Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
FASEB J ; 35(4): e21452, 2021 04.
Article in English | MEDLINE | ID: mdl-33749946

ABSTRACT

Despite the importance of germ cell (GC) differentiation for sexual reproduction, the gene networks underlying their fate remain unclear. Here, we comprehensively characterize the gene expression dynamics during sex determination based on single-cell RNA sequencing of 14 914 XX and XY mouse GCs between embryonic days (E) 9.0 and 16.5. We found that XX and XY GCs diverge transcriptionally as early as E11.5 with upregulation of genes downstream of the bone morphogenic protein (BMP) and nodal/Activin pathways in XY and XX GCs, respectively. We also identified a sex-specific upregulation of genes associated with negative regulation of mRNA processing and an increase in intron retention consistent with a reduction in mRNA splicing in XY testicular GCs by E13.5. Using computational gene regulation network inference analysis, we identified sex-specific, sequential waves of putative key regulator genes during GC differentiation and revealed that the meiotic genes are regulated by positive and negative master modules acting in an antagonistic fashion. Finally, we found that rare adrenal GCs enter meiosis similarly to ovarian GCs but display altered expression of master genes controlling the female and male genetic programs, indicating that the somatic environment is important for GC function. Our data are available on a web platform and provide a molecular roadmap of GC sex determination at single-cell resolution, which will serve as a valuable resource for future studies of gonad development, function, and disease.


Subject(s)
Gene Expression Profiling/methods , Sex Determination Processes , Animals , Female , Gene Expression Regulation, Developmental , Germ Cells , Male , Mice , Mice, Transgenic , Single-Cell Analysis , Time Factors , X Chromosome , Y Chromosome
2.
FASEB J ; 32(6): 3321-3335, 2018 06.
Article in English | MEDLINE | ID: mdl-29401624

ABSTRACT

The insulin family of growth factors (insulin, IGF1, and IGF2) are critical in sex determination, adrenal differentiation, and testicular function. Notably, the IGF system has been reported to mediate the proliferation of steroidogenic cells. However, the precise role and contribution of the membrane receptors mediating those effects, namely, insulin receptor (INSR) and type-I insulin-like growth factor receptor (IGF1R), have not, to our knowledge, been investigated. We show here that specific deletion of both Insr and Igf1r in steroidogenic cells in mice leads to severe alterations of adrenocortical and testicular development. Double-mutant mice display drastic size reduction of both adrenocortex and testes, with impaired corticosterone, testosterone, and sperm production. Detailed developmental analysis of the testes revealed that fetal Leydig cell (LC) function is normal, but there is a failure of adult LC maturation and steroidogenic function associated with accumulation of progenitor LCs (PLCs). Cell-lineage tracing revealed PLC enrichment is secondary to Insr and Igf1r deletion in differentiated adult LCs, suggesting a feedback mechanism between cells at different steps of differentiation. Taken together, these data reveal the cell-autonomous and nonautonomous roles of the IGF system for proper development and maintenance of steroidogenic lineages.-Neirijnck, Y., Calvel, P., Kilcoyne, K. R., Kühne, F., Stévant, I., Griffeth, R. J., Pitetti, J.-L., Andric, S. A., Hu, M.-C., Pralong, F., Smith, L. B., Nef, S. Insulin and IGF1 receptors are essential for the development and steroidogenic function of adult Leydig cells.


Subject(s)
Cell Differentiation , Leydig Cells/metabolism , Receptor, Insulin/metabolism , Receptors, Somatomedin/metabolism , Stem Cells/metabolism , Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Animals , Corticosterone/genetics , Corticosterone/metabolism , Leydig Cells/cytology , Male , Mice , Mice, Knockout , Receptor, Insulin/genetics , Receptors, Somatomedin/genetics , Stem Cells/cytology , Testosterone/genetics , Testosterone/metabolism
3.
Int J Mol Sci ; 20(18)2019 Sep 09.
Article in English | MEDLINE | ID: mdl-31505893

ABSTRACT

Persistent research over the past few decades has clearly established that the insulin-like family of growth factors, which is composed of insulin and insulin-like growth factors 1 (IGF1) and 2 (IGF2), plays essential roles in sexual development and reproduction of both males and females. Within the male and female reproductive organs, ligands of the family act in an autocrine/paracrine manner, in order to guide different aspects of gonadogenesis, sex determination, sex-specific development or reproductive performance. Although our knowledge has greatly improved over the last years, there are still several facets that remain to be deciphered. In this review, we first briefly outline the principles of sexual development and insulin/IGF signaling, and then present our current knowledge, both in rodents and humans, about the involvement of insulin/IGFs in sexual development and reproductive functions. We conclude by highlighting some interesting remarks and delineating certain unanswered questions that need to be addressed in future studies.


