Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
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
2.
Commun Biol ; 4(1): 645, 2021 05 31.
Article in English | MEDLINE | ID: mdl-34059773

ABSTRACT

WDR62 is a scaffold protein involved in centriole duplication and spindle assembly during mitosis. Mutations in WDR62 can cause primary microcephaly and premature ovarian insufficiency. We have generated a genetrap mouse model deficient in WDR62 and characterised the developmental effects of WDR62 deficiency during meiosis in the testis. We have found that WDR62 deficiency leads to centriole underduplication in the spermatocytes due to reduced or delayed CEP63 accumulation in the pericentriolar matrix. This resulted in prolonged metaphase that led to apoptosis. Round spermatids that inherited a pair of centrioles progressed through spermiogenesis, however, manchette removal was delayed in WDR62 deficient spermatids due to delayed Katanin p80 accumulation in the manchette, thus producing misshapen spermatid heads with elongated manchettes. In mice, WDR62 deficiency resembles oligoasthenoteratospermia, a common form of subfertility in men that is characterised by low sperm counts, poor motility and abnormal morphology. Therefore, proper WDR62 function is necessary for timely spermatogenesis and spermiogenesis during male reproduction.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/genetics , Nerve Tissue Proteins/metabolism , Spermatogenesis/genetics , Animals , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Centrioles/metabolism , Cytoskeleton/metabolism , Female , Male , Meiosis , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Spermatids/metabolism , Spermatogenesis/physiology , Spermatozoa/metabolism , Testis/metabolism
3.
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
4.
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
5.
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
6.
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
7.
Mol Cell Endocrinol ; 382(1): 488-497, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-24076097

ABSTRACT

Germ cells are unique in undergoing meiosis to generate oocytes and sperm. In mammals, meiosis onset is before birth in females, or at puberty in males, and recent studies have uncovered several regulatory steps involved in initiating meiosis in each sex. Evidence suggests that retinoic acid (RA) induces expression of the critical pre-meiosis gene Stra8 in germ cells of the fetal ovary, pubertal testis and adult testis. In the fetal testis, CYP26B1 degrades RA, while FGF9 further antagonises RA signalling to suppress meiosis. Failsafe mechanisms involving Nanos2 may further suppress meiosis in the fetal testis. Here, we draw together the growing knowledge relating to these meiotic control mechanisms, and present evidence that they are co-ordinately regulated and that additional factors remain to be identified. Understanding this regulatory network will illuminate not only how the foundations of mammalian reproduction are laid, but also how mis-regulation of these steps can result in infertility or germline tumours.


Subject(s)
Germ Cells/cytology , Mammals/metabolism , Meiosis , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Lineage/drug effects , Germ Cells/metabolism , Humans , Meiosis/drug effects , Tretinoin/pharmacology
8.
Development ; 139(22): 4123-32, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23034635

ABSTRACT

Germ cells, the embryonic precursors of sperm or oocytes, respond to molecular cues that regulate their sex-specific development in the fetal gonads. In males in particular, the balance between continued proliferation and cell fate commitment is crucial: defects in proliferation result in insufficient spermatogonial stem cells for fertility, but escape from commitment and prolonged pluripotency can cause testicular germ cell tumors. However, the factors that regulate this balance remain unidentified. Here, we show that signaling by the TGFß morphogen Nodal and its co-receptor Cripto is active during a crucial window of male germ cell development. The Nodal pathway is triggered when somatic signals, including FGF9, induce testicular germ cells to upregulate Cripto. Germ cells of mutant mice with compromised Nodal signaling showed premature differentiation, reduced pluripotency marker expression and a reduced ability to form embryonic germ (EG) cell colonies in vitro. Conversely, human testicular tumors showed upregulation of NODAL and CRIPTO that was proportional to invasiveness and to the number of malignant cells. Thus, Nodal signaling provides a molecular control mechanism that regulates male germ cell potency in normal development and testicular cancer.


Subject(s)
Epidermal Growth Factor/metabolism , Germ Cells/physiology , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Nodal Protein/metabolism , Signal Transduction , Spermatogenesis/physiology , Spermatogonia/metabolism , Testis/embryology , Animals , Cell Differentiation , Cell Proliferation , Fibroblast Growth Factor 9/metabolism , Germ Cells/cytology , Humans , Male , Mice , Neoplasms, Germ Cell and Embryonal/metabolism , Pluripotent Stem Cells/cytology , Spermatogonia/cytology , Testicular Neoplasms/metabolism , Transforming Growth Factor beta
9.
FASEB J ; 25(10): 3561-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21757499

ABSTRACT

Sex determination in fetal germ cells depends on a balance between exposure to retinoic acid (RA) and the degradation of RA achieved by the testis-specific expression of the catabolic cytochrome P450 enzyme, CYP26B1. Therefore, identification of factors regulating the expression of the Cyp26b1 gene is an important goal in reproductive biology. We used in situ hybridization to demonstrate that Cyp26b1 and transcription factor genes steroidogenic factor-1 (Sf1) and Sry-related HMG box 9 (Sox9) are coexpressed in Sertoli cells, whereas Cyp26b1 and Sf1 are coexpressed in Leydig cells in mouse fetal testes. In the mouse gonadal somatic cell line TM3, transfection of constructs expressing SOX9 and SF1 activated Cyp26b1 expression, independently of the positive regulator RA. In embryonic gonads deficient in SOX9 or SF1, Cyp26b1 expression was decreased relative to wild-type (WT) controls, as measured by quantitative RT-PCR (qRT-PCR). Furthermore, qRT-PCR showed that Cyp26b1 up-regulation by SOX9/SF1 was attenuated by the ovarian transcription factor Forkhead box L2 (FOXL2) in TM3 cells, whereas in Foxl2-null mice, Cyp26b1 expression in XX gonads was increased ∼20-fold relative to WT controls. These data support the hypothesis that SOX9 and SF1 ensure the male fate of germ cells by up-regulating Cyp26b1 and that FOXL2 acts to antagonize Cyp26b1 expression in ovaries.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , SOX9 Transcription Factor/metabolism , Testis/growth & development , Transcription Factors/metabolism , Animals , Cytochrome P-450 Enzyme System/genetics , DNA-Binding Proteins/genetics , Forkhead Box Protein L2 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental/physiology , Male , Mice , RNA Splicing Factors , Retinoic Acid 4-Hydroxylase , SOX9 Transcription Factor/genetics , Sex Determination Processes/physiology , Testis/metabolism , Transcription Factors/genetics , Up-Regulation
10.
Dev Cell ; 19(3): 440-9, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20833365

ABSTRACT

Sex determination of mammalian germ cells occurs during fetal development and depends on signals from gonadal somatic cells. Previous studies have established that retinoic acid (RA) triggers ovarian germ cells to enter meiosis and thereby commit to oogenesis, whereas in the developing testis, the enzyme CYP26B1 degrades RA and germ cells are not induced to enter meiosis. Using in vitro and in vivo models, we demonstrate that fibroblast growth factor 9 (FGF9) produced in the fetal testis acts directly on germ cells to inhibit meiosis; in addition, FGF9 maintains expression of pluripotency-related genes and upregulates markers associated with male germ cell fate. We conclude that two independent and mutually antagonistic pathways involving RA and FGF9 act in concert to determine mammalian germ cell sexual fate commitment and support a model in which the mitosis/meiosis switch is robustly controlled by both positive and negative regulatory factors.


Subject(s)
Fibroblast Growth Factor 9/physiology , Germ Cells/physiology , Meiosis/physiology , Pluripotent Stem Cells/metabolism , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Female , Fetus/cytology , Fetus/drug effects , Fetus/metabolism , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oogenesis/drug effects , Oogenesis/physiology , Ovary/embryology , Ovary/metabolism , RNA, Messenger/genetics , Retinoic Acid 4-Hydroxylase , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Testis/embryology , Testis/metabolism , Tretinoin/pharmacology
11.
Dev Dyn ; 238(9): 2430-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19653320

ABSTRACT

The development of the reproductive system in bufonids (true toads) is unique in several respects: sexual differentiation occurs later than in other anurans, and toads develop a Bidder's organ, a rudimentary ovary that can be manipulated in males to produce mature oocytes. To illuminate the genesis of this unusual reproductive system, we isolated from the cane toad (Bufo marinus) the orthologues of several known vertebrate sex-determining genes, determined their primary structure, and studied their expression by reverse transcriptase-polymerase chain reaction and in situ hybridization of tissue sections. We report here that cane toad Sox9, Dmrt1, and p450aromatase (Cyp19a1) are highly homologous to their counterparts in other vertebrates. They show profiles of expression that generally follow patterns observed in other taxa, but with some novel features. Our data suggest that these genes likely play key roles in sex determination and early gonad development in bufonids.


Subject(s)
Amphibian Proteins/genetics , Bufo marinus/embryology , Bufo marinus/genetics , Gene Expression Regulation, Developmental , Sexual Maturation/genetics , Animals , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Humans , In Situ Hybridization , Male , Reverse Transcriptase Polymerase Chain Reaction
12.
Dev Dyn ; 238(8): 2073-80, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19618468

ABSTRACT

Balanced production and degradation of retinoids is important in regulating development of several organ systems in the vertebrate embryo. Among these, it is known that retinoic acid (RA), and the retinoid-catabolyzing enzyme CYP26B1 together regulate the sex-specific behavior of germ cells in developing mouse gonads. We report here that the gene encoding a cytosolic class-1 aldehyde dehydrogenase, ALDH1A1, a weak catalyst of RA production, is strongly expressed in a male-specific manner in somatic cells of the developing mouse testis, beginning shortly after Sry expression is first detectable. This expression pattern is conserved in the developing male gonad of the chicken and is dependent on the testis-specific transcription factor SOX9. Our data suggest that low levels of RA may be required for early developmental events in the testis, or that Aldh1a1 expression in the fetus may prefigure a later requirement for ALDH1A1 in regulating spermatogenesis postnatally.


Subject(s)
Aldehyde Dehydrogenase/genetics , Isoenzymes/genetics , Retinoids/metabolism , Testis/embryology , Testis/enzymology , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Base Sequence , Chick Embryo , DNA Primers/genetics , Female , Gene Expression Regulation, Developmental , Genes, sry , Isoenzymes/metabolism , Male , Mice , Mice, Transgenic , Pregnancy , Retinal Dehydrogenase , SOX9 Transcription Factor/metabolism , Sex Differentiation , Testis/metabolism , Tretinoin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...