Germline development

Last updated

In developmental biology, the cells that give rise to the gametes are often set aside during embryonic cleavage. During development, these cells will differentiate into primordial germ cells, migrate to the location of the gonad, and form the germline of the animal.

Contents

Creation of germ plasm and primordial germ cells

Cleavage in most animals segregates cells containing germ plasm from other cells. The germ plasm effectively turns off gene expression to render the genome of the cell inert. Cells expressing germ plasm become primordial germ cells (PGCs) which will then give rise to the gametes. The germ line development in mammals, on the other hand, occurs by induction and not by an endogenous germ plasm.[ citation needed ]

Germ plasm in fruit fly

Germ plasm has been studied in detail in Drosophila. The posterior pole of the embryo contains necessary materials for the fertility of the fly. This cytoplasm, pole plasm, contains specialized materials called polar granules and the pole cells are the precursors to primordial germ cells.[ citation needed ]

Pole plasm is organized by and contains the proteins and mRNA of the posterior group genes (such as oskar, nanos gene, Tudor, vasa, and Valois). These genes play a role in germ line development to localize nanos mRNA to the posterior and localize germ cell determinants. Drosophila progeny with mutations in these genes fail to produce pole cells and are thus sterile, giving these mutations the name 'grandchildless'. The genes oskar, nanos and germ cell-less (gcl) have important roles. Oskar is sufficient to recruit the other genes to form functional germ plasm. Nanos is required to prevent mitosis and somatic differentiation and for the pole cells to migrate to function as PGCs (see next section). Gcl is necessary (but not sufficient) for pole cell formation. In addition to these genes, Pgc polar granule component blocks phosphorylation and consequently activation of RNA polymerase II and shuts down transcription.[ citation needed ]

Germ plasm in amphibians

Similar germ plasm has been identified in Amphibians in the polar cytoplasm at the vegetal pole. This cytoplasm moves to the bottom of the blastocoel and eventually ends up as its own subset of endodermal cells. While specified to produce germ cells, the germ plasm does not irreversibly commit these cells to produce gametes and no other cell type. [1] [2]

Migration of primordial germ cells

Fruit flies

The first phase of migration in Drosophila occurs when the pole cells move passively and infold into the midgut invagination. Active migration occurs through repellents and attractants. The expression of wunen in the endoderm repels the PGCs out. The expression of columbus and hedgehog attracts the PGCs to the mesodermal precursors of the gonad. Nanos is required during migration. Regardless of PGC injection site, PGCs are able to correctly migrate to their target sites.[ citation needed ]

Zebrafish

In zebrafish, the PGCs express two CXCR4 transmembrane receptor proteins. The signaling system involving this protein and its ligand, Sdf1, is necessary and sufficient to direct PGC migration in fish.

Frogs

In frogs, the PGCs migrate along the mesentery to the gonadal mesoderm facilitated by orientated extracellular matrix with fibronectin. There is also evidence for the CXCR4/Sdf1 system in frogs.[ citation needed ]

Birds

In birds, the PGCs arise from the epiblast and migrate to anteriorly of the primitive streak to the germinal crest. From there, they use blood vessels to find their way to the gonad. The CXCR4/Sdf1 system is also used, though may not be the only method necessary. [3]

Mammals

In the mouse, primordial germ cells (PGCs) arise in the posterior primitive streak of the embryo [4] and start to migrate around 6.25 days after conception. PGCs start to migrate to the embryonic endoderm and then to the hindgut and finally towards the future genital ridges where the somatic gonadal precursors reside. [4] [5] This migration requires a series of attractant and repellent cues as well as a number of adhesion molecules such as E-cadherin and β1-Integrin to guide the migration of PGCs. [4] Around 10 days post conception; the PGCs occupy the genital ridge [5] where they begin to lose their motility and polarized shape. [4]

Germline development in mammals

Mammalian PGCs are specified by signalling between cells (induction), rather than by the segregation of germ plasm as the embryo divides. [6] In mice, PGCs originate from the proximal epiblast, close to the extra-embryonic ectoderm (ExE), of the post-implantation embryo as early as embryonic day 6.5. [7] By E7.5 a founding population of approximately 40 PGCs are generated in this region of the epiblast in the developing mouse embryo. [8] [9] [10] The epiblast, however, also give rise to somatic cell lineages that make up the embryo proper; including the endoderm, ectoderm and mesoderm. [11] [12] [13] The specification of primordial germ cells in mammals is mainly attributed to the downstream functions of two signaling pathways; the BMP signaling pathway and the canonical WNT/β-catenin pathway. [7]

