Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
Int J Biol Sci ; 19(7): 2234-2255, 2023.
Article in English | MEDLINE | ID: mdl-37151878

ABSTRACT

In spermatozoa, the nuclear F-actin supports the acroplaxome, a subacrosomal structure involved in the correct exposure of several acrosomal membrane proteins; among them, the glycoprotein IZUMO1 is the major protein involved in sperm-oocyte fusion. Nuclear F-actin is also involved in sperm head shaping and chromosome compartmentalization. To date, few notions regarding the bivalent role of F-actin on sperm chromatin organization and IZUMO1 positioning have been reported. In our work, we characterized subcellular organization of F-actin in human high- and low-quality spermatozoa (A- and B-SPZ), respectively, showing that F-actin over-expression in sperm head of B-SPZ affected IZUMO1 localization. A correct IZUMO1 repositioning following in vitro induction of F-actin depolymerization, by cytochalasin D treatment, occurred. Interestingly, F-actin depolymerization was also associated with a correct acrosome repositioning, thus to favor a proper acrosome reaction onset, with changes in sperm nuclear size parameters and histone acetylation rate reaching high-quality conditions. In conclusion, the current work shows a key role of F-actin in the control of IZUMO1 localization as well as chromatin remodeling and acetylation events.


Subject(s)
Actins , Membrane Proteins , Male , Humans , Actins/metabolism , Cytochalasin D/pharmacology , Cytochalasin D/analysis , Cytochalasin D/metabolism , Membrane Proteins/metabolism , Semen/metabolism , Spermatozoa/metabolism , Immunoglobulins/metabolism
2.
Front Cell Dev Biol ; 10: 877270, 2022.
Article in English | MEDLINE | ID: mdl-35813201

ABSTRACT

Kisspeptins are involved in the regulation of hypothalamic-pituitary-gonadal axis, Leydig cell functions, and testosterone secretion, acting as endogenous ligands of the KISS1 receptor. ANKRD31 protein participates in male fertility, regulating meiotic progression, and epididymal sperm maturation. Here, we show that in Leydig cells, KISS1 receptor and ANKRD31 proteins physically interact; the formation of this protein complex is enhanced by Kisspeptin-10 that also modulates F-actin synthesis, favoring histone acetylation in chromatin and gene expression via the cytoskeletal-nucleoskeletal pathway. Kp/KISS1R system deregulation, expression impairment of cytoskeletal-nucleoskeletal mediators, Leydig gene targets, and the decreased testosterone secretion in Ankrd31 -/- testis strongly supported our hypothesis. Furthermore, cytochalasin D treatment subverted the gene expression induction dependent on Kisspeptin-10 action. In conclusion, the current work highlights a novel role for the Kisspeptin-10 in the induction of the cytoskeletal-nucleoskeletal route, downstream a physical interaction between KISS1 receptor and ANKRD31, with gene expression activation as final effect, in Leydig cells.

3.
J Clin Invest ; 131(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34850745

ABSTRACT

Large-cell calcifying Sertoli cell tumors (LCCSCTs) are among the most frequent lesions occurring in male Carney complex (CNC) patients. Although they constitute a key diagnostic criterion for this rare multiple neoplasia syndrome resulting from inactivating mutations of the tumor suppressor PRKAR1A, leading to unrepressed PKA activity, LCCSCT pathogenesis and origin remain elusive. Mouse models targeting Prkar1a inactivation in all somatic populations or separately in each cell type were generated to decipher the molecular and paracrine networks involved in the induction of CNC testis lesions. We demonstrate that the Prkar1a mutation was required in both stromal and Sertoli cells for the occurrence of LCCSCTs. Integrative analyses comparing transcriptomic, immunohistological data and phenotype of mutant mouse combinations led to the understanding of human LCCSCT pathogenesis and demonstrated PKA-induced paracrine molecular circuits in which the aberrant WNT4 signal production is a limiting step in shaping intratubular lesions and tumor expansion both in a mouse model and in human CNC testes.


