Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 7860, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36543770

ABSTRACT

Seminiferous tubules (STs) in the mammalian testes are connected to the rete testis (RT) via a Sertoli valve (SV). Spermatozoa produced in the STs are released into the tubular luminal fluid and passively transported through the SV into the RT. However, the physiological functions of the RT and SV remain unclear. Here, we identified the expression of Sox17 in RT epithelia. The SV valve was disrupted before puberty in RT-specific Sox17 conditional knockout (Sox17-cKO) male mice. This induced a backflow of RT fluid into the STs, which caused aberrant detachment of immature spermatids. RT of Sox17-cKO mice had reduced expression levels of various growth factor genes, which presumably support SV formation. When transplanted next to the Sox17+ RT, Sertoli cells of Sox17-cKO mice reconstructed the SV and supported proper spermiogenesis in the STs. This study highlights the novel and unexpected modulatory roles of the RT in SV valve formation and spermatogenesis in mouse testes, as a downstream action of Sox17.


Subject(s)
Rete Testis , SOXF Transcription Factors , Sexual Maturation , Spermatogenesis , Animals , Male , Mice , Epithelium , HMGB Proteins/metabolism , Mammals , Mice, Knockout , Rete Testis/metabolism , Sertoli Cells/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Spermatogenesis/genetics , Testis/metabolism
2.
Development ; 148(9)2021 05 01.
Article in English | MEDLINE | ID: mdl-33914868

ABSTRACT

In mammalian ovaries, immature oocytes are reserved in primordial follicles until their activation for potential ovulation. Precise control of primordial follicle activation (PFA) is essential for reproduction, but how this is achieved is unclear. Here, we show that canonical wingless-type MMTV integration site family (WNT) signaling is pivotal for pre-granulosa cell (pre-GC) activation during PFA. We identified several WNT ligands expressed in pre-GCs that act in an autocrine manner. Inhibition of WNT secretion from pre-GCs/GCs by conditional knockout (cKO) of the wntless (Wls) gene led to female infertility. In Wls cKO mice, GC layer thickness was greatly reduced in growing follicles, which resulted in impaired oocyte growth with both an abnormal, sustained nuclear localization of forkhead box O3 (FOXO3) and reduced phosphorylation of ribosomal protein S6 (RPS6). Constitutive stabilization of ß-catenin (CTNNB1) in pre-GCs/GCs induced morphological changes of pre-GCs from a squamous into a cuboidal form, though it did not influence oocyte activation. Our results reveal that canonical WNT signaling plays a permissive role in the transition of pre-GCs to GCs, which is an essential step to support oocyte growth.


Subject(s)
Fertility , Granulosa Cells/metabolism , Infertility, Female/metabolism , Ovary/metabolism , Wnt Signaling Pathway , Animals , Female , Mice , Mice, Knockout , Oocytes/metabolism , Oogenesis , Ovarian Follicle/metabolism , Ovulation , Transcriptome , WT1 Proteins/genetics , beta Catenin/genetics
3.
Sci Rep ; 11(1): 1110, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441739

ABSTRACT

In mammalian testes, undifferentiated spermatogonia (Aundiff) undergo differentiation in response to retinoic acid (RA), while their progenitor states are partially maintained by fibroblast growth factors (FGFs). Sertoli valve (SV) is a region located at the terminal end of seminiferous tubule (ST) adjacent to the rete testis (RT), where the high density of Aundiff is constitutively maintained with the absence of active spermatogenesis. However, the molecular and cellular characteristics of SV epithelia still remain unclear. In this study, we first identified the region-specific AKT phosphorylation in the SV Sertoli cells and demonstrated non-cell autonomous specialization of Sertoli cells in the SV region by performing a Sertoli cell ablation/replacement experiment. The expression of Fgf9 was detected in the RT epithelia, while the exogenous administration of FGF9 caused ectopic AKT phosphorylation in the Sertoli cells of convoluted ST. Furthermore, we revealed the SV region-specific expression of Cyp26a1, which encodes an RA-degrading enzyme, and demonstrated that the increased RA levels in the SV region disrupt its pool of Aundiff by inducing their differentiation. Taken together, RT-derived FGFs and low levels of RA signaling contribute to the non-cell-autonomous regionalization of the SV epithelia and its local maintenance of Aundiff in the SV region.