Bone morphogenetic protein 4 (BMP4) is released by the extra-embryonic ectoderm (ExE) at embryonic day 5.5 to 5.75 directly adjacent to the epiblast [6] and causes the region of the epiblast nearest to the ExE to express Blimp1 and Prdm14 in a dose-dependent manner. [14] This is evident as the number of PGCs forming in the epiblast decreases in proportion to the loss of BMP4 alleles. [15] BMP4 acts through its downstream intercellular transcription factors SMAD1 and SMAD5. [15] [16] [17] [18] [19] During approximately the same time, WNT3 starts to be expressed in the posterior visceral endoderm of the epiblast. [20] [21] WNT3 signalling has been shown to be essential in order for the epiblast to acquire responsiveness to the BMP4 signal from the ExE. [22] WNT3 mutants fail to establish a primordial germ cell population, but this can be restored with exogenous WNT activity. [23] The WNT3/β-catenin signalling pathway is essential for the expression of the transcription factor T (Brachyury), a transcription factor that was previously characterized somatic and mesoderm specific genes. [24] [25] T was recently found to be both necessary and sufficient to induce the expression of the known PGC specification genes Blimp1 and Prdm14. [23] The induction of Transcription Factor T was seen 12 hours after BMP/WNT signaling, as opposed to the 24 to 36 hours it took for Blimp1 and Prdm14 genes to be expressed. Transcription factor T acts upstream of BLIMP1 and Prdm14 in PGC specification by binding to the genes respective enhancer elements. [23] It is important to note that while T can activate the expression of Blimp1 and Prdm14 in the absence of both BMP4 and WNT3, pre-exposure of PGC progenitors to WNTs (without BMP4) prevents T from activating these genes. [23] Details on how BMP4 prevents T from inducing mesodermal genes, and only activate PGC specification genes, remain unclear.

Expression of Blimp1 is the earliest known marker of PGC specification. [26] A mutation in the Blimp1 gene results in the formation of PGC-like cells at embryonic day 8.5 that closely resemble their neighbouring somatic cells. [27] A central role of Blimp 1 is the induction of Tcfap2c, a helix-span helix transcription factor. [28] Tcfap2c mutants exhibited an early loss of primordial germ cells. [29] [30] Tcfap2c is thought to repress somatic gene expression, including the mesodermal marker Hoxb1. [30] So, Blimp1, Tcfap2c and Prdm14 together are able to activate and repress the transcription of all the necessary genes to regulate PGC specification. [14] Mutation of Prdm14 results in the formation of PGCs that are lost by embryonic day 11.5. [31] The loss of PGCs in the Prdm14 mutant is due to failure in global erasure of histone 3 methylation patterns. [32] Blimp1 and Prdm14 also elicit another epigenetic event that causes global DNA demethylation. [33]

Other notable genes positively regulated by Blimp1 and Prdm14 are: Sox2, Nanos3, Nanog, Stella and Fragilis. [14] At the same time, Blimp1 and Prdm14 also repress the transcription of programs that drive somatic differentiation by inhibiting transcription of the Hox family genes. [14] In this way, Blimp1 and Prdm14 drive PGC specification by promoting germ line development and potential pluripotency transcriptional programs while also keeping the cells from taking on a somatic fate. [14]

Generation of mammalian PGCs in vitro

With the vast knowledge about in-vivo PGC specification collected over the last few decades, several attempts to generate in-vitro PGCs from post-implantation epiblast were made. Various groups were able to successfully generate PGC-like cells, cultured in the presence of BMP4 and various cytokines. [15] The efficiency of this process was later enhanced by the addition of stem cell factor (SCF), epidermal growth factor (EGF), leukaemia inhibitory factor (LIF) and BMP8B. [34] PGC-like cells generated using this method can be transplanted into a gonad, where the differentiate, and are able to give viable gametes and offspring in vivo. [34] PGC-like cells can also be generated from naïve embryonic stem cells (ESCs) that are cultured for two days in the presence of FGF and Activin-A to adopt an epiblast-like state. These cells are then cultured with BMP4, BMP8B, EGF, LIF and SCF and various cytokines for four more days. [35] These in-vitro generated PGCs can also develop into viable gametes and offspring. [35]

