Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters










Publication year range
1.
Andrology ; 1(1): 160-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23258646

ABSTRACT

The testis-specific serine/threonine protein kinases TSSK1 and TSSK2 are known to be essential for male fertility, in mice. The enzymes are present in elongating spermatids, and targeted deletion of the two genes Tssk1 and Tssk2 results in dysregulation of spermiogenesis. The mouse genes are genetically closely linked, forming a Tssk1-Tssk2 tandem. In human, TSSK1 is present in the form of a pseudogene, TSSK1A, which is linked to an intact TSSK2 gene, and in the form of an intact gene, TSSK1B, which is not genetically linked to TSSK2. Studies on conservation of genes and gene function between mouse and human are relevant, to be able to use mouse models for studies on human infertility, and to evaluate possible targets for non-hormonal contraception targeting the male. Therefore, we have performed a detailed analysis of the evolution of genes encoding TSSK1 and TSSK2 among mammals, in particular among primates. This study includes functional analysis of replacement mutation K27R in TSSK2, which is frequently observed among humans. In primates, the kinase domains of TSSK1B and TSSK2 have evolved under negative selection, reflecting the importance to maintain their kinase activity. Positive selection was observed for the C-terminal domain of TSSK1B, which indicates that TSSK1B and TSSK2 may perform at least partly differential functions.


Subject(s)
Evolution, Molecular , Protein Serine-Threonine Kinases/genetics , Pseudogenes , Testis/enzymology , Animals , Conserved Sequence , DNA Mutational Analysis , Databases, Genetic , Genotype , Humans , Male , Mutation , Phenotype , Phosphorylation , Phylogeny , Primates , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Selection, Genetic , Sequence Analysis, Protein , Spermatogenesis/genetics
2.
Nucleic Acids Res ; 32(Database issue): D560-7, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14681481

ABSTRACT

GermOnline provides information and microarray expression data for genes involved in mitosis and meiosis, gamete formation and germ line development across species. The database has been developed, and is being curated and updated, by life scientists in cooperation with bioinformaticists. Information is contributed through an online form using free text, images and the controlled vocabulary developed by the GeneOntology Consortium. Authors provide up to three references in support of their contribution. The database is governed by an international board of scientists to ensure a standardized data format and the highest quality of GermOnline's information content. Release 2.0 provides exclusive access to microarray expression data from Saccharomyces cerevisiae and Rattus norvegicus, as well as curated information on approximately 700 genes from various organisms. The locus report pages include links to external databases that contain relevant annotation, microarray expression and proteome data. Conversely, the Saccharomyces Genome Database (SGD), S.cerevisiae GeneDB and Swiss-Prot link to the budding yeast section of GermOnline from their respective locus pages. GermOnline, a fully operational prototype subject-oriented knowledgebase designed for community annotation and array data visualization, is accessible at http://www.germonline.org. The target audience includes researchers who work on mitotic cell division, meiosis, gametogenesis, germ line development, human reproductive health and comparative genomics.


Subject(s)
Cell Differentiation/genetics , Databases, Genetic , Gene Expression Profiling , Germ Cells/cytology , Germ Cells/metabolism , Animals , Computational Biology , Genomics , Humans , Information Storage and Retrieval , Internet , Meiosis/genetics , Mitosis/genetics , Oligonucleotide Array Sequence Analysis , Proteins/metabolism , Proteome , Proteomics , Rats
4.
J Clin Endocrinol Metab ; 88(5): 2327-34, 2003 May.
Article in English | MEDLINE | ID: mdl-12727992

