Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Methods Mol Biol ; 2770: 37-52, 2024.
Article in English | MEDLINE | ID: mdl-38351445

ABSTRACT

Transcriptomic analyses of germ cells at different stages of differentiation have shed light on the transcriptional and post-transcriptional mechanisms regulating gene expression that ensure the correct progression of spermatogenesis and male fertility. In this chapter, we describe a method to isolate meiotic and post-meiotic germ cells, based on gravimetric sedimentation, starting from a testicular germ cell suspension isolated from a single adult mouse. We also describe how to assess the purity and quality of the collected fractions of germ cells and how to optimize the extraction from these samples of RNA for subsequent RNA-sequencing experiment. In our experience, this protocol is suitable for germ cell isolation and transcriptomic analysis for mouse models with spermatogenic defects, overcoming the limits that reduced fertility poses to the obtaining of experimental animals.


Subject(s)
Spermatogenesis , Testis , Mice , Male , Animals , Spermatogenesis/genetics , Germ Cells , Gene Expression Profiling , RNA/genetics
2.
Cell Death Dis ; 8(10): e3085, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28981118

ABSTRACT

Type 2 cannabinoid receptor (CB2R) has been proposed to promote in vitro meiotic entry of postnatal male germ cells and to maintain the temporal progression of spermatogenesis in vivo. However, no information is presently available on the role played by CB2R in male and female fetal gonads. Here we show that in vitro pharmacological stimulation with JWH133, a CB2R agonist, induced activation of the meiotic program in both male and female fetal gonads. Upon stimulation, gonocytes initiated the meiotic program but became arrested at early stages of prophase I, while oocytes showed an increased rate of meiotic entry and progression toward more advanced stage of meiosis. Acceleration of meiosis in oocytes was accompanied by a strong increase in the percentage of γ-H2AX-positive pachytene and diplotene cells, paralleled by an increase of TUNEL-positive cells, suggesting that DNA double-strand breaks were not correctly repaired during meiosis, leading to oocyte apoptosis. Interestingly, in vivo pharmacological stimulation of CB2R in fetal germ cells through JWH133 administration to pregnant females caused a significant reduction of primordial and primary follicles in the ovaries of newborns with a consequent depletion of ovarian reserve and reduced fertility in adult life, while no alterations of spermatogenesis in the testis of the offspring were detected. Altogether our findings highlight a pro-meiotic role of CB2R in male and female germ cells and suggest that the use of cannabis in pregnant female might represent a risk for fertility and reproductive lifespan in female offspring.


Subject(s)
Meiosis/genetics , Ovarian Follicle/drug effects , Receptor, Cannabinoid, CB2/genetics , Testis/drug effects , Apoptosis/drug effects , Cannabinoids/pharmacology , DNA Breaks, Double-Stranded/drug effects , Female , Fetus , Germ Cells/drug effects , Germ Cells/growth & development , Gonads/drug effects , Gonads/growth & development , Histones/genetics , Humans , Male , Meiosis/drug effects , Oocytes/drug effects , Oocytes/growth & development , Ovarian Follicle/growth & development , Ovarian Reserve/drug effects , Ovarian Reserve/genetics , Pregnancy , Receptor, Cannabinoid, CB2/agonists , Spermatogenesis/drug effects , Testis/growth & development
3.
Sci Rep ; 7(1): 3469, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28615629

ABSTRACT

Cytochrome P450-aromatase catalyzes estrogen biosynthesis from C19 steroids. In the testis, Sertoli cells express P450-aromatase and represent the primary source of estrogen during prepuberal age. This study focused on the effect of simulated microgravity (SM) on aromatase expression in primary mouse Sertoli cells. When cultured in Rotary Cell Culture System (RCCS), Sertoli cells, formed multicellular three dimensional spheroids (3D). Biological properties were first analyzed in terms of viability, cell cycle, expression of cytoskeletal components and growth factors in comparison to Sertoli cells cultured in spheroids at unit gravity (G). SM did not affect cell viability and proliferation, nor expression of the main cytoskeleton proteins and of growth factors like Kit Ligand (KL) and glial derived neurotrophic factor (GDNF). On the other hand, SM caused a strong increase in P450 aromatase mRNA and protein expression. Interestingly, P450-aromatase was no more inducible by 8-Br-cAMP. The presence of a functional aromatase was confirmed by enrichment of 17ß-estradiol released in the medium by androgen precursors. We concluded that SM causes a significant upregulation of aromatase gene expression in Sertoli cells, leading to a consequent increase in 17ß-estradiol secretion. High level of 17ß-estradiol in the testis could have potentially adverse effects on male fertility and testicular cancer.