Differentiation of primordial germ cells

Prior to their arrival at the gonads, PGCs express pluripotency factors, generate pluripotent cell lines in cell culture (known as EG cells, [36] [37] ) and can produce multi-lineage tumors, known as teratomas. [38] Similar findings in other vertebrates indicate that PGCs are not yet irreversibly committed to produce gametes, and no other cell type. [1] [39] [40] On arrival at the gonads, human and mouse PGCs activate widely conserved germ cell-specific factors, and subsequently down-regulate the expression of pluripotency factors. [41] This transition results in the determination of germ cells, a form of cell commitment that is no longer reversible. [42]

Prior to their occupation of the genital ridge, there is no known difference between XX and XY PGCs. [4] However, once migration is complete and germ cell determination has occurred, these germline cells begin to differentiate according to the gonadal niche.

Early male differentiation

Male PGCs become known as gonocytes once they cease migration and undergo mitosis. [43] The term gonocyte is generally used to describe all stages post PGC until the gonocytes differentiate into spermatogonia. [43] Anatomically, gonocytes can be identified as large, euchromatic cells that often have two nucleoli in the nucleus. [43]

In the male genital ridge, transient Sry expression causes supporting cells to differentiate into Sertoli cells which then act as the organizing center for testis differentiation. Point mutations or deletions in the human or mouse Sry coding region can lead to female development in XY individuals. [44] Sertoli cells also act to prevent gonocytes from differentiating prematurely. [45] They produce the enzyme CYP26B1 to counteract surrounding retinoic acid. Retinoic acid acts as a signal to the gonocytes to enter meiosis. [45] The gonocyte and Sertoli cells have been shown to form gap and desmosomelike junctions as well as adherins junctions composed of cadherins and connexins. [43] To differentiate into spermatogonia, the gonocytes must lose their junctions to Sertoli cells and become migratory once again. [43] They migrate to the basement membrane of the seminiferous cord [43] and differentiate.

Late differentiation

In the gonads, the germ cells undergo either spermatogenesis or oogenesis depending on whether the sex is male or female respectively.[ citation needed ]

Spermatogenesis

Mitotic germ stem cells, spermatogonia, divide by mitosis to produce spermatocytes committed to meiosis. The spermatocytes divide by meiosis to form spermatids. The post-meiotic spermatids differentiate through spermiogenesis to become mature and functional spermatozoa.[ citation needed ] Spermatogenic cells at different stages of development in the mouse have a frequency of mutation that is 5 to 10-fold lower than the mutation frequency in somatic cells. [46]

In Drosophila , the ability of premeiotic male germ line cells to repair double-strand breaks declines dramatically with age. [47] In mouse, spermatogenesis declines with advancing paternal age likely due to an increased frequency of meiotic errors. [48]

Oogenesis

Mitotic germ stem cells, oogonia, divide by mitosis to produce primary oocytes committed to meiosis. Unlike sperm production, oocyte production is not continuous. These primary oocytes begin meiosis but pause in diplotene of meiosis I while in the embryo. All of the oogonia and many primary oocytes die before birth. After puberty in primates, small groups of oocytes and follicles prepare for ovulation by advancing to metaphase II. Only after fertilization is meiosis completed. Meiosis is asymmetric producing polar bodies and oocytes with large amounts of material for embryonic development.[ citation needed ] The mutation frequency of female mouse germ line cells, like male germ line cells, is also lower than that of somatic cells. [49] Low germ line mutation frequency appears to be due, in part, to elevated levels of DNA repair enzymes that remove potentially mutagenic DNA damages. Enhanced genetic integrity may be a fundamental characteristic of germ line development. [49]

See also

Related Research Articles

<span class="mw-page-title-main">Mesoderm</span> Middle germ layer of embryonic development

The mesoderm is the middle layer of the three germ layers that develops during gastrulation in the very early development of the embryo of most animals. The outer layer is the ectoderm, and the inner layer is the endoderm.

<span class="mw-page-title-main">Gastrulation</span> Stage in embryonic development in which germ layers form

Gastrulation is the stage in the early embryonic development of most animals, during which the blastula, or in mammals the blastocyst is reorganized into a multilayered structure known as the gastrula. Before gastrulation, the embryo is a continuous epithelial sheet of cells; by the end of gastrulation, the embryo has begun differentiation to establish distinct cell lineages, set up the basic axes of the body, and internalized one or more cell types including the prospective gut.

