Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 534: 141-148, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33333437

ABSTRACT

Nuclear transporter Importin (Imp, Ipo) 13 is known to transport various mammalian cargoes into/out of the nucleus, but its role in directing cell-fate is unclear. Here we examine the role of Imp13 in the maintenance of pluripotency and differentiation of embryonic stem cells (ESCs) for the first time, using an embryonic body (EB)-based model. When induced to differentiate, Ipo13-/- ESCs displayed slow proliferation, reduced EB size, and lower expression of the proliferation marker KI67, concomitant with an increase in the number of TUNEL+ nuclei compared to wildtype ESCs. At days 5 and 10 of differentiation, Ipo13-/- EBs also showed enhanced loss of the pluripotency transcript OCT3/4, and barely detectable clusters of OCT3/4 positive cells. Day 5 Ipo13-/- EBs further exhibited reduced levels of the mesodermal markers Brachyury and Mixl1, correlating with reduced numbers of haemoglobinised cells generated. Our findings suggest that Imp13 is critical to ESC survival as well as early post-gastrulation differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Karyopherins/physiology , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cell Survival , Embryoid Bodies/metabolism , Gene Knockout Techniques , Karyopherins/genetics , Mesoderm/metabolism , Mice , Octamer Transcription Factor-3/metabolism
3.
Hum Reprod ; 35(9): 1991-2003, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32667987

ABSTRACT

STUDY QUESTION: Is WNT signalling functional in normal and/or neoplastic human male germ cells? SUMMARY ANSWER: Regulated WNT signalling component synthesis in human testes indicates that WNT pathway function changes during normal spermatogenesis and is active in testicular germ cell tumours (TGCTs), and that WNT pathway blockade may restrict seminoma growth and migration. WHAT IS KNOWN ALREADY: Regulated WNT signalling governs many developmental processes, including those affecting male fertility during early germ cell development at embryonic and adult (spermatogonial) ages in mice. In addition, although many cancers arise from WNT signalling alterations, the functional relevance and WNT pathway components in TGCT, including germ cell neoplasia in situ (GCNIS), are unknown. STUDY DESIGN, SIZE, DURATION: The cellular distribution of transcripts and proteins in WNT signalling pathways was assessed in fixed human testis sections with normal spermatogenesis, GCNIS and seminoma (2-16 individuals per condition). Short-term (1-7 h) ligand activation and long-term (1-5 days) functional outcomes were examined using the well-characterised seminoma cell line, TCam-2. Pathway inhibition used siRNA or chemical exposures over 5 days to assess survival and migration. PARTICIPANTS/MATERIALS, SETTING, METHODS: The cellular localisation of WNT signalling components was determined using in situ hybridisation and immunohistochemistry on Bouin's- and formalin-fixed human testis sections with complete spermatogenesis or germ cell neoplasia, and was also assessed in TCam-2 cells. Pathway function tests included exposure of TCam-2 cells to ligands, small molecules and siRNAs. Outcomes were measured by monitoring beta-catenin (CTNNB1) intracellular localisation, cell counting and gap closure measurements. MAIN RESULTS AND THE ROLE OF CHANCE: Detection of nuclear-localised beta-catenin (CTNNB1), and key WNT signalling components (including WNT3A, AXIN2, TCF7L1 and TCF7L2) indicate dynamic and cell-specific pathway activity in the adult human testis. Their presence in germ cell neoplasia and functional analyses in TCam-2 cells indicate roles for active canonical WNT signalling in TGCT relating to viability and migration. All data were analysed to determine statistical significance. LARGE SCALE DATA: No large-scale datasets were generated in this study. LIMITATIONS, REASONS FOR CAUTION: As TGCTs are rare and morphologically heterogeneous, functional studies in primary cancer cells were not performed. Functional analysis was performed with the only well-characterised, widely accepted seminoma-derived cell line. WIDER IMPLICATIONS OF THE FINDINGS: This study demonstrated the potential sites and involvement of the WNT pathway in human spermatogenesis, revealing similarities with murine testis that suggest the potential for functional conservation during normal spermatogenesis. Evidence that inhibition of canonical WNT signalling leads to loss of viability and migratory activity in seminoma cells suggests that potential treatments using small molecule or siRNA inhibitors may be suitable for patients with metastatic TGCTs. STUDY FUNDING AND COMPETING INTEREST(S): This study was funded by National Health and Medical Research Council of Australia (Project ID 1011340 to K.L.L. and H.E.A., and Fellowship ID 1079646 to K.L.L.) and supported by the Victorian Government's Operational Infrastructure Support Program. None of the authors have any competing interests.


