Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2677: 221-231, 2023.
Article in English | MEDLINE | ID: mdl-37464245

ABSTRACT

The fetal gonad contains a great variety of differentiating cell populations, of which germ cells make up a relatively small percentage. In order to study germ cell-specific gene and protein expression, as well as determine direct effects of signaling molecules, it is necessary to prepare enriched populations of germ cells and maintain them in culture for several hours to multiple days. The protocols in this chapter are designed to provide a guide for the isolation or enrichment of primordial germ cells (from 9.5 days post coitum (dpc) to 18.5 dpc) by flow cytometry (Subheading 3.1) or magnetic sorting (Subheading 3.2), followed by feeder-free primary germ cell culture (Subheading 3.3).


Subject(s)
Fetus , Germ Cells , Mice , Animals , Germ Cells/metabolism , Culture Techniques , Flow Cytometry , Gonads
2.
Genesis ; 61(1-2): e23511, 2023 03.
Article in English | MEDLINE | ID: mdl-36693128

ABSTRACT

Germline-specific Cre lines are useful for analyses of primordial germ cell, spermatogonial and oogonial development, but also for whole-body deletions when transmitted through subsequent generations. Several germ cell specific Cre mouse strains exist, with various degrees of specificity, efficiency, and temporal activation. Here, we describe the CRISPR/Cas9 targeted insertion of an improved Cre (iCre) sequence in-frame at the 3' end of the Ddx4 locus to generate the Ddx4-P2A-iCre allele. Our functional assessment of this new allele, designated Ddx4iCreJoBo , reveals that Cre activity begins in PGCs from at least E10.5, and that it achieves higher efficiency for early gonadal (E10.5-12.5) germline deletion when compared to the inducible Oct4CreERT2 line. We found the Ddx4iCreJoBo allele to be hypomorphic for Ddx4 expression and homozygous males, but not females, were infertile. Using two reporter lines (R26RLacZ and R26RtdTomato ) and a floxed gene of interest (Criptoflox ) we found ectopic activity in multiple organs; global recombination (a common feature of germline Cre alleles) varies from 10 to 100%, depending on the particular floxed allele. There is a strong maternal effect, and therefore it is preferable for Ddx4iCreJoBo to be inherited from the male parent if ubiquitous deletion is not desired. With these limitations considered, we describe the Ddx4iCreJoBo line as useful for germline studies in which early gonadal deletion is required.


Subject(s)
Germ Cells , Integrases , Animals , Male , Mice , Animals, Genetically Modified , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Germ Cells/metabolism , Integrases/genetics , Integrases/metabolism , Mice, Transgenic
3.
Biol Reprod ; 106(6): 1191-1205, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35243487

ABSTRACT

Members of the nuclear factor I (NFI) family are key regulators of stem cell biology during development, with well-documented roles for NFIA, NFIB, and NFIX in a variety of developing tissues, including brain, muscle, and lung. Given the central role these factors play in stem cell biology, we posited that they may be pivotal for spermatogonial stem cells or further developing spermatogonia during testicular development. Surprisingly, in stark contrast to other developing organ systems where NFI members are co-expressed, these NFI family members show discrete patterns of expression within the seminiferous tubules. Sertoli cells (spermatogenic supporting cells) express NFIA, spermatocytes express NFIX, round spermatids express NFIB, and peritubular myoid cells express each of these three family members. Further analysis of NFIX expression during the cycle of the seminiferous epithelium revealed expression not in spermatogonia, as we anticipated, but in spermatocytes. These data suggested a potential role for NFIX in spermatogenesis. To investigate, we analyzed mice with constitutive deletion of Nfix (Nfix-null). Assessment of germ cells in the postnatal day 20 (P20) testes of Nfix-null mice revealed that spermatocytes initiate meiosis, but zygotene stage spermatocytes display structural defects in the synaptonemal complex, and increased instances of unrepaired DNA double-strand breaks. Many developing spermatocytes in the Nfix-null testis exhibited multinucleation. As a result of these defects, spermatogenesis is blocked at early diplotene and very few round spermatids are produced. Collectively, these novel data establish the global requirement for NFIX in correct meiotic progression during the first wave of spermatogenesis.