Subject(s)
Carney Complex/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Sertoli Cells/cytology , Testicular Neoplasms/metabolism , Wnt4 Protein/metabolism , Animals , Apoptosis , Carney Complex/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Disease Models, Animal , Gene Expression Profiling , Genes, Tumor Suppressor , Humans , Male , Mice , Mice, Knockout , Mutation , Oligonucleotide Array Sequence Analysis , Paracrine Communication , Phenotype , Pigmentation , Seminiferous Tubules/metabolism , Testis/metabolism , Transcriptome
4.
Front Cell Dev Biol ; 9: 741975, 2021.
Article in English | MEDLINE | ID: mdl-34820371

ABSTRACT

Ankyrin proteins (ANKRD) are key mediators linking membrane and sub-membranous cytoskeletal proteins. Recent findings have highlighted a new role of ANKRD31 during spermatogenesis, elucidating its involvement in meiotic recombination and male germ cell progression. Following testicular differentiation, spermatozoa (SPZ) enter into the epididymis, where they undergo several biochemical and enzymatic changes. The epididymal epithelium is characterized by cell-to-cell junctions that are able to form the blood-epididymal barrier (BEB). This intricate epithelial structure provides the optimal microenvironment needed for epididymal sperm maturation. To date, no notions have been reported regarding a putative role of ANKRD31 in correct BEB formation. In our work, we generated an Ankrd31 knockout male mouse model (Ankrd31-/- ) and characterized its reproductive phenotype. Ankrd31-/- mice were infertile and exhibited oligo-astheno-teratozoospermia (a low number of immotile SPZ with abnormal morphological features). In addition, a complete deregulation of BEB was found in Ankrd31-/- , due to cell-to-cell junction anomalies. In order to suggest that BEB deregulation may depend on Ankrd31 gene deletion, we showed the physical interaction among ANKRD31 and some epithelial junction proteins in wild-type (WT) epididymides. In conclusion, the current work shows a key role of ANKRD31 in the control of germ cell progression as well as sperm and epididymal integrity.

6.
Genes (Basel) ; 12(5)2021 04 27.
Article in English | MEDLINE | ID: mdl-33925685

ABSTRACT

Nuclear architecture undergoes an extensive remodeling during spermatogenesis, especially at levels of spermatocytes (SPC) and spermatids (SPT). Interestingly, typical events of spermiogenesis, such as nuclear elongation, acrosome biogenesis, and flagellum formation, need a functional cooperation between proteins of the nuclear envelope and acroplaxome/manchette structures. In addition, nuclear envelope plays a key role in chromosome distribution. In this scenario, special attention has been focused on the LINC (linker of nucleoskeleton and cytoskeleton) complex, a nuclear envelope-bridge structure involved in the connection of the nucleoskeleton to the cytoskeleton, governing mechanotransduction. It includes two integral proteins: KASH- and SUN-domain proteins, on the outer (ONM) and inner (INM) nuclear membrane, respectively. The LINC complex is involved in several functions fundamental to the correct development of sperm cells such as head formation and head to tail connection, and, therefore, it seems to be important in determining male fertility. This review provides a global overview of the main LINC complex components, with a special attention to their subcellular localization in sperm cells, their roles in the regulation of sperm morphological maturation, and, lastly, LINC complex alterations associated to male infertility.


Subject(s)
Cell Nucleus/physiology , Cytoskeleton/metabolism , Cytoskeleton/physiology , Nuclear Envelope/metabolism , Nuclear Matrix/metabolism , Spermatozoa/metabolism , Spermatozoa/physiology , Animals , Cell Nucleus/metabolism , Humans , Infertility, Male/metabolism , Infertility, Male/physiopathology , Male , Mechanotransduction, Cellular/physiology , Nuclear Matrix/physiology , Spermatids/metabolism , Spermatids/physiology , Spermatocytes/metabolism , Spermatocytes/physiology
7.
J Mol Biol ; 433(7): 166805, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33450253

ABSTRACT

PiggyBac(PB)-like elements (pble) are members of a eukaryotic DNA transposon family. This family is of interest to evolutionary genomics because pble transposases have been domesticated at least 9 times in vertebrates. The amino acid sequence of pble transposases can be split into three regions: an acidic N-terminal domain (~100 aa), a central domain (~400 aa) containing a DD[D/E] catalytic triad, and a cysteine-rich domain (CRD; ~90 aa). Two recent reports suggested that a functional CRD is required for pble transposase activity. Here we found that two CRD-deficient pble transposases, a PB variant and an isoform encoded by the domesticated PB-derived vertebrate transposase gene 5 (pgbd5) trigger transposition of the Ifp2 pble. When overexpressed in HeLa cells, these CRD-deficient transposases can insert Ifp2 elements with proper and improper transposon ends, associated with deleterious effects on cells. Finally, we found that mouse CRD-deficient transposase Pgbd5, as well as PB, do not insert pbles at random into chromosomes. Transposition events occurred more often in genic regions, in the neighbourhood of the transcription start sites and were often found in genes predominantly expressed in the human central nervous system.