Subject(s)
Seminiferous Tubules/metabolism , Sertoli Cells/metabolism , Tretinoin/metabolism , Animals , Cell Differentiation , Epithelium/physiology , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/analysis , Regeneration , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Seminiferous Tubules/drug effects , Seminiferous Tubules/growth & development , Sertoli Cells/physiology , Sertoli Cells/transplantation , Signal Transduction , Spermatogenesis , Tretinoin/pharmacology , Up-Regulation
4.
Reproduction ; 154(2): 135-143, 2017 08.
Article in English | MEDLINE | ID: mdl-28559472

ABSTRACT

USP9X (ubiquitin-specific peptidase 9, X chromosome) is the mammalian orthologue of Drosophila deubiquitinase fat facets that was previously shown to regulate the maintenance of the germ cell lineage partially through stabilizing Vasa, one of the widely conserved factors crucial for gametogenesis. Here, we demonstrate that USP9X is expressed in the gonocytes and spermatogonia in mouse testes from newborn to adult stages. By using Vasa-Cre mice, germ cell-specific conditional deletion of Usp9x from the embryonic stage showed no abnormality in the developing testes by 1 week and no appreciable defects in the undifferentiated and differentiating spermatogonia at postnatal and adult stages. Interestingly, after 2 weeks, Usp9x-null spermatogenic cells underwent apoptotic cell death at the early spermatocyte stage, and then, caused subsequent aberrant spermiogenesis, which resulted in a complete infertility of Usp9x conditional knockout male mice. These data provide the first evidence of the crucial role of the spermatogonial USP9X during transition from the mitotic to meiotic phases and/or maintenance of early meiotic phase in Usp9x conditional knockout testes.


Subject(s)
Endopeptidases/metabolism , Fertility , Infertility, Male/enzymology , Spermatogenesis , Spermatogonia/enzymology , Testis/enzymology , Age Factors , Animals , Apoptosis , Endopeptidases/deficiency , Endopeptidases/genetics , Genotype , Infertility, Male/genetics , Infertility, Male/physiopathology , Male , Meiosis , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Spermatogonia/pathology , Testis/pathology , Testis/physiopathology , Ubiquitin Thiolesterase
5.
Biochem Biophys Res Commun ; 476(4): 546-552, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27255992

ABSTRACT

In mouse testes, spermatogonial stem cells (SSCs), a subpopulation of GFRα1 (GDNF family receptor-α1)-positive spermatogonia, are widely distributed along the convoluted seminiferous tubules. The proliferation and differentiation of the SSCs are regulated in part by local expression of GDNF (glial cell-derived neurotorphic factor), one of major niche factors for SSCs. However, the in vivo dynamics of the GDNF-stimulated GFRα1-positive spermatogonia remains unclear. Here, we developed a simple method for transplanting DiI-labeled and GDNF-soaked beads into the mouse testicular interstitium. By using this method, we examined the dynamics of GFRα1-positive spermatogonia in the tubular walls close to the transplanted GDNF-soaked beads. The bead-derived GDNF signals were able to induce the stratified aggregate formation of GFRα1-positive undifferentiated spermatogonia by day 3 post-transplantation. Each aggregate consisted of tightly compacted Asingle and marginal Apaired-Aaligned GFRα1-positive spermatogonia and was surrounded by Aaligned GFRα1-negative spermatogonia at more advanced stages. These data not only provide in vivo evidence for the inductive roles of GDNF in forming a rapid aggregation of GFRα1-positive spermatogonia but also indicate the usefulness of this in vivo assay system of various growth factors for the stem/progenitor spermatogonia in mammalian spermatogenesis.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Spermatogonia/metabolism , Animals , Cell Aggregation/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Drug Implants/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Signal Transduction , Spermatogenesis/drug effects , Spermatogenesis/physiology , Spermatogonia/drug effects , Stem Cell Niche/drug effects , Testis/cytology , Testis/drug effects , Testis/metabolism
6.
Sci Rep ; 6: 24171, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27053385