<span class="mw-page-title-main">Germ cell</span> Gamete-producing cell

A germ cell is any cell that gives rise to the gametes of an organism that reproduces sexually. In many animals, the germ cells originate in the primitive streak and migrate via the gut of an embryo to the developing gonads. There, they undergo meiosis, followed by cellular differentiation into mature gametes, either eggs or sperm. Unlike animals, plants do not have germ cells designated in early development. Instead, germ cells can arise from somatic cells in the adult, such as the floral meristem of flowering plants.

<span class="mw-page-title-main">Germline</span> Population of a multicellular organisms cells that pass on their genetic material to the progeny

In biology and genetics, the germline is the population of a multicellular organism's cells that pass on their genetic material to the progeny (offspring). In other words, they are the cells that form the egg, sperm and the fertilised egg. They are usually differentiated to perform this function and segregated in a specific place away from other bodily cells.

<span class="mw-page-title-main">Blastocyst</span> Structure formed around day 5 of mammalian embryonic development

The blastocyst is a structure formed in the early embryonic development of mammals. It possesses an inner cell mass (ICM) also known as the embryoblast which subsequently forms the embryo, and an outer layer of trophoblast cells called the trophectoderm. This layer surrounds the inner cell mass and a fluid-filled cavity known as the blastocoel. In the late blastocyst the trophectoderm is known as the trophoblast. The trophoblast gives rise to the chorion and amnion, the two fetal membranes that surround the embryo. The placenta derives from the embryonic chorion and the underlying uterine tissue of the mother.

A germ layer is a primary layer of cells that forms during embryonic development. The three germ layers in vertebrates are particularly pronounced; however, all eumetazoans produce two or three primary germ layers. Some animals, like cnidarians, produce two germ layers making them diploblastic. Other animals such as bilaterians produce a third layer between these two layers, making them triploblastic. Germ layers eventually give rise to all of an animal's tissues and organs through the process of organogenesis.

<span class="mw-page-title-main">Oct-4</span> Mammalian protein found in Homo sapiens

Oct-4, also known as POU5F1, is a protein that in humans is encoded by the POU5F1 gene. Oct-4 is a homeodomain transcription factor of the POU family. It is critically involved in the self-renewal of undifferentiated embryonic stem cells. As such, it is frequently used as a marker for undifferentiated cells. Oct-4 expression must be closely regulated; too much or too little will cause differentiation of the cells.

Gametogonium are stem cells for gametes located within the gonads. They originate from primordial germ cells, which have migrated to the gonads. Male gametogonia which are located within the testes during development and adulthood are called spermatogonium. Female gametogonia, known as oogonium, are found within the ovaries of the developing foetus and were thought to be depleted at or after birth. Spermatogonia and oogonia are classified as sexually differentiated germ cells.

An oogonium is a small diploid cell which, upon maturation, forms a primordial follicle in a female fetus or the female gametangium of certain thallophytes.

In biology, reprogramming refers to erasure and remodeling of epigenetic marks, such as DNA methylation, during mammalian development or in cell culture. Such control is also often associated with alternative covalent modifications of histones.

<span class="mw-page-title-main">Epiblast</span> Embryonic inner cell mass tissue that forms the embryo itself, through the three germ layers

In amniote embryonic development, the epiblast is one of two distinct cell layers arising from the inner cell mass in the mammalian blastocyst, or from the blastula in reptiles and birds, the other layer is the hypoblast. It drives the embryo proper through its differentiation into the three primary germ layers, ectoderm, mesoderm and endoderm, during gastrulation. The amniotic ectoderm and extraembryonic mesoderm also originate from the epiblast.

<span class="mw-page-title-main">Bone morphogenetic protein 4</span> Human protein and coding gene

Bone morphogenetic protein 4 is a protein that in humans is encoded by BMP4 gene. BMP4 is found on chromosome 14q22-q23.

<span class="mw-page-title-main">Fish development</span>

The development of fishes is unique in some specific aspects compared to the development of other animals.

<span class="mw-page-title-main">Hypoblast</span> Embryonic inner cell mass tissue that forms the yolk sac and, later, chorion

In amniote embryology, the hypoblast is one of two distinct layers arising from the inner cell mass in the mammalian blastocyst, or from the blastodisc in reptiles and birds. The hypoblast gives rise to the yolk sac, which in turn gives rise to the chorion.