Subject(s)
DNA Transposable Elements/genetics , Nerve Tissue Proteins/genetics , Protein Domains/genetics , Transposases/genetics , Animals , Chromosomes/genetics , HeLa Cells , Humans , Mice , Recombination, Genetic
8.
BMC Genomics ; 20(1): 734, 2019 Oct 14.
Article in English | MEDLINE | ID: mdl-31610792

ABSTRACT

BACKGROUND: More and more eukaryotic genomes are sequenced and assembled, most of them presented as a complete model in which missing chromosomal regions are filled by Ns and where a few chromosomes may be lacking. Avian genomes often contain sequences with high GC content, which has been hypothesized to be at the origin of many missing sequences in these genomes. We investigated features of these missing sequences to discover why some may not have been integrated into genomic libraries and/or sequenced. RESULTS: The sequences of five red jungle fowl cDNA models with high GC content were used as queries to search publicly available datasets of Illumina and Pacbio sequencing reads. These were used to reconstruct the leptin, TNFα, MRPL52, PCP2 and PET100 genes, all of which are absent from the red jungle fowl genome model. These gene sequences displayed elevated GC contents, had intron sizes that were sometimes larger than non-avian orthologues, and had non-coding regions that contained numerous tandem and inverted repeat sequences with motifs able to assemble into stable G-quadruplexes and intrastrand dyadic structures. Our results suggest that Illumina technology was unable to sequence the non-coding regions of these genes. On the other hand, PacBio technology was able to sequence these regions, but with dramatically lower efficiency than would typically be expected. CONCLUSIONS: High GC content was not the principal reason why numerous GC-rich regions of avian genomes are missing from genome assembly models. Instead, it is the presence of tandem repeats containing motifs capable of assembling into very stable secondary structures that is likely responsible.


Subject(s)
Base Composition , Chickens/genetics , Genomics/methods , Animals , DNA/chemistry , DNA/genetics , High-Throughput Nucleotide Sequencing/veterinary , Introns , Sequence Analysis, DNA/veterinary
9.
Chromosome Res ; 26(4): 297-306, 2018 12.
Article in English | MEDLINE | ID: mdl-30225548

ABSTRACT

The chicken genome was the third vertebrate to be sequenced. To date, its sequence and feature annotations are used as the reference for avian models in genome sequencing projects developed on birds and other Sauropsida species, and in genetic studies of domesticated birds of economic and evolutionary biology interest. Therefore, an accurate description of this genome model is important to a wide number of scientists. Here, we review the location and features of a very basic element, the centromeres of chromosomes in the galGal5 genome model. Centromeres are elements that are not determined by their DNA sequence but by their epigenetic status, in particular by the accumulation of the histone-like protein CENP-A. Comparison of data from several public sources (primarily marker probes flanking centromeres using fluorescent in situ hybridization done on giant lampbrush chromosomes and CENP-A ChIP-seq datasets) with galGal5 annotations revealed that centromeres are likely inappropriately mapped in 9 of the 16 galGal5 chromosome models in which they are described. Analysis of karyology data confirmed that the location of the main CENP-A peaks in chromosomes is the best means of locating the centromeres in 25 galGal5 chromosome models, the majority of which (16) are fully sequenced and assembled. This data re-analysis reaffirms that several sources of information should be examined to produce accurate genome annotations, particularly for basic structures such as centromeres that are epigenetically determined.


