):611-618
  69. 69. Mathieu J, Zhang Z, Nelson A, Lamba DA, Reh TA, Ware C, et al. Hypoxia induces re-entry of committed cells into pluripotency. Stem Cells. 2013 Sep;31(9):1737-1748
  70. 70. Simon MC, Keith B. The role of oxygen availability in embryonic development and stem cell function. Nature Reviews Molecular Cell Biology. 2008 Apr;9(4):285-296
  71. 71. Shaw RJ. Glucose metabolism and cancer. Current Opinion in Cell Biology. 2006;18(6):598-608
  72. 72. Yu Y, Liang D, Tian Q, Chen X, Jiang B, Chou BK, et al. Stimulation of somatic cell reprogramming by ERas-Akt-FoxO1 signaling axis. Stem Cells. 2014 Feb;32(2):349-363
  73. 73. Sterneckert J, Hoing S, Scholer HR. Concise review: Oct4 and more: The reprogramming expressway. Stem Cells. 2012 Jan;30(1):15-21
  74. 74. Mottet D, Dumont V, Deccache Y, Demazy C, Ninane N, Raes M, et al. Regulation of hypoxia-inducible factor-1alpha protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta pathway in HepG2 cells. The Journal of Biological Chemistry. 2003;278(33):31277-31285
  75. 75. Fujimoto T, Miyayama Y, Fuyuta M. The origin, migration and fine morphology of human primordial germ cells. The Anatomical Record. 1977 Jul;188(3):315-330
  76. 76. Radzisheuskaya A, Chia Gle B, dos Santos RL, Theunissen TW, Castro LF, Nichols J, et al. A defined Oct4 level governs cell state transitions of pluripotency entry and differentiation into all embryonic lineages. Nature Cell Biology. 2013 Jun;15(6):579-590
  77. 77. Karwacki-Neisius V, Goke J, Osorno R, Halbritter F, Ng JH, Weisse AY, et al. Reduced Oct4 expression directs a robust pluripotent state with distinct signaling activity and increased enhancer occupancy by Oct4 and Nanog. Cell Stem Cell. 2013 May 2;12(5):531-545
  78. 78. Kim H, Jang H, Kim TW, Kang BH, Lee SE, Jeon YK, et al. Core pluripotency factors directly regulate metabolism in embryonic stem cell to maintain pluripotency. Stem Cells. 2015 Sep;33(9):2699-2711
  79. 79. Forristal CE, Wright KL, Hanley NA, Oreffo RO, Houghton FD. Hypoxia inducible factors regulate pluripotency and proliferation in human embryonic stem cells cultured at reduced oxygen tensions. Reproduction. 2010 Jan;139(1):85-97
  80. 80. Liu X, Duan H, Zhou S, Liu Z, Wu D, Zhao T, et al. microRNA-199a-3p functions as tumor suppressor by regulating glucose metabolism in testicular germ cell tumors. Molecular Medicine Reports. 2016 Sep;14(3):2311-2320
  81. 81. Mandal S, Lindgren AG, Srivastava AS, Clark AT, Banerjee U. Mitochondrial function controls proliferation and early differentiation potential of embryonic stem cells. Stem Cells. 2011 Mar;29(3):486-495
  82. 82. Westfall SD, Sachdev S, Das P, Hearne LB, Hannink M, Roberts RM, et al. Identification of oxygen-sensitive transcriptional programs in human embryonic stem cells. Stem Cells and Development. 2008 Oct;17(5):869-881
  83. 83. Lengner CJ, Gimelbrant AA, Erwin JA, Cheng AW, Guenther MG, Welstead GG, et al. Derivation of pre-X inactivation human embryonic stem cells under physiological oxygen concentrations. Cell. 2010 May 28;141(5):872-883
  84. 84. Motta PM, Nottola SA, Makabe S, Heyn R. Mitochondrial morphology in human fetal and adult female germ cells. Human Reproduction. 2000 Jul;15(Suppl 2):129-147
  85. 85. Zhang H, Bosch-Marce M, Shimoda LA, Tan YS, Baek JH, Wesley JB, et al. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. The Journal of Biological Chemistry. 2008 Apr 18;283(16):10892-10903
  86. 86. Wang S, Xia P, Ye B, Huang G, Liu J, Fan Z. Transient activation of autophagy via Sox2-mediated suppression of mTOR is an important early step in reprogramming to pluripotency. Cell Stem Cell. 2013 Nov 07;13(5):617-625
  87. 87. Polesskaya A, Cuvellier S, Naguibneva I, Duquet A, Moss EG, Harel-Bellan A. Lin-28 binds IGF-2 mRNA and participates in skeletal myogenesis by increasing translation efficiency. Genes & Development. 2007 May 01;21(9):1125-1138
  88. 88. Peng S, Chen LL, Lei XX, Yang L, Lin H, Carmichael GG, et al. Genome-wide studies reveal that Lin28 enhances the translation of genes important for growth and survival of human embryonic stem cells. Stem Cells. 2011 Mar;29(3):496-504
  89. 89. Zhu H, Shyh-Chang N, Segre AV, Shinoda G, Shah SP, Einhorn WS, et al. The Lin28/let-7 axis regulates glucose metabolism. Cell. 2011 Sep 30;147(1):81-94
  90. 90. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007 Dec 21;318(5858):1917-1920
  91. 91. Qiu C, Ma Y, Wang J, Peng S, Huang Y. Lin28-mediated post-transcriptional regulation of Oct4 expression in human embryonic stem cells. Nucleic Acids Research. 2010 Mar;38(4):1240-1248
  92. 92. West JA, Viswanathan SR, Yabuuchi A, Cunniff K, Takeuchi A, Park IH, et al. A role for Lin28 in primordial germ-cell development and germ-cell malignancy. Nature. 2009 Aug 13;460(7257):909-913
  93. 93. Irie N, Weinberger L, Tang WW, Kobayashi T, Viukov S, Manor YS, et al. SOX17 is a critical specifier of human primordial germ cell fate. Cell. 2015 Jan 15;160(1-2):253-268
  94. 94. Guo F, Yan L, Guo H, Li L, Hu B, Zhao Y, et al. The transcriptome and DNA methylome landscapes of human primordial germ cells. Cell. 2015 Jun 04;161(6):1437-1452
  95. 95. Seki Y, Yamaji M, Yabuta Y, Sano M, Shigeta M, Matsui Y, et al. Cellular dynamics associated with the genome-wide epigenetic reprogramming in migrating primordial germ cells in mice. Development. 2007 Jul;134(14):2627-2638
  96. 96. Yabuta Y, Kurimoto K, Ohinata Y, Seki Y, Saitou M. Gene expression dynamics during germline specification in mice identified by quantitative single-cell gene expression profiling. Biology of Reproduction. 2006 Nov;75(5):705-716
  97. 97. Yamaji M, Seki Y, Kurimoto K, Yabuta Y, Yuasa M, Shigeta M, et al. Critical function of Prdm14 for the establishment of the germ cell lineage in mice. Nature Genetics. 2008 Aug;40(8):1016-1022
  98. 98. Sugawa F, Arauzo-Bravo MJ, Yoon J, Kim KP, Aramaki S, Wu G, et al. Human primordial germ cell commitment in vitro associates with a unique PRDM14 expression profile. The EMBO Journal. 2015 Apr 15;34(8):1009-1024
  99. 99. Sasaki H, Matsui Y. Epigenetic events in mammalian germ-cell development: Reprogramming and beyond. Nature Reviews Genetics. 2008 Feb;9(2):129-140
  100. 100. Ancelin K, Lange UC, Hajkova P, Schneider R, Bannister AJ, Kouzarides T, et al. Blimp1 associates with Prmt5 and directs histone arginine methylation in mouse germ cells. Nature Cell Biology. 2006 Jun;8(6):623-630
  101. 101. Hajkova P, Jeffries SJ, Lee C, Miller N, Jackson SP, Surani MA. Genome-wide reprogramming in the mouse germ line entails the base excision repair pathway. Science. 2010 Jul 02;329(5987):78-82
  102. 102. De Felici M. Nuclear reprogramming in mouse primordial germ cells: Epigenetic contribution. Stem Cells International. 2011;2011:425863
  103. 103. Hajkova P, Ancelin K, Waldmann T, Lacoste N, Lange UC, Cesari F, et al. Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature. 2008 Apr 17;452(7189):877-881
  104. 104. Gunesdogan U, Magnusdottir E, Surani MA. Primordial germ cell specification: A context-dependent cellular differentiation event [corrected]. Philosophical transactions of the Royal Society of London Series B, Biological sciences. 2014 Dec 5;369:1657-1665
  105. 105. Su ST, Ying HY, Chiu YK, Lin FR, Chen MY, Lin KI. Involvement of histone demethylase LSD1 in Blimp-1-mediated gene repression during plasma cell differentiation. Molecular and Cellular Biology. 2009 Mar;29(6):1421-1431
  106. 106. Perrett RM, Turnpenny L, Eckert JJ, O’Shea M, Sonne SB, Cameron IT, et al. The early human germ cell lineage does not express SOX2 during in vivo development or upon in vitro culture. Biology of Reproduction. 2008 May;78(5):852-858
  107. 107. Mansour AA, Gafni O, Weinberger L, Zviran A, Ayyash M, Rais Y, et al. The H3K27 demethylase Utx regulates somatic and germ cell epigenetic reprogramming. Nature. 2012 Aug 16;488(7411):409-413
  108. 108. Guibert S, Forne T, Weber M. Global profiling of DNA methylation erasure in mouse primordial germ cells. Genome Research. 2012 Apr;22(4):633-641
  109. 109. Seisenberger S, Andrews S, Krueger F, Arand J, Walter J, Santos F, et al. The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Molecular Cell. 2012 Dec 28;48(6):849-862
  110. 110. Stewart KR, Veselovska L, Kelsey G. Establishment and functions of DNA methylation in the germline. Epigenomics. 2016 Oct;8(10):1399-1413
  111. 111. Smallwood SA, Tomizawa S, Krueger F, Ruf N, Carli N, Segonds-Pichon A, et al. Dynamic CpG island methylation landscape in oocytes and preimplantation embryos. Nature Genetics. 2011 Jun 26;43(8):811-814
  112. 112. Eguizabal C, Herrera L, De Onate L, Montserrat N, Hajkova P, Izpisua Belmonte JC. Characterization of the epigenetic changes during human gonadal primordial germ cells reprogramming. Stem Cells. 2016 Sep;34(9):2418-2428
  113. 113. Hyldig SM, Ostrup O, Vejlsted M, Thomsen PD. Changes of DNA methylation level and spatial arrangement of primordial germ cells in embryonic day 15 to embryonic day 28 pig embryos. Biology of Reproduction. 2011 Jun;84(6):1087-1093
  114. 114. Wermann H, Stoop H, Gillis AJ, Honecker F, van Gurp RJ, Ammerpohl O, et al. Global DNA methylation in fetal human germ cells and germ cell tumours: Association with differentiation and cisplatin resistance. The Journal of Pathology. 2010 Aug;221(4):433-442
  115. 115. Almstrup K, Nielsen JE, Mlynarska O, Jansen MT, Jorgensen A, Skakkebaek NE, et al. Carcinoma in situ testis displays permissive chromatin modifications similar to immature foetal germ cells. British Journal of Cancer. 2010 Oct 12;103(8):1269-1276
  116. 116. Bartkova J, Moudry P, Hodny Z, Lukas J, Rajpert-De Meyts E, Bartek J. Heterochromatin marks HP1gamma, HP1alpha and H3K9me3, and DNA damage response activation in human testis development and germ cell tumours. International Journal of Andrology. 2011 Aug;34(4 Pt 2):e103-e113
  117. 117. Gkountela S, Zhang KX, Shafiq TA, Liao WW, Hargan-Calvopina J, Chen PY, et al. DNA demethylation dynamics in the human prenatal germline. Cell. 2015 Jun 04;161(6):1425-1436
  118. 118. Guo H, Hu B, Yan L, Yong J, Wu Y, Gao Y, et al. DNA methylation and chromatin accessibility profiling of mouse and human fetal germ cells. Cell Research. 2017 Feb;27(2):165-183
  119. 119. Tada M, Takahama Y, Abe K, Nakatsuji N, Tada T. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Current Biology. 2001 Oct 02;11(19):1553-1558
  120. 120. Labosky PA, Barlow DP, Hogan BL. Mouse embryonic germ (EG) cell lines: Transmission through the germline and differences in the methylation imprint of insulin-like growth factor 2 receptor (Igf2r) gene compared with embryonic stem (ES) cell lines. Development. 1994 Nov;120(11):3197-3204
  121. 121. Tada T, Tada M, Hilton K, Barton SC, Sado T, Takagi N, et al. Epigenotype switching of imprintable loci in embryonic germ cells. Development Genes and Evolution. 1998 Feb;207(8):551-561
  122. 122. Shovlin TC, Durcova-Hills G, Surani A, McLaren A. Heterogeneity in imprinted methylation patterns of pluripotent embryonic germ cells derived from pre-migratory mouse germ cells. Developmental Biology. 2008 Jan 15;313(2):674-681
  123. 123. Leitch HG, Tang WW, Surani MA. Primordial germ-cell development and epigenetic reprogramming in mammals. Current Topics in Developmental Biology. 2013;104:149-187
  124. 124. Durcova-Hills G, Ainscough J, McLaren A. Pluripotential stem cells derived from migrating primordial germ cells. Differentiation. 2001 Oct;68(4-5):220-226
  125. 125. Lee J, Inoue K, Ono R, Ogonuki N, Kohda T, Kaneko-Ishino T, et al. Erasing genomic imprinting memory in mouse clone embryos produced from day 11.5 primordial germ cells. Development. 2002 Apr;129(8):1807-1817
  126. 126. Sharova LV, Sharov AA, Piao Y, Shaik N, Sullivan T, Stewart CL, et al. Global gene expression profiling reveals similarities and differences among mouse pluripotent stem cells of different origins and strains. Developmental Biology. 2007 Jul 15;307(2):446-459
  127. 127. Saitou M, Yamaji M. Primordial germ cells in mice. Cold Spring Harbor perspectives in biology. 2012 Nov 01;4(11):1-19
  128. 128. Surani MA, Durcova-Hills G, Hajkova P, Hayashi K, Tee WW. Germ line, stem cells, and epigenetic reprogramming. Cold Spring Harbor Symposia on Quantitative Biology. 2008;73:9-15
  129. 129. Sainz de la Maza et al. Metabolic reprogramming, autophagy, and reactive oxygen species are necessary for primordial germ cell reprogramming into pluripotency. Oxidative Medicine and Cellular Longevity. 2017;2017:Article ID 4745252. p. 17. DOI:10.1155/2017/4745252
  130. 130. Meissner A. Epigenetic modifications in pluripotent and differentiated cells. Nature Biotechnology. 2010 Oct;28(10):1079-1088
  131. 131. Gaspar-Maia A, Alajem A, Meshorer E, Ramalho-Santos M. Open chromatin in pluripotency and reprogramming. Nature Reviews Molecular Cell Biology. 2011 Jan;12(1):36-47
  132. 132. Hou P, Li Y, Zhang X, Liu C, Guan J, Li H, et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science. 2013 Aug 9;341(6146):651-654
  133. 133. Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, Chen AE, et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nature Biotechnology. 2008 Jul;26(7):795-797
  134. 134. Tonge PD, Corso AJ, Monetti C, Hussein SM, Puri MC, Michael IP, et al. Divergent reprogramming routes lead to alternative stem-cell states. Nature. 2014 Dec 11;516(7530):192-197
  135. 135. Chen X, Zhai Y, Yu D, Cui J, Hu JF, Li W. Valproic acid enhances iPSC induction from human bone marrow-derived cells through the suppression of reprogramming-induced senescence. Journal of Cellular Physiology. 2016 Aug;231(8):1719-1727
  136. 136. Stadtfeld M, Apostolou E, Akutsu H, Fukuda A, Follett P, Natesan S, et al. Aberrant silencing of imprinted genes on chromosome 12qF1 in mouse induced pluripotent stem cells. Nature. 2010 May 13;465(7295):175-181
  137. 137. Onder TT, Kara N, Cherry A, Sinha AU, Zhu N, Bernt KM, et al. Chromatin-modifying enzymes as modulators of reprogramming. Nature. 2012 Mar 04;483(7391):598-602
  138. 138. Mikkelsen TS, Hanna J, Zhang X, Ku M, Wernig M, Schorderet P, et al. Dissecting direct reprogramming through integrative genomic analysis. Nature. 2008 Jul 03;454(7200):49-55
  139. 139. Shi Y, Desponts C, Do JT, Hahm HS, Scholer HR, Ding S. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell. 2008 Nov 6;3(5):568-574
  140. 140. Chia NY, Chan YS, Feng B, Lu X, Orlov YL, Moreau D, et al. A genome-wide RNAi screen reveals determinants of human embryonic stem cell identity. Nature. 2010 Nov 11;468(7321):316-320
  141. 141. Donovan PJ, Stott D, Cairns LA, Heasman J, Wylie CC. Migratory and postmigratory mouse primordial germ cells behave differently in culture. Cell. 1986;44(6):831-838

Written By

Maria P. De Miguel, Yago Alcaina and Diego Sainz de la Maza

Submitted: 31 October 2016 Reviewed: 31 May 2017 Published: 20 December 2017