Gonocytes are the precursors of spermatogonia that differentiate in the testis from primordial germ cells around week 7 of embryonic development and exist up until the postnatal period, when they become spermatogonia. Despite some uses of the term to refer to the precursors of oogonia, it was generally restricted to male germ cells. Germ cells operate as vehicles of inheritance by transferring genetic and epigenetic information from one generation to the next. Male fertility is centered around continual spermatogonia which is dependent upon a high stem cell population. Thus, the function and quality of a differentiated sperm cell is dependent upon the capacity of its originating spermatogonial stem cell (SSC).

<span class="mw-page-title-main">Cell potency</span> Ability of a cell to differentiate into other cell types

Cell potency is a cell's ability to differentiate into other cell types. The more cell types a cell can differentiate into, the greater its potency. Potency is also described as the gene activation potential within a cell, which like a continuum, begins with totipotency to designate a cell with the most differentiation potential, pluripotency, multipotency, oligopotency, and finally unipotency.

Vasa is an RNA binding protein with an ATP-dependent RNA helicase that is a member of the DEAD box family of proteins. The vasa gene is essential for germ cell development and was first identified in Drosophila melanogaster, but has since been found to be conserved in a variety of vertebrates and invertebrates including humans. The Vasa protein is found primarily in germ cells in embryos and adults, where it is involved in germ cell determination and function, as well as in multipotent stem cells, where its exact function is unknown.

<span class="mw-page-title-main">Spermatogonial stem cell</span> Spermatogonium that does not differentiate into a spermatocyte

A spermatogonial stem cell (SSC), also known as a type A spermatogonium, is a spermatogonium that does not differentiate into a spermatocyte, a precursor of sperm cells. Instead, they continue dividing into other spermatogonia or remain dormant to maintain a reserve of spermatogonia. Type B spermatogonia, on the other hand, differentiate into spermatocytes, which in turn undergo meiosis to eventually form mature sperm cells.

<span class="mw-page-title-main">Primordial germ cell migration</span>

Primordial germ cell (PGC) migration is the process of distribution of primordial germ cells throughout the embryo during embryogenesis.

The germ cell nest forms in the ovaries during their development. The nest consists of multiple interconnected oogonia formed by incomplete cell division. The interconnected oogonia are surrounded by somatic cells called granulosa cells. Later on in development, the germ cell nests break down through invasion of granulosa cells. The result is individual oogonia surrounded by a single layer of granulosa cells. There is also a comparative germ cell nest structure in the developing spermatogonia, with interconnected intracellular cytoplasmic bridges.