Subject(s)
NFI Transcription Factors , Spermatogonia , Testis , Animals , Male , Meiosis , Mice , Mice, Knockout , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism , Spermatocytes/metabolism , Spermatogenesis/genetics , Testis/metabolism
4.
Curr Res Toxicol ; 3: 100069, 2022.
Article in English | MEDLINE | ID: mdl-35345548

ABSTRACT

The Adverse Outcome Pathway (AOP) concept is an emerging tool in regulatory toxicology that uses simplified descriptions to show cause-effect relationships between stressors and toxicity outcomes in intact organisms. The AOP structure is a modular framework, with Key Event Relationships (KERs) representing the unit of causal relationship based on existing knowledge, describing the connection between two Key Events. Because KERs are the only unit to support inference it has been argued recently that KERs should be recognized as the core building blocks of knowledge assembly within the AOP-Knowledge Base. Herein, we present a first case to support this proposal and provide a full description of a KER linking decreased all-trans retinoic acid (atRA) levels in developing ovaries with disrupted meiotic entry of oogonia. We outline the evidence to support a role for atRA in inducing meiosis in oogonia across mammals; this is important because elements of the RA synthesis/degradation pathway are recognized targets for numerous environmental chemicals. The KER we describe will be used to support an intended AOP linking inhibition of the atRA producing ALDH1A enzymes with reduced fertility in women.

5.
Sex Dev ; 16(5-6): 342-354, 2022.
Article in English | MEDLINE | ID: mdl-35320803

ABSTRACT

BACKGROUND: Germ cells are critical for the survival of our species. They are the only cells that undergo meiosis - the reductive form of cell division that is necessary for genetic reassortment of chromosomes and production of the haploid gametes, the sperm and eggs. Remarkably, the initial female/male fate decision in fetal germ cells does not depend on whether they are chromosomally XX or XY; rather, initial sexual fate is imposed by influences from the surrounding tissue. In mammals, the female germline is particularly precious: despite recent suggestions that germline stem cells exist in the ovary, it is still generally accepted that the ovarian reserve is finite, and its size is dependant on germ cells of the fetal ovary initiating meiosis in a timely manner. SUMMARY: Prior to 2006, evidence suggested that gonadal germ cells initiate meiotic prophase I by default, but more recent data support a key role for the signalling molecule retinoic acid (RA) in instructing female germ cell fate. Newer findings also support a key meiosis-inducing role for another signalling molecule, bone morphogenic protein (BMP). Nonetheless, many questions remain. KEY MESSAGES: Here, we review knowledge thus far regarding extrinsic and intrinsic determinants of a female germ cell fate, focusing on the mouse model.

6.
Development ; 148(5)2021 03 09.
Article in English | MEDLINE | ID: mdl-33574039

ABSTRACT

In mice, the entry of germ cells into meiosis crucially depends on the expression of stimulated by retinoic acid gene 8 (Stra8). Stra8 is expressed specifically in pre-meiotic germ cells of females and males, at fetal and postnatal stages, respectively, but the mechanistic details of its spatiotemporal regulation are yet to be defined. In particular, there has been considerable debate regarding whether retinoic acid is required, in vivo, to initiate Stra8 expression in the mouse fetal ovary. We show that the distinctive anterior-to-posterior pattern of Stra8 initiation, characteristic of germ cells in the fetal ovary, is faithfully recapitulated when 2.9 kb of the Stra8 promoter is used to drive eGFP expression. Using in vitro transfection assays of cutdown and mutant constructs, we identified two functional retinoic acid responsive elements (RAREs) within this 2.9 kb regulatory element. We also show that the transcription factor DMRT1 enhances Stra8 expression, but only in the presence of RA and the most proximal RARE. Finally, we used CRISPR/Cas9-mediated targeted mutation studies to demonstrate that both RAREs are required for optimal Stra8 expression levels in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Germ Cells/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites , CRISPR-Cas Systems/genetics , Female , Fetal Development/genetics , Fetus/cytology , Fetus/metabolism , Gene Expression Regulation/drug effects , Germ Cells/cytology , Meiosis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis , Ovary/cytology , Ovary/metabolism , Promoter Regions, Genetic , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/pharmacology , Tretinoin/pharmacology
7.
Cancers (Basel) ; 12(3)2020 Mar 23.
Article in English | MEDLINE | ID: mdl-32210110