Subject(s)
Neoplasms, Germ Cell and Embryonal , Testicular Neoplasms , Adult , Animals , Australia , Humans , Male , Mice , Neoplasms, Germ Cell and Embryonal/genetics , Spermatogenesis , Testicular Neoplasms/genetics , Testis , Wnt Signaling Pathway
4.
Reproduction ; 160(1): 141-154, 2020 07.
Article in English | MEDLINE | ID: mdl-32484162

ABSTRACT

Disrupted fetal germline development underpins testicular germ cell neoplasia, which is increasing worldwide. The complex signaling milieu during normal testis development includes TGFß superfamily ligands; this study tests the hypothesis that, activin A, a TGFß superfamily member, can influence gonocyte development. The human seminoma-derived cell line, TCam-2, a model of fetal gonocytes, was cultured with activin A (1.25-25 ng/mL) for 48 h, or with 5 ng/mL activin A for short- (6, 24, and 48 h) and long-term (13 days) exposures, and downstream targets measured by qRT-PCR. Transcripts that exhibited significant dose-dependent responses to activin A included the early germ cell markers KIT, NODAL, and CRIPTO (NODALl co-receptor and activin inhibitor) which all increased and the differentiation marker DNMT3L which decreased. After 48 h, KIT, NODAL, and CRIPTO levels were significantly higher, while the differentiation marker NANOS2 was significantly lower. Interestingly, activin A exposure also significantly reduced both transcript and protein levels of the PIWI/piRNA pathway component DNMT3L. Because TCam-2 cells produce the activin inhibitor CRIPTO, CRIPTO was reduced using siRNA prior to activin A exposure. This selectively increased KIT in response to activin A. Other ligands present in the fetal testis (BMP4, FGF9, TGFß1, and TGFß2) induced distinct effects on germline marker expression. This study showed that activin A can directly modulate germline markers in this human gonocyte-like cell, promoting a less-differentiated phenotype. Additional findings indicate evidence of signaling crosstalk between activin A and NODAL, leading to target-specific effects on gonocyte differentiation.


Subject(s)
Activins/pharmacology , Cell Differentiation , Gene Expression Regulation/drug effects , Germ Cells/pathology , Nodal Protein/metabolism , Seminoma/pathology , Transforming Growth Factor beta/pharmacology , Gene Expression Profiling , Germ Cells/metabolism , Humans , Male , Nodal Protein/genetics , Seminoma/drug therapy , Seminoma/genetics , Testicular Neoplasms/drug therapy , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology
5.
Andrology ; 8(5): 1456-1470, 2020 09.
Article in English | MEDLINE | ID: mdl-32441446