Subject(s)
Centromere Protein A/metabolism , Centromere/ultrastructure , Chickens/genetics , Genome/genetics , Animals , Chromosomal Proteins, Non-Histone , Chromosome Mapping/standards , Epigenomics
10.
Article in English | MEDLINE | ID: mdl-29456523

ABSTRACT

G protein-coupled receptors (GPCRs) exert their physiological function by transducing a complex signaling network that coordinates gene expression and dictates the phenotype of highly differentiated cells. Much is known about the gene networks they transcriptionally regulate upon ligand exposure in a process that takes hours before a new protein is synthesized. However, far less is known about GPCR impact on the translational machinery and subsequent mRNA translation, although this gene regulation level alters the cell phenotype in a strikingly different timescale. In fact, mRNA translation is an early response kinetically connected to signaling events, hence it leads to the synthesis of a new protein within minutes following receptor activation. By these means, mRNA translation is responsive to subtle variations of the extracellular environment. In addition, when restricted to cell subcellular compartments, local mRNA translation contributes to cell micro-specialization, as observed in synaptic plasticity or in cell migration. The mechanisms that control where in the cell an mRNA is translated are starting to be deciphered. But how an extracellular signal triggers such local translation still deserves extensive investigations. With the advent of high-throughput data acquisition, it now becomes possible to review the current knowledge on the translatome that some GPCRs regulate, and how this information can be used to explore GPCR-controlled local translation of mRNAs.

11.
C R Biol ; 341(2): 65-74, 2018 Feb.
Article in French | MEDLINE | ID: mdl-29326051

ABSTRACT

With the advent of next-generation sequencing technologies, identifying the translatome, which includes genome-wide ribosome-associated mRNAs, provides new opportunities to define faithfully the protein repertoire of a cell, as opposed to transcriptomic approaches. In addition, the role that extracellular signals such as hormonal modulations could play on the translatome remains to be deciphered. In particular, the regulation of the translatome by G protein-coupled receptors (GPCR) is still poorly described, albeit the trophic role that many receptors of this family play in their target cells. Here, we provide an overview of the current methods that are used to study the translatome, applied to the GPCR receptor family.


Subject(s)
High-Throughput Nucleotide Sequencing , Receptors, G-Protein-Coupled/genetics , Transcriptome , Genome , Humans , Proteins/metabolism , RNA, Messenger/genetics
12.
Science ; 354(6314): 909-912, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27856912

ABSTRACT

DNA methylation is prevalent in mammalian genomes and plays a central role in the epigenetic control of development. The mammalian DNA methylation machinery is thought to be composed of three DNA methyltransferase enzymes (DNMT1, DNMT3A, and DNMT3B) and one cofactor (DNMT3L). Here, we describe the discovery of Dnmt3C, a de novo DNA methyltransferase gene that evolved via a duplication of Dnmt3B in rodent genomes and was previously annotated as a pseudogene. We show that DNMT3C is the enzyme responsible for methylating the promoters of evolutionarily young retrotransposons in the male germ line and that this specialized activity is required for mouse fertility. DNMT3C reveals the plasticity of the mammalian DNA methylation system and expands the scope of the mechanisms involved in the epigenetic control of retrotransposons.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Epigenesis, Genetic , Mutagenesis/genetics , Promoter Regions, Genetic , Retroelements , Spermatogonia/enzymology , Animals , Cell Line , DNA (Cytosine-5-)-Methyltransferases/classification , DNA (Cytosine-5-)-Methyltransferases/genetics , Ethylnitrosourea/pharmacology , Gene Knockout Techniques , Hypogonadism/chemically induced , Hypogonadism/genetics , Hypogonadism/pathology , Male , Mice , Phylogeny , Spermatogonia/drug effects , Testis/drug effects , Testis/pathology
13.
BMC Genomics ; 17(1): 659, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27542599