ABSTRACT

miR-371a-3p is currently the most informative reported biomarker for germ cell tumors (GCTs). Another developmental-related biomarker, CRIPTO, is involved in the regulation of pluripotency and germ cell fate commitment. We aimed to assess the value of CRIPTO as a diagnostic and prognostic biomarker of testicular GCTs (TGCTs) and also to assess its presence in seminal plasma samples, compared with miR-371a-3p. In total, 217 and 94 serum/seminal plasma samples were analyzed. CRIPTO was quantified using ELISA and miR-371a-3p using bead-based isolation followed by RT-qPCR. Methylation profiling (EPIC array) for the CRIPTO promoter region was undertaken in 35 TGCT tissues plus four (T)GCT cell lines. Significantly higher CRIPTO concentration was found in sera of non-seminomas compared to controls (p = 0.0297), and in stage II/III disease compared to stage I (p = 0.0052, p = 0.0097). CRIPTO concentration was significantly positively correlated with miR-371a-3p levels in serum (r = 0.16) and seminal plasma (r = 0.40). CRIPTO/miR-371a-3p levels were significantly higher in seminal plasma controls when compared to serum controls (p = 0.0001, p < 0.0001). CRIPTO/miR-371a-3p were detected both in normospermic and azoospermic males, and levels were higher in TGCTs compared to GCNIS-only. We have provided the largest dataset of evaluation of CRIPTO in serum and seminal plasma of GCTs, showing its potential value as a biomarker of the disease.

8.
Mol Hum Reprod ; 26(2): 111-116, 2020 02 29.
Article in English | MEDLINE | ID: mdl-31943113

ABSTRACT

An adverse outcome pathway (AOP) is a simplified description of the sequence of mechanistic events that lead to a particular toxicological effect, from initial trigger to adverse outcome. Although designed to inform regulatory risk assessors, the AOP framework also provides a platform for innovative collaborations between experts from relevant research fields and the regulatory community. The underpinning for any AOP is basic knowledge about molecular and developmental processes; such knowledge can only be attained by solid bioscientific research. Starting with this fundamental knowledge, the objective is to devise novel testing strategies that focus on key events in a causative pathway. It is anticipated that such a knowledge-based approach will ultimately alleviate many of the burdens associated with classical chemical testing strategies that typically involve large-scale animal toxicity regimens. This hails from the notion that a solid understanding of the underlying mechanisms will allow the development and use of alternative test methods, including both in vitro and in silico approaches. This review is specifically targeted at professionals working in bioscientific fields, such as developmental and reproductive biology, and aims to (i) inform on the existence of the AOP framework and (ii) encourage new cross-disciplinary collaborations. It is hoped that fundamental biological knowledge can thus be better exploited for applied purposes: firstly, an improved understanding of how our perpetual exposure to environmental chemicals is causing human reproductive disease and, secondly, new approaches to screen for harmful chemicals more efficiently. This is not an instructional manual on how to create AOPs; rather, we discuss how to harness fundamental knowledge from the biosciences to assist regulatory toxicologists in their efforts to protect humans against chemicals that harm human reproductive development and function.