ABSTRACT

BACKGROUND: Snail transcription factors mediate key cellular transitions in many developmental processes, including spermatogenesis, and their production can be regulated by TGF-ß superfamily signalling. SNAI1 and SNAI2 support many cancers of epithelial origin. Their functional relevance and potential regulation by TGF-ß superfamily ligands in germ cell neoplasia are unknown. METHODS: SNAI1, SNAI2 and importin 5 (IPO5; nuclear transporter that selectively mediates BMP signalling) cellular localization was examined in fixed normal adult human and/or neoplastic testes using in situ hybridization and/or immunohistochemistry. SNAI1 and SNAI2 functions were assessed using the well-characterized human seminoma cell line, TCam-2. Cell migration, adhesion/proliferation and survival were measured by scratch assay, xCELLigence and flow cytometry following siRNA-induced reduction of SNAI1 and SNAI2 in TCam-2 cells. The potential regulation of SNAI1 and SNAI2 in TCam-2 cells by TGF-ß signalling ligands, activin A and BMP4 was evaluated following 48 hours culture, including with siRNA regulation of IPO5 to selectively restrict BMP4 signalling. RESULTS: In normal testes, SNAI1 transcript was identified in some spermatogonia and in spermatocytes, and SNAI2 protein localized to nuclei of spermatogonia, spermatocytes and round spermatids. In neoplastic testes, both SNAI1 and SNAI2 were detected in GCNIS and in seminoma cells. SNAI1 and SNAI2 reduction in TCam-2 cells by siRNAs significantly inhibited migration and survival, respectively. Exposure to BMP4, but not activin A, significantly increased SNAI2 (~18-fold). IPO5 inhibition by siRNAs decreased BMP4-induced SNAI2 upregulation (~5-fold). Additionally, SNAI2 reduction using siRNAs inhibited BMP4-induced TCam-2 cell survival. CONCLUSIONS: This is the first evidence that SNAI1 and SNAI2 are involved in human spermatogenesis, with independent functions. These outcomes demonstrate that SNAI1 and SNAI2 inhibition leads to loss of migratory and viability capacities in seminoma cells. These findings show the potential for therapeutic treatments targeting SNAIL or BMP4 signalling for patients with metastatic testicular germ cell tumours.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Neoplasms, Germ Cell and Embryonal/metabolism , Snail Family Transcription Factors/metabolism , Testicular Neoplasms/metabolism , Bone Morphogenetic Protein 4/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , Neoplasms, Germ Cell and Embryonal/genetics , Signal Transduction/physiology , Snail Family Transcription Factors/genetics , Spermatogenesis/physiology , Testicular Neoplasms/genetics
6.
Endocrinology ; 161(7)2020 07 01.
Article in English | MEDLINE | ID: mdl-32274496

ABSTRACT

Activin A promotes fetal mouse testis development, including driving Sertoli cell proliferation and cord morphogenesis, but its mechanisms of action are undefined. We performed ribonucleic acid sequencing (RNA-seq) on testicular somatic cells from fetal activin A-deficient mice (Inhba KO) and wildtype littermates at embryonic day (E) E13.5 and E15.5. Analysis of whole gonads provided validation, and cultures with a pathway inhibitor discerned acute from chronic effects of altered activin A bioactivity. Activin A deficiency predominantly affects the Sertoli cell transcriptome. New candidate targets include Minar1, Sel1l3, Vnn1, Sfrp4, Masp1, Nell1, Tthy1 and Prss12. Importantly, the testosterone (T) biosynthetic enzymes present in fetal Sertoli cells, Hsd17b1 and Hsd17b3, were identified as activin-responsive. Activin-deficient testes contained elevated androstenedione (A4), displayed an Inhba gene dose-dependent A4/T ratio, and contained 11-keto androgens. The remarkable accumulation of lipid droplets in both Sertoli and germ cells at E15.5 indicated impaired lipid metabolism in the absence of activin A. This demonstrated for the first time that activin A acts on Sertoli cells to determine local steroid production during fetal testis development. These outcomes reveal how compounds that perturb fetal steroidogenesis can function through cell-specific mechanisms and can indicate how altered activin levels in utero may impact testis development.


Subject(s)
Activins/physiology , Gonadal Steroid Hormones/metabolism , Testis/embryology , Testis/metabolism , Animals , Female , Male , Mice , Mice, Knockout , Pregnancy , Sex Determination Processes
7.
Reproduction ; 158(3): 267-280, 2019 09.
Article in English | MEDLINE | ID: mdl-31299635

