Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35043944

ABSTRACT

Establishment of a healthy ovarian reserve is contingent upon numerous regulatory pathways during embryogenesis. Previously, mice lacking TBP-associated factor 4b (Taf4b) were shown to exhibit a diminished ovarian reserve. However, potential oocyte-intrinsic functions of TAF4b have not been examined. Here, we use a combination of gene expression profiling and chromatin mapping to characterize TAF4b-dependent gene regulatory networks in mouse oocytes. We find that Taf4b-deficient oocytes display inappropriate expression of meiotic, chromatin modification/organization, and X-linked genes. Furthermore, dysregulated genes in Taf4b-deficient oocytes exhibit an unexpected amount of overlap with dysregulated genes in oocytes from XO female mice, a mouse model of Turner Syndrome. Using Cleavage Under Targets and Release Using Nuclease (CUT&RUN), we observed TAF4b enrichment at genes involved in chromatin remodeling and DNA repair, some of which are differentially expressed in Taf4b-deficient oocytes. Interestingly, TAF4b target genes were enriched for Sp/Klf family and NFY target motifs rather than TATA-box motifs, suggesting an alternative mode of promoter interaction. Together, our data connect several gene regulatory nodes that contribute to the precise development of the mammalian ovarian reserve.


Subject(s)
Gene Regulatory Networks/genetics , Oogenesis , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Animals , DNA Repair , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Germ Cells/cytology , Germ Cells/metabolism , Meiosis , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oocytes/metabolism , Promoter Regions, Genetic , TATA-Binding Protein Associated Factors/deficiency , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/metabolism , X Chromosome/genetics , X Chromosome/metabolism
2.
PLoS Genet ; 16(1): e1008515, 2020 01.
Article in English | MEDLINE | ID: mdl-31914128

ABSTRACT

Germ cells undergo many developmental transitions before ultimately becoming either eggs or sperm, and during embryonic development these transitions include epigenetic reprogramming, quiescence, and meiosis. To begin understanding the transcriptional regulation underlying these complex processes, we examined the spatial and temporal expression of TAF4b, a variant TFIID subunit required for fertility, during embryonic germ cell development. By analyzing published datasets and using our own experimental system to validate these expression studies, we determined that both Taf4b mRNA and protein are highly germ cell-enriched and that Taf4b mRNA levels dramatically increase from embryonic day 12.5-18.5. Surprisingly, additional mRNAs encoding other TFIID subunits are coordinately upregulated through this time course, including Taf7l and Taf9b. The expression of several of these germ cell-enriched TFIID genes is dependent upon Dazl and/or Stra8, known regulators of germ cell development and meiosis. Together, these data suggest that germ cells employ a highly specialized and dynamic form of TFIID to drive the transcriptional programs that underlie mammalian germ cell development.


Subject(s)
Gametogenesis , Gene Expression Regulation, Developmental , Germ Cells/metabolism , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Deleted in Azoospermia 1 Protein/genetics , Deleted in Azoospermia 1 Protein/metabolism , Germ Cells/cytology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism
3.
PLoS Genet ; 12(6): e1006128, 2016 06.
Article in English | MEDLINE | ID: mdl-27341508

ABSTRACT

TAF4b is a gonadal-enriched subunit of the general transcription factor TFIID that is implicated in promoting healthy ovarian aging and female fertility in mice and humans. To further explore the potential mechanism of TAF4b in promoting ovarian follicle development, we analyzed global gene expression at multiple time points in the human fetal ovary. This computational analysis revealed coordinate expression of human TAF4B and critical regulators and effectors of meiosis I including SYCP3, YBX2, STAG3, and DAZL. To address the functional relevance of this analysis, we turned to the embryonic Taf4b-deficient mouse ovary where, for the first time, we demonstrate, severe deficits in prophase I progression as well as asynapsis in Taf4b-deficient oocytes. Accordingly, TAF4b occupies the proximal promoters of many essential meiosis and oogenesis regulators, including Stra8, Dazl, Figla, and Nobox, and is required for their proper expression. These data reveal a novel TAF4b function in regulating a meiotic gene expression program in early mouse oogenesis, and support the existence of a highly conserved TAF4b-dependent gene regulatory network promoting early oocyte development in both mice and women.


