Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 3386, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099644

ABSTRACT

During early pregnancy in the mouse, nidatory estrogen (E2) stimulates endometrial receptivity by activating a network of signaling pathways that is not yet fully characterized. Here, we report that bone morphogenetic proteins (BMPs) control endometrial receptivity via a conserved activin receptor type 2 A (ACVR2A) and SMAD1/5 signaling pathway. Mice were generated to contain single or double conditional deletion of SMAD1/5 and ACVR2A/ACVR2B receptors using progesterone receptor (PR)-cre. Female mice with SMAD1/5 deletion display endometrial defects that result in the development of cystic endometrial glands, a hyperproliferative endometrial epithelium during the window of implantation, and impaired apicobasal transformation that prevents embryo implantation and leads to infertility. Analysis of Acvr2a-PRcre and Acvr2b-PRcre pregnant mice determined that BMP signaling occurs via ACVR2A and that ACVR2B is dispensable during embryo implantation. Therefore, BMPs signal through a conserved endometrial ACVR2A/SMAD1/5 pathway that promotes endometrial receptivity during embryo implantation.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Embryo Implantation , Infertility, Female/genetics , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Biopsy , Disease Models, Animal , Endometrium/metabolism , Endometrium/pathology , Estrogens/metabolism , Female , Humans , Mice , Mice, Knockout , Pregnancy , Signal Transduction/physiology , Smad1 Protein/analysis , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/analysis , Smad5 Protein/genetics , Smad5 Protein/metabolism
2.
Proc Natl Acad Sci U S A ; 116(9): 3873-3882, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30651315

ABSTRACT

SMAD2 and SMAD3 are downstream proteins in the transforming growth factor-ß (TGF ß) signaling pathway that translocate signals from the cell membrane to the nucleus, bind DNA, and control the expression of target genes. While SMAD2/3 have important roles in the ovary, we do not fully understand the roles of SMAD2/3 in the uterus and their implications in the reproductive system. To avoid deleterious effects of global deletion, and given previous data showing redundant function of Smad2 and Smad3, a double-conditional knockout was generated using progesterone receptor-cre (Smad2/3 cKO) mice. Smad2/3 cKO mice were infertile due to endometrial hyperproliferation observed as early as 6 weeks of postnatal life. Endometrial hyperplasia worsened with age, and all Smad2/3 cKO mice ultimately developed bulky endometrioid-type uterine cancers with 100% mortality by 8 months of age. The phenotype was hormone-dependent and could be prevented with removal of the ovaries at 6 weeks of age but not at 12 weeks. Uterine tumor epithelium was associated with decreased expression of steroid biosynthesis genes, increased expression of inflammatory response genes, and abnormal expression of cell cycle checkpoint genes. Our results indicate the crucial role of SMAD2/3 in maintaining normal endometrial function and confirm the hormone-dependent nature of SMAD2/3 in the uterus. The hyperproliferation of the endometrium affected both implantation and maintenance of pregnancy. Our findings generate a mouse model to study the roles of SMAD2/3 in the uterus and serve to provide insight into the mechanism by which the endometrium can escape the plethora of growth regulatory proteins.


Subject(s)
Infertility/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , Uterine Neoplasms/genetics , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Endometrium/metabolism , Endometrium/pathology , Female , Gene Expression Regulation/genetics , Humans , Infertility/pathology , Mice , Mice, Knockout , Pregnancy , Receptors, Progesterone/genetics , Uterine Neoplasms/pathology , Uterus/metabolism , Uterus/pathology
3.
Development ; 145(15)2018 08 13.
Article in English | MEDLINE | ID: mdl-29980567

ABSTRACT

Throughout the male reproductive lifespan, spermatogonial stem cells (SSCs) produce committed progenitors that proliferate and then remain physically connected in growing clones via short cylindrical intercellular bridges (ICBs). These ICBs, which enlarge in meiotic spermatocytes, have been demonstrated to provide a conduit for postmeiotic haploid spermatids to share sex chromosome-derived gene products. In addition to ICBs, spermatogonia exhibit multiple thin cytoplasmic projections. Here, we have explored the nature of these projections in mice and find that they are dynamic, span considerable distances from their cell body (≥25 µm), either terminate or physically connect multiple adjacent spermatogonia, and allow for sharing of macromolecules. Our results extend the current model that subsets of spermatogonia exist as isolated cells or clones, and support a model in which spermatogonia of similar developmental fates are functionally connected through a shared dynamic cytoplasm mediated by thin cytoplasmic projections.