ABSTRACT

Expression profiles and subcellular localisations of core Drosophila behaviour/human splicing (DBHS) proteins (PSPC1, SFPQ and NONO) and NEAT1, a long noncoding RNA (lncRNA), are investigated in developing and adult mouse testes. Core DBHS proteins are markers for the distinct subnuclear domain termed paraspeckles, while a long NEAT1 isoform scaffold facilitates paraspeckle nucleation. Paraspeckles contain many proteins (>40) and are broadly involved in RNA metabolism, including transcriptional regulation by protein sequestration, nuclear retention of A-to-I edited RNA transcripts to regulate translation and promoting survival during cellular stress. Immunohistochemistry reveals cell-specific profiles for core DBHS paraspeckle protein expression, indicating their functional diversity. PSPC1 is an androgen receptor co-activator, and it is detected in differentiating Sertoli cell nuclei from day 15 onwards, as they develop androgen responsiveness. PSPC1 is nuclear in the most mature male germ cell type present at each age, from foetal to adult life. In adult mouse testes, PSPC1 and SFPQ are present in Sertoli cells, spermatocytes and round spermatids, while the NEAT1 lncRNA appears in the punctate nuclear foci delineating paraspeckles only within Leydig cells. Identification of NEAT1 in the cytoplasm of spermatogonia and spermatocytes must reflect non-paraspeckle-related functions. NONO was absent from germ cells but nuclear in Sertoli cells. Reciprocal nuclear profiles of PSPC1 and γ-H2AX in spermatogenic cells suggest that each performs developmentally regulated roles in stress responses. These findings demonstrate paraspeckles and paraspeckle-related proteins contribute to diverse functions during testis development and spermatogenesis.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Leydig Cells/metabolism , PTB-Associated Splicing Factor/metabolism , RNA-Binding Proteins/metabolism , Spermatogenesis/physiology , Testis/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Male , Mice , PTB-Associated Splicing Factor/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , Sertoli Cells/metabolism , Testis/growth & development
8.
Anat Sci Educ ; 12(1): 90-96, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30106512

ABSTRACT

The practical aspect of human developmental biology education is often limited to the observation and use of animal models to illustrate developmental anatomy. This is due in part to the difficulty of accessing human embryonic and fetal specimens, and the sensitivity inherent to presenting these specimens as teaching materials. This report presents a new approach using three-dimensional (3D) printed replicas of actual human materials in practical classes, thus allowing for the inclusion of accurate examples of human developmental anatomy in the educational context. A series of 3D prints have been produced from digital data collected by computed tomography (CT) imaging of an archived series of preserved human embryonic and fetal specimens. The final versions of 3D prints have been generated in a range of single or multiple materials to illustrate the progression of human development, including the development of internal anatomy. Furthermore, multiple copies of each replica have been printed for large group teaching. In addition to the educational benefit of examining accurate 3D replicas, this approach lessens the potential for adverse student reaction (due to cultural background or personal experience) to observing actual human embryonic/fetal anatomical specimens, and reduces the potential of damage or loss of original specimens. This approach, in combination with ongoing improvements in the management and analysis of digital data and advances in scanning technology, has enormous potential to allow embryology students access to both local and international collections of human gestational material. Anat Sci Educ 00: 000-000. © 2018 American Association of Anatomists.


Subject(s)
Embryology/education , Imaging, Three-Dimensional , Models, Anatomic , Printing, Three-Dimensional , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/diagnostic imaging , Fetus/anatomy & histology , Fetus/diagnostic imaging , Humans , Teaching , Tomography, X-Ray Computed
9.
Semin Cell Dev Biol ; 45: 94-103, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26500180

ABSTRACT

The TGF-ß ligand superfamily contains at least 40 members, many of which are produced and act within the mammalian testis to facilitate formation of sperm. Their progressive expression at key stages and in specific cell types determines the fertility of adult males, influencing testis development and controlling germline differentiation. BMPs are essential for the interactive instructions between multiple cell types in the early embryo that drive initial specification of gamete precursors. In the nascent foetal testis, several ligands including Nodal, TGF-ßs, Activins and BMPs, serve as key masculinizing switches by regulating male germline pluripotency, somatic and germline proliferation, and testicular vascularization and architecture. In postnatal life, local production of these factors determine adult testis size by regulating Sertoli cell multiplication and differentiation, in addition to specifying germline differentiation and multiplication. Because TGF-ß superfamily signaling is integral to testis formation, it affects processes that underlie testicular pathologies, including testicular cancer, and its potential to contribute to subfertility is beginning to be understood.