ABSTRACT

Tibolone, a synthetic steroid acting in a tissue-specific manner and used in hormone replacement therapy, is converted into three active metabolites: a Delta(4) isomer (exerting progestogenic and androgenic effects) and two hydroxy metabolites, 3 alpha-hydroxytibolone (3 alpha-OH-tibolone) and 3beta-OH-tibolone (exerting estrogenic effects). In the present study an endometrial carcinoma cell line (Ishikawa PRAB-36) was used to investigate the progestogenic properties of tibolone and its metabolites. This cell line contains progesterone receptors A and B, but lacks estrogen and androgen receptors. When tibolone was added to the cells, complete conversion into the progestogenic/androgenic Delta(4) isomer was observed within 6 d. Furthermore, when cells were cultured with tibolone or when the Delta(4) isomer or the established progestagen medroxyprogesterone acetate was added to the medium, marked inhibition of growth was observed. Interestingly, 3 beta-OH-tibolone also induces some inhibition of growth. These growth inhibitions were not observed in progesterone receptor-negative parental Ishikawa cells, and progestagen-induced growth inhibition of PRAB-36 cells could readily be reversed using the antiprogestagen Org-31489. Upon measuring the expression of two progesterone-regulated genes (fibronectin and IGF-binding protein-3), tibolone, the Delta(4) isomer and medroxyprogesterone acetate showed similar gene expression regulation. These results indicate that tibolone, the Delta(4) metabolite, and to some extent 3 beta-OH-tibolone exert progestogenic effects. Tibolone and most likely 3 beta-OH-tibolone are converted into the Delta(4) metabolite.


Subject(s)
Endometrial Neoplasms/metabolism , Norpregnenes/pharmacology , Progestins/metabolism , Cell Division/drug effects , Endometrial Neoplasms/chemistry , Endometrial Neoplasms/pathology , Female , Fibronectins/genetics , Gene Expression Regulation/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Medroxyprogesterone Acetate/pharmacology , Norpregnenes/metabolism , Progesterone/pharmacology , Progestins/antagonists & inhibitors , Receptors, Progesterone/analysis , Tumor Cells, Cultured
5.
Cytogenet Genome Res ; 103(3-4): 225-34, 2003.
Article in English | MEDLINE | ID: mdl-15051943

ABSTRACT

During the male meiotic prophase in mouse and man, pairing and recombination of homologous chromosomes is accompanied by changes in chromatin structure. In this review, the dynamics of assembly and disassembly of the chromatin-associated complexes that mediate sister chromatid cohesion (cohesin) and maintain chromosome pairing (the synaptonemal complex) are described. Special features of the meiotic S phase are discussed, and also the dynamics of several key players that act together after the S phase at sites of meiotic double-strand break DNA repair. Current knowledge on histone modifications that occur during the male meiotic prophase is discussed, with special attention for the inactive chromatin of the X and Y chromosomes that constitutes the sex body. Finally, it is discussed that in the future, it will be possible to view the true chromatin dynamics during male meiosis in time, in living cells, through analysis of fluorescent-tagged proteins expressed in transgenic mice, using advanced fluorescent microscopy techniques.


Subject(s)
Chromatin/ultrastructure , Spermatozoa/ultrastructure , Animals , Chromatin/metabolism , DNA Repair , DNA Replication , Histones/metabolism , Male , Meiosis , Mice , Prophase , Recombination, Genetic , Sex Chromosomes , Spermatozoa/metabolism , Synaptonemal Complex
7.
Endocrinology ; 142(11): 4891-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11606457

ABSTRACT

Although ovarian follicle growth is under the influence of many growth factors and hormones of which FSH remains one of the most prominent regulators. Therefore, factors affecting the sensitivity of ovarian follicles to FSH are also important for follicle growth. The aim of the present study was to investigate whether anti-Müllerian hormone (AMH) has an inhibitory effect on follicle growth by decreasing the sensitivity of ovarian follicles to FSH. Furthermore, the combined action of AMH and FSH on ovarian follicle development was examined. Three different experiments were performed. Using an in vitro follicle culture system it was shown that FSH-stimulated preantral follicle growth is attenuated in the presence of AMH. This observation was confirmed by an in vivo experiment showing that in immature AMH-deficient females, more follicles start to grow under the influence of exogenous FSH than in their wild-type littermates. In a third experiment, examination of the follicle population of 4-month-old wild-type, FSH beta-, AMH-, and AMH-/FSH beta-deficient females revealed that loss of FSH expression has no impact on the number of primordial and preantral follicles, but the loss of inhibitory action of AMH on the recruitment of primordial follicles in AMH-deficient mice is increased in the absence of FSH. In conclusion, these studies show that AMH inhibits FSH-stimulated follicle growth in the mouse, suggesting that AMH is one of the factors determining the sensitivity of ovarian follicles for FSH and that AMH is a dominant regulator of early follicle growth.