References

  1. 1 2 Wylie CC, Holwill S, O'Driscoll M, Snape A, Heasman J (1985-01-01). "Germ plasm and germ cell determination in Xenopus laevis as studied by cell transplantation analysis". Cold Spring Harbor Symposia on Quantitative Biology. 50: 37–43. doi:10.1101/SQB.1985.050.01.007. PMID   3868485.
  2. Strome S, Updike D (July 2015). "Specifying and protecting germ cell fate". Nature Reviews. Molecular Cell Biology. 16 (7): 406–16. doi:10.1038/nrm4009. PMC   4698964 . PMID   26122616.
  3. Lee, JH; Park, JW; Kim, SW; Park, J; Park, TS (15 December 2017). "C-X-C chemokine receptor type 4 (CXCR4) is a key receptor for chicken primordial germ cell migration". The Journal of Reproduction and Development. 63 (6): 555–562. doi: 10.1262/jrd.2017-067 . PMC   5735266 . PMID   28867677.
  4. 1 2 3 4 5 Richardson BE, Lehmann R (January 2010). "Mechanisms guiding primordial germ cell migration: strategies from different organisms". Nature Reviews. Molecular Cell Biology. 11 (1): 37–49. doi:10.1038/nrm2815. PMC   4521894 . PMID   20027186.
  5. 1 2 Svingen T, Koopman P (November 2013). "Building the mammalian testis: origins, differentiation, and assembly of the component cell populations". Genes & Development. 27 (22): 2409–26. doi:10.1101/gad.228080.113. PMC   3841730 . PMID   24240231.
  6. 1 2 Ewen-Campen B, Schwager EE, Extavour CG (January 2010). "The molecular machinery of germ line specification". Molecular Reproduction and Development. 77 (1): 3–18. doi: 10.1002/mrd.21091 . PMID   19790240. S2CID   11341985.
  7. 1 2 Magnúsdóttir E, Surani MA (Jan 21, 2014). "How to make a primordial germ cell". Electroencephalography and Clinical Neurophysiology. 66 (6): 529–538. doi:10.1016/0013-4694(87)90100-3. PMC   3896947 . PMID   2438119.
  8. Chiquoine AD (February 1954). "The identification, origin, and migration of the primordial germ cells in the mouse embryo". The Anatomical Record. 118 (2): 135–46. doi:10.1002/ar.1091180202. PMID   13138919. S2CID   31896844.
  9. Ginsburg M, Snow MH, McLaren A (October 1990). "Primordial germ cells in the mouse embryo during gastrulation". Development. 110 (2): 521–8. doi:10.1242/dev.110.2.521. PMID   2133553.
  10. Lawson KA, Hage WJ (1994). "Clonal analysis of the origin of primordial germ cells in the mouse". Ciba Foundation Symposium. Novartis Foundation Symposia. 182: 68–84, discussion 84–91. doi:10.1002/9780470514573.ch5. ISBN   978-0-470-51457-3. PMID   7835158.
  11. Lanner F (February 2014). "Lineage specification in the early mouse embryo". Experimental Cell Research. 321 (1): 32–9. doi:10.1016/j.yexcr.2013.12.004. PMID   24333597.
  12. Schrode N, Xenopoulos P, Piliszek A, Frankenberg S, Plusa B, Hadjantonakis AK (April 2013). "Anatomy of a blastocyst: cell behaviors driving cell fate choice and morphogenesis in the early mouse embryo". Genesis. 51 (4): 219–33. doi:10.1002/dvg.22368. PMC   3633705 . PMID   23349011.
  13. Gilbert, Scott F. (2013). Developmental biology (10th ed.). Sunderland: Sinauer Associates. ISBN   978-1-60535-173-5.[ page needed ]
  14. 1 2 3 4 5 Saitou M, Yamaji M (November 2012). "Primordial germ cells in mice". Cold Spring Harbor Perspectives in Biology. 4 (11): a008375. doi:10.1101/cshperspect.a008375. PMC   3536339 . PMID   23125014.
  15. 1 2 3 Lawson KA, Dunn NR, Roelen BA, Zeinstra LM, Davis AM, Wright CV, et al. (February 1999). "Bmp4 is required for the generation of primordial germ cells in the mouse embryo". Genes & Development. 13 (4): 424–36. doi:10.1101/gad.13.4.424. PMC   316469 . PMID   10049358.
  16. Hayashi K, Kobayashi T, Umino T, Goitsuka R, Matsui Y, Kitamura D (October 2002). "SMAD1 signaling is critical for initial commitment of germ cell lineage from mouse epiblast". Mechanisms of Development. 118 (1–2): 99–109. doi: 10.1016/S0925-4773(02)00237-X . PMID   12351174.
  17. Tam PP, Snow MH (August 1981). "Proliferation and migration of primordial germ cells during compensatory growth in mouse embryos". Journal of Embryology and Experimental Morphology. 64: 133–47. PMID   7310300.