Subject(s)
Signal Transduction , Spermatogenesis , Testis/metabolism , Transforming Growth Factor beta/physiology , Animals , Gene Expression Regulation, Developmental , Humans , Male , Sexual Development , Spermatozoa/physiology , Testis/cytology , Testis/growth & development
10.
Asian J Androl ; 17(4): 537-44, 2015.
Article in English | MEDLINE | ID: mdl-25994647

ABSTRACT

Importin proteins were originally characterized for their central role in protein transport through the nuclear pores, the only intracellular entry to the nucleus. This vital function must be tightly regulated to control access by transcription factors and other nuclear proteins to genomic DNA, to achieve appropriate modulation of cellular behaviors affecting cell fate. Importin-mediated nucleocytoplasmic transport relies on their specific recognition of cargoes, with each importin binding to distinct and overlapping protein subsets. Knowledge of importin function has expanded substantially in regard to three key developmental systems: embryonic stem cells, muscle cells and the germ line. In the decade since the potential for regulated nucleocytoplasmic transport to contribute to spermatogenesis was proposed, we and others have shown that the importins that ferry transcription factors into the nucleus perform additional roles, which control cell fate. This review presents key findings from studies of mammalian spermatogenesis that reveal potential new pathways by which male fertility and infertility arise. These studies of germline genesis illuminate new ways in which importin proteins govern cellular differentiation, including via directing proteins to distinct intracellular compartments and by determining cellular stress responses.


Subject(s)
Fertilization/physiology , Karyopherins/physiology , Spermatogenesis/physiology , Active Transport, Cell Nucleus , Animals , Female , Fertilization/genetics , Humans , Infertility/genetics , Infertility/physiopathology , Karyopherins/genetics , Male , Mammals , Spermatogenesis/genetics
11.
Biol Reprod ; 90(1): 3, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24258210

ABSTRACT

The importance of Wnt signaling for postnatal testis function has been previously studied in several mouse models, with chronic pathway disruption addressing its function in Sertoli cells and in postmeiotic germ cells. While chronic beta-catenin deletion in Sertoli cells does not profoundly affect testis development, new data indicate that Wnt signaling is required at multiple stages of spermatogenesis. We used two mouse models that allow acute disruption of Wnt signaling to explore the importance of regulated Wnt pathway activity for normal germ cell development in adult male mice. Short-term induction of mutations in Adenomatous polyposis coli (Apc) and beta-catenin (Ctnnbl), which increase and decrease Wnt signaling levels, were generated in AhCre Apc(fl/fl) and AhCre Ctnnb1(fl/fl) mice, respectively. Each exhibited a distinct phenotype of disrupted spermatogenesis that was evident within 24 h and persisted for up to 4 days. Outcomes included germ cell apoptosis and rapid loss and altered blood-testis barrier protein distribution and morphology. The functional significance of nuclear localized beta-catenin protein in spermatocytes and round spermatids, indicative of active Wnt signaling, was highlighted by the profound loss of postmitotic germ cells in both models. Developmentally regulated Wnt signaling mediators identified through transcriptional profiling of wild-type and AhCre Ctnnb1(fl/fl) mouse testes identified Wnt receptors (e.g., Fzd4) and ligands (e.g., Wnt3, Wnt3a, Wnt5b, Wnt7a, and Wnt8b). This demonstration that Wnt signaling control is essential for adult spermatogenesis supports the growing understanding that its disruption may underpin certain cases of male infertility.


Subject(s)
Spermatogenesis/genetics , Wnt Signaling Pathway/physiology , beta Catenin , Adult Stem Cells/physiology , Animals , Apoptosis/genetics , Genes, APC , Germ Cells/physiology , Male , Mice , Mice, Transgenic , Mutation , Spermatids/cytology , Spermatids/physiology , Spermatocytes/cytology , Spermatocytes/physiology , beta Catenin/genetics , beta Catenin/metabolism
12.
Biochim Biophys Acta ; 1833(10): 2348-56, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23773962

ABSTRACT

The importin (IMP) superfamily of nuclear transport proteins is essential to key developmental pathways, including in the murine testis where expression of the 6 distinct IMPα proteins is highly dynamic. Present predominantly from the spermatocyte stage onwards, IMPα4 is unique in showing a striking nuclear localization, a property we previously found to be linked to maintenance of pluripotency in embryonic stem cells and to the cellular stress response in cultured cells. Here we examine the role of IMPα4 in vivo for the first time using a novel transgenic mouse model in which we overexpress an IMPα4-EGFP fusion protein from the protamine 1 promoter to recapitulate endogenous testicular germ cell IMPα4 expression in spermatids. IMPα4 overexpression did not affect overall fertility, testis morphology/weight or spermatogenic progression under normal conditions, but conferred significantly (>30%) increased resistance to oxidative stress specifically in the spermatid subpopulation expressing the transgene. Consistent with a cell-specific role for IMPα4 in protecting against oxidative stress, haploid germ cells from IMPα4 null mice were significantly (c. 30%) less resistant to oxidative stress than wild type controls. These results from two unique and complementary mouse models demonstrate a novel protective role for IMPα4 in stress responses specifically within haploid male germline cells, with implications for male fertility and genetic integrity.


Subject(s)
Active Transport, Cell Nucleus/genetics , Cell Nucleus/genetics , Germ Cells/metabolism , Oxidative Stress , Spermatids/metabolism , Spermatogenesis , Testis/metabolism , alpha Karyopherins/physiology , Animals , Blotting, Southern , Blotting, Western , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , DNA/genetics , Fertility , Flow Cytometry , Germ Cells/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Promoter Regions, Genetic/genetics , Protamines/genetics , Real-Time Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spermatids/cytology , Testis/cytology
13.
FASEB J ; 25(11): 3958-65, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21840941

ABSTRACT

The cellular repertoire of importin (IMP) proteins that mediates nuclear import of transcription factors and chromatin remodeling agents is critical to processes such as differentiation and transformation. This study identifies for the first time independent roles for specific IMPαs in murine embryonic stem cells (mESCs), showing that mESC differentiation is accompanied by dynamic changes in the levels of transcripts encoding the IMPs, IMPα3, IMPα4, IMPß1, and IPO5. Of these, only IMPα4 was maintained at higher levels in differentiating mESCs, correlating with the finding that IMPα4 overexpression induced a significant decrease in Oct3/4 protein levels compared to control transfections. In parallel, IMPα4 protein showed a unique and striking shift in subcellular localization from the nucleus to the cytoplasm during differentiation, which is consistent with activation of a role in nuclear import of differentiation factors. Overexpression of a dominant-negative IMPα2 isoform, when assessed against adjacent untransfected or IMPα2 transfected cells, led to both a significant reduction in endogenous Oct3/4 protein levels and inhibition of Oct3/4 nuclear localization, suggesting that IMPα2-mediated delivery of Oct3/4 to the nucleus contributes directly to maintenance of mESC pluripotency. These findings implicate IMPα2 and IMPα4 in specific but distinct roles in the fate choice between pluripotency and commitment to differentiation.


Subject(s)
Embryonic Stem Cells/metabolism , Karyopherins/physiology , Nuclear Proteins/physiology , Octamer Transcription Factor-3/metabolism , alpha Karyopherins/physiology , Active Transport, Cell Nucleus/genetics , Animals , Cell Differentiation/physiology , Cell Line , Mice , Nuclear Proteins/biosynthesis , Octamer Transcription Factor-3/biosynthesis , alpha Karyopherins/biosynthesis , beta Karyopherins/physiology
14.
Biol Reprod ; 82(2): 390-401, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19846600

ABSTRACT

Bone morphogenetic protein (BMP) signaling is critical for germline establishment during mouse embryogenesis. To exploit its importance for induction of germline precursors in vitro, mouse embryonic stem cells (mESCs) were cultured as embryoid body (EB) aggregates with combinations of BMP2, BMP4, and BMP8B for 3-10 days. At Day 10 of culture, well-delineated clusters of POU5F1-positive (POU5F1+) cells were visible in BMP4-treated and BMP2-treated EBs; these were rarely detected in untreated and BMP8B-treated cultures. Quantitative mRNA analysis revealed that a significant elevation of markers associated with primordial germ cell development had occurred in the presence of BMP4 by Day 10, including late germline markers such as Ddx4 (Mvh). Reasoning that germline specification was established by Day 10, we surveyed earlier time points for altered levels of germline marker mRNAs. A peak of early markers, Prdm1 (Blimp1), Ifitm3 (Fragilis), and Dppa3 (Stella), was measured in Day 3 to Day 4 EBs grown in BMP4, followed by a decrease at Day 5. In contrast, other markers, Pou5f1, Nanog, Dazl, and Ddx4, progressively increased from Day 3 to Day 5. Transforming growth factor beta superfamily signaling components Acvr1 (ALK2), Smad1, and Smad5 remained relatively constant. Isolated POU5F1+ cells from BMP4-treated Day 5 EBs contained significantly elevated germline markers compared with POU5F1-negative cells, with a transcript profile differing from mESCs, verifying their unique identity. These results demonstrate that signaling by BMP2 and BMP4, but not BMP8B, enhances germline marker expression within EBs and identify Day 3 to Day 5 in EB differentiation as a window for specification of germ cells in vitro.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Germ Cells/cytology , Animals , Biomarkers/analysis , Bone Morphogenetic Protein 2/administration & dosage , Bone Morphogenetic Protein 4/administration & dosage , Bone Morphogenetic Proteins/administration & dosage , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , DEAD-box RNA Helicases/genetics , Gene Expression/drug effects , Germ Cells/chemistry , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Mice , Octamer Transcription Factor-3/analysis , Octamer Transcription Factor-3/genetics , RNA, Messenger/analysis , Recombinant Proteins/administration & dosage , Time Factors
15.
Int J Biochem Cell Biol ; 36(4): 555-67, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15010323

ABSTRACT

Embryonic stem (ES) cells are in vitro cultivated pluripotent cells derived from the inner cell mass (ICM) of the embryonic blastocyst. Attesting to their pluripotency, ES cells can be differentiated into representative derivatives of all three embryonic germ layers (endoderm, ectoderm and mesoderm) both in vitro and in vivo. Although mouse ES cells have been studied for many years, human ES cells have only more recently been derived and successfully propagated. Many biochemical differences and culture requirements between mouse and human ES cells have been described, yet despite these differences the study of murine ES cells has provided important insights into methodologies aimed at generating a greater and more in depth understanding of human ES cell biology. One common feature of both mouse and human ES cells is their capacity to undergo controlled differentiation into spheroid structures termed embryoid bodies (EBs). EBs recapitulate several aspects of early development, displaying regional-specific differentiation programs into derivatives of all three embryonic germ layers. For this reason, EB formation has been utilised as an initial step in a wide range of studies aimed at differentiating both mouse and human ES cells into a specific and desired cell type. Recent reports utilising specific growth factor combinations and cell-cell induction systems have provided alternative strategies for the directed differentiation of cells into a desired lineage. According to each one of these strategies, however, a relatively high cell lineage heterogeneity remains, necessitating subsequent purification steps including mechanical dissection, selective media or fluorescent or magnetic activated cell sorting (FACS and MACS, respectively). In the future, the ability to specifically direct differentiation of human ES cells at 100% efficiency into a desired lineage will allow us to fully explore the potential of these cells in the analysis of early human development, drug discovery, drug testing and repair of damaged or diseased tissues via transplantation.


Subject(s)
Embryo, Mammalian/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Gene Transfer Techniques , Humans , Mice , Pluripotent Stem Cells/physiology , Spheroids, Cellular
SELECTION OF CITATIONS
SEARCH DETAIL
...