Subject(s)
Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Reproduction/physiology , Sex Differentiation/physiology , Signal Transduction/physiology , Animals , Female , Humans , Male
4.
Kidney Int ; 93(5): 1142-1153, 2018 05.
Article in English | MEDLINE | ID: mdl-29459093

ABSTRACT

Congenital abnormalities of the kidney and the urinary tract (CAKUT) belong to the most common birth defects in human, but the molecular basis for the majority of CAKUT patients remains unknown. Here we show that the transcription factor SOX11 is a crucial regulator of kidney development. SOX11 is expressed in both mesenchymal and epithelial components of the early kidney anlagen. Deletion of Sox11 in mice causes an extension of the domain expressing Gdnf within rostral regions of the nephrogenic cord and results in duplex kidney formation. On the molecular level SOX11 directly binds and regulates a locus control region of the protocadherin B cluster. At later stages of kidney development, SOX11 becomes restricted to the intermediate segment of the developing nephron where it is required for the elongation of Henle's loop. Finally, mutation analysis in a cohort of patients suffering from CAKUT identified a series of rare SOX11 variants, one of which interferes with the transactivation capacity of the SOX11 protein. Taken together these data demonstrate a key role for SOX11 in normal kidney development and may suggest that variants in this gene predispose to CAKUT in humans.


Subject(s)
Kidney/abnormalities , Mutation , SOXC Transcription Factors/genetics , Ureter/abnormalities , Urogenital Abnormalities/genetics , Vesico-Ureteral Reflux/genetics , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , Genetic Association Studies , Genetic Predisposition to Disease , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Kidney/metabolism , Male , Mice, Knockout , Morphogenesis , Phenotype , Risk Factors , SOXC Transcription Factors/deficiency , Ureter/metabolism , Urogenital Abnormalities/metabolism , Urogenital Abnormalities/pathology , Vesico-Ureteral Reflux/metabolism , Vesico-Ureteral Reflux/pathology
5.
Stem Cells ; 31(7): 1252-65, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23533157

ABSTRACT

Stem cell-like properties of glioma initiating cells (GiCs) fuel glioblastoma (GBM) development by providing the different cell types that comprise the tumor. It is therefore likely that the molecular circuitries that regulate their decision to self-renew or commit to a more differentiated state may offer targets for future innovative therapies. In previous micro-RNA profiling studies to search for regulators of stem cell plasticity, we identified miR-18a* as a potential candidate and its expression correlated with the stemness state. Here, using human GiCs we found that miR-18a* expression promotes clonal proliferation in vitro and tumorigenicity in vivo. Mechanistically, ERK-dependent induction of miR-18a* directly represses expression of DLL3, an autocrine inhibitor of NOTCH, thus enhancing the level of activated NOTCH-1. Activated NOTCH-1 in turn is required for sustained ERK activation. This feed-forward loop, driven by miR-18a*, is required to turn on the SHH-GLI-NANOG network, essential for GiC self-renewal. Hence, by tightly regulating expression of DLL3, miR-18a* constitutes an important signaling mediator for fine tuning the level of GiC self-renewal.


Subject(s)
Glioma/genetics , MicroRNAs/genetics , Receptor, Notch1/metabolism , Aged , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Down-Regulation , Glioma/metabolism , Glioma/pathology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred NOD , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Middle Aged , Receptor, Notch1/genetics , Transfection
6.
Elife ; 122024 Mar 05.
Article in English | MEDLINE | ID: mdl-38441556