Subject(s)
Adverse Outcome Pathways , Developmental Biology/methods , Noxae/adverse effects , Reproduction/drug effects , Reproductive Medicine/methods , Toxicology/methods , Anal Canal/embryology , Androgens/physiology , Animals , Endocrine Disruptors/toxicity , Genitalia/embryology , Humans , Interdisciplinary Communication , Internet , Models, Animal , Nipples/embryology , Noxae/toxicity , Reproduction/physiology , Tretinoin/toxicity
9.
Curr Top Dev Biol ; 134: 253-288, 2019.
Article in English | MEDLINE | ID: mdl-30999978

ABSTRACT

Germ cells are the stem cells of the species. Thus, it is critical that we have a good understanding of how they are specified, how the somatic cells instruct and support them, how they commit to one or other sex, and how they ultimately develop into functional gametes. Here, we focus on specifics of how sexual fate is determined during fetal life. Because the majority of relevant experimental work has been done using the mouse model, we focus on that species. We review evidence regarding the identity of instructive signals from the somatic cells, and the molecular responses that occur in germ cells in response to those extrinsic signals. In this way we aim to clarify progress to date regarding the mechanisms underlying the mitotic to meiosis switch in germ cells of the fetal ovary, and those involved in adopting and securing male fate in germ cells of the fetal testis.


Subject(s)
Cell Differentiation , Germ Cells/cytology , Mammals/genetics , Meiosis , Sex Characteristics , Sex Determination Processes/physiology , Animals , Female , Germ Cells/physiology , Male , Mice , Oogenesis , Signal Transduction , Spermatogenesis
10.
Cell Rep ; 24(5): 1330-1341, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067986

ABSTRACT

Mammalian sex determination depends on a complex interplay of signals that promote the bipotential fetal gonad to develop as either a testis or an ovary, but the details are incompletely understood. Here, we investigated whether removal of the signaling molecule retinoic acid (RA) by the degradative enzyme CYP26B1 is necessary for proper development of somatic cells of the testes. Gonadal organ culture experiments suggested that RA promotes expression of some ovarian markers and suppresses expression of some testicular markers, acting downstream of Sox9. XY Cyp26b1-null embryos, in which endogenous RA is not degraded, develop mild ovotestes, but more important, steroidogenesis is impaired and the reproductive tract feminized. Experiments involving purified gonadal cells showed that these effects are independent of germ cells and suggest the direct involvement of the orphan nuclear receptor DAX1. Our results reveal that active removal of endogenous RA is required for normal testis development in the mouse.


Subject(s)
Sex Determination Processes , Testis/metabolism , Tretinoin/pharmacology , Animals , Cells, Cultured , DAX-1 Orphan Nuclear Receptor/genetics , DAX-1 Orphan Nuclear Receptor/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , SOX9 Transcription Factor/metabolism , Testis/drug effects , Testis/embryology
11.
Mol Cell Endocrinol ; 478: 84-96, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30053582

ABSTRACT

Mammalian sex determination hinges on sexually dimorphic transcriptional programs in developing fetal gonads. A comprehensive view of these programs is crucial for understanding the normal development of fetal testes and ovaries and the etiology of human disorders of sex development (DSDs), many of which remain unexplained. Using strand-specific RNA-sequencing, we characterized the mouse fetal gonadal transcriptome from 10.5 to 13.5 days post coitum, a key time window in sex determination and gonad development. Our dataset benefits from a greater sensitivity, accuracy and dynamic range compared to microarray studies, allows global dynamics and sex-specificity of gene expression to be assessed, and provides a window to non-transcriptional events such as alternative splicing. Spliceomic analysis uncovered female-specific regulation of Lef1 splicing, which may contribute to the enhanced WNT signaling activity in XX gonads. We provide a user-friendly visualization tool for the complete transcriptomic and spliceomic dataset as a resource for the field.