Subject(s)
Aromatase/genetics , Sertoli Cells/metabolism , Weightlessness , Animals , Aromatase/metabolism , Biomarkers , Cells, Cultured , Male , Mice
4.
Dev Cell ; 41(1): 82-93.e4, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28366282

ABSTRACT

Global transcriptome reprogramming during spermatogenesis ensures timely expression of factors in each phase of male germ cell differentiation. Spermatocytes and spermatids require particularly extensive reprogramming of gene expression to switch from mitosis to meiosis and to support gamete morphogenesis. Here, we uncovered an extensive alternative splicing program during this transmeiotic differentiation. Notably, intron retention was largely the most enriched pattern, with spermatocytes showing generally higher levels of retention compared with spermatids. Retained introns are characterized by weak splice sites and are enriched in genes with strong relevance for gamete function. Meiotic intron-retaining transcripts (IRTs) were exclusively localized in the nucleus. However, differently from other developmentally regulated IRTs, they are stable RNAs, showing longer half-life than properly spliced transcripts. Strikingly, fate-mapping experiments revealed that IRTs are recruited onto polyribosomes days after synthesis. These studies reveal an unexpected function for regulated intron retention in modulation of the timely expression of select transcripts during spermatogenesis.


Subject(s)
Cell Differentiation/genetics , Introns/genetics , Meiosis/genetics , Spermatozoa/cytology , Spermatozoa/metabolism , Alternative Splicing/genetics , Animals , Cell Nucleus/genetics , Gene Ontology , Male , Mice, Inbred C57BL , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Spermatogenesis/genetics , Transcription, Genetic , Transcriptome/genetics
5.
FASEB J ; 30(4): 1453-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26671998

ABSTRACT

Type 2 cannabinoid receptor (CB2) has been proposed to play a pivotal role in meiotic entry of male germ cells, similar to retinoic acid (RA). In this study, we showed that activation of CB2with the specific agonist JWH133 [3-(1',1'-dimethylbutyl)-1-deoxy-8-THC] (IC5010(-6)M) mimics epigenetic events induced by RA (IC5010(-7)M) in spermatogonia. Both JWH133 and RA treatments stimulate the expression of the meiotic genes c-KitandStra8, by up-regulating H3K4me3 and down-regulating H3K9me2 levels in genomic regions flanking the transcription start site. Moreover, both agents increase the expression ofPrdm9, the gene encoding a meiosis-specific histone, H3K4me3 methyltransferase, which marks hotspots of recombination in prophase I, thus resulting in a global increase in H3K4me3. Notably, prolonged administration of JWH133 to immature 7 dpp CD-1 mice induced an acceleration of the onset of spermatogenesis, whereas the specific CB2antagonist delayed germ cell differentiation. Thus, both hyper- and hypostimulation of CB2disrupted the temporal dynamics of the spermatogenic cycle. These findings highlight the importance of proper CB2signaling for the maintenance of a correct temporal progression of spermatogenesis and suggest a possible adverse effect of cannabis in deregulating this process.-Di Giacomo, D., De Domenico, E., Sette, C., Geremia, R., Grimaldi, P. Type 2 cannabinoid receptor contributes to the physiological regulation of spermatogenesis.


Subject(s)
Cell Differentiation/physiology , Receptor, Cannabinoid, CB2/metabolism , Signal Transduction/physiology , Spermatogenesis/physiology , Spermatogonia/physiology , Animals , Blotting, Western , Cannabinoids/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Gene Expression/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Indoles/pharmacology , Lysine/metabolism , Male , Meiosis/drug effects , Meiosis/genetics , Methylation/drug effects , Mice , Microscopy, Fluorescence , Promoter Regions, Genetic/genetics , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Spermatogenesis/drug effects , Spermatogenesis/genetics , Spermatogonia/cytology , Spermatogonia/drug effects , Tretinoin/pharmacology
6.
Front Endocrinol (Lausanne) ; 4: 192, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24379805

ABSTRACT

Spermatogenesis is a complex process in which male germ cells undergo a mitotic phase followed by meiosis and by a morphogenetic process to form mature spermatozoa. Spermatogenesis is under the control of gonadotropins, steroid hormones and it is modulated by a complex network of autocrine and paracrine factors. These modulators ensure the correct progression of germ cell differentiation to form mature spermatozoa. Recently, it has been pointed out the relevance of endocannabinoids as critical modulators of male reproduction. Endocannabinoids are natural lipids able to bind to cannabinoid receptors and whose levels are regulated by specific biosynthetic and degradative enzymes. Together with their receptors and metabolic enzymes, they form the "endocannabinoid system" (ECS). In male reproductive tracts, they affect Sertoli cell activities, Leydig cell proliferation, germ cell differentiation, sperm motility, capacitation, and acrosome reaction. The ECS interferes with the pituitary-gonadal axis, and an intricate crosstalk between ECS and steroid hormones has been highlighted. This mini-review will focus on the involvement of the ECS in the control of spermatogenesis and on the interaction between ECS and steroid hormones.

7.
Cell Mol Life Sci ; 69(24): 4177-90, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22802127

ABSTRACT

Estrogen (E(2)) regulates spermatogenesis, yet its direct target genes have not been identified in the testis. Here, we cloned the proximal 5' flanking region of the mouse fatty acid amide hydrolase (faah) gene upstream of the luciferase reporter gene, and demonstrated its promoter activity and E(2) inducibility in primary mouse Sertoli cells. Specific mutations in the E(2) response elements (ERE) of the faah gene showed that two proximal ERE sequences (ERE2/3) are essential for E(2)-induced transcription, and chromatin immunoprecipitation experiments showed that E(2) induced estrogen receptor ß binding at ERE2/3 sites in the faah promoter in vivo. Moreover, the histone demethylase LSD1 was found to be associated with ERE2/3 sites and to play a role in mediating E(2) induction of FAAH expression. E(2) induced epigenetic modifications at the faah proximal promoter compatible with transcriptional activation by remarkably decreasing methylation of both DNA at CpG site and histone H3 at lysine 9. Finally, FAAH silencing abolished E(2) protection against apoptosis induced by the FAAH substrate anandamide. Taken together, our results identify FAAH as the first direct target of E(2).


Subject(s)
Amidohydrolases/genetics , Estrogens/pharmacology , Gene Expression Regulation , Oxidoreductases, N-Demethylating/physiology , Sertoli Cells/metabolism , Amidohydrolases/chemistry , Amidohydrolases/physiology , Animals , Apoptosis , Base Sequence , DNA Methylation/drug effects , Estrogen Receptor beta/metabolism , Estrogen Receptor beta/physiology , Histone Demethylases , Histones/metabolism , Male , Methylation , Mice , Molecular Sequence Data , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Promoter Regions, Genetic , Sertoli Cells/drug effects
8.
PLoS One ; 7(6): e39729, 2012.
Article in English | MEDLINE | ID: mdl-22745822

ABSTRACT

The chromatoid body (CB) is a unique structure of male germ cells composed of thin filaments that condense into a perinuclear organelle after meiosis. Due to the presence of proteins involved in different steps of RNA metabolism and of different classes of RNAs, including microRNAs (miRNAs), the CB has been recently suggested to function as an RNA processing centre. Herein, we show that the RNA binding protein SAM68 transiently localizes in the CB, in concomitance with the meiotic divisions of mouse spermatocytes. Precise staging of the seminiferous tubules and co-localization studies with MVH and MILI, two well recognized CB markers, documented that SAM68 transiently associates with the CB in secondary spermatocytes and early round spermatids. Furthermore, although SAM68 co-immunoprecipitated with MVH in secondary spermatocytes, its ablation did not affect the proper localization of MVH in the CB. On the other hand, ablation of the CB constitutive component MIWI did not impair association of SAM68 with the CB. Isolation of CBs from Sam68 wild type and knockout mouse testes and comparison of their protein content by mass spectrometry indicated that Sam68 ablation did not cause overall alterations in the CB proteome. Lastly, we found that SAM68 interacts with DROSHA and DICER in secondary spermatocytes and early round spermatids and that a subset of miRNAs were altered in Sam68(-/-) germ cells. These results suggest a novel role for SAM68 in the miRNA pathway during spermatogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Germ Cells/metabolism , MicroRNAs/genetics , RNA-Binding Proteins/metabolism , Spermatids/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Fluorescent Antibody Technique , Germ Cells/ultrastructure , Immunoprecipitation , Male , Mass Spectrometry , Mice , Microscopy, Electron, Transmission , RNA-Binding Proteins/genetics , Real-Time Polymerase Chain Reaction
9.
Endocr Relat Cancer ; 18(5): 541-54, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21712346

ABSTRACT

Pancreatic endocrine tumours (PETs) are rare and heterogeneous neoplasms, often diagnosed at metastatic stage, for which no cure is currently available. Recently, activation of two pathways that support proliferation and invasiveness of cancer cells, the Src family kinase (SFK) and mammalian target of rapamycin (mTOR) pathways, was demonstrated in PETs. Since both pathways represent suitable targets for therapeutic intervention, we investigated their possible interaction in PETs. Western blot and immunofluorescence analyses indicated that SFK and mTOR activity correlate in PET cell lines. We also found that SFKs coordinate cell adhesion and spreading with activation of the mTOR pathway in PET cells. Live cell metabolic labelling and biochemical studies demonstrated that SFK activity enhance mTOR-dependent translation initiation. Furthermore, microarray analysis of the mRNAs associated with polyribosomes revealed that SFKs regulate mTOR-dependent translation of specific transcripts, with an enrichment in mRNAs encoding cell cycle proteins. Importantly, a synergic inhibition of proliferation was observed in PET cells concomitantly treated with SFK and mTOR inhibitors, without activation of the phosphatidylinositol 3-kinase/AKT pro-survival pathway. Tissue microarray analysis revealed activation of Src and mTOR in some PET samples, and identified phosphorylation of 4E-BP1 as an independent marker of poor prognosis in PETs. Thus, our work highlights a novel link between the SFK and mTOR pathways, which regulate the translation of mRNAs for cell cycle regulators, and suggest that crosstalk between these pathways promotes PET cell proliferation.


Subject(s)
Cell Adhesion/physiology , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , src-Family Kinases/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Humans , Neuroendocrine Tumors/enzymology , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , Tissue Array Analysis
10.
Nucleic Acids Res ; 39(12): 4961-74, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21355037

ABSTRACT

Sam68 plays an essential role in mouse spermatogenesis and male fertility. Herein, we report an interaction between Sam68 and the phosphorylated forms of the RNA polymerase II (RNAPII) in meiotic spermatocytes. RNase treatment decreased but did not abolish the interaction, consistently with in vitro binding of RNAPII to the Sam68 carboxyl-terminal region. Sam68 retention in the spermatocyte nucleus was dependent on the integrity of cellular RNAs, suggesting that the protein is recruited to transcriptionally active chromatin. Mouse knockout models characterized by stage-specific arrest of spermatogenesis and staining with the phosphorylated form of RNAPII documented that Sam68 expression is confined to the transcriptionally active stages of spermatogenesis. Furthermore, Sam68 associates with splicing regulators in germ cells and we report that alternative splicing of Sgce exon 8 is regulated in a Sam68-dependent manner during spermatogenesis. RNA and chromatin crosslink immunoprecipitation experiments showed that Sam68 binds in vivo to sequences surrounding the intron 7/exon 8 boundary, thereby affecting the recruitment of the phosphorylated RNAPII and of the general splicing factor U2AF65. These results suggest that Sam68 regulates alternative splicing at transcriptionally active sites in differentiating germ cells and provide new insights into the regulation of Sam68 expression during spermatogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alternative Splicing , RNA-Binding Proteins/metabolism , Spermatogenesis/genetics , Spermatozoa/metabolism , Transcription, Genetic , Animals , Male , Meiotic Prophase I/genetics , Mice , Mice, Knockout , RNA Polymerase II/metabolism , Sarcoglycans/genetics , Sarcoglycans/metabolism , Spermatocytes/enzymology , Spermatocytes/metabolism
11.
J Cell Sci ; 124(Pt 1): 91-9, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21147852

ABSTRACT

Meiosis is a crucial process for the production of functional gametes. However, the biological significance of many genes expressed during the meiotic phase remains poorly understood, mainly because of the lethal phenotypes of the knockout mice. Functional analysis of such genes using the conditional knockout approach is hindered by the lack of suitable Cre transgenic lines. We describe here the generation of transgenic mice expressing Cre recombinase under the control of the meiotic Spo11 gene. Using LacZ-R26(loxP) and EYFP-R26(loxP) reporter mice, we show the specific expression and activity of Cre during meiosis in males and females. Spo11(Cre) mice were then crossed with floxed Nbs1 and JAM-C mice to produce conditional knockouts. A strong reduction of Nbs1 and JAM-C protein levels was found in the testis. Although Nbs1-deleted mice developed minor gonadal abnormalities, JAM-C-knockout mice showed a spermiogenetic arrest, as previously described for the null mice. These results provide strong evidence that Spo11(Cre) transgenic mice represent a powerful tool for deleting genes of interest specifically in meiotic and/or in postmeiotic germ cells.


Subject(s)
Endodeoxyribonucleases/genetics , Gene Deletion , Gene Targeting/methods , Germ Cells/enzymology , Integrases/metabolism , Animals , Endodeoxyribonucleases/metabolism , Female , Germ Cells/cytology , Integrases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Miosis , Promoter Regions, Genetic
12.
Hum Mol Genet ; 19(24): 4886-94, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20881015

ABSTRACT

Sam68 is a multifunctional RNA-binding protein highly expressed in the gonads, whose ablation causes male infertility. Herein, we have investigated Sam68 expression in the adult ovary and its function in female fertility. Immunohistochemistry showed that Sam68 was localized in the nucleus of oocytes and follicular cells at all stages of folliculogenesis. Sam68(-/-) females were severely subfertile, and they showed a delay in the age of first pregnancy, increased breeding time for successful pregnancy and yielded smaller litters. Morphological analyses indicated a significant reduction in the number of secondary and pre-antral follicles in the ovary. These defects were associated with alteration of oestrous cycles and a reduced number of ovulated oocytes, which were only partially restored by the administration of exogenous gonadotropins. Crosslinking/immunoprecipitation experiments showed that Sam68 directly binds the mRNAs for the follicle-stimulating hormone (FSH) and the luteinizing hormone receptors (Fshr and Lhcgr), which were downregulated in ovaries of adult knockout females. Stimulation of immature females with FSH-like pregnant mare serum gonadotropin (PMSG), or of follicular cells with the FSH second messenger analogue 8Br-cAMP, caused the upregulation of Sam68. The increase in Sam68 levels paralleled that of the Fshr and Lhcgr mRNAs in the pre-ovulatory follicle and was required to allow accumulation of these transcripts in follicular cells. These studies identify a new crucial function for Sam68 in the regulation of female fertility and indicate that this protein is required to insure proper expression of the gonadotropin receptor transcripts in pre-ovulatory follicles in adult ovary.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Deletion , Gonadotropins, Equine/pharmacology , Infertility, Female/pathology , Ovarian Follicle/drug effects , Ovarian Follicle/growth & development , RNA-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Estrous Cycle/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Infertility, Female/genetics , Male , Mice , Ovarian Follicle/pathology , Ovarian Follicle/physiopathology , Pregnancy , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Receptors, Gonadotropin/genetics , Receptors, Gonadotropin/metabolism , Up-Regulation/drug effects
13.
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
14.
Biol Reprod ; 83(4): 607-15, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20574055

ABSTRACT

Translation of stored mRNAs accounts for protein synthesis during the transcriptionally inactive stages of spermatogenesis. A key step in mRNA translation is the assembly of the initiation complex EIF4F, which is regulated by the MTOR (mammalian target of rapamycin) and MNK1/2 (MAP kinase-interacting kinase 1 and 2) pathways. We investigated the expression and activity of regulatory proteins of these pathways in male germ cells at different stages of differentiation. All translation factors analyzed were expressed in germ cells throughout spermatogenesis. However, while EIF4G and PABP1 (poly[A]-binding protein 1) were more abundant in postmeiotic cells, MTOR and its target EIF4EBP1 (4E-BP1) decreased steadily during spermatogenesis. In vivo labeling showed that pachytene spermatocytes display higher rates of protein synthesis, which are partially dependent on MTOR and MNK activity. By contrast, haploid spermatids are characterized by lower levels of protein synthesis, which are independent of the activity of these pathways. Accordingly, MTOR and MNK activity enhanced formation of the EIF4F complex in pachytene spermatocytes but not in round spermatids. Moreover, external cues differentially modulated the activity of these pathways in meiotic and haploid cells. Heat shock decreased MTOR and MNK activity in pachytene spermatocytes, whereas round spermatids were much less sensitive. On the other hand, treatment with the phosphatase inhibitor okadaic acid activated MTOR and MNK in both cell types. These results indicate that translational regulation is differentially dependent on the MTOR and MNK pathways in mouse spermatocytes and spermatids and suggest that the late stages of germ cell differentiation display constitutive assembly of the translation initiation complex.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Meiosis/physiology , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , Spermatocytes/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Cell Differentiation/physiology , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-4F/metabolism , Immunohistochemistry , Male , Mice , Okadaic Acid/pharmacology , Protein Biosynthesis , RNA, Messenger/metabolism , Signal Transduction , Spermatocytes/cytology , Spermatocytes/metabolism , Spermatogenesis/physiology
15.
In Vitro Cell Dev Biol Anim ; 46(7): 619-23, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20376706

ABSTRACT

The importance of obtaining stem cells through alternative methods has increased progressively in the recent years due to the potential role that embryonic stem (ES) cells play in the field of regenerative medicine. In this regard, generation of parthenogenetic blastocysts allows the production of ethic-free ES cells without the need to manipulate normal embryos. Our work was aimed at clarifying whether variations in the method adopted to generate diploid parthenogenetic blastocysts could determine differences in the quality of blastocysts produced. In vitro development of mouse oocytes activated with three protocols, using Sr2+ and cytochalasin for different time, was compared with that of in vivo fertilized embryos. We have evaluated the efficiency of blastocyst formation and analysed the expression pattern of the stemness markers OCT4, CDX2, and NANOG. Our results indicate that the yield of diploid parthenogenotes and the segregation of the stemness marker OCT4 in the developing blastocyst are influenced by the parthenogenetic protocol adopted. Particularly, even if all methods tested allowed the production of blastocysts in vitro, the correct segregation of OCT4 occurred only in blastocysts developed from oocytes concomitantly treated for 4 h with Sr2+ and cytochalasin D. Our results indicate that the protocol employed to develop parthenogenetic blastocysts in vitro affects the quality of cells in the inner cell mass.


Subject(s)
Blastocyst/metabolism , Diploidy , Parthenogenesis , Research Embryo Creation/methods , Stem Cells/metabolism , Animals , Biomarkers/metabolism , Blastocyst/cytology , Embryonic Development , Fertilization , Mice , Octamer Transcription Factor-3/metabolism
16.
PLoS One ; 5(2): e9064, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-20140225

ABSTRACT

A critical step of spermatogenesis is the entry of mitotic spermatogonia into meiosis. Progresses on these topics are hampered by the lack of an in vitro culture system allowing mouse spermatogonia differentiation and entry into meiosis. Previous studies have shown that mouse pachytene spermatocytes cultured in simulated microgravity (SM) undergo a spontaneous meiotic progression. Here we report that mouse mitotic spermatogonia cultured under SM with a rotary cell culture system (RCCS) enter into meiosis in the absence of any added exogenous factor or contact with somatic cells. We found that isolated Kit-positive spermatogonia under the RCCS condition enter into the prophase of the first meiotic division (leptotene stage), as monitored by chromosomal organization of the synaptonemal complex 3 protein (Scp3) and up-regulation of several pro-meiotic genes. SM was found to activate the phosphatidyl inositol 3 kinase (PI3K) pathway and to induce in Kit-positive spermatogonia the last round of DNA replication, typical of the preleptotene stage. A PI3K inhibitor abolished Scp3 induction and meiotic entry stimulated by RCCS conditions. A positive effect of SM on germ cell differentiation was also observed in undifferentiated (Kit-negative) spermatogonia, in which RCCS conditions stimulate the expression of Kit and Stra8. In conclusion, SM is an artificial environmental condition which promotes postnatal male germ cell differentiation and might provide a tool to study the molecular mechanisms underlying the switch from mitosis to meiosis in mammals.