  18. Ying Y, Zhao GQ (April 2001). "Cooperation of endoderm-derived BMP2 and extraembryonic ectoderm-derived BMP4 in primordial germ cell generation in the mouse". Developmental Biology. 232 (2): 484–92. doi: 10.1006/dbio.2001.0173 . PMID   11401407.
  19. Ying Y, Liu XM, Marble A, Lawson KA, Zhao GQ (July 2000). "Requirement of Bmp8b for the generation of primordial germ cells in the mouse". Molecular Endocrinology. 14 (7): 1053–63. doi: 10.1210/mend.14.7.0479 . PMID   10894154. S2CID   18854728.
  20. Liu P, Wakamiya M, Shea MJ, Albrecht U, Behringer RR, Bradley A (August 1999). "Requirement for Wnt3 in vertebrate axis formation". Nature Genetics. 22 (4): 361–5. doi:10.1038/11932. PMID   10431240. S2CID   22195563.
  21. Rivera-Pérez JA, Magnuson T (December 2005). "Primitive streak formation in mice is preceded by localized activation of Brachyury and Wnt3". Developmental Biology. 288 (2): 363–71. doi: 10.1016/j.ydbio.2005.09.012 . PMID   16289026.
  22. Tanaka SS, Nakane A, Yamaguchi YL, Terabayashi T, Abe T, Nakao K, et al. (2013). "Dullard/Ctdnep1 modulates WNT signalling activity for the formation of primordial germ cells in the mouse embryo". PLOS ONE. 8 (3): e57428. Bibcode:2013PLoSO...857428T. doi: 10.1371/journal.pone.0057428 . PMC   3587611 . PMID   23469192.
  23. 1 2 3 4 Aramaki S, Hayashi K, Kurimoto K, Ohta H, Yabuta Y, Iwanari H, et al. (December 2013). "A mesodermal factor, T, specifies mouse germ cell fate by directly activating germline determinants". Developmental Cell. 27 (5): 516–29. doi: 10.1016/j.devcel.2013.11.001 . PMID   24331926.
  24. Herrmann BG, Labeit S, Poustka A, King TR, Lehrach H (February 1990). "Cloning of the T gene required in mesoderm formation in the mouse". Nature. 343 (6259): 617–22. Bibcode:1990Natur.343..617H. doi:10.1038/343617a0. PMID   2154694. S2CID   4365020.
  25. Naiche LA, Harrelson Z, Kelly RG, Papaioannou VE (2005). "T-box genes in vertebrate development". Annual Review of Genetics. 39: 219–39. doi:10.1146/annurev.genet.39.073003.105925. PMID   16285859. S2CID   6347720.
  26. Cinalli RM, Rangan P, Lehmann R (February 2008). "Germ cells are forever". Cell. 132 (4): 559–62. doi: 10.1016/j.cell.2008.02.003 . PMID   18295574.
  27. Ohinata Y, Payer B, O'Carroll D, Ancelin K, Ono Y, Sano M, et al. (July 2005). "Blimp1 is a critical determinant of the germ cell lineage in mice". Nature. 436 (7048): 207–13. Bibcode:2005Natur.436..207O. doi:10.1038/nature03813. PMID   15937476. S2CID   4399840.
  28. Werling U, Schorle H (May 2002). "Transcription factor gene AP-2 gamma essential for early murine development". Molecular and Cellular Biology. 22 (9): 3149–56. doi:10.1128/mcb.22.9.3149-3156.2002. PMC   133770 . PMID   11940672.
  29. Magnúsdóttir E, Dietmann S, Murakami K, Günesdogan U, Tang F, Bao S, et al. (August 2013). "A tripartite transcription factor network regulates primordial germ cell specification in mice". Nature Cell Biology. 15 (8): 905–15. doi:10.1038/ncb2798. PMC   3796875 . PMID   23851488.
  30. 1 2 Weber S, Eckert D, Nettersheim D, Gillis AJ, Schäfer S, Kuckenberg P, et al. (January 2010). "Critical function of AP-2 gamma/TCFAP2C in mouse embryonic germ cell maintenance". Biology of Reproduction. 82 (1): 214–23. doi: 10.1095/biolreprod.109.078717 . PMID   19776388.
  31. Hajkova P, Ancelin K, Waldmann T, Lacoste N, Lange UC, Cesari F, et al. (April 2008). "Chromatin dynamics during epigenetic reprogramming in the mouse germ line". Nature. 452 (7189): 877–81. Bibcode:2008Natur.452..877H. doi:10.1038/nature06714. PMC   3847605 . PMID   18354397.
  32. Hajkova P, Jeffries SJ, Lee C, Miller N, Jackson SP, Surani MA (July 2010). "Genome-wide reprogramming in the mouse germ line entails the base excision repair pathway". Science. 329 (5987): 78–82. Bibcode:2010Sci...329...78H. doi:10.1126/science.1187945. PMC   3863715 . PMID   20595612.
  33. Hackett JA, Sengupta R, Zylicz JJ, Murakami K, Lee C, Down TA, Surani MA (January 2013). "Germline DNA demethylation dynamics and imprint erasure through 5-hydroxymethylcytosine". Science. 339 (6118): 448–52. Bibcode:2013Sci...339..448H. doi:10.1126/science.1229277. PMC   3847602 . PMID   23223451.