Subject(s)
Meiosis/genetics , Oocytes/metabolism , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Animals , Female , Gene Expression/genetics , Gene Regulatory Networks/genetics , Humans , Male , Mice , Oogenesis/genetics , Ovary/metabolism , Promoter Regions, Genetic/genetics
4.
Development ; 142(15): 2554-63, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26243868

ABSTRACT

The adult mammalian ovary is devoid of definitive germline stem cells. As such, female reproductive senescence largely results from the depletion of a finite ovarian follicle pool that is produced during embryonic development. Remarkably, the crucial nature and regulation of follicle assembly and survival during embryogenesis is just coming into focus. This developmental pathway involves the coordination of meiotic progression and the breakdown of germ cell cysts into individual oocytes housed within primordial follicles. Recent evidence also indicates that genetic and environmental factors can specifically perturb primordial follicle assembly. Here, we review the cellular and molecular mechanisms by which the mammalian ovarian reserve is established, highlighting the presence of a crucial checkpoint that allows survival of only the highest-quality oocytes.


Subject(s)
Germ Cells/growth & development , Mammals/embryology , Meiosis/physiology , Oocytes/cytology , Ovarian Follicle/embryology , Ovarian Reserve/physiology , Signal Transduction/physiology , Adult , Animals , Female , Humans , Models, Biological , Signal Transduction/genetics
5.
Stem Cells ; 33(4): 1267-76, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25727968

ABSTRACT

Long-term mammalian spermatogenesis requires proper development of spermatogonial stem cells (SSCs) that replenish the testis with germ cell progenitors during adult life. TAF4b is a gonadal-enriched component of the general transcription factor complex, TFIID, which is required for the maintenance of spermatogenesis in the mouse. Successful germ cell transplantation assays into adult TAF4b-deficient host testes suggested that TAF4b performs an essential germ cell autonomous function in SSC establishment and/or maintenance. To elucidate the SSC function of TAF4b, we characterized the initial gonocyte pool and rounds of spermatogenic differentiation in the context of the Taf4b-deficient mouse testis. Here, we demonstrate a significant reduction in the late embryonic gonocyte pool and a deficient expansion of this pool soon after birth. Resulting from this reduction of germ cell progenitors is a developmental delay in meiosis initiation, as compared to age-matched controls. While GFRα1+ spermatogonia are appropriately present as Asingle and Apaired in wild-type testes, TAF4b-deficient testes display an increased proportion of long and clustered chains of GFRα1+ cells. In the absence of TAF4b, seminiferous tubules in the adult testis either lack germ cells altogether or are found to have missing generations of spermatogenic progenitor cells. Together these data indicate that TAF4b-deficient spermatogenic progenitor cells display a tendency for differentiation at the expense of self-renewal and a renewing pool of SSCs fail to establish during the critical window of SSC development.


Subject(s)
Adult Stem Cells/physiology , Cell Differentiation/physiology , Spermatogenesis/physiology , Spermatogonia/growth & development , TATA-Binding Protein Associated Factors/biosynthesis , Transcription Factor TFIID/biosynthesis , Animals , Animals, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Dev Biol ; 392(1): 42-51, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24836512

ABSTRACT

Primary ovarian insufficiency (POI) affects 1% of women under the age of 40 and is associated with premature ovarian follicle depletion. TAF4b deficiency in adult female mouse models results in hallmarks of POI including stereotyped gonadotropin alterations indicative of early menopause, poor oocyte quality, and infertility. However, the precise developmental mechanisms underlying these adult deficits remain unknown. Here we show that TAF4b is required for the initial establishment of the primordial follicle reserve at birth. Ovaries derived from TAF4b-deficient mice at birth exhibit delayed germ cell cyst breakdown and a significant increase in Activated Caspase 3 staining compared to control ovaries. Culturing neonatal TAF4b-deficient ovaries with the pan-caspase inhibitor ZVAD-FMK suppresses the excessive loss of these oocytes around the time of birth. These data reveal a novel TAF4b function in orchestrating the correct timing of germ cell cyst breakdown and establishment of the primordial follicle reserve during a critical window of development.


Subject(s)
Estradiol/pharmacology , Oocytes/cytology , Oogenesis/physiology , Ovarian Follicle/embryology , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/biosynthesis , Caspase 3/genetics , Caspase Inhibitors/pharmacology , Cell Survival , Embryonic Development , Enzyme Activation , Female , Mice , Mice, Knockout , Oocytes/physiology , Oogenesis/genetics , Ovarian Follicle/physiology , Primary Ovarian Insufficiency/enzymology , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics
7.
Biol Reprod ; 89(5): 116, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24068106

ABSTRACT

Estrogen signaling in the ovary is a fundamental component of normal ovarian function, and evidence also indicates that excessive estrogen is a risk factor for ovarian cancer. We have previously demonstrated that the gonadally enriched TFIID subunit TAF4B, a paralog of the general transcription factor TAF4A, is required for fertility in mice and for the proliferation of ovarian granulosa cells following hormonal stimulation. However, the relationship between TAF4B and estrogen signaling in the normal ovary or during ovarian tumor initiation and progression has yet to be defined. Herein, we show that Taf4b mRNA and TAF4B protein, but not Taf4a mRNA or TAF4A protein, are increased in whole ovaries and granulosa cells of the ovary after exposure to 17beta-estradiol or the synthetic estrogen diethylstilbestrol and that this response occurs within hours after stimulation. Furthermore, this increase occurs via nuclear estrogen receptors both in vivo and in a mouse granulosa cancer cell line, NT-1. We observe a significant increase in Taf4b mRNA in estrogen-supplemented mouse ovarian tumors, which correlates with diminished survival of these mice. These data highlight the novel response of the general transcription factor TAF4B to estrogen in the normal ovary and during ovarian tumor progression in the mouse, suggesting its potential role in regulating actions downstream of estrogen stimulation.


Subject(s)
Estradiol/pharmacology , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Ovary/drug effects , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Animals , Carcinoma, Ovarian Epithelial , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Ovary/metabolism , Receptors, Estrogen/agonists , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism
8.
Front Cell Dev Biol ; 11: 1270408, 2023.
Article in English | MEDLINE | ID: mdl-37900284

ABSTRACT

Prospermatogonia (ProSpg) link the embryonic development of male primordial germ cells to the healthy establishment of postnatal spermatogonia and spermatogonial stem cells. While these spermatogenic precursor cells undergo the characteristic transitions of cycling and quiescence, the transcriptional events underlying these developmental hallmarks remain unknown. Here, we investigated the expression and function of TBP-associated factor 4b (Taf4b) in the timely development of quiescent mouse ProSpg using an integration of gene expression profiling and chromatin mapping. We find that Taf4b mRNA expression is elevated during the transition of mitotic-to-quiescent ProSpg and Taf4b-deficient ProSpg are delayed in their entry into quiescence. Gene ontology, protein network analysis, and chromatin mapping demonstrate that TAF4b is a direct and indirect regulator of chromatin and cell cycle-related gene expression programs during ProSpg quiescence. Further validation of these cell cycle mRNA changes due to the loss of TAF4b was accomplished via immunostaining for proliferating cell nuclear antigen (PCNA). Together, these data indicate that TAF4b is a key transcriptional regulator of the chromatin and quiescent state of the developing mammalian spermatogenic precursor lineage.

9.
Med Oncol ; 39(5): 71, 2022 May 15.
Article in English | MEDLINE | ID: mdl-35568777

ABSTRACT

Human epididymis protein-4 (HE4/WFDC2) has been well-studied as an ovarian cancer clinical biomarker. To improve our understanding of its functional role in high grade serous ovarian cancer, we determined transcriptomic differences between ovarian tumors with high- versus low-WFDC2 mRNA levels in The Cancer Genome Atlas dataset. High-WFDC2 transcript levels were significantly associated with reduced survival in stage III/IV serous ovarian cancer patients. Differential expression and correlation analyses revealed secretory leukocyte peptidase inhibitor (SLPI/WFDC4) as the gene most positively correlated with WFDC2, while A kinase anchor protein-12 was most negatively correlated. WFDC2 and SLPI were strongly correlated across many cancers. Gene ontology analysis revealed enrichment of oxidative phosphorylation in differentially expressed genes associated with high-WFDC2 levels, while extracellular matrix organization was enriched among genes associated with low-WFDC2 levels. Immune cell subsets found to be positively correlated with WFDC2 levels were B cells and plasmacytoid dendritic cells, while neutrophils and endothelial cells were negatively correlated with WFDC2. Results were compared with DepMap cell culture gene expression data. Gene ontology analysis of k-means clustering revealed that genes associated with low-WFDC2 were also enriched in extracellular matrix and adhesion categories, while high-WFDC2 genes were enriched in epithelial cell proliferation and peptidase activity. These results support previous findings regarding the effect of HE4/WFDC2 on ovarian cancer pathogenesis in cell lines and mouse models, while adding another layer of complexity to its potential functions in ovarian tumor tissue. Further experimental explorations of these findings in the context of the tumor microenvironment are merited.


Subject(s)
Computational Biology , Ovarian Neoplasms , WAP Four-Disulfide Core Domain Protein 2 , Animals , Biomarkers, Tumor/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Matrix/metabolism , Female , Gene Expression , Humans , Mice , Ovarian Neoplasms/pathology , Proteins/metabolism , Tumor Microenvironment , WAP Four-Disulfide Core Domain Protein 2/genetics
10.
Front Cell Dev Biol ; 9: 666508, 2021.
Article in English | MEDLINE | ID: mdl-34568311

ABSTRACT

The development of multicellular organisms and the uniqueness of each cell are achieved by distinct transcriptional programs. Multiple processes that regulate gene expression converge at the core promoter region, an 80 bp region that directs accurate transcription initiation by RNA polymerase II (Pol II). In recent years, it has become apparent that the core promoter region is not a passive DNA component, but rather an active regulatory module of transcriptional programs. Distinct core promoter compositions were demonstrated to result in different transcriptional outputs. In this mini-review, we focus on the role of the core promoter, particularly its downstream region, as the regulatory hub for developmental genes. The downstream core promoter element (DPE) was implicated in the control of evolutionarily conserved developmental gene regulatory networks (GRNs) governing body plan in both the anterior-posterior and dorsal-ventral axes. Notably, the composition of the basal transcription machinery is not universal, but rather promoter-dependent, highlighting the importance of specialized transcription complexes and their core promoter target sequences as key hubs that drive embryonic development, differentiation and morphogenesis across metazoan species. The extent of transcriptional activation by a specific enhancer is dependent on its compatibility with the relevant core promoter. The core promoter content also regulates transcription burst size. Overall, while for many years it was thought that the specificity of gene expression is primarily determined by enhancers, it is now clear that the core promoter region comprises an important regulatory module in the intricate networks of developmental gene expression.

11.
Biochim Biophys Acta ; 1789(3): 161-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19437618

ABSTRACT

Through the reductive divisions of meiosis, sexually reproducing organisms have gained the ability to produce specialized haploid cells called germ cells that fuse to establish the diploid genome of the resulting progeny. The totipotent nature of these germ cells is highlighted by their ability to provide a single fertilized egg cell with all the genetic information necessary to develop the complete repertoire of cell types of the future organism. Thus, the production of these germ cells must be tightly regulated to ensure the continued success of the germ line in future generations. One surprising germ cell development mechanism utilizes variation of the global transcriptional machinery, such as TFIID and TFIIA. Like histone variation, general transcription factor variation serves to produce gonadal-restricted or -enriched expression of selective transcriptional regulatory factors required for establishing and/or maintaining the germ line of diverse organisms. This strategy is observed among invertebrates and vertebrates, and perhaps plants, suggesting that a common theme in germ cell evolution is the diversification of selective promoter initiation factors to regulate critical gonadal-specific programs of gene expression required for sexual reproduction. This review discusses the identification and characterization of a subset of these specialized general transcription factors in diverse organisms that share a common goal of germ line regulation through transcriptional control at its most fundamental level.


Subject(s)
Gene Expression Regulation, Developmental , Germ Cells/metabolism , Transcription Factors/metabolism , Animals , Transcription Factor TFIIA/genetics , Transcription Factor TFIIA/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factors/genetics
12.
Biochim Biophys Acta ; 1789(5): 413-21, 2009 May.
Article in English | MEDLINE | ID: mdl-19362612

ABSTRACT

Chromatin modifications are essential for directing transcription during embryonic development. Bromodomain-containing protein 2 (Brd2; also called RING3 and Fsrg1) is one of four BET (bromodomain and extra-terminal domain) family members known to selectively bind acetylated histones H3 and H4. Brd2 associates with multiple subunits of the transcriptional apparatus including the mediator, TFIID and Swi/Snf multiprotein complexes. While molecular interactions of Brd2 are known, the functions of Brd2 in mammalian embryogenesis remain unknown. In developing a mouse model deficient in Brd2, we find that Brd2 is required for the completion of embryogenesis and proper neural tube closure during development. Embryos lacking Brd2 expression survive up to embryonic day 13.5, soon after mid-gestation, and display fully penetrant neurulation defects that largely result in exencephaly of the developing hindbrain. In this study, we find that highest expression of Brd2 is detected in the developing neural tube, correlating with the neural tube defects found in Brd2-null embryos. Additionally, embryos lacking Brd2 expression display altered gene expression programs, including the mis-expression of multiple genes known to guide neuronal development. Together these results implicate essential roles for Brd2 as a critical integrator of chromatin structure and transcription during mammalian embryogenesis and neurogenesis.