Subject(s)
Follicle Stimulating Hormone/antagonists & inhibitors , Follicle Stimulating Hormone/pharmacology , Glycoproteins , Growth Inhibitors/physiology , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Testicular Hormones/physiology , Animals , Anti-Mullerian Hormone , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/genetics , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Growth Inhibitors/genetics , In Vitro Techniques , Inhibins/blood , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Organ Size/drug effects , Ovarian Follicle/anatomy & histology , Ovary/anatomy & histology , Testicular Hormones/genetics , Uterus/anatomy & histology
8.
Reproduction ; 121(1): 31-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11226027

ABSTRACT

In mammals, there is a complex and intriguing relationship between DNA repair and gametogenesis. DNA repair mechanisms are involved not only in the repair of different types of DNA damage in developing germline cells, but also take part in the meiotic recombination process. Furthermore, the DNA repair mechanisms should tolerate mutations occurring during gametogenesis, to a limited extent. In the present review, several gametogenic aspects of DNA mismatch repair, homologous recombination repair and postreplication repair are discussed. In addition, the role of DNA damage-induced cell cycle checkpoint control is considered briefly. It appears that many genes encoding proteins that take part in DNA repair mechanisms show enhanced or specialized expression during mammalian gametogenesis, and several gene knockout mouse models show male or female infertility. On the basis of such knowledge and models, future experiments may provide more information about the precise relationship between DNA repair, chromatin dynamics, and genomic stability versus instability during gametogenesis.


Subject(s)
DNA Repair , Gametogenesis , Animals , Cell Cycle/genetics , DNA Damage , Female , Humans , Male , Mice , Mice, Knockout , Spermatogenesis/genetics
9.
Endocrinology ; 141(12): 4720-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108287

ABSTRACT

The gubernaculum connects the gonad to the inguinoscrotal region and is involved in testis descent. It rapidly develops in the male fetus, whereas development in the female fetus is lacking. Possible factors involved in gubernaculum development are androgens, anti-Müllerian hormone (AMH), and insulin-like factor (Insl3). Sexual dimorphism in gubernaculum development correlated with the mitotic activity of cells in the gubernacular bulbs from male and female fetuses. Androgen receptor expression was restricted to the mesenchymal core of the gubernacular bulb, whereas skeletal muscle was detected in its outer layer. In an organ culture system devised to further study gubernaculum development in vitro, morphology of gubernacular explants grown in the presence of testes was comparable with that of gubernacula developed in vivo. Testicular tissue or medium containing R1881, a synthetic androgen, had a growth stimulatory effect on gubernacular explants compared with ovarian tissue or basal medium only. Moreover, Amh-/-, Amh+/-, and Insl3+/- testes stimulated the growth of gubernacular explants to the same extent as control testes. Insl3-/- testes, however, did not produce such an activity. This study reveals an essential role for both androgen and Insl3 in the gubernaculum outgrowth during transabdominal testis descent.


Subject(s)
Androgens/physiology , Genitalia, Male/embryology , Glycoproteins , Growth Inhibitors/physiology , Proteins/physiology , Testicular Hormones/physiology , Testis/embryology , Animals , Anti-Mullerian Hormone , Cell Division , Female , Genitalia, Male/chemistry , Genitalia, Male/cytology , Growth Inhibitors/deficiency , Immunohistochemistry , Insulin , Male , Metribolone/pharmacology , Mice , Mice, Mutant Strains , Mitosis , Models, Biological , Organ Culture Techniques , Ovary/physiology , Proteins/genetics , Rats , Rats, Wistar , Receptors, Androgen/analysis , Sex Characteristics , Testicular Hormones/deficiency , Testis/physiology
10.
Article in English | MEDLINE | ID: mdl-11097779

ABSTRACT

Mammalian spermatogenesis shows a strict control of many specific molecular and cellular events. This control involves Sertoli cell-germ cell interaction, as well as a programmed performance of changes in chromatin structure and gene expression in the developing germ cells. In recent years, much knowledge about the functions of defined genes in spermatogenesis has been gained by making use of mouse transgenic and gene knockout models. Several of these models are discussed in this brief overview, with an emphasis on genes encoding proteins involved in the control of gene transcription, mRNA translation, DNA repair and protein ubiquitination. A better understanding of the molecular and cellular biology of spermatogenesis in the mouse may provide concepts that can improve our understanding of human male infertility and may also lead to the identification of novel targets for contraceptive intervention.


Subject(s)
Sertoli Cells/physiology , Spermatogenesis/physiology , Spermatozoa/physiology , Animals , Chromatin/physiology , Gene Expression/physiology , Gene Expression Regulation , Humans , Male , Meiosis/physiology , Spermatogenesis/genetics
11.
Hum Reprod ; 15(12): 2504-11, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11098018

ABSTRACT

Atresia, a degenerative process through which many follicles are removed from the growing pool, involves apoptotic changes in the follicular granulosa cells. To identify histochemical markers of early stages of atresia, an in-vivo rat model was used which allowed the study of atresia of pre-ovulatory follicles in a synchronized and chronological order. By blocking the pre-ovulatory luteinizing hormone surge with a gonadotrophin-releasing hormone (GnRH) antagonist, ovulation of the pre-ovulatory follicles is prevented, after which these follicles became atretic. The first morphological sign of atresia (pyknotic granulosa cell nuclei) was found 27 h after injection of GnRH antagonist. Since the pre-ovulatory follicles gradually become atretic in a synchronous fashion, this model provided an opportunity to study and define markers of future atresia in pre-ovulatory follicles. Atresia involves apoptosis of granulosa cells, and therefore internucleosomal DNA fragmentation was examined. Using the terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labelling (TUNEL) assay it was found that the first sign of internucleosomal DNA fragmentation in granulosa cells of pre-ovulatory follicles was detectable 24 h after GnRH antagonist treatment. In order to find an upstream marker of atresia, the 5-bromo-deoxyuridine (BrdU) labelling index was used as a measure of proliferation. Already at 14 h after GnRH antagonist treatment, when morphological signs of atresia were not yet present, a clear decrease in BrdU labelling index was found in the granulosa cells.


Subject(s)
Apoptosis , Cell Division , Follicular Atresia , Ovulation , Animals , Bromodeoxyuridine/metabolism , Cell Nucleus/ultrastructure , DNA Fragmentation , Female , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Granulosa Cells/ultrastructure , Immunohistochemistry , In Situ Nick-End Labeling , Luteinizing Hormone/metabolism , Meiosis , Oocytes/cytology , Ovulation/drug effects , Proestrus , Rats , Rats, Wistar
12.
J Endocrinol Invest ; 23(9): 597-604, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11079455

ABSTRACT

The ubiquitin system is involved in numerous cellular processes, regulating the amounts and/or activities of specific proteins through posttranslational coupling with ubiquitin or ubiquitin-like proteins. In spermatogenesis, there appears to be a special requirement for certain components of the ubiquitin system, as exemplified in human and mouse by mutation of USP9Y and HR6B, respectively. Both genes encode proteins which take part in the ubiquitin system and are ubiquitously expressed, but their mutation generates no apparent phenotype other than male infertility. Different phases of mammalian spermatogenesis probably require different specialized activities of the ubiquitin system. It is anticipated that ubiquitination activities similar to those required during mitotic cell cycle regulation will play some role in control of the meiotic divisions. In spermatocytes, there is an intricate link among DNA repair, the ubiquitin system, and regulation of meiotic chromatin structure, as indicated by the co-localization of proteins involved in these processes on meiotic recombination complexes. HR6B and its nearly identical homolog HR6A are multiple function proteins, with ubiquitin-conjugating activity and essential roles in post-replication DNA repair. HR6B, possibly together with the ubiquitin-ligating enzyme mRAD1 8Sc, is most likely involved in chromatin re-organization during the meiotic and post-meiotic phases of spermatogenesis. Biochemical data indicate that, in particular during spermiogenesis, the general activity of the ubiquitin system is high, which most likely is related to the high requirement for massive breakdown of cytoplasmatic and nuclear proteins during this last phase of spermatogenesis.


Subject(s)
Spermatogenesis , Ubiquitins/physiology , Animals , Cyclins/metabolism , DNA Repair , Humans , Male , Meiosis , Mitosis , Spermatogenesis/genetics , Ubiquitins/genetics
13.
Genomics ; 69(1): 86-94, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11013078

ABSTRACT

The RAD18 gene of the yeast Saccharomyces cerevisiae encodes a protein with ssDNA binding activity that interacts with the ubiquitin-conjugating enzyme RAD6 and plays an important role in postreplication repair. We identified and characterized the putative mouse homolog of RAD18, designated mRAD18Sc. The mRAD18Sc open reading frame encodes a 509-amino-acid polypeptide that is strongly conserved in size and sequence between yeast and mammals, with specific conservation of the RING-zinc-finger and the classic zinc-finger domain. The degree of sequence conservation between mRAD18Sc, RAD18, and homologous sequences identified in other species (NuvA from Aspergillus nidulans and Uvs-2 from Neurospora crassa) is entirely consistent with the evolutionary relationship of these organisms, strongly arguing that these genes are one another's homologs. Consistent with the presence of a nuclear translocation signal in the amino acid sequence, we observed the nuclear localization of GFP-tagged mRAD18Sc after stable transfection to HeLa cells. mRNA expression of mRAD18Sc in the mouse was observed in thymus, spleen, brain, and ovary, but was most pronounced in testis, with the highest level of expression in pachytene-stage primary spermatocytes, suggesting that mRAD18Sc plays a role in meiosis of spermatogenesis. Finally, we mapped the mRAD18Sc gene on mouse chromosome 6F.


Subject(s)
DNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , COS Cells , Chromosome Mapping , DNA, Complementary/chemistry , DNA, Complementary/genetics , Female , Fungal Proteins/genetics , Gene Expression , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution , Ubiquitin-Protein Ligases
14.
Endocrinology ; 141(2): 846-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10650968

ABSTRACT

Recently, it has been shown that targeted inactivation of the Insl3 gene in male mice results in cryptorchidism. The Insl3 gene encodes insulin-like factor 3 (Insl3), which is expressed in fetal Leydig cells. The testicular factor Insl3 appears to play an important role in the transabdominal phase of testis descent, which involves development of the gubernaculum. Other studies have demonstrated that in utero exposure to diethylstilbestrol (DES), a synthetic estrogen, can lead to cryptorchidism both in humans and in animal models. The present study was undertaken to investigate whether prenatal DES-exposure might interfere with testicular Insl3 mRNA expression. Furthermore, the effect of DES on steroidogenic factor 1 (SF-1) mRNA expression level was determined, since it has been shown that SF-1 plays an essential role in transcriptional activation of the Insl3 gene promoter. Timed pregnant mice were treated with DES (100 microg/kg body weight) or vehicle alone on days E9 (gestational day 9) through E17. Control and DES-exposed mouse fetuses were collected at E16, E17 and E18, when transabdominal testis descent is taking place. Lack of gubernaculum development in DES-exposed animals was confirmed by histological analyses at E17. Expression of Insl3 and SF-1 mRNAs was studied in testes of control and DES-exposed fetuses at E16 and E18 by RNase protection assay. Prenatal DES-exposure resulted in a three-fold decrease in Insl3 mRNA expression level (P<0.005), at both E16 and E18. In contrast, DES treatment had no effect on the expression of SF-1 mRNA. These results support our hypothesis that DES may interfere with gubernaculum development by altering Insl3 mRNA expression, providing a possible mechanism by which DES may cause cryptorchidism.


Subject(s)
Cryptorchidism/chemically induced , Cryptorchidism/embryology , Diethylstilbestrol/toxicity , Gene Expression Regulation, Developmental , Proteins/physiology , Testis/embryology , Animals , Cryptorchidism/pathology , DNA-Binding Proteins/genetics , Female , Fushi Tarazu Transcription Factors , Gene Expression Regulation, Developmental/drug effects , Gestational Age , Homeodomain Proteins , Hormones/physiology , Insulin , Leydig Cells/physiology , Male , Mice , Mice, Inbred Strains , Pregnancy , Proteins/genetics , Receptors, Cytoplasmic and Nuclear , Steroidogenic Factor 1 , Testis/drug effects , Testis/metabolism , Transcription Factors/genetics , Transcription, Genetic
15.
Endocrinology ; 140(12): 5789-96, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10579345

ABSTRACT

The dimeric glycoprotein anti-Müllerian hormone (AMH) is a member of the transforming growth factor-beta superfamily of growth and differentiation factors. During male fetal sex differentiation, AMH is produced by Sertoli cells and induces degeneration of the Müllerian ducts, which form the anlagen of part of the internal female genital system. In females, AMH is produced by the ovary, but only postnatally. The function of AMH in the ovary is, however, still unknown. Female AMH null mice were reported to be fertile, with normal litter size, but this does not exclude a more subtle function for ovarian AMH. To investigate the function of AMH in the ovary, the complete follicle population was determined in AMH null mice, in mice heterozygous for the AMH null mutation, and in wild-type mice of different ages: 25 days, 4 months, and 13 months. In the present study we found that ovaries of 25-day- and 4-month-old AMH null females, compared to those of wild-type females, contain more preantral and small antral follicles. In addition, in 4- and 13-month-old AMH null females, smaller numbers of primordial follicles were found. Actually, in 13-month-old AMH null females, almost no primordial follicles could be detected, coinciding with a reduced number of preantral and small antral follicles in these females. In almost all females heterozygous for the AMH null mutation the number of follicles fell in between the numbers found in wild-type and AMH null females. In 4-month-old AMH null females serum inhibin levels were higher and FSH levels were lower compared to those in wild-type females. In contrast, inhibin levels were lower in 13-month-old AMH null females, and FSH levels were unchanged compared to those in wild-type females. Furthermore, the weight of the ovaries was twice as high in the 4-month-old AMH null females as in age-matched wild-type females. We conclude that AMH plays an important role in primordial follicle recruitment, such that more primordial follicles are recruited in AMH null mice than in wild-type mice; the mice heterozygous for the AMH null mutation take an in-between position. Consequently, the ovaries of AMH null females and those of females heterozygous for the AMH null mutation will show a relatively early depletion of their stock of primordial follicles. The female AMH null mouse may thus provide a useful model to study regulation of primordial follicle recruitment and the relation between follicular dynamics and ovarian aging.


Subject(s)
Glycoproteins , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Ovarian Follicle/physiology , Testicular Hormones/genetics , Testicular Hormones/physiology , Aging , Animals , Anti-Mullerian Hormone , Corpus Luteum/anatomy & histology , Estrus , Female , Follicle Stimulating Hormone/blood , Inhibins/blood , Male , Mice , Mice, Knockout , Organ Size , Ovarian Follicle/anatomy & histology , Ovary/anatomy & histology , Ovary/growth & development , Uterus/anatomy & histology
16.
Mol Cell Endocrinol ; 151(1-2): 5-16, 1999 May 25.
Article in English | MEDLINE | ID: mdl-10411315

ABSTRACT

Ubiquitin is a ubiquitous and highly conserved protein of 76 amino acid residues, that can be covalently attached to cellular acceptor proteins. The attachment of ubiquitin to target proteins is achieved through a multi-step enzymatic pathway, which involves activities of ubiquitin-activating E1 enzymes, ubiquitin-conjugating E2 enzymes, and ligating E3 enzymes. Mono- or poly-ubiquitination of proteins can lead to protein degradation or modification of protein activity. Many components of the complex ubiquitin system show remarkable evolutionary conservation, from yeast to mammalian species. The ubiquitin system is essential to all eukaryotic cells. Among others, several signal transduction cascades show involvement of the ubiquitin system, but there are currently little data supporting a specific role of the ubiquitin system in hormonal control of reproduction. Interestingly, during gametogenesis, many specialized and important aspects of the ubiquitin system become apparent. Components of the ubiquitin system appear to be involved in different steps and processes during gametogenesis, including control of meiosis, and reorganization of chromatin structure.


Subject(s)
Signal Transduction/physiology , Spermatogenesis/physiology , Ubiquitins/physiology , Animals , Humans , Male
17.
J Androl ; 20(3): 399-406, 1999.
Article in English | MEDLINE | ID: mdl-10386820

ABSTRACT

The in vitro response of Sertoli cells isolated from adult rat testes to testosterone (T) and follicle-stimulating hormone (FSH) treatment was investigated. Sertoli cells from >70-day-old Sprague-Dawley rats were isolated by a combined enzymatic treatment followed by the removal of the majority of contaminating germ cells with immobilized peanut agglutinin lectin. Sertoli cells were then cultured for 6-10 days, forming a confluent layer with a cell viability of >83% and 74-77% purity. The contaminating cells were peritubular cells (4-6%), pachytene spermatocytes (4-5%), round spermatids (<2%), elongated spermatids (<1%), and degenerating germ cells (14.8%). The proportion of degenerating germ cells decreased from 14.8% to 8.6% between days 6 and 10 in culture. After a prestimulation culture period of 4 days, FSH treatment over a 2-day period resulted in a dose-related increase of inhibin with a median effective dose (ED50) value of 36.7+/-20.4 ng/ml in comparison with an ED50 value of 4.4+/-0.9 ng/ml obtained with immature Sertoli cell cultures from 20-day-old rats. Mature Sertoli cells, in contrast to immature Sertoli cells, were unresponsive to combined FSH + T treatment for the production of the cell adhesion protein N-cadherin. FSH treatment promoted the in vitro binding of round spermatids isolated from adult testis to adult Sertoli cells in coculture. It is concluded that mature Sertoli cells in culture are responsive to FSH in terms of inhibin production and round-spermatid binding. The lack of an FSH + T-induced increase in N-cadherin or round spermatid binding is attributed to either a reduced sensitivity, or an alteration in the regulation of mature Sertoli cells by FSH + T.


Subject(s)
Cadherins/metabolism , Follicle Stimulating Hormone/pharmacology , Inhibins/metabolism , Sertoli Cells/drug effects , Spermatids/metabolism , Testosterone/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Sertoli Cells/cytology , Sertoli Cells/metabolism
18.
Dev Biol ; 207(2): 322-33, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10068466

ABSTRACT

Male infertility in HR6B knockout mice is associated with impairment of spermatogenesis. The HR6B gene is a mammalian, autosomal homolog of the Saccharomyces cerevisiae gene Rad6 encoding a ubiquitin-conjugating enzyme. In addition, X-chromosomal HR6A has been identified, in human and mouse. RAD6 in yeast is required for a variety of cellular functions, including sporulation, DNA repair, and mutagenesis. Since RAD6 and its mammalian homologs can ubiquitinate histones in vitro, we have investigated the pattern of histone ubiquitination in mouse testis. By immunoblot and immunohistochemical analysis of wild-type mouse testis, a high amount of ubiquitinated H2A (uH2A) was detected in pachytene spermatocytes. This signal became undetectable in round spermatids, but then increased again during a relatively short developmental period, in elongating spermatids. No other ubiquitinated histones were observed. In the HR6B knockout mice, we failed to detect an overt defect in the overall pattern of histone ubiquitination. For somatic cell types, it has been shown that histone ubiquitination is associated with destabilization of nucleosomes, in relation to active gene transcription. Unexpectedly, the most intense uH2A signal in pachytene spermatocytes was detected in the sex body, an inactive nuclear structure that contains the heterochromatic X and Y chromosomes. The postmeiotic uH2A immunoexpression in elongating spermatids indicates that nucleosome destabilization induced by histone ubiquitination may play a facilitating role during histone-to-protamine replacement.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Ligases/genetics , Saccharomyces cerevisiae Proteins , Spermatogenesis/genetics , Testis/enzymology , Ubiquitins/metabolism , Animals , Germ Cells/classification , Male , Mice , Mice, Knockout , Nuclear Proteins/analysis , Spermatids/metabolism , Spermatocytes/metabolism , Ubiquitin-Conjugating Enzymes
19.
Endocrinology ; 139(10): 4244-51, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9751506

ABSTRACT

The clinical use of diethylstilbestrol (DES) by pregnant women has resulted in an increased incidence of genital carcinoma in the daughters born from these pregnancies. Also, in the so-called DES-sons abnormalities were found, mainly, the presence of Müllerian duct remnants, which indicates that fetal exposure to DES may have an effect on male sex differentiation. Fetal regression of the Müllerian ducts is under testicular control through anti-Müllerian hormone (AMH). In male mice, treated in utero with DES, the Müllerian ducts do not regress completely, although DES-exposed testes do produce AMH. We hypothesized that incomplete regression in DES-exposed males is caused by a diminished sensitivity of the Müllerian ducts to AMH. Therefore, the effect of DES on temporal aspects of Müllerian duct regression and AMH type II receptor (AMHRII) messenger RNA (mRNA) expression in male mouse fetuses was studied. It was observed that Müllerian duct regression was incomplete at E19 (19 days post coitum), upon DES administration during pregnancy from E9 through E16. Furthermore, analysis of earlier time points of fetal development revealed that the DES treatment had clearly delayed the onset of Müllerian duct formation by approximately 2 days; in untreated fetuses, Müllerian duct formation was complete by E13, whereas fully formed Müllerian ducts were not observed in DES-treated male fetuses until E15. Using in situ hybridization, no change in the localization of AMH and AMHRII mRNA expression was observed in DES-exposed male fetuses. The mRNA expression was quantified using ribonuclease protection assay, showing an increased expression level of AMH and AMHRII mRNAs at E 13 in DES-exposed male fetuses. Furthermore, the mRNA expression levels of Hoxa 11 and steroidogenic factor-1 (SF-1) were determined as a marker for fetal development. Prenatal DES exposure had no effect on Hoxa 11 mRNA expression, indicating that DES did not exert an overall effect on the rate of fetal development. In DES-exposed male fetuses, SF-1 showed a similar increase in mRNA expression as AMH, in agreement with the observations that the AMH gene promoter requires an intact SF-1 DNA binding site for time- and cell-specific expression, although an effect of DES on SF-1 expression in other tissues, such as the adrenal and pituitary gland, cannot be excluded. However, the increased expression levels of AMH and AMHRII mRNAs do not directly explain the decreased sensitivity of the Müllerian ducts to AMH. Therefore, it is concluded that prenatal DES exposure of male mice delays the onset of Müllerian duct development, which may result in an asynchrony in the timing of Müllerian duct formation, with respect to the critical period of Müllerian duct regression, leading to persistence of Müllerian duct remnants in male mice.


Subject(s)
Diethylstilbestrol/toxicity , Fetus/drug effects , Glycoproteins , Mullerian Ducts/drug effects , Animals , Anti-Mullerian Hormone , Female , Growth Inhibitors/genetics , Male , Mice , Pregnancy , Promoter Regions, Genetic , RNA, Messenger/analysis , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta , Testicular Hormones/genetics
20.
Hum Reprod ; 13(6): 1559-66, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9688392

ABSTRACT

During mammalian spermatogenesis, the chromatin of the spermatogenic cells is profoundly reorganized. Somatic histones are partly replaced by testis-specific histones. These histones are then replaced by transition proteins and finally by protamines. This series of nucleoprotein rearrangements results in a highly condensed sperm cell nucleus. In contrast to spermatozoa from other species, human spermatozoa still contain a significant amount of histones, including testis-specific histone 2B (TH2B). In the present study it is shown that an antibody targeting tyrosine hydroxylase, which has been found previously to cross-react with rat TH2B, also specifically immunoreacts with human TH2B on Western blots, in immunohistochemistry of human testis tissue, and in immunocytochemistry of decondensed human spermatozoa. In human testis tissue, TH2B immunostaining first apparent in spermatogonia, shows marked variation, especially at the pachytene spermatocyte stage, and then reaches an intense signal in round spermatids. Shortly before spermatid elongation, a portion of the spermatid nucleus, corresponding to the acrosomal region, loses its immunoreactivity. During condensation of the spermatid nucleus, the immunodetectability of TH2B disappears gradually, from the anterior region of the nucleus onwards. At the final stages of spermiogenesis, the immunostaining is completely absent. Immunocytochemical staining of spermatozoa revealed no TH2B immunosignal, but immunostaining was observed when spermatozoa obtained from semen were decondensed to make nuclear proteins accessible to the antibody. There was, however, a striking intercellular variability in the intensity of staining of spermatozoa within an ejaculate. In a population of 35 men attending our Andrology Clinic, we observed interindividual differences in total sperm TH2B content, which showed a significant, although not very pronounced, negative correlation with normal morphology (P = 0.05).


Subject(s)
Histones/biosynthesis , Nuclear Proteins/biosynthesis , Spermatozoa/physiology , Testis/physiology , Animals , Antibodies/immunology , Histones/immunology , Humans , Immunohistochemistry , Male , Nuclear Proteins/immunology , Rats , Rats, Wistar , Spermatogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...