Subject(s)
Alternative Splicing/genetics , Gene Expression Profiling , RNA, Messenger/genetics , Sex Determination Processes/genetics , Animals , Female , Fetus/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Gonads/embryology , Gonads/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Mice , RNA, Messenger/metabolism , Reproducibility of Results , Sequence Analysis, RNA , Sex Characteristics , Time Factors , Transcriptional Activation/genetics
12.
Sci Rep ; 7(1): 17619, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29247201

ABSTRACT

Male infertility is a major and growing problem and, in most cases, the specific root cause is unknown. Here we show that the transcription factor SOX30 plays a critical role in mouse spermatogenesis. Sox30-null mice are healthy and females are fertile, but males are sterile. In the absence of Sox30 meiosis initiates normally in both sexes but, in males, germ cell development arrests during the post-meiotic round spermatid period. In the mutant testis, acrosome and axoneme development are aberrant, multinucleated germ cells (symplasts) form and round spermatids unable to process beyond step 3 of spermiogenesis. No elongated spermatids nor spermatozoa are produced. Thus, Sox30 represents a rare example of a gene for which loss of function results in a complete arrest of spermatogenesis at the onset of spermiogenesis. Our results suggest that SOX30 mutations may underlie some instances of unexplained non-obstructive azoospermia in humans.


Subject(s)
Azoospermia/genetics , Infertility, Male/genetics , Oocytes/growth & development , SOX Transcription Factors/genetics , Spermatogenesis/genetics , Acrosome/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Axoneme/physiology , Female , Male , Meiosis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovary/embryology , Spermatids/cytology , Testis/embryology
13.
Annu Rev Genet ; 51: 265-285, 2017 11 27.
Article in English | MEDLINE | ID: mdl-28853925

ABSTRACT

Sexual reproduction crucially depends on the production of sperm in males and oocytes in females. Both types of gamete arise from the same precursor, the germ cells. We review the events that characterize the development of germ cells during fetal life as they commit to, and prepare for, oogenesis or spermatogenesis. In females, fetal germ cells enter meiosis, whereas in males they delay meiosis and instead lose pluripotency, activate an irreversible program of prospermatogonial differentiation, and temporarily cease dividing. Both pathways involve sex-specific molecular signals from the somatic cells of the developing gonads and a suite of intrinsic receptors, signal transducers, transcription factors, RNA stability factors, and epigenetic modulators that act in complex, interconnected positive and negative regulatory networks. Understanding these networks is important in the contexts of the etiology, diagnosis, and treatment of infertility and gonadal cancers, and in efforts to augment human and animal fertility using stem cell approaches.


Subject(s)
Infertility, Female/genetics , Infertility, Male/genetics , Oogenesis/genetics , Sex Determination Processes , Sex Differentiation/genetics , Spermatogenesis/genetics , Animals , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Infertility, Female/metabolism , Infertility, Female/pathology , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Meiosis , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Ovum/cytology , Ovum/growth & development , Ovum/metabolism , Signal Transduction , Spermatozoa/cytology , Spermatozoa/growth & development , Spermatozoa/metabolism
14.
Stem Cell Res ; 24: 174-180, 2017 10.
Article in English | MEDLINE | ID: mdl-28754604

ABSTRACT

There is now substantial evidence that members of the transforming growth factor-ß (TGFß family) regulate germ cell development in the mouse fetal testis. Correct development of germ cells during fetal life is critical for establishment of effective spermatogenesis and for avoiding the formation of testicular germ cell cancer in later life. Here we consider the evidence for involvement of various TGFß family members, attempt to reconcile discrepancies and clarify what we believe to be the likely in vivo roles of these factors.


Subject(s)
Fetus/cytology , Germ Cells/cytology , Germ Cells/metabolism , Transforming Growth Factor beta/metabolism , Animals , Humans , Male , Signal Transduction
15.
Int J Biochem Cell Biol ; 86: 22-25, 2017 05.
Article in English | MEDLINE | ID: mdl-28288913

ABSTRACT

Germ cell neoplasia in situ is the non-invasive precursor cell of origin for type II testicular germ cell tumors. It has long been postulated that germ cell neoplasia in situ is derived from defective germ cell development during embryonic life, and although it is impossible to trace in vivo the progression from fetal germ cell to germ cell neoplasia in situ to tumor, there is a large volume of evidence supporting this theory. Current studies focus on understanding how germ cell neoplasia in situ forms, how these cells are activated at puberty and how they transform to invasive tumors of various subtypes. Such information is informing novel diagnostic and therapeutic options.