Subject(s)
Cytoplasm/metabolism , Mammals/metabolism , Spermatogonia/metabolism , Animals , Cell Differentiation , Cytoplasm/ultrastructure , Diffusion , Green Fluorescent Proteins/metabolism , Intracellular Space/metabolism , Macromolecular Substances/metabolism , Male , Meiosis , Mice, Transgenic , Papio , Rats , Spermatocytes/cytology , Spermatocytes/metabolism , Spermatogonia/cytology , Spermatogonia/ultrastructure
4.
Cell ; 150(4): 673-84, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22901802

ABSTRACT

A pharmacologic approach to male contraception remains a longstanding challenge in medicine. Toward this objective, we explored the spermatogenic effects of a selective small-molecule inhibitor (JQ1) of the bromodomain and extraterminal (BET) subfamily of epigenetic reader proteins. Here, we report potent inhibition of the testis-specific member BRDT, which is essential for chromatin remodeling during spermatogenesis. Biochemical and crystallographic studies confirm that occupancy of the BRDT acetyl-lysine binding pocket by JQ1 prevents recognition of acetylated histone H4. Treatment of mice with JQ1 reduced seminiferous tubule area, testis size, and spermatozoa number and motility without affecting hormone levels. Although JQ1-treated males mate normally, inhibitory effects of JQ1 evident at the spermatocyte and round spermatid stages cause a complete and reversible contraceptive effect. These data establish a new contraceptive that can cross the blood:testis boundary and inhibit bromodomain activity during spermatogenesis, providing a lead compound targeting the male germ cell for contraception.


Subject(s)
Azepines/pharmacology , Contraceptive Agents, Male/pharmacology , Nuclear Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , Azepines/chemistry , Blood-Testis Barrier , Contraceptive Agents, Male/chemistry , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Models, Molecular , Nuclear Proteins/chemistry , Protein Structure, Tertiary , Sperm Count , Sperm Motility/drug effects , Spermatozoa/drug effects , Testis/cytology , Testis/drug effects , Triazoles/chemistry
5.
PLoS Genet ; 7(10): e1002320, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028666

ABSTRACT

The transforming growth factor ß (TGFß) superfamily proteins are principle regulators of numerous biological functions. Although recent studies have gained tremendous insights into this growth factor family in female reproduction, the functions of the receptors in vivo remain poorly defined. TGFß type 1 receptor (TGFBR1), also known as activin receptor-like kinase 5, is the major type 1 receptor for TGFß ligands. Tgfbr1 null mice die embryonically, precluding functional characterization of TGFBR1 postnatally. To study TGFBR1-mediated signaling in female reproduction, we generated a mouse model with conditional knockout (cKO) of Tgfbr1 in the female reproductive tract using anti-Müllerian hormone receptor type 2 promoter-driven Cre recombinase. We found that Tgfbr1 cKO females are sterile. However, unlike its role in growth differentiation factor 9 (GDF9) signaling in vitro, TGFBR1 seems to be dispensable for GDF9 signaling in vivo. Strikingly, we discovered that the Tgfbr1 cKO females develop oviductal diverticula, which impair embryo development and transit of embryos to the uterus. Molecular analysis further demonstrated the dysregulation of several cell differentiation and migration genes (e.g., Krt12, Ace2, and MyoR) that are potentially associated with female reproductive tract development. Moreover, defective smooth muscle development was also revealed in the uteri of the Tgfbr1 cKO mice. Thus, TGFBR1 is required for female reproductive tract integrity and function, and disruption of TGFBR1-mediated signaling leads to catastrophic structural and functional consequences in the oviduct and uterus.


Subject(s)
Embryonic Development/genetics , Growth Differentiation Factor 9/metabolism , Muscle, Smooth/growth & development , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reproductive Physiological Phenomena/genetics , Uterus/embryology , Animals , Cells, Cultured , Diverticulum/genetics , Diverticulum/pathology , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Female , Fertility/genetics , Growth Differentiation Factor 9/genetics , HEK293 Cells , Humans , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth/metabolism , Progesterone/blood , Receptor, Transforming Growth Factor-beta Type I , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Uterus/abnormalities
6.
PLoS Biol ; 8(8)2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20808952

ABSTRACT

During gametogenesis and pre-implantation development, the mammalian epigenome is reprogrammed to establish pluripotency in the epiblast. Here we show that the histone 3 lysine 4 (H3K4) methyltransferase, MLL2, controls most of the promoter-specific chromatin modification, H3K4me3, during oogenesis and early development. Using conditional knockout mutagenesis and a hypomorph model, we show that Mll2 deficiency in oocytes results in anovulation and oocyte death, with increased transcription of p53, apoptotic factors, and Iap elements. MLL2 is required for (1) bulk H3K4me3 but not H3K4me1, indicating that MLL2 controls most promoters but monomethylation is regulated by a different H3K4 methyltransferase; (2) the global transcriptional silencing that preceeds resumption of meiosis but not for the concomitant nuclear reorganization into the surrounded nucleolus (SN) chromatin configuration; (3) oocyte survival; and (4) normal zygotic genome activation. These results reveal that MLL2 is autonomously required in oocytes for fertility and imply that MLL2 contributes to the epigenetic reprogramming that takes place before fertilization. We propose that once this task has been accomplished, MLL2 is not required until gastrulation and that other methyltransferases are responsible for bulk H3K4me3, thereby revealing an unexpected epigenetic control switch amongst the H3K4 methyltransferases during development.


Subject(s)
Gene Expression Regulation, Developmental , Histones/metabolism , Lysine/metabolism , Methyltransferases/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oocytes/enzymology , Animals , Epigenomics , Female , Histone-Lysine N-Methyltransferase , Methylation , Methyltransferases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/metabolism , Oogenesis
7.
Reprod Biol Endocrinol ; 8: 69, 2010 Jun 21.
Article in English | MEDLINE | ID: mdl-20565978

ABSTRACT

BACKGROUND: Inhibin is a tumor-suppressor and activin antagonist. Inhibin-deficient mice develop gonadal tumors and a cachexia wasting syndrome due to enhanced activin signaling. Because activins signal through SMAD2 and SMAD3 in vitro and loss of SMAD3 attenuates ovarian tumor development in inhibin-deficient females, we sought to determine the role of SMAD2 in the development of ovarian tumors originating from the granulosa cell lineage. METHODS: Using an inhibin alpha null mouse model and a conditional knockout strategy, double conditional knockout mice of Smad2 and inhibin alpha were generated in the current study. The survival rate and development of gonadal tumors and the accompanying cachexia wasting syndrome were monitored. RESULTS: Nearly identical to the controls, the Smad2 and inhibin alpha double knockout mice succumbed to weight loss, aggressive tumor progression, and death. Furthermore, elevated activin levels and activin-induced pathologies in the liver and stomach characteristic of inhibin deficiency were also observed in these mice. Our results indicate that SMAD2 ablation does not protect inhibin-deficient females from the development of ovarian tumors or the cachexia wasting syndrome. CONCLUSIONS: SMAD2 is not required for mediating tumorigenic signals of activin in ovarian tumor development caused by loss of inhibin.


Subject(s)
Carcinoma/genetics , Inhibins/genetics , Ovarian Neoplasms/genetics , Smad2 Protein/physiology , Testicular Neoplasms/genetics , Activins/blood , Activins/metabolism , Animals , Carcinoma/metabolism , Carcinoma/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Smad2 Protein/genetics , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
8.
Mol Reprod Dev ; 76(5): 453-9, 2009 May.
Article in English | MEDLINE | ID: mdl-18951373

ABSTRACT

Tektins are evolutionarily conserved flagellar (and ciliary) filamentous proteins present in the axoneme and peri-axonemal structures in diverse metazoan species. We have previously shown that tektin 3 (TEKT3) and tektin 4 (TEKT4) are male germ cell-enriched proteins, and that TEKT4 is essential for coordinated and progressive sperm motility in mice. Here we report that male mice null for TEKT3 produce sperm with reduced motility (47.2% motility) and forward progression, and increased flagellar structural bending defects. Male TEKT3-null mice however maintain normal fertility in two different genetic backgrounds tested, in contrast to TEKT4-null mice. Furthermore, male mice null for both TEKT3 and TEKT4 show subfertility on a mixed B6;129 genetic background, significantly different from either single knockouts, suggesting partial nonredundant roles for these two proteins in sperm physiology. Our results suggest that tektins are potential candidate genes for nonsyndromic asthenozoospermia in humans.


Subject(s)
Fertility , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , Sperm Motility , Spermatozoa/physiology , Analysis of Variance , Animals , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Fertility/genetics , Flagella/physiology , Male , Mice , Mice, Knockout , Spermatozoa/pathology
9.
Biol Reprod ; 80(3): 449-57, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19020301

ABSTRACT

A conserved feature of germ cell cytokinesis is the formation of stable intercellular bridges between daughter cells. These intercellular bridges are seen in diverse species from Drosophila melanogaster to Homo sapiens and have been shown to have roles in communication of large numbers of germ cells. In testis expressed gene 14 (Tex14) knockout mice, intercellular bridges do not form during spermatogenesis, and male mice are sterile, demonstrating an essential role for intercellular bridges in postnatal spermatogenesis in mammals. Intercellular bridges also form between dividing germ cells in both male and female embryos. However, little is known about the formation or role of the embryonic intercellular bridges in mammals. In females, embryonic intercellular bridges have been proposed to have a role in development of the presumptive oocyte. Herein, we show that TEX14 is an essential component of male and female embryonic intercellular bridges. In addition, we demonstrate that mitotic kinesin-like protein 1 (MKLP1, official symbol KIF23), which we have discovered is a component of intercellular bridges during spermatogenesis, is also a component of male and female embryonic intercellular bridges. Germ cell intercellular bridges are readily identified by KIF23 immunofluorescence between the gonocytes and oogonia of control mice but are absent between germ cells of Tex14-null mice. Furthermore, by electron microscopy, intercellular bridges are present in all control newborn ovaries but are absent in the Tex14 knockout ovaries. Despite the absence of embryonic intercellular bridges in the Tex14-null mice, male mice initiate spermatogenesis, and female mice are fertile. Although fewer oocytes were present in Tex14-null neonatal ovaries, folliculogenesis was still active at 1 yr of age. Thus, while TEX14 and intercellular bridges have an essential role in postnatal spermatogenesis, they are not required in the embryo.


Subject(s)
Cell Communication/physiology , Fertility/physiology , Germ Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , Cytokinesis/physiology , Female , Germ Cells/cytology , Kinesins/metabolism , Male , Mice , Mice, Knockout , Oocytes/cytology , Oogenesis/physiology , Ovary/cytology , Ovary/embryology , Ovary/metabolism , Protein Serine-Threonine Kinases/genetics , Spermatogenesis/physiology , Testis/cytology , Testis/embryology , Testis/metabolism , Transcription Factors/genetics
10.
Mol Endocrinol ; 22(10): 2336-52, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18687735

ABSTRACT

Dicer is an evolutionarily conserved ribonuclease III that is necessary for microRNA (miRNA) processing and the synthesis of small interfering RNAs from long double-stranded RNA. Although it has been shown that Dicer plays important roles in the mammalian germline and early embryogenesis, the functions of Dicer-dependent pathways in the somatic cells of the female reproductive tract are unknown. Using a transgenic line in which Cre recombinase is driven by the anti-Müllerian hormone receptor type 2 promoter, we conditionally inactivated Dicer1 in the mesenchyme of the developing Müllerian ducts and postnatally in ovarian granulosa cells and mesenchyme-derived cells of the oviducts and uterus. Deletion of Dicer in these cell types results in female sterility and multiple reproductive defects including decreased ovulation rates, compromised oocyte and embryo integrity, prominent bilateral paratubal (oviductal) cysts, and shorter uterine horns. The paratubal cysts act as a reservoir for spermatozoa and oocytes and prevent embryos from transiting the oviductal isthmus and passing the uterotubal junction to enter the uterus for implantation. Deep sequencing of small RNAs in oviduct revealed down-regulation of specific miRNAs in Dicer conditional knockout females compared with wild type. The majority of these differentially expressed miRNAs are predicted to regulate genes important for Müllerian duct differentiation and mesenchyme-derived structures, and several of these putative target genes were significantly up-regulated upon conditional deletion of Dicer1. Thus, our findings reveal diverse and critical roles for Dicer and its miRNA products in the development and function of the female reproductive tract.


Subject(s)
DEAD-box RNA Helicases/metabolism , Endoribonucleases/metabolism , Genitalia, Female/physiology , Infertility/genetics , Animals , DEAD-box RNA Helicases/genetics , Embryo, Mammalian/abnormalities , Embryo, Mammalian/anatomy & histology , Endoribonucleases/genetics , Estradiol/blood , Estrous Cycle/physiology , Female , Follicle Stimulating Hormone/blood , Genitalia, Female/anatomy & histology , Male , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Oocytes/pathology , Oviducts/anatomy & histology , Oviducts/pathology , Ovulation/physiology , Pregnancy , Promoter Regions, Genetic , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Ribonuclease III , Spermatozoa/cytology
11.
Mol Cell Endocrinol ; 294(1-2): 19-28, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18657590

ABSTRACT

The inhibins are secreted alpha:beta heterodimers of the TGF-beta superfamily that are mainly synthesized in Sertoli cells and granulosa cells, and are critical regulators of testicular and ovarian development and function. Mice homozygous for a targeted deletion of the inhibin alpha subunit gene (Inha(-/-)) develop sex cord-stromal tumors in a gonadotropin-dependent manner. Here, we determine the contribution of LH to gonadal tumorigenesis by generating mice deficient in both inhibins and LH. Inha(-/-)Lhb(-/-) mice have increased survival and delayed tumor progression, and these observations correlate with lower serum FSH and estradiol levels compared to Inha(-/-) controls. Double mutant testicular tumors demonstrate decreased expression of cyclin D2, while double mutant ovarian tumors have elevated expression of p15(INK4b) and trend toward higher levels of p27(Kip1). We conclude that LH is not required for tumor formation in the absence of inhibins but promotes tumor progression, likely through alterations in serum hormone levels and cell cycle regulators.


Subject(s)
Inhibins/deficiency , Luteinizing Hormone/metabolism , Ovarian Neoplasms/pathology , Testicular Neoplasms/pathology , Animals , Cachexia/pathology , Disease Progression , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Gene Expression Regulation, Neoplastic , Inhibins/metabolism , Longevity , Luteinizing Hormone/deficiency , Male , Mice , Mutation/genetics , Organ Specificity , Ovarian Neoplasms/genetics , Survival Analysis , Syndrome , Testicular Neoplasms/genetics , Testosterone/blood
12.
Mol Endocrinol ; 21(10): 2458-71, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17609433

ABSTRACT

Activins have diverse roles in multiple physiological processes including reproduction. Mutations and loss of heterozygosity at the human activin receptor ACVR1B and ACVR2 loci are observed in pituitary, pancreatic, and colorectal cancers. Functional studies support intraovarian roles for activins, although clarifying the in vivo roles has remained elusive due to the perinatal death of activin betaA knockout mice. To study the roles of activins in ovarian growth, differentiation, and cancer, a tissue-specific knockout system was designed to ablate ovarian production of activins. Mice lacking ovarian activin betaA were intercrossed to Inhbb homozygous null mice to produce double activin knockouts. Whereas ovarian betaA knockout females are subfertile, betaB/betaA double mutant females are infertile. Strikingly, the activin betaA and betaB/betaA-deficient ovaries contain increased numbers of functional corpora lutea but do not develop ovarian tumors. Microarray analysis of isolated granulosa cells identifies significant changes in expression for a number of genes with known reproductive roles, including Kitl, Taf4b, and Ghr, as well as loss of expression of the proto-oncogene, Myc. Thus, in contrast to the known tumor suppressor role of activins in some tissues, our data indicate that activin betaA and betaB function redundantly in a growth stimulatory pathway in the mammalian ovary.


Subject(s)
Fertility/genetics , Inhibin-beta Subunits/physiology , Ovary/metabolism , Animals , Carrier Proteins/genetics , Corpus Luteum/metabolism , Female , Gene Expression , Granulosa Cells/metabolism , Inhibin-beta Subunits/genetics , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Ovary/cytology , Proto-Oncogene Mas , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics
13.
FASEB J ; 21(4): 1013-25, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17244819

ABSTRACT

Sperm flagellar motion is the outcome of a dynamic interplay between the axonemal cytoskeleton, the peri-axonemal accessory structures, and multiple regulatory networks that coordinate to produce flagellar beat and waveform. Tektins are conserved components of the flagellar proteome in evolutionarily diverse species and are believed to play essential roles in the mechanics of sperm motility. Using database mining, we identified multiple new paralogs of previously annotated tektins, including tektin 4 (TEKT4), which shares 77.1% identity with its nearest human homologue. Mouse Tekt4 is a germ cell-enriched gene, most abundantly expressed in haploid round spermatids in the testis, and the protein is localized to the sperm flagella. Male mice lacking TEKT4 on a 129S5/SvEvBrd inbred background are subfertile. Tekt4-null sperm exhibit drastically reduced forward progressive velocity and uncoordinated waveform propagation along the flagellum. In Tekt4-null sperm, flagellar ultrastructure is grossly unaltered as revealed by transmission electron microscopy. However, the ineffective flagellar strokes lead to approximately 10-fold higher consumption of intracellular ATP in Tekt4-null sperm as compared to wild-type, and null spermatozoa rapidly lose progressive motility when incubated for > or = 1.5 h. Our studies demonstrate that TEKT4 is necessary for the proper coordinated beating of the sperm flagellum and male reproductive physiology.


Subject(s)
Asthenozoospermia/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Gene Expression Regulation , Microtubule Proteins/genetics , Microtubule Proteins/physiology , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Asthenozoospermia/metabolism , Cytoskeleton/metabolism , Flagella/metabolism , Humans , Male , Mice , Mice, Knockout , Molecular Sequence Data , Sequence Homology, Amino Acid , Spermatozoa/metabolism
14.
Proc Natl Acad Sci U S A ; 103(13): 4982-7, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-16549803

ABSTRACT

Cytokinesis in somatic cells concludes with the formation of a midbody, which is abscised to form individual daughter cells. In contrast, germ cell cytokinesis results in a permanent intercellular bridge connecting the daughter cells through a large cytoplasmic channel. During spermatogenesis, proposed roles for the intercellular bridge include germ cell communication, synchronization, and chromosome dosage compensation in haploid cells. Although several essential components of the midbody have recently been identified, essential components of the vertebrate germ cell intercellular bridge have until now not been described. Herein, we show that testis-expressed gene 14 (TEX14) is a novel protein that localizes to germ cell intercellular bridges. In the absence of TEX14, intercellular bridges are not observed by using electron microscopy and other markers. Spermatogenesis in Tex14(-/-) mice progresses through the transit amplification of diploid spermatogonia and the expression of early meiotic markers but halts before the completion of the first meiotic division. Thus, TEX14 is required for intercellular bridges in vertebrate germ cells, and these studies provide evidence that the intercellular bridge is essential for spermatogenesis and fertility.


Subject(s)
Fertility/physiology , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Alleles , Animals , Cell Communication , Immunohistochemistry , Male , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Spermatogenesis , Testis/cytology , Testis/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
15.
Proc Natl Acad Sci U S A ; 102(16): 5755-60, 2005 Apr 19.
Article in English | MEDLINE | ID: mdl-15824319

ABSTRACT

MSY2, a germ-cell-specific member of the Y-box family of DNA-/RNA-binding proteins, is proposed to function as a coactivator of transcription in the nucleus and to stabilize and store maternal and paternal mRNAs in the cytoplasm. In mice lacking Msy2, a normal Mendelian ratio is observed after matings between heterozygotes with equal numbers of phenotypically normal but sterile male and female homozygotes (Msy2-/-). Spermatogenesis is disrupted in postmeiotic null germ cells with many misshapen and multinucleated spermatids, and no spermatozoa are detected in the epididymis. Apoptosis is increased in the testes of homozygotes, and real-time RT-PCR assays reveal large reductions in the mRNA levels of postmeiotic male germ cell mRNAs and smaller reductions of meiotic germ cell transcripts. In females, there is no apparent decrease in either the number of follicles or their morphology in ovaries obtained from 2- and 8-day-old Msy2-/- mice. In contrast, follicle number and progression are reduced in 21-day-old Msy2-/- ovaries. In adult Msy2-/- females, oocyte loss increases, anovulation is observed, and multiple oocyte and follicle defects are seen. Thus, Msy2 represents one of a small number of germ-cell-specific genes whose deletion leads to the disruption of both spermatogenesis and oogenesis.


Subject(s)
DNA-Binding Proteins/metabolism , Infertility , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Female , Gene Targeting , In Situ Nick-End Labeling , Male , Mice , Mice, Knockout , Oocytes/cytology , Oocytes/physiology , Oogenesis/physiology , Ovary/abnormalities , Ovary/cytology , Ovary/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Spermatogenesis/physiology , Spermatozoa/abnormalities , Spermatozoa/cytology , Spermatozoa/physiology , Testis/cytology , Testis/physiology , Transcription Factors/genetics
16.
Mol Cell Endocrinol ; 212(1-2): 19-27, 2003 Dec 30.
Article in English | MEDLINE | ID: mdl-14654247

ABSTRACT

To examine in vivo, the local effects of inhibins and activins within the anterior pituitary, independent of their endocrine effects exerted from the gonad, in mediating FSH homeostasis, we used castrated knockout mice lacking either inhibin alpha or activin receptor II (ACVR2) alone or in combination. Compared to castrated wild-type (WT) mice, FSHbeta mRNA levels in the pituitaries of Acvr2 null mice were significantly downregulated in the absence of gonadal feedback. FSHbeta mRNA levels were not significantly higher in the pituitaries of castrated inhibin alpha null mice compared to those in Acvr2 null mice and remained the same in the pituitaries of castrated double mutant mice lacking both inhibin and ACVR2. In contrast to FSHbeta mRNA expression changes, pituitary FSH content was significantly reduced in Acvr2 null mice whereas it was only slightly upregulated in inhibin alpha null mice. Combined absence of both ACVR2 signaling and inhibins caused a decrease in FSH content compared to that in the absence of inhibins alone. These changes in pituitary content were in parallel to those in serum FSH levels in these three groups of castrated mice, suggesting that the unopposed actions of locally produced inhibins are dominant over those effects mediated by ACVR2 signaling to regulate FSH biosynthesis and secretion. Thus, our in vivo results demonstrate that within the pituitary, locally produced activins and inhibins exert their actions at distinct phases of FSH homeostasis. In an independent set of experiments, we tested whether in vivo signaling via ACVR2 is necessary for hypothalamic GnRH biosynthesis and for GnRH receptor expression. Our results demonstrate that in contrast to previous in vitro studies, signaling through ACVR2 is neither required for hypothalamic synthesis of GnRH peptide nor for expression of GnRH receptors in the anterior pituitary. We conclude that within the hypothalamic-pituitary short loop, ACVR2 signaling is critical only for FSH homeostasis and not for GnRH biosynthesis or induction of pituitary GnRH receptor expression. Our studies confirm the importance of using in vivo genetic models for studying regulation of the hypothalamic-pituitary-gonadal axis.


Subject(s)
Activin Receptors, Type II/metabolism , Follicle Stimulating Hormone, beta Subunit/metabolism , Gene Expression Regulation , Receptors, LHRH/metabolism , Activin Receptors, Type II/genetics , Activins/metabolism , Animals , Castration , Follicle Stimulating Hormone, beta Subunit/genetics , Gonadotropin-Releasing Hormone/metabolism , Homeostasis , Hypothalamus/metabolism , Inhibins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pituitary Gland/cytology , Pituitary Gland/metabolism , Receptors, LHRH/genetics , Signal Transduction/physiology
17.
Mol Endocrinol ; 17(10): 2053-69, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12855747

ABSTRACT

Inhibins are heterodimeric (alpha:betaA and alpha:betaB) endocrine, paracrine, and autocrine factors of the TGFbeta superfamily that are produced predominantly by ovarian granulosa cells in females and testicular Sertoli cells in males. Control of granulosa and Sertoli cell proliferation is lost in the inhibin alpha (Inhalpha) knockout mouse model, leading to gonadotropin-dependent gonadal tumors of the granulosa/Sertoli cell lineage in both females and males. Castrate Inhalpha knockout mice develop sex steroidogenic tumors of the adrenal cortex. Physiological control of granulosa/Sertoli cell cycle progression depends on p27Kip1 and cyclin D2, which function in the G1-->S phase transition. To study the cell cycle-regulatory factors involved in ovarian, testicular, and adrenal tumor development in vivo, we have bred Inhalpha mutant mice to mice with targeted disruptions of the p27 and cyclin D2 genes. Our previous studies demonstrated that inhibins act cooperatively with p27 to negatively regulate granulosa cell proliferation, as double mutant mice lacking inhibins and p27 develop and succumb to ovarian tumors more rapidly than Inhalpha knockout mice. Here, we report that cyclin D2 antagonizes this inhibition and is key in promoting gonadal growth and tumor development, and tumor development is markedly suppressed in double-mutant mice. We found that double-knockout females lacking cyclin D2 and Inhalpha lived longer than mice lacking inhibins alone; the majority of these double-knockout mice lived longer than 17 wk, as opposed to inhibin alpha single-knockout females with 50% survival at between 12 and 13 wk of age. Moreover, 95% of inhibin alpha knockout males succumb to testicular tumor development by 12 wk of age, whereas double knockouts were protected from early signs of tumor development and had a 50% survival of 40 wk. Interestingly, the results of these studies reflect tissue-specific consequences of loss of these cell cycle regulators. In castrate mice, loss of p27 has little effect on adrenal cortical tumor progression in the absence of inhibins, whereas loss of cyclin D2 prolongs the lifespan of cyclin D2, Inhalpha double knockouts. After gonadectomy, 50% of cyclin D2, Inhalpha double-knockout males live to more than 46 wk of age, 10 wk longer than 50% of littermates lacking only inhibins. Similarly, 50% of female cyclin D2, inhibin alpha double knockouts live to 47 wk of age before succumbing to adrenal tumor development, in contrast to the 50% survival of Inhalpha single-knockout females at between 27 and 28 wk. Thus, identification of genetic modifiers of the Inhalpha knockout tumor phenotype has led us to a better appreciation of how specific components of the cell cycle machinery contribute to tumorigenesis in the ovary, testis, and adrenal gland.


Subject(s)
Adrenal Cortex Neoplasms/physiopathology , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Ovarian Neoplasms/physiopathology , Testicular Neoplasms/physiopathology , Tumor Suppressor Proteins/metabolism , Adrenal Cortex Neoplasms/pathology , Animals , Cachexia/physiopathology , Castration , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Division , Cell Transformation, Neoplastic , Cyclin D2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/genetics , Cyclins/biosynthesis , Cyclins/genetics , Disease Progression , Female , Follicle Stimulating Hormone/antagonists & inhibitors , Follicle Stimulating Hormone/blood , Granulosa Cells/metabolism , Granulosa Cells/pathology , Inhibins/genetics , Inhibins/metabolism , Male , Mice , Mice, Knockout , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , Sertoli Cell Tumor/pathology , Sertoli Cell Tumor/physiopathology , Testicular Neoplasms/pathology , Testis/metabolism , Testis/pathology , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
18.
Mol Endocrinol ; 17(10): 2039-52, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12855748

ABSTRACT

Sex steroids control cellular phenotypes by binding to receptor proteins that in turn regulate downstream gene expression. They are important tropic factors in hormonally responsive tissues and have been implicated in the pathogenesis of both benign proliferations and malignancies at some of these sites. Knockout mice lacking inhibins, alpha:beta heterodimeric peptide hormones of the TGFbeta superfamily, develop gonadal tumors that produce sex steroids and depend on pituitary gonadotropin hormones. To better appreciate how sex steroid receptor signaling pathways contribute to the loss of granulosa/Sertoli cell proliferation in the ovary and testis of inhibin alpha (Inhalpha) knockout mice, we are using both pharmacologic and genetic approaches. Roles of androgens in testicular tumor development have been investigated in our previous studies using double-mutant mice lacking inhibins and carrying the null testicular feminization (tfm) mutation of the androgen receptor. Herein, we report that androgens also participate in the development of ovarian tumors, as tumor development is forestalled in mice treated with flutamide, a nonsteroidal inhibitor of androgen actions. Additionally, we generated double-mutant mice lacking estrogen receptor alpha (ERalpha) and Inhalpha or ERbeta and Inhalpha, as well as triple-mutant mice lacking ERalpha, ERbeta, and Inhalpha to determine the effects of individual and combined ER signaling pathways on tumor development. Although estrogens may have proliferative effects during follicle development and are important in specifying the granulosa cell phenotype, ERalpha and ERbeta signaling are not essential for timely granulosa cell tumor development or granulosa cell-like morphological features in ovarian tumors. However, redundant ER signaling through ERalpha and ERbeta in males is critical for testicular tumor formation, as triple-knockout, but not double-knockout, males are protected from early Sertoli cell tumorigenesis and death. Together, these studies indicate important and sexually dimorphic functions of estrogens and androgens in tumor development in this mouse model and indicate, for the first time, overlapping functions of ERalpha and ERbeta in Sertoli cell pathophysiology.


Subject(s)
Androgens/physiology , Ovarian Neoplasms/metabolism , Receptors, Estrogen/physiology , Testicular Neoplasms/metabolism , Animals , Body Weight/drug effects , Cachexia/physiopathology , Cyclin D2 , Cyclins/genetics , Cyclins/metabolism , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Flutamide/pharmacology , Genotype , Granulosa Cell Tumor/metabolism , Granulosa Cell Tumor/pathology , Inhibins/genetics , Inhibins/physiology , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Ovarian Neoplasms/pathology , Ovary/cytology , Ovary/drug effects , Ovary/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Estrogen/genetics , Sertoli Cell Tumor/metabolism , Sertoli Cell Tumor/pathology , Sex Characteristics , Signal Transduction , Testicular Neoplasms/pathology , Testis/cytology , Testis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...