ABSTRACT

From a cohort of 167 infertile patients suffering from multiple morphological abnormalities of the flagellum (MMAF), pathogenic bi-allelic mutations were identified in the CCDC146 gene. In somatic cells, CCDC146 is located at the centrosome and at multiple microtubule-related organelles during mitotic division, suggesting that it is a microtubule-associated protein (MAP). To decipher the molecular pathogenesis of infertility associated with CCDC146 mutations, a Ccdc146 knock-out (KO) mouse line was created. KO male mice were infertile, and sperm exhibited a phenotype identical to CCDC146 mutated patients. CCDC146 expression starts during late spermiogenesis. In the spermatozoon, the protein is conserved but is not localized to centrioles, unlike in somatic cells, rather it is present in the axoneme at the level of microtubule doublets. Expansion microscopy associated with the use of the detergent sarkosyl to solubilize microtubule doublets suggests that the protein may be a microtubule inner protein (MIP). At the subcellular level, the absence of CCDC146 impacted all microtubule-based organelles such as the manchette, the head-tail coupling apparatus (HTCA), and the axoneme. Through this study, a new genetic cause of infertility and a new factor in the formation and/or structure of the sperm axoneme were characterized.


Subject(s)
Abnormalities, Multiple , Infertility, Male , Animals , Humans , Male , Mice , Centrioles , Infertility, Male/genetics , Mice, Knockout , Microtubule-Associated Proteins/genetics , Semen
7.
Hum Mol Genet ; 20(6): 1143-53, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21212101

ABSTRACT

Congenital abnormalities of the kidney and urinary tract are some of the most common defects detected in the unborn child. Kidney growth is controlled by the GDNF/RET signalling pathway, but the molecular events required for the activation of RET downstream targets are still poorly understood. Here we show that SOX9, a gene involved in campomelic dysplasia (CD) in humans, together with its close homologue SOX8, plays an essential role in RET signalling. Expression of SOX9 can be found from the earliest stages of renal development within the ureteric tip, the ureter mesenchyme and in a segment-specific manner during nephrogenesis. Using a tissue-specific knockout approach, we show that, in the ureteric tip, SOX8 and SOX9 are required for ureter branching, and double-knockout mutants exhibit severe kidney defects ranging from hypoplastic kidneys to renal agenesis. Further genetic analysis shows that SOX8/9 are required downstream of GDNF signalling for the activation of RET effector genes such as Sprouty1 and Etv5. At later stages of development, SOX9 is required to maintain ureteric tip identity and SOX9 ablation induces ectopic nephron formation. Taken together, our study shows that SOX9 acts at multiple steps during kidney organogenesis and identifies SOX8 and SOX9 as key factors within the RET signalling pathway. Our results also explain the aetiology of kidney hypoplasia found in a proportion of CD patients.


Subject(s)
Campomelic Dysplasia/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Kidney/embryology , Proto-Oncogene Proteins c-ret/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction , Animals , Campomelic Dysplasia/embryology , Campomelic Dysplasia/genetics , Cells, Cultured , Disease Models, Animal , Female , Humans , Kidney/metabolism , Male , Mice , Mice, Knockout , Organogenesis , Proto-Oncogene Proteins c-ret/genetics , SOX9 Transcription Factor/genetics , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism
8.
Cell Rep ; 42(3): 112191, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36862551

ABSTRACT

Adrenal cortex and gonads represent the two major steroidogenic organs in mammals. Both tissues are considered to share a common developmental origin characterized by the expression of Nr5a1/Sf1. The precise origin of adrenogonadal progenitors and the processes driving differentiation toward the adrenal or gonadal fate remain, however, elusive. Here, we provide a comprehensive single-cell transcriptomic atlas of early mouse adrenogonadal development including 52 cell types belonging to twelve major cell lineages. Trajectory reconstruction reveals that adrenogonadal cells emerge from the lateral plate rather than the intermediate mesoderm. Surprisingly, we find that gonadal and adrenal fates have already diverged prior to Nr5a1 expression. Finally, lineage separation into gonadal and adrenal fates involves canonical versus non-canonical Wnt signaling and differential expression of Hox patterning genes. Thus, our study provides important insights into the molecular programs of adrenal and gonadal fate choice and will be a valuable resource for further research into adrenogonadal ontogenesis.


Subject(s)
Gonads , Transcriptome , Mice , Animals , Transcriptome/genetics , Gonads/metabolism , Cell Lineage/genetics , Cell Differentiation/genetics , Gene Expression Profiling , Mammals
9.
Science ; 382(6670): 600-606, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37917714

ABSTRACT

Sex determination in mammals depends on the differentiation of the supporting lineage of the gonads into Sertoli or pregranulosa cells that govern testis and ovary development, respectively. Although the Y-linked testis-determining gene Sry has been identified, the ovarian-determining factor remains unknown. In this study, we identified -KTS, a major, alternatively spliced isoform of the Wilms tumor suppressor WT1, as a key determinant of female sex determination. Loss of -KTS variants blocked gonadal differentiation in mice, whereas increased expression, as found in Frasier syndrome, induced precocious differentiation of ovaries independently of their genetic sex. In XY embryos, this antagonized Sry expression, resulting in male-to-female sex reversal. Our results identify -KTS as an ovarian-determining factor and demonstrate that its time of activation is critical in gonadal sex differentiation.


Subject(s)
Ovary , Sex Determination Processes , WT1 Proteins , Animals , Female , Male , Mice , Ovary/growth & development , Sex Determination Processes/genetics , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Testis/growth & development , WT1 Proteins/genetics , WT1 Proteins/metabolism , Protein Isoforms
10.
Cell Rep ; 39(11): 110935, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35705036

ABSTRACT

Leydig cells (LCs) are the major androgen-producing cells in the testis. They arise from steroidogenic progenitors (SPs), whose origins, maintenance, and differentiation dynamics remain largely unknown. Single-cell transcriptomics reveal that the mouse steroidogenic lineage is specified as early as embryonic day 12.5 (E12.5) and has a dual mesonephric and coelomic origin. SPs specifically express the Wnt5a gene and evolve rapidly. At E12.5 and E13.5, they give rise first to an intermediate population of pre-LCs, and finally to fetal LCs. At E16.5, SPs possess the characteristics of the dormant progenitors at the origin of adult LCs and are also transcriptionally closely related to peritubular myoid cells (PMCs). In agreement with our in silico analysis, in vivo lineage tracing indicates that Wnt5a-expressing cells are bona fide progenitors of PMCs as well as fetal and adult LCs, contributing to most of the LCs present in the fetal and adult testis.


Subject(s)
Leydig Cells , Testis , Androgens , Animals , Cell Differentiation , Fetus , Male , Mice
11.
Elife ; 112022 04 22.
Article in English | MEDLINE | ID: mdl-35451961

ABSTRACT

Male infertility is an important health concern that is expected to have a major genetic etiology. Although high-throughput sequencing has linked gene defects to more than 50% of rare and severe sperm anomalies, less than 20% of common and moderate forms are explained. We hypothesized that this low success rate could at least be partly due to oligogenic defects - the accumulation of several rare heterozygous variants in distinct, but functionally connected, genes. Here, we compared fertility and sperm parameters in male mice harboring one to four heterozygous truncating mutations of genes linked to multiple morphological anomalies of the flagellum (MMAF) syndrome. Results indicated progressively deteriorating sperm morphology and motility with increasing numbers of heterozygous mutations. This first evidence of oligogenic inheritance in failed spermatogenesis strongly suggests that oligogenic heterozygosity could explain a significant proportion of asthenoteratozoospermia cases. The findings presented pave the way to further studies in mice and man.


Subject(s)
Abnormalities, Multiple , Asthenozoospermia , Infertility, Male , Abnormalities, Multiple/genetics , Asthenozoospermia/genetics , Humans , Infertility, Male/genetics , Male , Multifactorial Inheritance , Mutation , Sperm Tail , Spermatozoa
12.
Nat Commun ; 13(1): 4412, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35906245

ABSTRACT

Gonadal sexual fate in mammals is determined during embryonic development and must be actively maintained in adulthood. In the mouse ovary, oestrogen receptors and FOXL2 protect ovarian granulosa cells from transdifferentiation into Sertoli cells, their testicular counterpart. However, the mechanism underlying their protective effect is unknown. Here, we show that TRIM28 is required to prevent female-to-male sex reversal of the mouse ovary after birth. We found that upon loss of Trim28, ovarian granulosa cells transdifferentiate to Sertoli cells through an intermediate cell type, different from gonadal embryonic progenitors. TRIM28 is recruited on chromatin in the proximity of FOXL2 to maintain the ovarian pathway and to repress testicular-specific genes. The role of TRIM28 in ovarian maintenance depends on its E3-SUMO ligase activity that regulates the sex-specific SUMOylation profile of ovarian-specific genes. Our study identifies TRIM28 as a key factor in protecting the adult ovary from the testicular pathway.


Subject(s)
Ovary , Sumoylation , Animals , Female , Male , Mammals/metabolism , Mice , Ovary/metabolism , Sertoli Cells/metabolism , Testis/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tripartite Motif-Containing Protein 28/genetics , Tripartite Motif-Containing Protein 28/metabolism
13.
Sci Adv ; 8(21): eabm0972, 2022 May 27.
Article in English | MEDLINE | ID: mdl-35613264

ABSTRACT

Gonadal sex determination represents a unique model for studying cell fate decisions. However, a complete understanding of the different cell lineages forming the developing testis and ovary remains elusive. Here, we investigated the origin, specification, and subsequent sex-specific differentiation of a previously uncharacterized population of supporting-like cells (SLCs) in the developing mouse gonads. The SLC lineage is closely related to the coelomic epithelium and specified as early as E10.5, making it the first somatic lineage to be specified in the bipotential gonad. SLC progenitors are localized within the genital ridge at the interface with the mesonephros and initially coexpress Wnt4 and Sox9. SLCs become sexually dimorphic around E12.5, progressively acquire a more Sertoli- or pregranulosa-like identity and contribute to the formation of the rete testis and rete ovarii. Last, we found that WNT4 is a crucial regulator of the SLC lineage and is required for normal development of the rete testis.

14.
Dev Dyn ; 239(6): 1867-78, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20503382

ABSTRACT

WTX/AMER1 is a novel negative regulator of the WNT/beta-catenin pathway with mutations detected in Wilms' tumors and an X-linked sclerosing bone dysplasia. WTX/AMER1 (Fam123b) shares several domains of homology with two other recently identified proteins: AMER2 (Fam123a) and AMER3 (Fam123c). Here, we describe an in-depth expression analysis of all three members of this gene family during mouse embryonic development. All genes were strongly expressed in the central as well as the peripheral nervous system, thus suggesting important roles of this gene family during neurogenesis. Specific expression was found in the retina, inner ear, and nasal epithelium. Outside of the nervous system Wtx/Amer1 showed the broadest expression domains including cephalic and limb mesenchyme, skeletal muscle, bladder, gonads, lung bud, salivary glands, and the kidneys. The widespread expression pattern of Wtx/Amer1, together with its role as a modulator of the Wnt signaling pathway, suggest that Wtx/Amer1 serves pleiotropic roles during mammalian organogenesis.


Subject(s)
Embryonic Development/genetics , Animals , Embryo, Mammalian , Female , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mutation , Nervous System/metabolism , Peripheral Nervous System/metabolism , Pregnancy , Proteins/genetics , Signal Transduction/genetics , Wilms Tumor/genetics , Wilms Tumor/metabolism , beta Catenin/genetics , beta Catenin/metabolism
15.
Front Cell Dev Biol ; 9: 695546, 2021.
Article in English | MEDLINE | ID: mdl-34262907

ABSTRACT

Leydig cells (LC) are the main testicular androgen-producing cells. In eutherian mammals, two types of LCs emerge successively during testicular development, fetal Leydig cells (FLCs) and adult Leydig cells (ALCs). Both display significant differences in androgen production and regulation. Using bulk RNA sequencing, we compared the transcriptomes of both LC populations to characterize their specific transcriptional and functional features. Despite similar transcriptomic profiles, a quarter of the genes show significant variations in expression between FLCs and ALCs. Non-transcriptional events, such as alternative splicing was also observed, including a high rate of intron retention in FLCs compared to ALCs. The use of single-cell RNA sequencing data also allowed the identification of nine FLC-specific genes and 50 ALC-specific genes. Expression of the corticotropin-releasing hormone 1 (Crhr1) receptor and the ACTH receptor melanocortin type 2 receptor (Mc2r) specifically in FLCs suggests a dual regulation of steroidogenesis. The androstenedione synthesis by FLCs is stimulated by luteinizing hormone (LH), corticotrophin-releasing hormone (CRH), and adrenocorticotropic hormone (ACTH) whereas the testosterone synthesis by ALCs is dependent exclusively on LH. Overall, our study provides a useful database to explore LC development and functions.

16.
Cells ; 11(1)2021 12 30.
Article in English | MEDLINE | ID: mdl-35011680

ABSTRACT

The genetic landscape of male infertility is highly complex. It is estimated that at least 4000 genes are involved in human spermatogenesis, but only few have so far been extensively studied. In this study, we investigated by whole exome sequencing two cases of idiopathic non-obstructive azoospermia (NOA) due to severe hypospermatogenesis. After variant filtering and prioritizing, we retained for each patient a homozygous loss-of-function (LoF) variant in a testis-specific gene, C1orf185 (c.250C>T; p.Gln84Ter) and CCT6B (c.615-2A>G), respectively. Both variants are rare according to the gnomAD database and absent from our local control cohort (n = 445). To verify the implication of these candidate genes in NOA, we used the CRISPR/Cas9 system to invalidate the mouse orthologs 4930522H14Rik and Cct6b and produced two knockout (KO) mouse lines. Sperm and testis parameters of homozygous KO adult male mice were analyzed and compared with those of wild-type animals. We showed that homozygous KO males were fertile and displayed normal sperm parameters and a functional spermatogenesis. Overall, these results demonstrate that not all genes highly and specifically expressed in the testes are essential for spermatogenesis, and in particular, we conclude that bi-allelic variants of C1orf185 and CCT6B are most likely not to be involved in NOA and male fertility.


Subject(s)
Azoospermia/etiology , CRISPR-Cas Systems/genetics , Chaperonin Containing TCP-1/genetics , Exome Sequencing/methods , Testis/metabolism , Azoospermia/physiopathology , Humans , Male
17.
Endocrinology ; 160(2): 387-398, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30576429

ABSTRACT

The IGFs are the major intratesticular factors regulating immature Sertoli cell proliferation and are, therefore, critical to establish the magnitude of sperm production. However, the intratesticular source of IGF production and the downstream signaling pathway mediating IGF-dependent Sertoli cell proliferation remain unclear. Single-cell RNA sequencing on mouse embryonic testis revealed a robust expression of Igf1 and Igf2 in interstitial steroidogenic progenitors, suggesting that IGFs exert paracrine actions on immature Sertoli cells. To elucidate the intracellular signaling mechanism that underlies the proliferative effects of IGFs on immature Sertoli cells, we have generated mice with Sertoli cell-specific deletion of the Pten gene, a negative regulator of the phosphatidylinositol-3 kinase (PI3K)/AKT pathway, alone or together with the insulin receptor (Insr) and the IGF1 receptor (Igf1r). Although ablation of Pten appears dispensable for Sertoli cell proliferation and spermatogenesis, inactivation of Pten in the absence of Insr and Igf1r rescued the Sertoli cell proliferation rate during late fetal development, testis size, and sperm production. Overall, these findings suggest that IGFs secreted by interstitial progenitor cells act in a paracrine fashion to promote the proliferation of immature Sertoli cells through the IGF/PTEN/PI3K pathway.


Subject(s)
PTEN Phosphohydrolase/metabolism , Somatomedins/metabolism , Testis/metabolism , Animals , Cell Proliferation , Male , Mice , PTEN Phosphohydrolase/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Sertoli Cells/physiology , Spermatogenesis , Testis/growth & development
18.
Cell Rep ; 22(6): 1589-1599, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29425512

ABSTRACT

The gonad is a unique biological system for studying cell-fate decisions. However, major questions remain regarding the identity of somatic progenitor cells and the transcriptional events driving cell differentiation. Using time-series single-cell RNA sequencing on XY mouse gonads during sex determination, we identified a single population of somatic progenitor cells prior to sex determination. A subset of these progenitors differentiates into Sertoli cells, a process characterized by a highly dynamic genetic program consisting of sequential waves of gene expression. Another subset of multipotent cells maintains their progenitor state but undergoes significant transcriptional changes restricting their competence toward a steroidogenic fate required for the differentiation of fetal Leydig cells. Our findings confirm the presence of a unique multipotent progenitor population in the gonadal primordium that gives rise to both supporting and interstitial lineages. These also provide the most granular analysis of the transcriptional events occurring during testicular cell-fate commitment.


Subject(s)
Cell Differentiation/physiology , Leydig Cells/cytology , Sertoli Cells/cytology , Sex Determination Processes/physiology , Testis/cytology , Animals , Cell Lineage , Male , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL