Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 261: 115821, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37776573

ABSTRACT

Reported here are the synthesis and in vitro evaluation of a series of 26 retinoic acid analogs based on dihydronaphthalene and chromene scaffolds using a transactivation assay. Chromene amide analog 21 was the most potent and selective retinoic acid receptor α antagonist identified from this series. In vitro evaluation indicated that 21 has favorable physicochemical properties and a favorable pharmacokinetic PK profile in vivo with significant oral bioavailability, metabolic stability, and testes exposure. Compound 21 was evaluated for its effects on spermatogenesis and disruption of fertility in a mouse model. Oral administration of compound 21 at low doses showed reproducibly characteristic albeit modest effects on spermatogenesis, but no effects on fertility were observed in mating studies. The inhibition of spermatogenesis could not be enhanced by raising the dose and lengthening the duration of dosing. Thus, 21 may not be a good candidate to pursue further for effects on male fertility.


Subject(s)
Contraception , Testis , Mice , Animals , Male , Retinoic Acid Receptor alpha/metabolism , Benzopyrans/pharmacology
2.
Arch Pharm (Weinheim) ; 356(7): e2300031, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37154197

ABSTRACT

Retinoic acid receptor alpha (RARα) antagonist ER-50891 and 15 analogs were prepared and tested in vitro for potency and selectivity at RARα, RARß, and RARγ using transactivation assays. Minor modifications to the parent molecule such as the introduction of a C4 tolyl group in place of the C4 phenyl group on the quinoline moiety slightly increased the RARα selectivity but larger substituents significantly decreased the potency. Replacement of the pyrrole moiety of ER-50891 with triazole, amides, or a double bond produced inactive compounds. ER-50891 was found to be stable in male mouse liver microsomes and was tested in male mice to assess its effects on spermatogenesis. Characteristic, albeit modest and transient, effects on spermatogenesis were observed.


Subject(s)
Contraception , Male , Mice , Animals , Retinoic Acid Receptor alpha , Structure-Activity Relationship
3.
Mol Reprod Dev ; 88(2): 141-157, 2021 02.
Article in English | MEDLINE | ID: mdl-33469999

ABSTRACT

BRDT, a member of the BET family of double bromodomain-containing proteins, is essential for spermatogenesis in the mouse and has been postulated to be a key regulator of transcription in meiotic and post-meiotic cells. To understand the function of BRDT in these processes, we first characterized the genome-wide distribution of the BRDT binding sites, in particular within gene units, by ChIP-Seq analysis of enriched fractions of pachytene spermatocytes and round spermatids. In both cell types, BRDT binding sites were mainly located in promoters, first exons, and introns of genes. BRDT binding sites in promoters overlapped with several histone modifications and histone variants associated with active transcription, and were enriched for consensus sequences for specific transcription factors, including MYB, RFX, ETS, and ELF1 in pachytene spermatocytes, and JunD, c-Jun, CRE, and RFX in round spermatids. Subsequent integration of the ChIP-seq data with available transcriptome data revealed that stage-specific gene expression programs are associated with BRDT binding to their gene promoters, with most of the BDRT-bound genes being upregulated. Gene Ontology analysis further identified unique sets of genes enriched in diverse biological processes essential for meiosis and spermiogenesis between the two cell types, suggesting distinct developmentally stage-specific functions for BRDT. Taken together, our data suggest that BRDT cooperates with different transcription factors at distinctive chromatin regions within gene units to regulate diverse downstream target genes that function in male meiosis and spermiogenesis.


Subject(s)
Epigenomics , Gene Expression Regulation, Developmental , Nuclear Proteins/physiology , Spermatogenesis/genetics , Transcription Factors/physiology , Animals , Binding Sites , Chromatin Immunoprecipitation Sequencing , DNA/metabolism , Male , Meiosis/genetics , Meiosis/physiology , Mice , Promoter Regions, Genetic , Spermatids/physiology , Spermatogenesis/physiology
4.
Biol Reprod ; 103(2): 390-399, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32671394

ABSTRACT

Retinoic acid receptor alpha (RARA), a nuclear receptor protein, has been validated as a target for male contraception by gene knockout studies and also pharmacologically using a pan-retinoic acid receptor antagonist. Retinoic acid receptor alpha activity is indispensable for the spermatogenic process, and therefore its antagonists have potential as male contraceptive agents. This review discusses the effects of systematic dosing regimen modifications of the orally bioavailable and reversible pan-antagonist BMS-189453 as well as studies with the alpha-selective antagonists BMS-189532 and BMS-189614 in a murine model. We also provide an overview of structure-activity studies of retinoic acid receptor alpha antagonists that provide insight for the design of novel alpha-selective ligands.


Subject(s)
Contraception/methods , Receptors, Retinoic Acid/antagonists & inhibitors , Spermatogenesis/drug effects , Animals , Humans , Mice , Signal Transduction/drug effects
5.
Biol Reprod ; 103(2): 378-389, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32678439

ABSTRACT

Spermiation is a multiple-step process involving profound cellular changes in both spermatids and Sertoli cells. We have observed spermiation defects, including abnormalities in spermatid orientation, translocation and release, in mice deficient in the retinoic acid receptor alpha (RARA) and upon treatment with a pan-RAR antagonist. To elucidate the role of retinoid signaling in regulating spermiation, we first characterized the time course of appearance of spermiogenic defects in response to treatment with the pan-RAR antagonist. The results revealed that defects in spermiation are indeed among the earliest abnormalities in spermatogenesis observed upon inhibition of retinoid signaling. Using fluorescent dye-conjugated phalloidin to label the ectoplasmic specialization (ES), we showed for the first time that these defects involved improper formation of filamentous actin (F-actin) bundles in step 8-9 spermatids and a failure of the actin-surrounded spermatids to move apically to the lumen and to disassemble the ES. The aberrant F-actin organization is associated with diminished nectin-3 expression in both RARA-deficient and pan-RAR antagonist-treated testes. An abnormal localization of both tyrosinated and detyrosinated tubulins was also observed during spermatid translocation in the seminiferous epithelium in drug-treated testes. These results highlight a crucial role of RAR receptor-mediated retinoid signaling in regulating microtubules and actin dynamics in the cytoskeleton rearrangements, required for proper spermiation. This is critical to understand in light of ongoing efforts to inhibit retinoid signaling as a novel approach for male contraception and may reveal spermiation components that could also be considered as new targets for male contraception.


Subject(s)
Actins/metabolism , Retinoids/metabolism , Signal Transduction/physiology , Spermatids/metabolism , Spermatogenesis/physiology , Testis/metabolism , Actin Cytoskeleton/metabolism , Animals , Male , Mice , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism
6.
Cell Cycle ; 19(13): 1654-1664, 2020 07.
Article in English | MEDLINE | ID: mdl-32420805

ABSTRACT

In mammals, male gonocytes are derived from primordial germ cells during embryogenesis, enter a period of mitotic proliferation, and then become quiescent until birth. After birth, the gonocytes proliferate and migrate from the center of testicular cord toward the basement membrane to form the pool of spermatogonial stem cells (SSCs) and establish the SSC niche architecture. However, the molecular mechanisms underlying gonocyte proliferation, migration and differentiation are largely unknown. Cyclin A2 is a key component of the cell cycle and required for cell proliferation. Here, we show that cyclin A2 is required in mouse male gonocyte development and the establishment of spermatogenesis in the neonatal testis. Loss of cyclin A2 function in embryonic gonocytes by targeted gene disruption affected the regulation of the male gonocytes to SSC transition, resulting in the disruption of SSC pool formation, imbalance between SSC self-renewal and differentiation, and severely abnormal spermatogenesis in the adult testis.