Subject(s)
Cell Differentiation/physiology , Meiosis/physiology , Spermatogonia/metabolism , Weightlessness Simulation , Animals , Blotting, Western , Cell Culture Techniques , Cell Differentiation/genetics , Cells, Cultured , DNA/biosynthesis , Enzyme Activation , Male , Meiosis/genetics , Mice , Microscopy, Confocal , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Spermatocytes/cytology , Spermatocytes/metabolism , Spermatogonia/cytology
17.
J Cell Sci ; 123(Pt 6): 871-80, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20159962

ABSTRACT

In the mouse, three genes that are homologous to the Drosophila Nanos (Nos) gene have been identified. Deletion of one of these genes, Nanos2, results in male sterility, owing to loss of germ cells during fetal life. Before apoptosis, Nanos2-null gonocytes enter meiosis, suggesting that Nanos2 functions as a meiotic repressor. Here, we show that Nanos2 is continuously expressed in male germ cells from fetal gonocytes to postnatal spermatogonial stem cells. We observed that the promeiotic factor AtRA, an analog of retinoic acid (RA), downregulates NANOS2 levels, in both fetal and postnatal gonocytes, while promoting meiosis. Interestingly, FGF9, a growth factor crucial for sex differentiation and survival of fetal gonocytes, upregulates levels of NANOS2 in both male and female primordial germ cells (PGCs) and in premeiotic spermatogonia. This effect was paralleled by an impairment of meiotic entry, suggesting that FGF9 acts as an inhibitor of meiosis through the upregulation of Nanos2. We found that NANOS2 interacts with PUM2, and that these two proteins colocalize in the ribonucleoparticle and polysomal fractions on sucrose gradients, supporting the notion that they bind RNA. Finally, we found that recombinant NANOS2 binds to two spermatogonial mRNAs, Gata2 and Taf7l, which are involved in germ-cell differentiation.


Subject(s)
Carrier Proteins/genetics , Fibroblast Growth Factor 9/pharmacology , Germ Cells/cytology , Germ Cells/metabolism , Meiosis/drug effects , Tretinoin/pharmacology , Animals , Animals, Newborn , Carrier Proteins/metabolism , Down-Regulation/drug effects , Female , Fetus/cytology , Fetus/drug effects , Fetus/metabolism , Gene Expression Regulation, Developmental/drug effects , Germ Cells/drug effects , Male , Mice , Ovum/cytology , Ovum/drug effects , Ovum/metabolism , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Ribonucleoproteins/metabolism , Spermatozoa/cytology , Spermatozoa/drug effects , Spermatozoa/metabolism , Testis/cytology , Testis/drug effects , Testis/metabolism , Up-Regulation/drug effects
18.
Nucleic Acids Res ; 38(9): 3005-18, 2010 May.
Article in English | MEDLINE | ID: mdl-20110258

ABSTRACT

DNA-damaging agents cause a multifaceted cellular stress response. Cells set in motion either repair mechanisms or programmed cell death pathways, depending on the extent of the damage and on their ability to withstand it. The RNA-binding protein (RBP) Sam68, which is up-regulated in prostate carcinoma, promotes prostate cancer cell survival to genotoxic stress. Herein, we have investigated the function of Sam68 in this cellular response. Mitoxantrone (MTX), a topoisomerase II inhibitor, induced relocalization of Sam68 from the nucleoplasm to nuclear granules, together with several other RBPs involved in alternative splicing, such as TIA-1, hnRNP A1 and the SR proteins SC35 and ASF/SF2. Sam68 accumulation in nuclear stress granules was independent of signal transduction pathways activated by DNA damage. Using BrU labelling and immunofluorescence, we demonstrate that MTX-induced nuclear stress granules are transcriptionally active foci where Sam68 and the phosphorylated form of RNA polymerase II accumulate. Finally, we show that MTX-induced relocalization of Sam68 correlates with changes in alternative splicing of its mRNA target CD44, and that MTX-induced CD44 splicing depends on Sam68 expression. These results strongly suggest that Sam68 is part of a RNA-mediated stress response of the cell that modulates alternative splicing in response to DNA damage.


Subject(s)
Adaptor Proteins, Signal Transducing/analysis , Alternative Splicing , Antineoplastic Agents/toxicity , Chromatin/genetics , DNA Damage , DNA-Binding Proteins/analysis , RNA-Binding Proteins/analysis , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cell Line , Cell Nucleus/chemistry , Cytoplasm/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Male , Mitoxantrone/toxicity , Mutagens/toxicity , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Structure, Tertiary , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription, Genetic
19.
Methods Mol Biol ; 558: 299-321, 2009.
Article in English | MEDLINE | ID: mdl-19685332

ABSTRACT

The studies of molecular events that occur in single cell types within a tissue often require the disaggregation of the tissue into a single cell suspension, followed by isolation of distinct cell populations. The germinal epithelium of mammals is composed of several cell types, which divide mitotically, before entering meiosis. In this chapter, we describe the isolation of five mouse germ-cell fractions by centrifugal elutriation, and characterize them by their DNA content (flow cytometry), cell morphology (DAPI staining of nuclei, Giemsa staining of squashed cells) and deposition of stage-specific meiotic markers (SYCP3, H1t, SAM68) on chromosome spreads and whole cells. Within 2 h it is possible to obtain enriched populations of elongated spermatids (up to approximately 50% of the fraction), round spermatids (up to approximately 80%), primary spermatocytes (up to approximately 89%), and secondary spermatocytes (up to approximately 17%). Furthermore, most of the collected spermatocytes of the primary spermatocyte fraction are in early-mid pachytene stage as judged by chromosome spreads, enriched up to approximately 89%. Elutriation and techniques used for characterization of germ cell fractions are described.


Subject(s)
Cell Separation/methods , Fractionation, Field Flow/methods , Germ Cells/cytology , Animals , Centrifugation/instrumentation , Centrifugation/methods , Fractionation, Field Flow/instrumentation , Male , Mice , Models, Biological
20.
Proc Natl Acad Sci U S A ; 106(27): 11131-6, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19541620

ABSTRACT

The exact role of the endocannabinoid system (ECS) during spermatogenesis has not been clarified. We used purified germ cell fractions representative of all phases of spermatogenesis and primary cultures of spermatogonia. This approach allowed the precise quantification of the cannabinoid receptor ligands, anandamide and 2-arachidonoylglycerol, and of the expression at transcriptional and transductional levels of their metabolic enzymes and receptors. Our data indicate that male mouse germ cells possess an active and complete ECS, which is modulated during meiosis, and suggest the presence of an autocrine endocannabinoid signal during spermatogenesis. Mitotic cells possess higher levels of 2-arachidonoylglycerol, which decrease in spermatocytes and spermatids. Accordingly, spermatogonia express higher and lower levels of 2-arachidonoylglycerol biosynthetic and degrading enzymes, respectively, as compared to meiotic and postmeiotic cells. This endocannabinoid likely plays a pivotal role in promoting the meiotic progression of germ cells by activating CB(2) receptors. In fact, we found that the selective CB(2) receptor agonist, JWH133, induced the Erk 1/2 MAPK phosphorylation cascade in spermatogonia and their progression toward meiosis, because it increased the number of cells positive for SCP3, a marker of meiotic prophase, and the expression of early meiotic prophase genes.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Receptor, Cannabinoid, CB2/metabolism , Spermatogenesis , Animals , Arachidonic Acids/biosynthesis , Cannabinoid Receptor Modulators/biosynthesis , Cannabinoids/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Fluorescent Antibody Technique , Glycerides/biosynthesis , MAP Kinase Signaling System/drug effects , Male , Meiotic Prophase I/drug effects , Mice , Polyunsaturated Alkamides , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/agonists , Spermatogenesis/drug effects , Spermatogonia/cytology , Spermatogonia/drug effects , Spermatogonia/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...