Subject(s)
Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/pathology , Animals , Cell Transformation, Neoplastic , Humans , Neoplasm Invasiveness , Neoplasms, Germ Cell and Embryonal/therapy , Risk Factors , Testicular Neoplasms/therapy
16.
PLoS One ; 12(1): e0170576, 2017.
Article in English | MEDLINE | ID: mdl-28107452

ABSTRACT

Fetal germ cell development is tightly regulated by the somatic cell environment, and is characterised by cell cycle states that differ between XY and XX gonads. In the testis, gonocytes enter G1/G0 arrest from 12.5 days post coitum (dpc) in mice and maintain cell cycle arrest until after birth. Failure to correctly maintain G1/G0 arrest can result in loss of germ cells or, conversely, germ cell tumours. High mobility group box containing transcription factor 1 (HBP1) is a transcription factor that was previously identified in fetal male germ cells at the time of embryonic cell cycle arrest. In somatic cells, HBP1 is classified as a tumour suppressor protein, known to regulate proliferation and senescence. We therefore investigated the possible role of HBP1 in the initiation and maintenance of fetal germ cell G1/G0 arrest using the mouse model. We identified two splice variants of Hbp1, both of which are expressed in XY and XX fetal gonads, but only one of which is localised to the nucleus in in vitro assays. To investigate Hbp1 loss of function, we used embryonic stem (ES) cells carrying a Genetrap mutation for Hbp1 to generate mice lacking Hbp1 function. We found that Hbp1-genetrap mouse mutant germ cells proliferated correctly throughout development, and adult males were viable and fertile. Multiple Hbp1-LacZ reporter mouse lines were generated, unexpectedly revealing Hbp1 embryonic expression in hair follicles, eye and limbs. Lastly, in a model of defective germ cell G1/G0 arrest, the Rb1-knockout model, we found no evidence for Hbp1 mis-regulation, suggesting that the reported RB1-HBP1 interaction is not critical in the germline, despite co-expression.


Subject(s)
Fertility/genetics , Germ Cells/physiology , High Mobility Group Proteins/physiology , Repressor Proteins/physiology , Animals , Cell Line , Germ Cells/metabolism , In Situ Hybridization , Male , Mice/embryology , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction
17.
Methods Mol Biol ; 1463: 173-183, 2017.
Article in English | MEDLINE | ID: mdl-27734356

ABSTRACT

The fetal gonad contains a great variety of differentiating cell populations, of which germ cells make up a small percentage. In order to study germ cell-specific gene and protein expression, as well as determine direct effects of signaling molecules, it is necessary to prepare enriched populations of germ cells and maintain them in culture for several hours to multiple days. The protocols in this chapter are designed to provide a guide for the isolation or enrichment of mouse primordial germ cells (from 9.5 days postcoitum (dpc) to 18.5 dpc) by flow cytometry (Subheading 3.1) or magnetic sorting (Subheading 3.2), followed by primary germ cell culture (Subheading 3.3).