  34. 1 2 Ohinata Y, Ohta H, Shigeta M, Yamanaka K, Wakayama T, Saitou M (May 2009). "A signaling principle for the specification of the germ cell lineage in mice". Cell. 137 (3): 571–84. doi: 10.1016/j.cell.2009.03.014 . PMID   19410550.
  35. 1 2 Hayashi K, Ohta H, Kurimoto K, Aramaki S, Saitou M (August 2011). "Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells". Cell. 146 (4): 519–32. doi: 10.1016/j.cell.2011.06.052 . PMID   21820164.
  36. Resnick JL, Bixler LS, Cheng L, Donovan PJ (October 1992). "Long-term proliferation of mouse primordial germ cells in culture". Nature. 359 (6395): 550–1. Bibcode:1992Natur.359..550R. doi:10.1038/359550a0. PMID   1383830. S2CID   4315359.
  37. Matsui Y, Zsebo K, Hogan BL (September 1992). "Derivation of pluripotential embryonic stem cells from murine primordial germ cells in culture". Cell. 70 (5): 841–7. doi:10.1016/0092-8674(92)90317-6. PMID   1381289. S2CID   37453479.
  38. Stevens LC, Little CC (November 1954). "Spontaneous Testicular Teratomas in an Inbred Strain of Mice". Proceedings of the National Academy of Sciences of the United States of America. 40 (11): 1080–7. Bibcode:1954PNAS...40.1080S. doi: 10.1073/pnas.40.11.1080 . PMC   1063969 . PMID   16578442.
  39. Gross-Thebing T, Yigit S, Pfeiffer J, Reichman-Fried M, Bandemer J, Ruckert C, et al. (December 2017). "The Vertebrate Protein Dead End Maintains Primordial Germ Cell Fate by Inhibiting Somatic Differentiation". Developmental Cell. 43 (6): 704–715.e5. doi: 10.1016/j.devcel.2017.11.019 . PMID   29257950.
  40. Chatfield J, O'Reilly MA, Bachvarova RF, Ferjentsik Z, Redwood C, Walmsley M, et al. (June 2014). "Stochastic specification of primordial germ cells from mesoderm precursors in axolotl embryos". Development. 141 (12): 2429–40. doi:10.1242/dev.105346. PMC   4050694 . PMID   24917499.
  41. Nicholls PK, Schorle H, Naqvi S, Hu YC, Fan Y, Carmell MA, et al. (November 2019). "Mammalian germ cells are determined after PGC colonization of the nascent gonad". Proceedings of the National Academy of Sciences of the United States of America. 116 (51): 25677–25687. Bibcode:2019PNAS..11625677N. doi: 10.1073/pnas.1910733116 . PMC   6925976 . PMID   31754036.
  42. Slack, J. M. W. (May 1991). From Egg to Embryo: Regional Specification in Early Development. Cambridge Core. doi:10.1017/cbo9780511525322. ISBN   9780521401081 . Retrieved 2019-11-27.
  43. 1 2 3 4 5 6 Culty M (August 2013). "Gonocytes, from the fifties to the present: is there a reason to change the name?". Biology of Reproduction. 89 (2): 46. doi: 10.1095/biolreprod.113.110544 . PMID   23843237.
  44. Sekido R, Lovell-Badge R (2013). "Genetic control of testis development". Sexual Development. 7 (1–3): 21–32. doi: 10.1159/000342221 . PMID   22964823.
  45. 1 2 Rossi P, Dolci S (November 2013). "Paracrine mechanisms involved in the control of early stages of Mammalian spermatogenesis". Frontiers in Endocrinology. 4: 181. doi: 10.3389/fendo.2013.00181 . PMC   3840353 . PMID   24324457.
  46. Walter CA, Intano GW, McCarrey JR, McMahan CA, Walter RB (August 1998). "Mutation frequency declines during spermatogenesis in young mice but increases in old mice". Proceedings of the National Academy of Sciences of the United States of America. 95 (17): 10015–9. Bibcode:1998PNAS...9510015W. doi: 10.1073/pnas.95.17.10015 . PMC   21453 . PMID   9707592.
  47. Delabaere L, Ertl HA, Massey DJ, Hofley CM, Sohail F, Bienenstock EJ, et al. (April 2017). "Aging impairs double-strand break repair by homologous recombination in Drosophila germ cells". Aging Cell. 16 (2): 320–328. doi:10.1111/acel.12556. PMC   5334535 . PMID   28000382.
  48. Vrooman LA, Nagaoka SI, Hassold TJ, Hunt PA (February 2014). "Evidence for paternal age-related alterations in meiotic chromosome dynamics in the mouse". Genetics. 196 (2): 385–96. doi:10.1534/genetics.113.158782. PMC   3914612 . PMID   24318536.
  49. 1 2 Murphey P, McLean DJ, McMahan CA, Walter CA, McCarrey JR (January 2013). "Enhanced genetic integrity in mouse germ cells". Biology of Reproduction. 88 (1): 6. doi:10.1095/biolreprod.112.103481. PMC   4434944 . PMID   23153565.