Subject(s)
Cell Differentiation , Cyclin A2/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , Aging/pathology , Animals , Animals, Newborn , Basement Membrane/metabolism , Cell Count , Cell Movement , Cyclin A2/genetics , Embryo, Mammalian/cytology , Gene Deletion , Gene Expression Regulation, Developmental , Male , Mice , Models, Biological , Phenotype , Spermatogenesis , Stem Cells/cytology , Testis/pathology
7.
Biol Reprod ; 101(3): 591-601, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31078132

ABSTRACT

Cyclins and cyclin-dependent kinases (CDKs) are key regulators of the cell cycle. Most of our understanding of their functions has been obtained from studies in single-cell organisms and mitotically proliferating cultured cells. In mammals, there are more than 20 cyclins and 20 CDKs. Although genetic ablation studies in mice have shown that most of these factors are dispensable for viability and fertility, uncovering their functional redundancy, CCNA2, CCNB1, and CDK1 are essential for embryonic development. Cyclin/CDK complexes are known to regulate both mitotic and meiotic cell cycles. While some mechanisms are common to both types of cell divisions, meiosis has unique characteristics and requirements. During meiosis, DNA replication is followed by two successive rounds of cell division. In addition, mammalian germ cells experience a prolonged prophase I in males or a long period of arrest in prophase I in females. Therefore, cyclins and CDKs may have functions in meiosis distinct from their mitotic functions and indeed, meiosis-specific cyclins, CCNA1 and CCNB3, have been identified. Here, we describe recent advances in the field of cyclins and CDKs with a focus on meiosis and early embryogenesis.


Subject(s)
Cyclin-Dependent Kinases/physiology , Cyclins/physiology , Gametogenesis/genetics , Germ Cells/physiology , Animals , Embryo, Mammalian , Female , Humans , Male , Mammals , Meiosis , Mice
8.
PLoS Genet ; 14(3): e1007209, 2018 03.
Article in English | MEDLINE | ID: mdl-29513658

ABSTRACT

The double bromodomain and extra-terminal domain (BET) proteins are critical epigenetic readers that bind to acetylated histones in chromatin and regulate transcriptional activity and modulate changes in chromatin structure and organization. The testis-specific BET member, BRDT, is essential for the normal progression of spermatogenesis as mutations in the Brdt gene result in complete male sterility. Although BRDT is expressed in both spermatocytes and spermatids, loss of the first bromodomain of BRDT leads to severe defects in spermiogenesis without overtly compromising meiosis. In contrast, complete loss of BRDT blocks the progression of spermatocytes into the first meiotic division, resulting in a complete absence of post-meiotic cells. Although BRDT has been implicated in chromatin remodeling and mRNA processing during spermiogenesis, little is known about its role in meiotic processes. Here we report that BRDT is an essential regulator of chromatin organization and reprograming during prophase I of meiosis. Loss of BRDT function disrupts the epigenetic state of the meiotic sex chromosome inactivation in spermatocytes, affecting the synapsis and silencing of the X and Y chromosomes. We also found that BRDT controls the global chromatin organization and histone modifications of the chromatin attached to the synaptonemal complex. Furthermore, the homeostasis of crossover formation and localization during pachynema was altered, underlining a possible epigenetic mechanism by which crossovers are regulated and differentially established in mammalian male genomes. Our observations reveal novel findings about the function of BRDT in meiosis and provide insight into how epigenetic regulators modulate the progression of male mammalian meiosis and the formation of haploid gametes.


Subject(s)
Chromatin/genetics , Epigenesis, Genetic/genetics , Meiosis/physiology , Nuclear Proteins/genetics , Sex Chromosomes/genetics , Animals , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromosome Pairing/genetics , Crossing Over, Genetic , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Infertility, Male/genetics , Male , Mice, Knockout , Nuclear Proteins/metabolism , Spermatocytes/pathology , Spermatocytes/physiology , Testis/cytology , Testis/physiology
9.
PLoS One ; 12(3): e0173926, 2017.
Article in English | MEDLINE | ID: mdl-28301569

ABSTRACT

Cyclin A1 (Ccna1), a member of the mammalian A type cyclins, is most abundantly expressed in spermatocytes and is essential for spermatogenesis in the mouse. Ccna1- deficient spermatocytes arrest at late meiotic prophase and undergo apoptosis. To further delineate the mechanisms and key factors involved in this process, we have examined changes in expression of genes involved in both intrinsic and extrinsic signaling pathways that trigger apoptosis in the mutant spermatocytes. Our results show that both pathways are involved, and that the factors involved in the intrinsic pathway were expressed earlier than those involved in the extrinsic pathway. We have also begun to identify in vivo Ccna1-interacting proteins, using an unbiased biochemical approach, and identified 14-3-3, a key regulator of apoptosis, as a Ccna1-interacting protein. Expression levels of 14-3-3 proteins remain unchanged between wild type and mutant testes but there were differences in the subcellular distribution. In wild type control, 14-3-3 is detected in both cytosolic and nuclear fractions whereas it is restricted to the cytoplasm in mutant testes. This differential distribution of 14-3-3 may contribute to the induction of apoptosis in Ccna1-deficient spermatocytes. These results provide insight into the apoptotic mechanisms and pathways that are triggered when progression through the meiotic cell cycle is defective in male gametogenesis.


Subject(s)
14-3-3 Proteins/metabolism , Apoptosis , Cyclin A1/genetics , Meiosis , Signal Transduction , Spermatocytes/metabolism , Animals , Female , Male , Mice , Mice, Transgenic , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
10.
Endocrinology ; 157(4): 1601-12, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26812157

ABSTRACT

We have previously shown that oral administration of a pan-retinoic acid receptor antagonist in mice daily at 2.5 mg/kg for 4 weeks reversibly inhibited spermatogenesis, with no detectable side effects. To elucidate the lowest dose and the longest dosing regimen that inhibits spermatogenesis but results in complete restoration of fertility upon cessation of administration of the drug, we examined the effects of daily doses as low as 1.0 mg/kg with dosing periods of 4, 8, and 16 weeks. We observed 100% sterility in all regimens, with restoration of fertility upon cessation of the drug treatment even for as long as 16 weeks. There was no change in testosterone levels in these males and the progeny examined from 2 of the recovered males were healthy and fertile, with normal testicular weight and testicular histology. Strikingly, a more rapid recovery, as assessed by mating studies, was observed at the lower dose and longer dosing periods. Insight into possible mechanisms underlying this rapid recovery was obtained at 2 levels. First, histological examination revealed that spermatogenesis was not as severely disrupted at the lower dose and with the longer treatment regimens. Second, gene expression analysis revealed that the more rapid recovery may involve the interplay of ATP-binding cassette efflux and solute carrier influx transporters in the testes.


Subject(s)
Membrane Transport Proteins/genetics , Receptors, Retinoic Acid/antagonists & inhibitors , Retinoids/pharmacology , Spermatogenesis/drug effects , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Administration, Oral , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dose-Response Relationship, Drug , Female , Fertility/drug effects , Fertility/physiology , Gene Expression/drug effects , Immunohistochemistry , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Mice , Microscopy, Fluorescence , Organ Size/drug effects , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Cation Transport Proteins , Receptors, Retinoic Acid/metabolism , Retinoids/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Spermatogenesis/physiology , Symporters , Testis/anatomy & histology , Testis/metabolism , Testis/physiology , Testosterone/metabolism , Time Factors
11.
J Cell Biochem ; 117(6): 1429-38, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26565999

ABSTRACT

The expression of BRDT, a member of the BET sub-family of double bromodomain-containing proteins, is restricted to the male germ line, specifically to pachytene-diplotene spermatocytes and early spermatids. We previously showed that loss of the first bromodomain of BRDT by targeted mutagenesis (Brdt(ΔBD1) ) resulted in sterility and abnormalities in spermiogenesis, but little is known about BRDT's function at the molecular level. As part of studies designed to identify BRDT-interacting proteins we stably introduced a FLAG-tagged BRDT cDNA into 293T cells, which do not normally express BRDT. Affinity-purification of FLAG-tagged BRDT complexes indicated that BRDT has novel interactions with the histone deacetylase HDAC1, the arginine-specific histone methyltransferase 5 PRMT5, and the Tripartite motif-containing 28 protein TRIM28. Immunofluorescent microscopy revealed that BRDT co-localized with each of these proteins in round spermatids and co-immunoprecipitation of testicular extracts showed that these proteins interact with BRDT. Furthermore, they bind the promoter of H1t, a putative target of BRDT-containing complexes. This binding of H1t was lost in mice expressing the Brdt(ΔBD1) mutant protein and concomitantly, H1t expression was elevated in round spermatids. Our study reveals a role for BRDT-containing complexes in the repression of gene expression in vivo that correlates with dramatic effects on chromatin remodeling and the progression of spermiogenesis.


Subject(s)
Histone Deacetylase 1/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Spermatogenesis , Transcription, Genetic , Animals , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , Multiprotein Complexes/metabolism , Nuclear Proteins/genetics , Protein-Arginine N-Methyltransferases/metabolism , Spermatids/metabolism
12.
Chromosoma ; 125(2): 253-64, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26712234

ABSTRACT

We have shown that E-type cyclins are key regulators of mammalian male meiosis. Depletion of cyclin E2 reduced fertility in male mice due to meiotic defects, involving abnormal pairing and synapsis, unrepaired DNA, and loss of telomere structure. These defects were exacerbated by additional loss of cyclin E1, and complete absence of both E-type cyclins produces a meiotic catastrophe. Here, we investigated the involvement of E-type cyclins in maintaining telomere integrity in male meiosis. Spermatocytes lacking cyclin E2 and one E1 allele (E1+/-E2-/-) displayed a high rate of telomere abnormalities but can progress to pachytene and diplotene stages. We show that their telomeres exhibited an aberrant DNA damage repair response during pachynema and that the shelterin complex proteins TRF2 and RAP2 were significantly decreased in the proximal telomeres. Moreover, the insufficient level of these proteins correlated with an increase of γ-H2AX foci in the affected telomeres and resulted in telomere associations involving TRF1 and telomere detachment in later prophase-I stages. These results suggest that E-type cyclins are key modulators of telomere integrity during meiosis by, at least in part, maintaining the balance of shelterin complex proteins, and uncover a novel role of E-type cyclins in regulating chromosome structure during male meiosis.


Subject(s)
Cyclin E/metabolism , Cyclins/metabolism , Meiosis , Oncogene Proteins/metabolism , Spermatocytes/cytology , Telomere/metabolism , Animals , Cyclin E/genetics , Cyclins/genetics , DNA Damage , DNA Repair , Female , Male , Mice , Oncogene Proteins/genetics , Pachytene Stage , Spermatocytes/metabolism , Telomere/genetics , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , rap GTP-Binding Proteins/genetics , rap GTP-Binding Proteins/metabolism
13.
Cell Cycle ; 14(12): 1948-60, 2015.
Article in English | MEDLINE | ID: mdl-25785996

ABSTRACT

It remains poorly understood how the haematopoietic stem/progenitor cells (HSPC) are attracted to their niches and the functional consequences of such interaction. In the present study, we show that the cell cycle regulator cyclin A1 in association with vascular endothelial growth factor receptor 1 (VEGFR1), is required for HSPC and their niches to maintain their function and proper interaction. In the absence of cyclin A1, the HSPC in the BM are increased in their frequency and display an increased migratory and homing ability. Concomitantly, the ability of the endosteal and central BM niche zones to attract and home the wild-type HSPC is significantly reduced in cyclin A1-null mice as compared to the wild-type controls. The impaired proliferation and homing of HSPC in the BM of cyclin A1-null mice are attributed to the increased density of microvessels in the endosteal and central BM niche zones, which is associated with the increased VEGFR1 expression. Thus, modulation of cyclin A1 and VEGFR1 in HSPC and their niches may provide new insights into therapeutic approaches.


Subject(s)
Cyclin A1/metabolism , Hematopoietic Stem Cells/cytology , Stem Cell Niche/physiology , Animals , Bone Marrow Transplantation , Cell Membrane/metabolism , Cell Movement , Cell Proliferation , Cell Separation , Female , Fibronectins/metabolism , Genotype , Homozygote , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Vascular Endothelial Growth Factor Receptor-1/metabolism
14.
J Cell Sci ; 127(Pt 11): 2554-64, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24695857

ABSTRACT

Bromodomain-containing protein 2 (Brd2) is a BET family chromatin adaptor required for expression of cell-cycle-associated genes and therefore involved in cell cycle progression. Brd2 is expressed in proliferating neuronal progenitors, displays cell-cycle-stimulating activity and, when overexpressed, impairs neuronal differentiation. Paradoxically, Brd2 is also detected in differentiating neurons. To shed light on the role of Brd2 in the transition from cell proliferation to differentiation, we had previously looked for proteins that interacted with Brd2 upon induction of neuronal differentiation. Surprisingly, we identified the growth factor pleiotrophin (Ptn). Here, we show that Ptn antagonized the cell-cycle-stimulating activity associated with Brd2, thus enhancing induced neuronal differentiation. Moreover, Ptn knockdown reduced neuronal differentiation. We analyzed Ptn-mediated antagonism of Brd2 in a cell differentiation model and in two embryonic processes associated with the neural tube: spinal cord neurogenesis and neural crest migration. Finally, we investigated the mechanisms of Ptn-mediated antagonism and determined that Ptn destabilizes the association of Brd2 with chromatin. Thus, Ptn-mediated Brd2 antagonism emerges as a modulation system accounting for the balance between cell proliferation and differentiation in the vertebrate nervous system.


Subject(s)
Carrier Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cytokines/metabolism , Neural Crest/physiology , Neurons/physiology , Spinal Cord/physiology , Animals , Carrier Proteins/genetics , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Chromosomal Proteins, Non-Histone/genetics , Cytokines/genetics , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice , Neurogenesis/genetics , Protein Binding/genetics , Protein Engineering , RNA, Small Interfering/genetics , Transcription Factors
15.
PLoS Genet ; 10(2): e1004165, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586195

ABSTRACT

Loss of function of cyclin E1 or E2, important regulators of the mitotic cell cycle, yields viable mice, but E2-deficient males display reduced fertility. To elucidate the role of E-type cyclins during spermatogenesis, we characterized their expression patterns and produced additional deletions of Ccne1 and Ccne2 alleles in the germline, revealing unexpected meiotic functions. While Ccne2 mRNA and protein are abundantly expressed in spermatocytes, Ccne1 mRNA is present but its protein is detected only at low levels. However, abundant levels of cyclin E1 protein are detected in spermatocytes deficient in cyclin E2 protein. Additional depletion of E-type cyclins in the germline resulted in increasingly enhanced spermatogenic abnormalities and corresponding decreased fertility and loss of germ cells by apoptosis. Profound meiotic defects were observed in spermatocytes, including abnormal pairing and synapsis of homologous chromosomes, heterologous chromosome associations, unrepaired double-strand DNA breaks, disruptions in telomeric structure and defects in cyclin-dependent-kinase 2 localization. These results highlight a new role for E-type cyclins as important regulators of male meiosis.


Subject(s)
Cyclin E/genetics , Cyclin-Dependent Kinase 2/genetics , Cyclins/biosynthesis , Oncogene Proteins/genetics , Animals , Chromosome Pairing/genetics , Cyclin E/biosynthesis , Cyclin-Dependent Kinase 2/metabolism , Cyclins/genetics , DNA Breaks, Double-Stranded , Gene Expression Regulation, Developmental , Humans , Male , Meiosis , Mice , Oncogene Proteins/biosynthesis , Spermatocytes/metabolism , Spermatogenesis/genetics , Telomere/genetics , Testis/metabolism
16.
ACS Med Chem Lett ; 4(5): 446-450, 2013 May 09.
Article in English | MEDLINE | ID: mdl-24040487

ABSTRACT

Oral administration of a retinoic acid receptor (RAR) pan-antagonist reversibly inhibits spermatogenesis. Given the importance of RARα in regulating spermatogenesis, we identified two RARα-selective antagonists by transactivation and transactivation competition assays and asked whether they effectively inhibit spermatogenesis. Although these two antagonists were potent in vitro, they displayed poor in vivo activity in mice when administered orally. Testicular weights were normal and morphological analysis revealed normal spermatid alignment and sperm release. In vitro drug property analyses were performed with one of these antagonists and compared with the pan-antagonist. We showed that the discrepancies may be explained by several factors, including high plasma protein binding, faster hepatic metabolism relative to the pan-antagonist, and only moderate permeability. The conclusion of poor oral bioavailability was supported by more pronounced defects in mice when the antagonist was administered intravenously versus intraperitoneally. These results are crucial for designing new RARα-selective antagonists for pharmaceutical application.

17.
Cell Cycle ; 12(19): 3135-45, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24013428

ABSTRACT

The RAD9A-RAD1-HUS1 (9-1-1) complex is a PCNA-like heterotrimeric clamp that binds damaged DNA to promote cell cycle checkpoint signaling and DNA repair. While various 9-1-1 functions in mammalian somatic cells have been established, mounting evidence from lower eukaryotes predicts critical roles in meiotic germ cells as well. This was investigated in 2 recent studies in which the 9-1-1 complex was disrupted specifically in the mouse male germline through conditional deletion of Rad9a or Hus1. Loss of these clamp subunits led to severely impaired fertility and meiotic defects, including faulty DNA double-strand break repair. While 9-1-1 is critical for ATR kinase activation in somatic cells, these studies did not reveal major defects in ATR checkpoint pathway signaling in meiotic cells. Intriguingly, this new work identified separable roles for 9-1-1 subunits, namely RAD9A- and HUS1-independent roles for RAD1. Based on these studies and the high-level expression of the paralogous proteins RAD9B and HUS1B in testis, we propose a model in which multiple alternative 9-1-1 clamps function during mammalian meiosis to ensure genome maintenance in the germline.


Subject(s)
Cell Cycle Proteins/metabolism , Exonucleases/metabolism , Meiosis , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , DNA Repair , Fertility , Germ Cells/metabolism , Male , Mice , Recombination, Genetic , Signal Transduction
18.
Int J Dev Biol ; 57(2-4): 159-68, 2013.
Article in English | MEDLINE | ID: mdl-23784826

ABSTRACT

Cyclins are key regulators of the mammalian cell cycle, functioning primarily in concert with their catalytic partners, the cyclin-dependent kinases (Cdks). While their function during mitosis in somatic cells has been extensively documented, their function during both mitosis and meiosis in the germ line is poorly understood. From the perspective of cell cycle regulation there are several aspects of mammalian spermatogenesis that suggest unique modes of regulation and hence, possible unique functions for the cyclins. This review will summarize our current understanding of cyclin expression and function in the male germ line, with particular focus on the A and E type cyclins in the mouse model. While the focus is on mammalian spermatogenesis, we note contrasts with similar functions in the female germ line when relevant and also draw upon observations in other model systems to provide further insight.


Subject(s)
Cell Cycle/physiology , Cyclins/metabolism , Germ Cells/cytology , Spermatogenesis/physiology , Animals , Female , Germ Cells/metabolism , Humans , Male , Mice
19.
J Cell Sci ; 126(Pt 17): 3927-38, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23788429

ABSTRACT

In mitotic cells, RAD9A functions in repairing DNA double-strand breaks (DSBs) by homologous recombination and facilitates the process by cell cycle checkpoint control in response to DNA damage. DSBs occur naturally in the germline during meiosis but whether RAD9A participates in repairing such breaks is not known. In this study, we determined that RAD9A is indeed expressed in the male germ line with a peak of expression in late pachytene and diplotene stages, and the protein was found associated with the XY body. As complete loss of RAD9A is embryonic lethal, we constructed and characterized a mouse strain with Stra8-Cre driven germ cell-specific ablation of Rad9a beginning in undifferentiated spermatogonia in order to assess its role in spermatogenesis. Adult mutant male mice were infertile or sub-fertile due to massive loss of spermatogenic cells. The onset of this loss occurs during meiotic prophase, and there was an increase in the numbers of apoptotic spermatocytes as determined by TUNEL. Spermatocytes lacking RAD9A usually arrested in meiotic prophase, specifically in pachytene. The incidence of unrepaired DNA breaks increased, as detected by accumulation of γH2AX and DMC1 foci on the axes of autosomal chromosomes in pachytene spermatocytes. The DNA topoisomerase IIß-binding protein 1 (TOPBP1) was still localized to the sex body, albeit with lower intensity, suggesting that RAD9A may be dispensable for sex body formation. We therefore show for the first time that RAD9A is essential for male fertility and for repair of DNA DSBs during meiotic prophase I.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , Fertility/genetics , Meiotic Prophase I/physiology , Recombinational DNA Repair/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/genetics , Carrier Proteins/metabolism , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/biosynthesis , Histones/biosynthesis , Histones/metabolism , Male , Meiotic Prophase I/genetics , Mice , Mice, Transgenic , Nuclear Proteins/biosynthesis , Nuclear Proteins/metabolism , Phosphate-Binding Proteins , Sequence Deletion/genetics , Spermatocytes/cytology , Spermatogenesis/genetics , Testis
20.
Curr Top Dev Biol ; 102: 293-326, 2013.
Article in English | MEDLINE | ID: mdl-23287038

ABSTRACT

The double bromodomain-containing BET (bromodomain and extra terminal) family of proteins is highly conserved from yeast to humans and consists not just of transcriptional regulators but also histone-interacting chromatin remodelers. The four mammalian BET genes are each expressed at unique times during spermatogenesis, and the testis-specific gene Brdt is essential for spermatogenesis. Loss of the first bromodomain of BRDT results in improper/incomplete spermatid elongation and severely morphologically defective sperm. The elongation defects observed in mutant spermatids can be directly tied to altered postmeiotic chromatin architecture. BRDT is required for creation/maintenance of the chromocenter of round spermatids, a structure that forms just after completion of meiosis. The chromocenter creates a defined topology in spermatids, and the presence of multiple chromocenters rather than a single intact chromocenter correlates with loss of spermatid polarity, loss of heterochromatin foci at the nuclear envelope, and loss of proper spermatid elongation. BRDT is not only essential for proper chromatin organization but also involved in regulation of transcription and in cotranscriptional processing. That is, transcription and alternative splicing are altered in spermatocytes and spermatids that lack full-length BRDT. Additionally, the transcription of mRNAs with short 3' UTRs, which is characteristic of round spermatids, is also altered. Examination of the genes regulated by BRDT yields many possible targets that could in part explain the morphologically abnormal sperm produced by the BRDT mutant testes. Thus, BRDT and possibly the other BET genes are required for proper spermatogenesis, which opens up the possibility that the recently discovered small molecule inhibitors of the BET family could be useful as reversible male contraceptives.


Subject(s)
Mammals/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Spermatogenesis/genetics , Animals , Contraceptive Agents/pharmacology , Fertility/drug effects , Fertility/genetics , Gene Expression Regulation/drug effects , Humans , Nuclear Proteins/antagonists & inhibitors , Spermatogenesis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...