Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Development ; 144(19): 3430-3439, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28827392

ABSTRACT

The human spermatogonial compartment is essential for daily production of millions of sperm. Despite this crucial role, the molecular signature, kinetic behavior and regulation of human spermatogonia are poorly understood. Using human testis biopsies with normal spermatogenesis and by studying marker protein expression, we have identified for the first time different subpopulations of spermatogonia. MAGE-A4 marks all spermatogonia, KIT marks all B spermatogonia and UCLH1 all Apale-dark (Ap-d) spermatogonia. We suggest that at the start of the spermatogenic lineage there are Ap-d spermatogonia that are GFRA1High, likely including the spermatogonial stem cells. Next, UTF1 becomes expressed, cells become quiescent and GFRA1 expression decreases. Finally, GFRA1 expression is lost and subsequently cells differentiate into B spermatogonia, losing UTF1 and acquiring KIT expression. Strikingly, most human Ap-d spermatogonia are out of the cell cycle and even differentiating type B spermatogonial proliferation is restricted. A novel scheme for human spermatogonial development is proposed that will facilitate further research in this field, the understanding of cases of infertility and the development of methods to increase sperm output.


Subject(s)
Spermatogonia/cytology , Spermatogonia/metabolism , Adult , Aged , Cell Count , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Kinetics , Male , Middle Aged , Models, Biological , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Young Adult
2.
Biol Reprod ; 89(3): 60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23946533

ABSTRACT

To date, in the human seminiferous epithelium, only six associations of cell types have been distinguished, subdividing the epithelial cycle into six stages of very different duration. This hampers comparisons between studies on human and laboratory animals in which the cycle is usually subdivided into 12 stages. We now propose a new stage classification on basis of acrosomal development made visible by immunohistochemistry (IHC) for (pro)acrosin. IHC for acrosin gives results that are comparable to periodic acid Schiff staining. In the human too, we now distinguish 12 stages that differ from each other in duration by a factor of two at most. B spermatogonia are first apparent in stage I, preleptotene spermatocytes are formed in stage V, leptonema starts in stage VII, and spermiation takes place at the end of stage VI. A similar timing was previously observed in several monkeys. Stage identification by way of IHC for acrosin appeared possible for tissue fixed in formalin, Bouin fixative, diluted Bouin fixative, Cleland fluid, and modified Davidson fixative, indicating a wide applicability. In addition, it is also possible to distinguish the 12 stages in glutaraldehyde/osmium-tetroxide fixed/plastic embedded testis material without IHC for acrosin. The new stage classification will greatly facilitate research on human spermatogenesis and enable a much better comparison with results from work on experimental animals than hitherto possible. In addition, it will enable a highly focused approach to evaluate spermatogenic impairments, such as germ cell maturation arrests or defects, and to study details of germ cell differentiation.


Subject(s)
Acrosome/classification , Acrosome/physiology , Spermatogenesis/physiology , Spermatogonia/classification , Adult , Aged , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Spermatids/physiology , Spermatogonia/cytology , Spermatogonia/physiology , Young Adult
3.
PLoS One ; 8(4): e59431, 2013.
Article in English | MEDLINE | ID: mdl-23613711

ABSTRACT

In mammals, the biological activity of the stem/progenitor compartment sustains production of mature gametes through spermatogenesis. Spermatogonial stem cells and their progeny belong to the class of undifferentiated spermatogonia, a germ cell population found on the basal membrane of the seminiferous tubules. A large body of evidence has demonstrated that glial cell line-derived neurotrophic factor (GDNF), a Sertoli-derived factor, is essential for in vivo and in vitro stem cell self-renewal. However, the mechanisms underlying this activity are not completely understood. In this study, we show that GDNF induces dose-dependent directional migration of freshly selected undifferentiated spermatogonia, as well as germline stem cells in culture, using a Boyden chamber assay. GDNF-induced migration is dependent on the expression of the GDNF co-receptor GFRA1, as shown by migration assays performed on parental and GFRA1-transduced GC-1 spermatogonial cell lines. We found that the actin regulatory protein vasodilator-stimulated phosphoprotein (VASP) is specifically expressed in undifferentiated spermatogonia. VASP belongs to the ENA/VASP family of proteins implicated in actin-dependent processes, such as fibroblast migration, axon guidance, and cell adhesion. In intact seminiferous tubules and germline stem cell cultures, GDNF treatment up-regulates VASP in a dose-dependent fashion. These data identify a novel role for the niche-derived factor GDNF, and they suggest that GDNF may impinge on the stem/progenitor compartment, affecting the actin cytoskeleton and cell migration.


Subject(s)
Cell Movement/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Spermatogonia/cytology , Stem Cell Niche , Stem Cells/cytology , Stem Cells/drug effects , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Cell Adhesion Molecules/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Rats , Stem Cells/metabolism
4.
Hum Reprod ; 27(8): 2267-76, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22647452

ABSTRACT

BACKGROUND: Clusterin, a heterodimeric glycoprotein found at several sites in the human male reproductive tract, could be a marker of morphologically abnormal spermatozoa, while TUNEL positivity indicates DNA fragmentation. Metabolic disorders such as diabetes mellitus and obesity may compromise sperm quality and fertility of men; however, little evidence specifically links hypertension with the impairment of male reproductive function. METHODS: By flow cytometric, immunofluorescence (TUNEL assay and clusterin immunolabeling) and immunohistochemical (peroxidase-streptavidin method) analyses, we have compared both clusterin- and TUNEL labeling in ejaculated spermatozoa from healthy normotensive donors and hypertensive subjects with the purpose to reveal possible differences between the two conditions. RESULTS: Data analysis from the normotensive (n=25) and hypertensive subjects (n=25) demonstrate a significant correlation between high levels of clusterin immunolabeling and the presence of sperm DNA damage, which is often associated with abnormal morphology. In the normotensive subjects, a low percentage (15.3±4.5) of spermatozoa positive for high levels of clusterin was detected; however, this percentage significantly increased (30.9±13.0) (P<0.01) in hypertensive subjects. Standard semen evaluations does not reveal any significant differences between the two groups of subjects, except for a reduced forward motility and lower sperm vitality in the hypertensive subjects. CONCLUSIONS: This pilot study strongly suggests a relationship between hypertension and markers indicative of poor sperm quality. In hypertensive subjects, high levels of clusterin immunolabeling identified a consistent fraction of ejaculated spermatozoa carrying both DNA fragmentation and strong morphological alterations, which was not correlated with age or with sperm cell mortality. The alternative possibility that sperm damage observed is due to adverse effects of anti-hypertensive drugs does not find support in the literature nor in the drug data sheets. The relationship observed between hypertension and human semen represents a novel and possibly relevant information to be considered in the study of male fertility.


Subject(s)
Clusterin/chemistry , DNA Damage , Hypertension/metabolism , Spermatozoa/metabolism , Adult , Case-Control Studies , Cohort Studies , DNA Fragmentation , Flow Cytometry/methods , Glycoproteins/chemistry , Humans , Hypertension/pathology , In Situ Nick-End Labeling , Male , Microscopy, Fluorescence/methods , Middle Aged , Regression Analysis , Semen/metabolism , Spermatozoa/pathology
5.
Reproduction ; 143(3): 325-32, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22143971

ABSTRACT

In mice and other mammals, spermatogenesis is maintained by spermatogonial stem cells (SSCs), a cell population belonging to undifferentiated type A spermatogonia. In the accepted model of SSC self-renewal, Asingle (As) spermatogonia are the stem cells, whereas paired (Apaired (Apr)) and chained (Aaligned (Aal)) undifferentiated spermatogonia are committed to differentiation. This model has been recently challenged by evidence that As and chained (Apr and Aal), undifferentiated spermatogonia are heterogeneous in terms of gene expression and function. The expression profile of several markers, such as GFRA1 (the GDNF co-receptor), is heterogeneous among As, Apr and Aal spermatogonia. In this study, we have analysed and quantified the distribution of GFRA1-expressing cells within the different stages of the seminiferous epithelial cycle. We show that in all stages, GFRA1+ chained spermatogonia (Apr to Aal) are more numerous than GFRA1+ As spermatogonia. Numbers of chained GFRA1+ spermatogonia are sharply reduced in stages VII-VIII when Aal differentiate into A1 spermatogonia. GFRA1 expression is regulated by GDNF and in cultures of isolated seminiferous tubules, we found that GDNF expression and secretion by Sertoli cells is stage-dependent, being maximal in stages II-VI and decreasing thereafter. Using qRT-PCR analysis, we found that GDNF regulates the expression of genes such as Tex14, Sohlh1 and Kit (c-Kit) known to be involved in spermatogonial differentiation. Expression of Kit was upregulated by GDNF in a stage-specific manner. Our data indicate that GDNF, besides its crucial role in the self-renewal of stem cells also functions in the differentiation of chained undifferentiated spermatogonia.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Spermatogonia/metabolism , Testis/metabolism , Age Factors , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Promyelocytic Leukemia Zinc Finger Protein , Seminiferous Epithelium/drug effects , Seminiferous Epithelium/metabolism , Spermatogonia/drug effects , Testis/cytology , Testis/drug effects , Tissue Distribution
6.
Int J Dev Biol ; 55(2): 209-14, 2011.
Article in English | MEDLINE | ID: mdl-21553385

ABSTRACT

In mammals, the stem cells of spermatogenesis are derived from an embryonic cell population called primordial germ cells (PGCs). Spermatogonial stem cells displaying the "side population" (SP) phenotype have been identified in the immature and adult mouse testis, but noting is known about the expression of the SP phenotype during prenatal development of germ cells. The SP phenotype, defined as the ability of cells to efflux fluorescent dyes such as Hoechst, is common to several stem/progenitor cell types. In the present study, we analyzed and characterized the Hoechst SP via cytofluorimetric analysis of disaggregated gonads at different time points during embryonic development in mice. To directly test the hypothesis that the SP phenotype is a feature of germ cell lineage, experiments were performed on transgenic animals expressing enhanced green fluorescent protein (EGFP) under the control of the Oct4 promoter, to identify early germ cells up to PGCs. We found that prenatal gonads contain a fraction of SP cells at each stage analyzed, and the percentage of cells in the SP fraction decreases as development proceeds. Surprisingly, more than 50% of the PGCs displayed the SP phenotype at 11.5 dpc (days post coitum). The percentage of germ cells with the SP phenotype decreased steadily with development, to less than 1% at 18.5 dpc. Cytofluorimetric analysis along with immunocytochemistry performed on sorted cells indicated that the SP fraction of prenatal gonads, as in the adult testis, was heterogeneous, being composed of both somatic and germ cells. Both cell types expressed the ABC transporters Abcg2, Abcb1a, Abcb1b and Abcc1. These findings provide evidence that the SP phenotype is a common feature of PGCs and identifies a subpopulation of fetal testis cells including prospermatogonia whose differentiation fate remains to be investigated.


Subject(s)
Germ Cells/cytology , Side-Population Cells/cytology , Testis/cytology , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Cell Differentiation , Cell Lineage , Flow Cytometry , Germ Cells/metabolism , Male , Mice , Mice, Transgenic , Octamer Transcription Factor-3/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Side-Population Cells/metabolism , Spermatogenesis , Spermatogonia/cytology , Spermatogonia/metabolism , Testis/embryology , Testis/metabolism
7.
Hum Reprod ; 25(9): 2188-202, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20601678

ABSTRACT

BACKGROUND: TR-KIT, a truncated form of KIT (the KITL receptor), corresponding to the c-terminal half of the intracellular split tyrosine kinase domain, is expressed during the haploid stages of mouse spermatogenesis, and is one of the candidate sperm factors possibly involved in egg activation at fertilization. METHODS: Immunocytochemistry of adult human testis, and studies of human semen samples from volunteer donors through immunofluorescence, confocal microscopy, flow cytometry, western blot and RT-PCR analyses were performed. RESULTS: We show that the TR-KIT is expressed during spermiogenesis in the human testis, and that it is maintained in human ejaculated spermatozoa. TR-KIT is localized both in the equatorial segment and in the sub-acrosomal region of the human sperm head. The equatorial localization of the TR-KIT persists after the spontaneous acrosome reaction. Cytometric analysis of several sperm samples from volunteer donors, showed variable degrees of the TR-KIT-specific immunolabeling, and a significant inverse correlation (Pearson's coefficient, r = -0.76, P < 0.0001, n = 23) of the TR-KIT positivity with markers of sperm damage, i.e. DNA fragmentation, as revealed by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-nick end labeling (TUNEL) analysis and the intense clusterin positivity. We also found less significant inverse correlation with altered head morphology (r = -0.47, P < 0.05, n = 23) and direct correlation with sperm forward motility parameters (r = 0.59, P < 0.01, n = 23). CONCLUSIONS: The TR-KIT is present in the equatorial region of human spermatozoa, which is the first sperm component entering into the oocyte cytoplasm after fusion with the egg. This localization is consistent with the function previously proposed for this protein in mice. In addition, the TR-KIT represents a potential predictive parameter of human sperm quality.


Subject(s)
DNA Fragmentation , Gene Expression , Proto-Oncogene Proteins c-kit/metabolism , Spermatozoa/chemistry , Spermatozoa/metabolism , Acrosome Reaction , Adult , Aged , Biomarkers/metabolism , Cell Shape , Clusterin/metabolism , Humans , Male , Middle Aged , Organ Specificity , Protein Isoforms/metabolism , Protein Transport , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger , Semen Analysis , Sperm Head/metabolism , Sperm Head/pathology , Spermatozoa/pathology , Testis/cytology , Testis/metabolism , Young Adult
8.
Mol Cell Endocrinol ; 320(1-2): 34-44, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20138961

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is transiently expressed in preovulatory follicles of different species and positively affects parameters correlated with the ovulatory process. It has also been shown to be expressed in the interstitial tissue and in interstitial glandular cells in the proximity of primordial and preantral follicles. The aim of the present study was to investigate whether PACAP influences the recruitment of primordial follicles and the growth and differentiation of preantral follicles. Rat ovaries from 2-day-old animals were cultured for 5 days in the presence of PACAP. This treatment significantly inhibited the primordial to primary follicle transition. PACAP inhibited granulosa cell proliferation without affecting cell viability. PACAP also inhibited the growth of isolated preantral follicles cultured under basal conditions or in the presence of follicle-stimulating hormone (FSH). These results suggest that PACAP is significantly involved in the cyclic recruitment of primordial follicles and in the FSH-dependent growth of preantral follicles.


Subject(s)
Ovarian Follicle/drug effects , Ovarian Follicle/growth & development , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation, Developmental/drug effects , Humans , In Situ Hybridization , Mice , Organ Culture Techniques , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
9.
Stem Cells ; 27(12): 3043-52, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19711452

ABSTRACT

Spermatogenesis is maintained by a pool of spermatogonial stem cells (SSCs). Analyses of the molecular profile of SSCs have revealed the existence of subsets, indicating that the stem cell population is more heterogeneous than previously believed. However, SSC subsets are poorly characterized. In rodents, the first steps in spermatogenesis have been extensively investigated, both under physiological conditions and during the regenerative phase that follows germ cell damage. In the widely accepted model, the SSCs are type Asingle (As) spermatogonia. Here, we tested the hypothesis that As spermatogonia are phenotypically heterogeneous by analyzing glial cell line-derived neurotrophic factor (GDNF) family receptor alpha1 (GFRA1) expression in whole-mounted seminiferous tubules, via cytofluorimetric analysis and in vivo colonogenic assays. GFRA1 is a coreceptor for GDNF, a Sertoli cell-derived factor essential for SSC self-renewal and proliferation. Morphometric analysis demonstrated that 10% of As spermatogonia did not express GFRA1 but were colonogenic, as shown by germ cell transplantation assay. In contrast, cells selected for GFRA1 expression were not colonogenic in vivo. In human testes, GFRA1 was also heterogeneously expressed in Adark and in Apale spermatogonia, the earliest spermatogonia. In vivo 5-bromo-2'-deoxyuridine administration showed that both GFRA1(+) and GFRA1(-) As spermatogonia were engaged in the cell cycle, a finding supported by the lack of long-term label-retaining As spermatogonia. GFRA1 expression was asymmetric in 5% of paired cells, suggesting that As subsets may be generated by asymmetric cell division. Our data support the hypothesis of the existence of SSC subsets and reveal a previously unrecognized heterogeneity in the expression profile of As spermatogonia in vivo.


Subject(s)
Cell Shape , Spermatogonia/cytology , Stem Cells/cytology , Aging , Animals , Cell Separation , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Spermatogonia/metabolism , Stem Cells/metabolism
10.
Differentiation ; 78(2-3): 131-6, 2009.
Article in English | MEDLINE | ID: mdl-19541401

ABSTRACT

Spermatogonial stem cells (SSC) ensure continuous production of mammalian male gametes. In rodents, the SSC are Asingle spermatogonia (As). Gene loss and gain-of-function mutations have provided some clues into SSC function, but genetic dissection of SSC physiology has not yet been accomplished. The adaptor protein Numb is an evolutionarily conserved protein originally implicated in the control of the fate of sibling cells. Mice homozygous for deficient Numb die before embryonic day 11.5, hampering the analysis of its inactivation in postnatal male germline. Here, we have developed an experimental strategy to conditionally inactivate Numb and its homolog Numblike in the postnatal germline by in vitro delivery of cell-permeant Cre recombinase. Cre-transduced SSC isolated from wild-type mice retained their ability to self-renew and to differentiate in vivo, as shown by their ability to give rise to normal spermatogenic colonies when transplanted in recipient testes. Cre-transduced SSC from conditional mutant mouse line were able to colonize recipient testes upon transplantation. Inactivation of either Numb or Numblike in SSC did not impair the development of normal donor-derived spermatogenesis. However, compared to single-null SSC, double-null SSC generated shorter colonies in which the germ cells failed to differentiate beyond the round spermatid stage, underscoring the essential roles of both Numb and Numblike in spermatogenesis. We demonstrate the feasibility of gene inactivation in adult SSC by ex vivo Cre delivery. This provides a means to analyze the function of genes that operate on a cell-autonomous basis or those that are coupled to signals that SSC receive from bystander cells.


Subject(s)
Gene Expression Regulation/physiology , Gene Silencing/physiology , Integrases/physiology , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Spermatogonia/cytology , Stem Cells/cytology , Animals , Cell Differentiation/physiology , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatogonia/metabolism , Stem Cells/metabolism , Testis/cytology , Testis/physiology , Transduction, Genetic
11.
J Cell Biol ; 185(2): 235-49, 2009 Apr 20.
Article in English | MEDLINE | ID: mdl-19380878

ABSTRACT

Sam68 is a KH-type RNA-binding protein involved in several steps of RNA metabolism with potential implications in cell differentiation and cancer. However, its physiological roles are still poorly understood. Herein, we show that Sam68(-/-) male mice are infertile and display several defects in spermatogenesis, demonstrating an essential role for Sam68 in male fertility. Sam68(-/-) mice produce few spermatozoa, which display dramatic motility defects and are unable to fertilize eggs. Expression of a subset of messenger mRNAs (mRNAs) is affected in the testis of knockout mice. Interestingly, Sam68 is associated with polyadenylated mRNAs in the cytoplasm during the meiotic divisions and in round spermatids, when it interacts with the translational machinery. We show that Sam68 is required for polysomal recruitment of specific mRNAs and for accumulation of the corresponding proteins in germ cells and in a heterologous system. These observations demonstrate a novel role for Sam68 in mRNA translation and highlight its essential requirement for the development of a functional male gamete.


Subject(s)
Germ Cells/physiology , Protein Biosynthesis , RNA, Messenger/metabolism , Spermatogenesis/physiology , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Female , Fertility/physiology , Gene Expression Regulation , Genes, Reporter , Germ Cells/cytology , Male , Meiosis/physiology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Pregnancy , RNA, Messenger/genetics , Spermatozoa/cytology , Spermatozoa/physiology , Testis/cytology , Testis/physiology
12.
Reproduction ; 134(2): 281-92, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17660238

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) positively affect several parameters correlated with the ovulatory process. PACAP is transiently expressed in rat preovulatory follicles, while VIP is present in nerve fibres at all stages of development. These two peptides act by interacting with three types of receptors: PACAP type I receptor (PAC1-R), which binds with higher affinity to PACAP, and two VIP receptors (VPAC1-R and VPAC2-R), which bind to PACAP and VIP with equal affinity. The aim of the present study was to characterise the PACAP/VIP/receptor system in the mouse ovary. Results obtained by RT-PCR, immunohistochemistry and in situ hybridisation showed that PACAP was transiently expressed in granulosa cells of preovulatory follicles after human chorionic gonadotrophin (hCG) stimulation, while VIP mRNA was never observed. All the receptors were present in 22-day-old untreated mice. In preovulatory follicles, PAC1-R was expressed both in granulosa cells and in residual ovarian tissue but was stimulated by hCG mainly in granulosa cells; VPAC2-R was present in both the cell compartments and was only mildly stimulated; VPAC1-R was present mainly in the residual ovarian tissue and was downregulated by hCG. PACAP and VIP were equipotent in inhibiting apoptosis in granulosa cells, confirming the presence of functional PACAP/VIP receptors. The contemporary induction by hCG of PACAP and PAC1-R in granulosa cells of preovulatory follicles suggests that, also in mouse ovary, PACAP may play a significant role around the time of ovulation. Moreover, the presence of PACAP/VIP receptors in the untreated ovary suggests a possible role for PACAP and VIP during follicle development.


Subject(s)
Ovary/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/analysis , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/analysis , Receptors, Vasoactive Intestinal Peptide/analysis , Vasoactive Intestinal Peptide/analysis , Animals , Chorionic Gonadotropin/pharmacology , Female , Gene Expression , Granulosa Cells/chemistry , Granulosa Cells/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred Strains , Ovary/chemistry , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , RNA, Messenger/analysis , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/analysis , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/analysis , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Culture Techniques , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
13.
Endocrinology ; 148(9): 4302-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17540721

ABSTRACT

To investigate the physiological effects of mitochondrial phospholipid hydroperoxide glutathione peroxidase (mPHGPx) overexpression during early male germ cell differentiation, we have generated transgenic mice bearing the rat mPhgpx coding sequence driven by the mouse synaptonemal complex protein 1 promoter, allowing the transgene to be specifically activated in the testis from the zygotene to diplotene stages of the first meiotic division. Northern/Western blotting and immunocytochemical analyses of endogenous mPHGPx expression during spermatogenesis showed a low enzyme level in middle-late pachytene spermatocytes, but not in earlier meiotic stages, and a significant increase in mPHGPx content in round spermatids. The histological and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling analysis of transgenic testes revealed a number of spermatogenetic defects, including primary spermatocyte apoptosis, haploid cell loss, and seminiferous epithelium disorganization. In line with these features, adult transgenic male mice also displayed a reduction in fertility. Results obtained in this study suggest that mPHGPx expression is tightly regulated in pachytene spermatocytes, with any spatial-temporal increase in mPHGPx expression resulting in damage to spermatogenesis and eventual loss of haploid cells. Present findings in the mouse may be of interest to human male fertility.


Subject(s)
Glutathione Peroxidase/genetics , Mitochondria/enzymology , Spermatozoa/enzymology , Animals , Cell Differentiation , Glutathione Peroxidase/metabolism , Haploidy , Immunohistochemistry , In Situ Nick-End Labeling , Infertility, Male/enzymology , Male , Meiosis , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Phospholipid Hydroperoxide Glutathione Peroxidase , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Spermatids/enzymology , Spermatocytes/enzymology , Spermatogenesis , Spermatozoa/cytology
14.
Reproduction ; 132(6): 887-97, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17127749

ABSTRACT

Numb is an adaptor protein that is asymmetrically inherited at mitosis and controls the fate of sibling cells in different species. The role of m-Numb (mammalian Numb) as an important cell fate-determining factor has extensively been described mostly in neural tissues, particularly in progenitor cells, in the mouse. Biochemical and genetic analyses have shown that Numb acts as an inhibitor of the Notch signaling pathway, an evolutionarily conserved pathway involved in the control of cell proliferation, differentiation, and apoptosis. In the present study, we sought to determine m-Numb distribution in germ cells in the postnatal mouse testis. We show that all four m-Numb isoforms are widely expressed during postnatal testis development. By reverse transcriptase-PCR and western blot analyses, we further identify p71 as the predominantly expressed isoform in germ cells. Moreover, we demonstrate through co-immunoprecipitation studies that m-Numb physically associates with Ap2a1, a component of the endocytotic clathrin-coated vesicles. Finally, we employed confocal immunofluorescence microscopy of whole mount seminiferous tubules and isolated germ cells to gain more insight into the subcellular localization of m-Numb. These morphological analyses confirmed m-Numb and Ap2a1 co-localization. However, we did not observe asymmetric localization of m-Numb neither in mitotic spermatogonial stem cells nor in more differentiated spermatogonial cells, suggesting that spermatogonial stem cell fate in the mouse does not rely on asymmetric partitioning of m-Numb.


Subject(s)
Membrane Proteins/analysis , Nerve Tissue Proteins/analysis , Protein Isoforms/analysis , Spermatozoa/chemistry , Testis/chemistry , Adaptor Protein Complex 2/analysis , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/analysis , Adaptor Protein Complex alpha Subunits/metabolism , Alternative Splicing , Animals , Blotting, Northern/methods , Blotting, Western/methods , Immunoprecipitation/methods , Male , Membrane Proteins/genetics , Mice , Microscopy, Confocal , Nerve Tissue Proteins/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Seminiferous Tubules/chemistry , Spermatogenesis/physiology , Spermatogonia/chemistry , Testis/growth & development
15.
J Endocrinol ; 191(1): 287-99, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17065411

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a bioactive peptide transiently expressed in preovulatory follicles. PACAP acts by interacting with three types of PACAP receptors. PACAP type I receptor (PAC(1)-R), which binds specifically to both PACAPs and vasoactive intestinal polypeptide (VIP), although with lower affinity, and two VIP receptors, VPAC(1)-R and VPAC(2)-R, which bind to PACAP and VIP with equal affinity. In the present study, we showed the expression of all three receptors in whole ovaries obtained from juvenile and gonadotropin-treated immature rats. A more detailed analysis on cells from preovulatory follicles showed that PAC(1)-R and VPAC(2)-R were expressed in granulosa cells, whereas only VIP receptors were expressed in theca/interstitial (TI) cells and fully grown oocytes presented only PAC(1)-R. The distribution of the VIP receptors was confirmed by immunofluorescence. HCG treatment induced stimulation of PAC(1)-R in granulosa cells and VPAC(2)-R in TI cells. The presence of functional PACAP/VIP receptors was also supported by metabolic studies. We further evaluated the presence of PACAP and VIP receptors by testing the effect of these peptides on apoptosis in granulosa cells cultured, isolated or in whole follicles. Treatment of follicles with PACAP and VIP dose-dependently inhibited apoptosis, while only PACAP significantly inhibited isolated granulosa cells. These results demonstrate a different expression of PACAP/VIP receptors in the various follicle compartments and suggest a possible role for PACAP and VIP on granulosa and TI cells, both during follicle development and ovulation.


Subject(s)
Ovarian Follicle/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/analysis , Receptors, Vasoactive Intestinal Peptide/analysis , Animals , Calcium/analysis , Chorionic Gonadotropin/pharmacology , Cyclic AMP/analysis , Cyclic AMP/metabolism , Cytoplasm/chemistry , DNA Primers , Dose-Response Relationship, Drug , Female , Fluorescent Antibody Technique , Granulosa Cells/chemistry , Oocytes/chemistry , Ovarian Follicle/metabolism , Phosphatidylinositols/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/analysis , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/analysis , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical , Theca Cells/chemistry , Vasoactive Intestinal Peptide/pharmacology
16.
J Cell Physiol ; 206(1): 238-45, 2006 Jan.
Article in English | MEDLINE | ID: mdl-15991248

ABSTRACT

Estrogens have been postulated to exert a detrimental effect on spermatogenesis in vivo. Since mouse male germ cells express estrogen receptors, we have investigated whether molecular pathways are activated by estrogen stimulation of these cells. Our results demonstrate that estrogen receptor beta is expressed in mitotic and meiotic male germ cells as well as in the spermatogonia derived GC-1 cell line. By using this cell line, we show that 17-beta-estradiol triggers activation of a transcriptional response that requires a functional estrogen receptor. Moreover, GC-1 cells respond to estrogens by transiently activating a signal transduction pathway that impinges on the mitogen-activated protein kinases (MAPK) ERK1 and -2. A similar dose-dependent transient activation of ERKs was also observed in primary mouse spermatocytes in culture. Activation by the estrogen was specific because other steroids such as progesterone and dihydrotestosterone were ineffective and because it could be blocked by the selective inhibitor of the ERK pathway and by competitive inhibitors of the estrogen receptor. Finally, we observed that 17-beta-estradiol does not affect spontaneous or induced apoptosis in cultured mouse spermatocytes, indicating that the apoptotic effects observed in vivo require additional testicular components.


Subject(s)
Estradiol/pharmacology , Germ Cells/drug effects , Spermatocytes/drug effects , Animals , Apoptosis/physiology , Cell Line , Cells, Cultured , Enzyme Activation , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, Reporter , Germ Cells/cytology , Germ Cells/physiology , Humans , MAP Kinase Signaling System/physiology , Male , Mice , Spermatocytes/cytology , Spermatocytes/physiology , Transcriptional Activation
17.
Contraception ; 72(4): 291-3, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16181973

ABSTRACT

Phospholipid hydroperoxide glutathione peroxidase (PHGPx) is a unique intracellular enzyme that directly reduces lipid hydroperoxides in membranes and has the ability to use protein thiol groups as donor substrates. Three isoforms of PHGPx have so far been identified, namely, a mitochondrial, a cytosolic and a nuclear variant. This article is focused on recent evidence demonstrating that (1) mitochondrial and nuclear PHGPx isoforms display a different pattern of expression during male germ cell differentiation; (2) different PHGPx isoforms play specific and independent functions during sperm maturation. The data are discussed in light of the idea that PHGPx is a moonlighting protein, changing roles depending on the intracellular localization, expression in a specific cell type and different partners which it interacts with.


Subject(s)
Glutathione Peroxidase/physiology , Spermatogenesis/physiology , Animals , Fertility , Glutathione Peroxidase/genetics , Humans , Isoenzymes/genetics , Isoenzymes/physiology , Male , Phospholipid Hydroperoxide Glutathione Peroxidase , Rats
18.
FASEB J ; 19(14): 2048-50, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16174786

ABSTRACT

Induction of human sperm chemotaxis is an established phenomenon, though signaling systems physiologically involved have not been identified. Recently, it has been demonstrated that RANTES is present in the follicular fluid and that this molecule is a chemoactractant for human spermatozoa. However, the presence of beta-chemokine receptors on human spermatozoa has never been reported. By cytometric, Western blotting and immunofluorescence analysis, we demonstrate the presence of CCR5 and CCR3 on ejaculated spermatozoa from healthy subjects. CCR5 was detected in the periacrosomal region of the sperm surface, whereas CCR3 was also present in the postacrosomal cap. Individual variability was observed on CCR5 and CCR3 positive sperm percentages. Presence of Delta32+/-) mutation was demonstrated in two subjects expressing CCR5 in half of the ejaculated spermatozoa. Our findings represent the missing information in favor of the possibility that beta-chemokines and their receptors are involved in sperm chemotaxis. Identification of molecular mechanisms of sperm chemotaxis may allow us to identify predictive parameters of sperm fertilizing ability in hypofertile or infertile subjects. Finally, both CCR5 and CCR3 expressed on the sperm cell surface may be involved in HIV-1 adhesion to spermatozoa, thus allowing these cells to perform as virion cellular carriers during sexual transmission of HIV-1 infection.


Subject(s)
Receptors, CCR5/biosynthesis , Receptors, Chemokine/biosynthesis , Sperm Motility , Spermatozoa/metabolism , Blotting, Western , Chemotaxis , Ejaculation , Gene Expression Regulation , Genotype , HIV-1/metabolism , Haplotypes , Humans , Male , Microscopy, Fluorescence , Mutation , Receptors, CCR3 , Receptors, CCR5/physiology , Receptors, Chemokine/physiology , Spermatozoa/virology
19.
AIDS ; 19(13): 1424-6, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-16103775

ABSTRACT

Viruses adhering to the sperm surface are described in the semen of HIV-1-infected individuals, although viral adhesion mechanisms have yet to be fully understood. We demonstrate, by cytometric analysis and immunofluorescence microscopy, the presence of beta-chemokine receptor 5 (CCR5) on the periacrosomal region of ejaculated spermatozoa. CCR5 expressed on the sperm cell surface may allow sperm to act as virion cellular carriers during the sexual transmission of HIV-1 infection.


Subject(s)
HIV-1/metabolism , Receptors, CCR5/metabolism , Spermatozoa/metabolism , Flow Cytometry , Humans , Male , Microscopy, Fluorescence , Spermatozoa/virology
20.
Eur J Endocrinol ; 151(4): 511-20, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15476453

ABSTRACT

AIM: The aim of this study was to determine the action of bone morphogenetic proteins (BMPs) on testicular cell proliferation during early postnatal life, a definite developmental time at which crucial changes in germ cell and Sertoli cell maturation occur. METHODS: We investigated the effect of BMP2 and BMP7, two factors which belong to the relatively distant decapentaplegic (DPP) and 60 A classes of the large BMP family, upon spermatogonial and Sertoli cell proliferation, and we examined the expression of activin/BMP type II and type I receptors. We used in vitro cultured testis fragments from 7-day-old mice, highly purified populations of somatic and germ cells and total testes from mice of different ages. Cell proliferation was assessed by BrdU labelling and [3H]-thymidine incorporation. Ribonuclease protection assays and Northern blotting were performed to analyse receptor expression. RESULTS AND CONCLUSIONS: We have demonstrated a stimulatory action of BMP2 and BMP7 in spermatogonia and Sertoli cell proliferation respectively. ActRIIB is the type II receptor expressed most in spermatogonia, whereas Sertoli cells specifically expressed BMPRIIB, in addition to ActRIIB. By contrast, the presence of ActRIIA was undetectable in either germ or somatic cells. The type I receptors ActRIA, ActRIB and BMPRIA were all found in both cell types, indicating that the observed effect of BMP2 and BMP7 on testicular cell proliferation may be mediated by a number of combinatorial interactions in the receptor complexes. These findings suggest that BMPs are involved in physiological paracrine signalling during the first wave of spermatogenesis.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Sertoli Cells/cytology , Spermatogonia/cytology , Transforming Growth Factor beta/metabolism , Activin Receptors/genetics , Activin Receptors/metabolism , Age Factors , Animals , Antimetabolites , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Protein Receptors , Bone Morphogenetic Proteins/pharmacology , Bromodeoxyuridine , Cell Division/drug effects , Cell Division/physiology , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred Strains , Organ Culture Techniques , Paracrine Communication/physiology , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Spermatogonia/drug effects , Spermatogonia/metabolism , Testis/cytology , Testis/growth & development , Thymidine/pharmacokinetics , Transforming Growth Factor beta/pharmacology , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...