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Fetus/cytology , Germ Cells/cytology , Animals , Cell Differentiation , Culture Media, Serum-Free , Fetus/metabolism , Flow Cytometry , Germ Cells/metabolism , Mice , Signal Transduction
18.
Nat Commun ; 7: 10845, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26892828

ABSTRACT

Substantial evidence exists that during fetal ovarian development in mammals, retinoic acid (RA) induces germ cells to express the pre-meiotic marker Stra8 and enter meiosis, and that these effects are prevented in the fetal testis by the RA-degrading P450 enzyme CYP26B1. Nonetheless, the role of RA has been disputed principally because germ cells in embryos lacking two major RA-synthesizing enzymes, ALDH1A2 and ALDH1A3, remain able to enter meiosis. Here we show that a third RA-synthesizing enzyme, ALDH1A1, is expressed in fetal ovaries, providing a likely source of RA in the absence of ALDH1A2 and ALDH1A3. In ovaries lacking ALDH1A1, the onset of germ cell meiosis is delayed. Our data resolve the conundrum posed by conflicting published data sets and reconfirm the model that meiosis is triggered by endogenous RA in the developing ovary.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Meiosis , Ovary/embryology , Ovary/enzymology , Tretinoin/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovary/cytology , Ovary/metabolism , Retinal Dehydrogenase
19.
Dev Dyn ; 245(4): 433-44, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26637965

ABSTRACT

BACKGROUND: It is widely accepted that, during the development of testes in the mammalian embryo, male germ cells are influenced by signals from the surrounding somatic cells, but not vice versa, so that germ cells are dispensable for the formation of testes. RESULTS: We now demonstrate that development of the mouse fetal testis is compromised in the absence of germ cells. Using two- and three-dimensional imaging techniques, we reveal that W(e)/W(e) mutant testes devoid of germ cells have misshapen and poorly organized cords. We also found that mutant gonads have fewer Sertoli cells than normal and that the Leydig cells express key markers at higher than normal levels. CONCLUSIONS: These observations point to the existence of germ cell-derived signals that directly or indirectly affect the Sertoli and Leydig cell populations, and provide a new paradigm for the organogenesis of the mammalian testes.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Germ Cells/metabolism , Leydig Cells/metabolism , Spermatic Cord/embryology , Animals , Male , Mice , Mice, Transgenic
20.
Mol Oncol ; 10(4): 526-37, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26654129

ABSTRACT

Type II germ cell tumors arise after puberty from a germ cell that was incorrectly programmed during fetal life. Failure of testicular germ cells to properly differentiate can lead to the formation of germ cell neoplasia in situ of the testis; this precursor cell invariably gives rise to germ cell cancer after puberty. The Nodal co-receptor Cripto is expressed transiently during normal germ cell development and is ectopically expressed in non-seminomas that arise from germ cell neoplasia in situ, suggesting that its aberrant expression may underlie germ cell dysregulation and hence germ cell cancer. Here we investigated methylation of the Cripto promoter in mouse germ cells and human germ cell cancer and correlated this with the level of CRIPTO protein expression. We found hypomethylation of the CRIPTO promoter in undifferentiated fetal germ cells, embryonal carcinoma and seminomas, but hypermethylation in differentiated fetal germ cells and the differentiated types of non-seminomas. CRIPTO protein was strongly expressed in germ cell neoplasia in situ along with embryonal carcinoma, yolk sac tumor and seminomas. Further, cleaved CRIPTO was detected in media from seminoma and embryonal carcinoma cell lines, suggesting that cleaved CRIPTO may provide diagnostic indication of germ cell cancer. Accordingly, CRIPTO was detectable in serum from 6/15 patients with embryonal carcinoma, 5/15 patients with seminoma, 4/5 patients with germ cell neoplasia in situ cells only and in 1/15 control patients. These findings suggest that CRIPTO expression may be a useful serological marker for diagnostic and/or prognostic purposes during germ cell cancer management.


Subject(s)
Carcinoma, Embryonal , Epidermal Growth Factor , Epigenesis, Genetic , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins , Neoplasm Proteins , Testicular Neoplasms , Animals , Carcinoma, Embryonal/blood , Carcinoma, Embryonal/diagnosis , Carcinoma, Embryonal/genetics , Epidermal Growth Factor/biosynthesis , Epidermal Growth Factor/genetics , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Testicular Neoplasms/blood , Testicular Neoplasms/diagnosis , Testicular Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...