ABSTRACT

BACKGROUND: The program RepeatMasker and the database Repbase-ISB are part of the most widely used strategy for annotating repeats in animal genomes. They have been used to show that avian genomes have a lower repeat content (8-12 %) than the sequenced genomes of many vertebrate species (30-55 %). However, the efficiency of such a library-based strategies is dependent on the quality and completeness of the sequences in the database that is used. An alternative to these library based methods are methods that identify repeats de novo. These alternative methods have existed for a least a decade and may be more powerful than the library based methods. We have used an annotation strategy involving several complementary de novo tools to determine the repeat content of the model genome galGal4 (1.04 Gbp), including identifying simple sequence repeats (SSRs), tandem repeats and transposable elements (TEs). RESULTS: We annotated over one Gbp. of the galGal4 genome and showed that it is composed of approximately 19 % SSRs and TEs repeats. Furthermore, we estimate that the actual genome of the red jungle fowl contains about 31-35 % repeats. We find that library-based methods tend to overestimate TE diversity. These results have a major impact on the current understanding of repeats distributions throughout chromosomes in the red jungle fowl. CONCLUSIONS: Our results are a proof of concept of the reliability of using de novo tools to annotate repeats in large animal genomes. They have also revealed issues that will need to be resolved in order to develop gold-standard methodologies for annotating repeats in eukaryote genomes.


Subject(s)
Chickens/genetics , Genome , Genomics , Tandem Repeat Sequences , Animals , Chromosome Mapping , Computational Biology/methods , CpG Islands , DNA Transposable Elements , Data Mining , Genomics/methods , Microsatellite Repeats , Molecular Sequence Annotation , Software
14.
Mol Cell Endocrinol ; 423: 96-112, 2016 Mar 05.
Article in English | MEDLINE | ID: mdl-26772142

ABSTRACT

The AMP-activated protein kinase (AMPK) is an important regulator of cellular energy homeostasis which plays a role in fertility. Complete disruption of the AMPK catalytic subunit α1 gene (α1AMPK KO) in male mice results in a decrease in litter size which is associated with the production of altered sperm morphology and motility. Because of the importance of Sertoli cells in the formation of germ cells, we have chosen to selectively disrupt α1AMPK only in the Sertoli cells in mice (Sc-α1AMPK-KO mice). Specific deletion of the α1AMPK gene in Sertoli cells resulted in a 25% reduction in male fertility associated with abnormal spermatozoa with a thin head. No clear alterations in testis morphology or modification in the number of Sertoli cells in vivo were observed, but a dysregulation in energy metabolism in Sertoli cells occurred. We have reported an increase in lactate production, in lipid droplets, and a reduction in ATP production in Sc-α1AMPK-KO Sertoli cells. These perturbations were associated with lower expression of mitochondrial markers (cytochrome c and PGC1-α). In addition another metabolic sensor, the deacetylase SIRT1, had a reduction in expression which is correlated with a decline in deacetylase activity. Finally, expression and localization of junctions forming the blood-testis barrier between Sertoli cells themselves and with germ cells were deregulated in Sc-α1AMPK-KO. In conclusion, these results suggest that dysregulation of the energy sensing machinery exclusively through disruption of α1AMPK in Sertoli cells translates to a reduction in the quality of germ cells and fertility.


Subject(s)
AMP-Activated Protein Kinases/genetics , Acrosome/enzymology , Sertoli Cells/enzymology , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , Female , Gene Expression , Infertility, Male/enzymology , Infertility, Male/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Transport , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sperm Motility
15.
Hum Mol Genet ; 24(17): 4792-808, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26045466

ABSTRACT

Somatic cell cytokinesis was shown to involve the insertion of sphingolipids (SLs) to midbodies prior to abscission. Spermatogenic midbodies transform into stable intercellular bridges (ICBs) connecting clonal daughter cells in a syncytium. This process requires specialized SL structures. (1) Using high resolution-mass spectrometric imaging, we show in situ a biphasic pattern of SL synthesis with testis-specific anchors. This pattern correlates with and depends on ceramide synthase 3 (CerS3) localization in both, pachytene spermatocytes until completion of meiosis and elongating spermatids. (2) Blocking the pathways to germ cell-specific ceramides (CerS3-KO) and further to glycosphingolipids (glucosylceramide synthase-KO) in mice highlights the need for special SLs for spermatid ICB stability. In contrast to somatic mitosis these SLs require ultra-long polyunsaturated anchors with unique physico-chemical properties, which can only be provided by CerS3. Loss of these anchors causes enhanced apoptosis during meiosis, formation of multinuclear giant cells and spermatogenic arrest. Hence, testis-specific SLs, which we also link to CerS3 in human testis, are quintessential for male fertility.


Subject(s)
Cell Membrane/metabolism , Cytokinesis , Meiosis/physiology , Sphingolipids/metabolism , Sphingosine N-Acyltransferase/metabolism , Animals , Apoptosis/genetics , Fatty Acids/metabolism , Gene Expression , Germ Cells/metabolism , Humans , Infertility , Male , Mice , RNA, Messenger/genetics , Spermatogenesis , Sphingolipids/biosynthesis , Sphingosine N-Acyltransferase/genetics , Testis/metabolism , Testis/pathology
16.
PLoS One ; 9(8): e105687, 2014.
Article in English | MEDLINE | ID: mdl-25144714

ABSTRACT

The Sertoli cells are critical regulators of testis differentiation and development. In the adult, however, their known function is restricted largely to maintenance of spermatogenesis. To determine whether the Sertoli cells regulate other aspects of adult testis biology we have used a novel transgenic mouse model in which Amh-Cre induces expression of the receptor for Diphtheria toxin (iDTR) specifically within Sertoli cells. This causes controlled, cell-specific and acute ablation of the Sertoli cell population in the adult animal following Diphtheria toxin injection. Results show that Sertoli cell ablation leads to rapid loss of all germ cell populations. In addition, adult Leydig cell numbers decline by 75% with the remaining cells concentrated around the rete and in the sub-capsular region. In the absence of Sertoli cells, peritubular myoid cell activity is reduced but the cells retain an ability to exclude immune cells from the seminiferous tubules. These data demonstrate that, in addition to support of spermatogenesis, Sertoli cells are required in the adult testis both for retention of the normal adult Leydig cell population and for support of normal peritubular myoid cell function. This has implications for our understanding of male reproductive disorders and wider androgen-related conditions affecting male health.


Subject(s)
Cell Communication/physiology , Leydig Cells/metabolism , Seminiferous Tubules/metabolism , Sertoli Cells/metabolism , Spermatocytes/metabolism , Animals , Cell Communication/drug effects , Diphtheria Toxin/toxicity , Leydig Cells/cytology , Male , Mice , Mice, Transgenic , Seminiferous Tubules/cytology , Sertoli Cells/cytology , Spermatocytes/cytology
17.
Development ; 141(10): 2096-107, 2014 May.
Article in English | MEDLINE | ID: mdl-24803656

ABSTRACT

The four related mammalian MEX-3 RNA-binding proteins are evolutionarily conserved molecules for which the in vivo functions have not yet been fully characterized. Here, we report that male mice deficient for the gene encoding Mex3b are subfertile. Seminiferous tubules of Mex3b-deficient mice are obstructed as a consequence of the disrupted phagocytic capacity of somatic Sertoli cells. In addition, both the formation and the integrity of the blood-testis barrier are compromised owing to mislocalization of N-cadherin and connexin 43 at the surface of Sertoli cells. We further establish that Mex3b acts to regulate the cortical level of activated Rap1, a small G protein controlling phagocytosis and cell-cell interaction, through the activation and transport of Rap1GAP. The active form of Rap1 (Rap1-GTP) is abnormally increased at the membrane cortex and chemically restoring Rap1-GTP to physiological levels rescues the phagocytic and adhesion abilities of Sertoli cells. Overall, these findings implicate Mex3b in the spatial organization of the Rap1 pathway that orchestrates Sertoli cell functions.


Subject(s)
RNA-Binding Proteins/physiology , Sertoli Cells/physiology , rap1 GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Female , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Knockout , RNA-Binding Proteins/genetics , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , Signal Transduction , Tissue Distribution/genetics , rap1 GTP-Binding Proteins/genetics
18.
Development ; 141(10): 2139-49, 2014 May.
Article in English | MEDLINE | ID: mdl-24803659

ABSTRACT

Sertoli cells (SCs) regulate testicular fate in the differentiating gonad and are the main regulators of spermatogenesis in the adult testis; however, their role during the intervening period of testis development, in particular during adult Leydig cell (ALC) differentiation and function, remains largely unknown. To examine SC function during fetal and prepubertal development we generated two transgenic mouse models that permit controlled, cell-specific ablation of SCs in pre- and postnatal life. Results show that SCs are required: (1) to maintain the differentiated phenotype of peritubular myoid cells (PTMCs) in prepubertal life; (2) to maintain the ALC progenitor population in the postnatal testis; and (3) for development of normal ALC numbers. Furthermore, our data show that fetal LCs function independently from SC, germ cell or PTMC support in the prepubertal testis. Together, these findings reveal that SCs remain essential regulators of testis development long after the period of sex determination. These findings have significant implications for our understanding of male reproductive disorders and wider androgen-related conditions affecting male health.


Subject(s)
Cell Differentiation , Leydig Cells/physiology , Seminiferous Epithelium/cytology , Sertoli Cells/physiology , Sexual Maturation/physiology , Testis/cytology , Testis/growth & development , Animals , Animals, Newborn , Male , Mice , Mice, Nude , Mice, Transgenic , Seminiferous Tubules/cytology
19.
PLoS One ; 8(9): e74015, 2013.
Article in English | MEDLINE | ID: mdl-24040148

ABSTRACT

Among T3 receptors, TRα1 is ubiquitous and its deletion or a specific expression of a dominant-negative TRα1 isoform in Sertoli cell leads to an increase in testis weight and sperm production. The identification of a 43-kDa truncated form of the nuclear receptor TRα1 (p43) in the mitochondrial matrix led us to test the hypothesis that this mitochondrial transcription factor could regulate Sertoli cell proliferation. Here we report that p43 depletion in mice increases testis weight and sperm reserve. In addition, we found that p43 deletion increases Sertoli cell proliferation in postnatal testis at 3 days of development. Electron microscopy studies evidence an alteration of mitochondrial morphology observed specifically in Sertoli cells of p43-/- mice. Moreover, gene expression studies indicate that the lack of p43 in testis induced an alteration of the mitochondrial-nuclear cross-talk. In particular, the up-regulation of Cdk4 and c-myc pathway in p43-/- probably explain the extended proliferation recorded in Sertoli cells of these mice. Our finding suggests that T3 limits post-natal Sertoli cell proliferation mainly through its mitochondrial T3 receptor p43.


Subject(s)
Gene Deletion , Mitochondria/metabolism , Sertoli Cells/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Animals , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Proliferation , Female , Gene Expression Regulation , Male , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/ultrastructure , Organ Size , Protein Isoforms , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sertoli Cells/pathology , Sertoli Cells/ultrastructure , Testis/metabolism , Testis/pathology
20.
Mol Endocrinol ; 27(5): 814-27, 2013 May.
Article in English | MEDLINE | ID: mdl-23518924

ABSTRACT

Testis size and sperm production are directly correlated to the total number of adult Sertoli cells (SCs). Although the establishment of an adequate number of SCs is crucial for future male fertility, the identification and characterization of the factors regulating SC survival, proliferation, and maturation remain incomplete. To investigate whether the IGF system is required for germ cell (GC) and SC development and function, we inactivated the insulin receptor (Insr), the IGF1 receptor (Igf1r), or both receptors specifically in the GC lineage or in SCs. Whereas ablation of insulin/IGF signaling appears dispensable for GCs and spermatogenesis, adult testes of mice lacking both Insr and Igf1r in SCs (SC-Insr;Igf1r) displayed a 75% reduction in testis size and daily sperm production as a result of a reduced proliferation rate of immature SCs during the late fetal and early neonatal testicular period. In addition, in vivo analyses revealed that FSH requires the insulin/IGF signaling pathway to mediate its proliferative effects on immature SCs. Collectively, these results emphasize the essential role played by growth factors of the insulin family in regulating the final number of SCs, testis size, and daily sperm output. They also indicate that the insulin/IGF signaling pathway is required for FSH-mediated SC proliferation.


Subject(s)
Follicle Stimulating Hormone/metabolism , Receptor, Insulin/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Animals , Cell Count , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation , Cell Shape/drug effects , Female , Fetus/cytology , Fetus/embryology , Gene Expression Profiling , Germ Cells/cytology , Germ Cells/drug effects , Germ Cells/metabolism , Humans , Leydig Cells/cytology , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Organ Size/drug effects , Organ Size/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Seminiferous Tubules/cytology , Seminiferous Tubules/drug effects , Seminiferous Tubules/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Spermatogenesis/drug effects , Spermatogenesis/genetics , Spermatozoa/cytology , Spermatozoa/drug effects , Spermatozoa/metabolism , Thyroid Hormones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...