ABSTRACT

Embryonic implantation comprises a dynamic and complicated series of events, which takes place only when the maternal uterine endometrium is in a receptive state. Blastocysts reaching the uterus communicate with the uterine endometrium to implant within a narrow time window. Interplay among various signalling molecules and transcription factors under the control of ovarian hormones is necessary for successful establishment of pregnancy. However, the molecular mechanisms that allow embryonic implantation in the receptive endometrium are still largely unknown. Here, we show that Sry-related HMG box gene-17 (Sox17) heterozygous mutant female mice exhibit subfertility due to implantation failure. Sox17 was expressed in the oviduct, uterine luminal epithelium, and blood vessels. Sox17 heterozygosity caused no appreciable defects in ovulation, fertilisation, blastocyst formation, and gross morphology of the oviduct and uterus. Another group F Sox transcription factor, Sox7, was also expressed in the uterine luminal and glandular epithelium relatively weakly. Despite uterine Sox7 expression, a significant reduction in the number of implantation sites was observed in Sox17 heterozygous mutant females due to haploinsufficiency. Our findings revealed a novel role of Sox17 in uterine receptivity to embryo implantation.


Subject(s)
Embryo Implantation/genetics , HMGB Proteins/genetics , Haploinsufficiency , Infertility, Female/genetics , SOXF Transcription Factors/genetics , Animals , Blastocyst/metabolism , Blotting, Western , Embryonic Development/genetics , Epithelium/metabolism , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HMGB Proteins/metabolism , Infertility, Female/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Oviducts/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SOXF Transcription Factors/metabolism , Uterus/metabolism
7.
Proc Natl Acad Sci U S A ; 113(11): E1489-97, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929341

ABSTRACT

Spermatogonial stem cells (SSCs) fuel the production of male germ cells but the mechanisms behind SSC self-renewal, proliferation, and differentiation are still poorly understood. Using the Wnt target gene Axin2 and genetic lineage-tracing experiments, we found that undifferentiated spermatogonia, comprising SSCs and transit amplifying progenitor cells, respond to Wnt/ß-catenin signals. Genetic elimination of ß-catenin indicates that Wnt/ß-catenin signaling promotes the proliferation of these cells. Signaling is likely initiated by Wnt6, which is uniquely expressed by neighboring Sertoli cells, the only somatic cells in the seminiferous tubule that support germ cells and act as a niche for SSCs. Therefore, unlike other stem cell systems where Wnt/ß-catenin signaling is implicated in self-renewal, the Wnt pathway in the testis specifically contributes to the proliferation of SSCs and progenitor cells.


Subject(s)
Spermatogonia/cytology , Testis/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Axin Protein/genetics , Axin Protein/metabolism , Cell Differentiation , Cell Proliferation , Male , Mice, Mutant Strains , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Sertoli Cells/metabolism , Spermatogonia/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Testis/cytology , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics
8.
Genes Dev ; 27(2): 169-81, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23322300

ABSTRACT

The liver is a unique organ with a remarkably high potential to regenerate upon injuries. In severely damaged livers where hepatocyte proliferation is impaired, facultative liver progenitor cells (LPCs) proliferate and are assumed to contribute to regeneration. An expansion of LPCs is often observed in patients with various types of liver diseases. However, the underlying mechanism of LPC activation still remains largely unknown. Here we show that a member of the fibroblast growth factor (FGF) family, FGF7, is a critical regulator of LPCs. Its expression was induced concomitantly with LPC response in the liver of mouse models as well as in the serum of patients with acute liver failure. Fgf7-deficient mice exhibited markedly depressed LPC expansion and higher mortality upon toxin-induced hepatic injury. Transgenic expression of FGF7 in vivo led to the induction of cells with characteristics of LPCs and ameliorated hepatic dysfunction. We revealed that Thy1(+) mesenchymal cells produced FGF7 and appeared in close proximity to LPCs, implicating a role for those cells as the functional LPC niche in the regenerating liver. These findings provide new insights into the cellular and molecular basis for LPC regulation and identify FGF7 as a potential therapeutic target for liver diseases.


Subject(s)
Fibroblast Growth Factor 7/metabolism , Hepatocytes/cytology , Liver Regeneration/physiology , Signal Transduction , Stem Cells/cytology , Animals , Cell Proliferation , Fibroblast Growth Factor 7/genetics , Hepatocytes/metabolism , Liver Diseases/physiopathology , Mice , Stem Cells/metabolism , Thy-1 Antigens/metabolism , Up-Regulation
9.
Dev Comp Immunol ; 35(5): 611-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21237196

ABSTRACT

The molecular mechanisms underlying nodule formation and melanization, an important pathogen defense mechanism in insects, are poorly understood. In this study, we investigated the role of BmSPH-1, a catalytically inactive Bombyx mori serine protease homolog, in nodule melanization induced by injection of Escherichia coli cells into the B. mori larval hemocoel. Addition of the melanization substrate L-3,4-dihydroxyphenylalanine (DOPA) to newly formed nodules prompted nodule melanization, confirming that nodules contain activated prophenoloxidase needed for melanization. Immunoprecipitation and immunoblot studies demonstrated that BmSPH-1 interacts with BmLBP, a C-type lectin that binds Gram-negative bacteria, and that BmSPH-1 is present in a truncated, putatively activated form at the E. coli cell surface in nodules. Pretreatment of larvae with anti-BmSPH-1 serum inhibited nodule melanization in E. coli-injected larvae. These results suggest that BmSPH-1 regulates nodule melanization and is recruited into nodules from the hemolymph by BmLBP.


Subject(s)
Bombyx/immunology , Escherichia coli , Serine Proteases/analysis , Animals , Bombyx/enzymology , Bombyx/growth & development , Bombyx/microbiology , Hemolymph/immunology , Larva/enzymology , Larva/immunology , Larva/microbiology , Melanins/metabolism , Monophenol Monooxygenase/metabolism , Serine Proteases/metabolism
10.
Dev Comp Immunol ; 33(6): 789-800, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19201380

ABSTRACT

C-type lectins can act as pattern recognition receptors (PRRs) in innate immunity. Previously, we identified two C-type lectins from silkworm (Bombyx mori), BmLBP and BmMBP, as PRRs. In the present study, we identified three homologs of these lectins by searching the silkworm genome database. These novel B. mori low-expression lectins were designated BmLEL-1, BmLEL-2, and BmLEL-3. Although Western-blot analysis failed to detect BmLEL-1, -2, or -3 in plasma, affinity precipitation of larval plasma with various microorganisms revealed that BmLEL-1 and -2 bind to rough and smooth strains of Gram-negative bacteria, respectively. BmLEL-1, -2, and -3 were found to be expressed in testis and ovary, where BmLEL-2 expression was up-regulated after bacteria infection. These results indicate that the novel C-type lectins might play a role in the innate immunity in these tissues as PRRs. Here, we discuss the roles and members of the C-type lectins as primary PRRs in B. mori cellular immunity.


Subject(s)
Bombyx/immunology , Lectins, C-Type/immunology , Amino Acid Sequence , Animals , Bacteria/metabolism , Bombyx/microbiology , Female , Genes, Insect , Hemolymph/immunology , Immunity, Innate , Insect Proteins/chemistry , Insect Proteins/classification , Insect Proteins/genetics , Insect Proteins/metabolism , Larva , Lectins, C-Type/chemistry , Lectins, C-Type/classification , Lectins, C-Type/genetics , Male , Molecular Sequence Data , Ovary/metabolism , Phylogeny , Sequence Alignment , Testis/metabolism , Yeasts/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...