Subject(s)
Chromatin/genetics , Embryonic Development/genetics , Neural Tube Defects/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Apoptosis/genetics , Cell Growth Processes/genetics , Cell Line , Chromatin/metabolism , Chromosomal Proteins, Non-Histone , Embryo, Mammalian , Embryonic Stem Cells/physiology , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mutation , Neural Crest/embryology , Neural Crest/pathology , Neural Tube/embryology , Neural Tube/pathology , Neural Tube Defects/embryology , Neural Tube Defects/pathology , Polymerase Chain Reaction , Pregnancy , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Transcription Factors
13.
Biol Reprod ; 82(1): 23-34, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19684329

ABSTRACT

The mammalian ovary is unique in that its reproductive life span is limited by oocyte quantity and quality. Oocytes are recruited from a finite pool of primordial follicles that are usually exhausted from the ovary during midadult life. If regulation of this pool is perturbed, the reproductive capacity of the ovary is compromised. TAF4B is a gonad-enriched subunit of the TFIID complex required for female fertility in mice. Previous characterization of TAF4B-deficient ovaries revealed several reproductive deficits that collectively result in infertility. However, the etiology of such fertility defects remains unknown. By assaying estrous cycle, ovarian pathology, and gene expression changes in young Taf4b-null female mice, we show that TAF4B-deficient female mice exhibit premature reproductive senescence. The rapid decline of ovarian function in Taf4b-null mice begins in early postnatal life, and follicle depletion is completed by 16 wk of age. To uncover differences in gene expression that may underlie accelerated ovarian aging, we compared genome-wide expression profiles of 3-wk-old, prepubescent Taf4b-null and wild-type ovaries. At 3 wk of age, decreased gene expression in Taf4b-null ovaries is similar to that seen in aged ovaries, revealing several molecular signatures of premature reproductive senescence, including reduced Smc1b. One significantly reduced transcript in the young TAF4B-null ovary codes for MOV10L1, a putative germline-specific RNA helicase that is related to the Drosophila RNA interference protein, armitage. We show here that Mov10l1 is expressed in mouse oocytes and that its expression is sensitive to TAF4B level, linking TAF4B to the posttranscriptional control of ovarian gene expression.


Subject(s)
Aging/physiology , Estrous Cycle , Ovary/physiology , RNA Helicases/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Down-Regulation , Female , Follistatin/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Homeodomain Proteins/metabolism , Infertility, Female/physiopathology , Mad2 Proteins , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Oocytes/physiology , Ovarian Cysts/pathology , Ovary/pathology , PTEN Phosphohydrolase/metabolism , Pituitary Gland/metabolism , Proto-Oncogene Proteins c-akt/metabolism
14.
Mol Cell Biol ; 40(7)2020 03 16.
Article in English | MEDLINE | ID: mdl-31932482

ABSTRACT

TAF4b is a subunit of the TFIID complex that is highly expressed in the ovary and testis and required for mouse fertility. TAF4b-deficient male mice undergo a complex series of developmental defects that result in the inability to maintain long-term spermatogenesis. To decipher the transcriptional mechanisms upon which TAF4b functions in spermatogenesis, we used two-hybrid screening to identify a novel TAF4b-interacting transcriptional cofactor, ZFP628. Deletion analysis of both proteins reveals discrete and novel domains of ZFP628 and TAF4b protein that function to bridge their direct interaction in vitro Moreover, coimmunoprecipitation of ZFP628 and TAF4b proteins in testis-derived protein extracts supports their endogenous association. Using CRISPR-Cas9, we disrupted the expression of ZFP628 in the mouse and uncovered a postmeiotic germ cell arrest at the round spermatid stage in the seminiferous tubules of the testis in ZFP628-deficient mice that results in male infertility. Coincident with round spermatid arrest, we find reduced mRNA expression of transition protein (Tnp1 and Tnp2) and protamine (Prm1 and Prm2) genes, which are critical for the specialized maturation of haploid male germ cells called spermiogenesis. These data delineate a novel association of two transcription factors, TAF4b and ZFP628, and identify ZFP628 as a novel transcriptional regulator of stage-specific spermiogenesis.


Subject(s)
DNA-Binding Proteins/metabolism , Infertility, Male/genetics , Spermatogenesis/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Transcription Factors/metabolism , Animals , Apoptosis/genetics , CRISPR-Cas Systems/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovary/metabolism , Protamines/genetics , Protamines/metabolism , Protein Domains/genetics , Testis/metabolism , Transcription Factors/genetics , Transcriptional Activation/genetics , Two-Hybrid System Techniques
15.
Toxicol Sci ; 167(2): 546-558, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30329139

ABSTRACT

Exposure to excess retinoic acid (RA) disrupts the development of the mammalian testicular seminiferous cord. However, the molecular events surrounding RA-driven loss of cord structure have not previously been examined. To investigate the mechanisms associated with this adverse developmental effect, fetal rat testes were isolated on gestational day 15, after testis determination and the initiation of cord development, and cultured in media containing all-trans RA (ATRA; 10-8 to 10-6 M) or vehicle for 3 days. ATRA exposure resulted in a concentration-dependent decrease in the number of seminiferous cords per testis section and number of germ cells, assessed by histopathology and immunohistochemistry. Following 1 day of culture, genome-wide expression profiling by microarray demonstrated that ATRA exposure altered biological processes related to retinoid metabolism and gonadal sex determination. Real-time RT-PCR analysis confirmed that ATRA enhanced the expression of the key ovarian development gene Wnt4 and the antitestis gene Nr0b1 in a concentration-dependent manner. After 3 days of culture, ATRA-treated testes contained both immunohistochemically DMRT1-positive and FOXL2-positive somatic cells, providing evidence of disrupted testicular cell fate maintenance following ATRA exposure. We conclude that exogenous RA disrupts seminiferous cord development in ex vivo cultured fetal rat testes, resulting in a reduction in seminiferous cord number, and interferes with maintenance of somatic cell fate by enhancing expression of factors that promote ovarian development.


Subject(s)
Fetal Organ Maturity/drug effects , Leydig Cells/drug effects , Sertoli Cells/drug effects , Testis/drug effects , Tretinoin/toxicity , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Gestational Age , Male , Rats , Seminiferous Tubules/drug effects , Testis/embryology
16.
Oncotarget ; 10(36): 3315-3327, 2019 May 21.
Article in English | MEDLINE | ID: mdl-31164954

ABSTRACT

Dual specificity phosphatase 6 (DUSP6) is a protein phosphatase that deactivates extracellular-signal-regulated kinase (ERK). Since the ovarian cancer biomarker human epididymis protein 4 (HE4) interacts with the ERK pathway, we sought to determine the relationship between DUSP6 and HE4 and elucidate DUSP6's role in epithelial ovarian cancer (EOC). Viability assays revealed a significant decrease in cell viability with pharmacological inhibition of DUSP6 using (E/Z)-BCI hydrochloride in ovarian cancer cells treated with carboplatin or paclitaxel, compared to treatment with either agent alone. Quantitative PCR was used to evaluate levels of ERK pathway response genes to BCI in combination with recombinant HE4 (rHE4), carboplatin, and paclitaxel. Expression of EGR1, a promoter of apoptosis, was higher in cells co-treated with BCI and paclitaxel or carboplatin than in cells treated with chemotherapeutic agents alone, while expression of the proto-oncogene c-JUN was decreased with co-treatment. The effect of BCI on the expression of these two genes opposed that of rHE4. Pathway focused quantitative PCR also revealed suppression of ERBB3 in cells co-treated with BCI plus carboplatin or paclitaxel. Finally, expression levels of DUSP6 in EOC tissue were evaluated by immunohistochemistry, revealing significantly increased levels of DUSP6 in serous EOC tissue compared to adjacent normal tissue. A positive correlation between HE4 and DUSP6 levels was determined by Spearman Rank correlation. In conclusion, DUSP6 inhibition sensitizes ovarian cancer cells to chemotherapeutic agents and alters gene expression of ERK response genes, suggesting that DUSP6 could plausibly function as a novel therapeutic target to reduce chemoresistance in EOC.

18.
Mol Cell Biol ; 22(18): 6564-72, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12192054

ABSTRACT

Regulated gene expression is a complex process achieved through the function of multiple protein factors acting in concert at a given promoter. The transcription factor TFIID is a central component of the machinery regulating mRNA synthesis by RNA polymerase II. This large multiprotein complex is composed of the TATA box binding protein (TBP) and several TBP-associated factors (TAF(II)s). The recent discovery of multiple TBP-related factors and tissue-specific TAF(II)s suggests the existence of specialized TFIID complexes that likely play a critical role in regulating transcription in a gene- and tissue-specific manner. The tissue-selective factor TAF(II)105 was originally identified as a component of TFIID derived from a human B-cell line. In this report we demonstrate the specific induction of TAF(II)105 in cultured B cells in response to bacterial lipopolysaccharide (LPS). To examine the in vivo role of TAF(II)105, we have generated TAF(II)105-null mice by homologous recombination. Here we show that B-lymphocyte development is largely unaffected by the absence of TAF(II)105. TAF(II)105-null B cells can proliferate in response to LPS, produce relatively normal levels of resting antibodies, and can mount an immune response by producing antigen-specific antibodies in response to immunization. Taken together, we conclude that the function of TAF(II)105 in B cells is likely redundant with the function of other TAF(II)105-related cellular proteins.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/physiology , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Transcription Factors/physiology , Animals , Blotting, Western , Cell Division , Cell Separation , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Erythrocytes/metabolism , Flow Cytometry , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Lipopolysaccharides/metabolism , Mice , Precipitin Tests , RNA, Messenger/metabolism , Spleen/metabolism , Spleen/pathology , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation
19.
Mol Cancer Res ; 13(1): 78-85, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25169943

ABSTRACT

UNLABELLED: Ovarian cancer is a lethal disease with the majority of diagnosed women having distant metastases. Interestingly, although Notch3 overexpression has been correlated with poor survival in epithelial ovarian cancer (EOC), little is known about its mechanism of action. Data show that Notch3 specifically promotes anoikis resistance. In addition, data indicate a positive role for focal adhesion kinase (FAK) as well as downstream signaling kinases such as Akt and Erk 1/2 in promoting anchorage-independent growth. Mechanistically, both mRNA transcript and protein levels of type IV collagen (COL4A2) are reduced when Notch3 levels are decreased and exogenous collagen IV supplementation reverses the anoikis sensitivity. Reduction of COL4A2 expression by RNAI-mediated knockdown induces cell death. Finally, elevated Notch3 expression levels correlate with higher COL4A2 expression in human ovarian tumor specimens. IMPLICATIONS: These data highlight type IV collagen as a novel therapeutic target for metastatic EOC. Visual Overview: http://mcr.aacrjournals.org/content/early/2014/11/25/1541-7786.MCR-14-0334/F1.large.jpg


Subject(s)
Anoikis/genetics , Collagen Type IV/biosynthesis , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Receptors, Notch/biosynthesis , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Collagen Type IV/genetics , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Neoplasm Metastasis , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , RNA, Messenger/biosynthesis , Receptor, Notch3 , Receptors, Notch/genetics
20.
J Steroid Biochem Mol Biol ; 143: 160-73, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24565562

ABSTRACT

Resistance to anti-estrogen therapies is a prominent challenge in the treatment of ovarian cancer. Tumors develop endocrine resistance by acquiring adaptations that help them rely on alternative oncogenic signaling cascades, which crosstalk with estrogen signaling pathways. An understanding of estrogen signaling crosstalk with these growth promoting cascades is essential in order to maximize efficacy of anti-estrogen treatments in ovarian cancer. Herein, we provide an overview of estrogen signaling in ovarian cancer and discuss the major challenges associated with anti-estrogen therapies. We also review what is currently known about how genomic and non-genomic estrogen signaling pathways crosstalk with several major oncogenic signaling cascades. The insights provided here illustrate existing strategies for targeting endocrine resistant ovarian tumors and may help identify new strategies to improve the treatment of this disease.


Subject(s)
Drug Resistance, Neoplasm , Endocrine System/drug effects , Estrogens/pharmacology , Ovarian Neoplasms/drug therapy , Signal Transduction/drug effects , Female , Humans , Ovarian Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL