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










Publication year range
1.
Protein Cell ; 13(10): 721-741, 2022 10.
Article in English | MEDLINE | ID: mdl-34731408

ABSTRACT

Understanding the regulatory networks for germ cell fate specification is necessary to developing strategies for improving the efficiency of germ cell production in vitro. In this study, we developed a coupled screening strategy that took advantage of an arrayed bi-molecular fluorescence complementation (BiFC) platform for protein-protein interaction screens and epiblast-like cell (EpiLC)-induction assays using reporter mouse embryonic stem cells (mESCs). Investigation of candidate interaction partners of core human pluripotent factors OCT4, NANOG, KLF4 and SOX2 in EpiLC differentiation assays identified novel primordial germ cell (PGC)-inducing factors including BEN-domain (BEND/Bend) family members. Through RNA-seq, ChIP-seq, and ATAC-seq analyses, we showed that Bend5 worked together with Bend4 and helped mark chromatin boundaries to promote EpiLC induction in vitro. Our findings suggest that BEND/Bend proteins represent a new family of transcriptional modulators and chromatin boundary factors that participate in gene expression regulation during early germline development.


Subject(s)
Chromatin , Embryonic Stem Cells , Animals , Cell Differentiation/genetics , Chromatin/metabolism , Germ Cells/metabolism , Germ Layers/metabolism , Mice
2.
PLoS Genet ; 17(11): e1009925, 2021 11.
Article in English | MEDLINE | ID: mdl-34813587

ABSTRACT

Drosophila chromosomes are elongated by retrotransposon attachment, a process poorly understood. Here we characterized a mutation affecting the HipHop telomere-capping protein. In mutant ovaries and the embryos that they produce, telomere retrotransposons are activated and transposon RNP accumulates. Genetic results are consistent with that this hiphop mutation weakens the efficacy of HP1-mediated silencing while leaving piRNA-based mechanisms largely intact. Remarkably, mutant females display normal fecundity suggesting that telomere de-silencing is compatible with germline development. Moreover, unlike prior mutants with overactive telomeres, the hiphop stock does not over-accumulate transposons for hundreds of generations. This is likely due to the loss of HipHop's abilities both to silence transcription and to recruit transposons to telomeres in the mutant. Furthermore, embryos produced by mutant mothers experience a checkpoint activation, and a further loss of maternal HipHop leads to end-to-end fusion and embryonic arrest. Telomeric retroelements fulfill an essential function yet maintain a potentially conflicting relationship with their Drosophila host. Our study thus showcases a possible intermediate in this arm race in which the host is adapting to over-activated transposons while maintaining genome stability. Our results suggest that the collapse of such a relationship might only occur when the selfish element acquires the ability to target non-telomeric regions of the genome. HipHop is likely part of this machinery restricting the elements to the gene-poor region of telomeres. Lastly, our hiphop mutation behaves as a recessive suppressor of PEV that is mediated by centric heterochromatin, suggesting its broader effect on chromatin not limited to telomeres.


Subject(s)
DNA Transposable Elements/genetics , Drosophila Proteins/genetics , Retroelements/genetics , Silencer Elements, Transcriptional/genetics , Telomere/genetics , Animals , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Drosophila melanogaster/genetics , Female , Genomic Instability/genetics , Germ Cells/metabolism , Heterochromatin/genetics , Mutation , RNA, Small Interfering/genetics
3.
Biochem Biophys Res Commun ; 574: 63-69, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34438348

ABSTRACT

Human extended pluripotent stem (hEPS) cell is a newly established human embryonic stem cell (hESC) line with the capacity of chimerizing both embryonic and extraembryonic tissues compared with primed hESCs which are inefficient to contribute to the inner cell mass (ICM). The molecular mechanism underlying the pluripotency of hEPS cells is still not clear. We conducted RNA-seq and ATAC-seq analysis to investigate the differential expression profiling and genomic chromatin accessibility features. According to our data, more than 2000 genes were specially up-regulated in hEPS cells. Furthermore, the open chromatin regions in these two human embryonic stem cell lines were quite different. In hEPS cells, transcriptional factors binding motifs associated with pluripotency maintenance were enriched in chromatin accessible regions. Integrating the results from ATAC-seq and RNA-seq, we identified new regulatory features which were important for pluripotency maintenance and cell development in hEPS cells. Together, these results provided a new perspective on the understanding of molecular features of hESCs in different pluripotent states and a novel resource for further studies on regenerative medicine by using hEPS cells.


Subject(s)
Chromatin/metabolism , Pluripotent Stem Cells/metabolism , Cells, Cultured , Humans
4.
Stem Cells ; 37(6): 743-753, 2019 06.
Article in English | MEDLINE | ID: mdl-30801858

ABSTRACT

The scaffold protein Symplekin (Sympk) is involved in cytoplasmic RNA polyadenylation, transcriptional modulation, and the regulation of epithelial differentiation and proliferation via tight junctions. It is highly expressed in embryonic stem cells (ESCs), in which its role remains unknown. In this study, we found Sympk overexpression in mouse ESCs significantly increased colony formation, and Sympk deletion via CRISPR/Cas9 decreased colony formation. Sympk promoted ESC growth and its overexpression sustained ESC pluripotency, as assessed by teratoma and chimeric mouse formation. Genomic stability was preserved in these cells after long-term passage. The domain of unknown function 3453 (DUF3453) in Sympk was required for its interaction with the key pluripotent factor Oct4, and its depletion led to impaired colony formation. Sympk activated proliferation-related genes and suppressed differentiation-related genes. Our results indicate that Sympk interacts with Oct4 to promote self-renewal and pluripotency in ESCs and preserves genome integrity; accordingly, it has potential value for stem cell therapies. Stem Cells 2019;37:743-753.


Subject(s)
Cytoskeletal Proteins/genetics , Gene Expression Regulation, Developmental , Genome , Membrane Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/genetics , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/metabolism , Animals , CRISPR-Cas Systems , Cell Differentiation , Cell Line , Cell Proliferation , Cytoskeletal Proteins/deficiency , Gene Deletion , Gene Expression Profiling , Genes, Reporter , Genomic Instability , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Membrane Proteins/deficiency , Mice , Mouse Embryonic Stem Cells/cytology , Nuclear Proteins/deficiency , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Signal Transduction , Teratoma/genetics , Teratoma/metabolism , Teratoma/pathology , Tight Junctions/metabolism
6.
Theriogenology ; 89: 365-373.e2, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27919445

ABSTRACT

Germ cell transplantation has facilitated spermatogonial stem cell (SSC) and spermatogenesis research and shown great potential in the seed-breeding of domestic livestock. However, little progress has been made in large animals, primarily reflecting the difficulties in preparing sterile recipients. Here, we developed a novel protocol to prepare recipient pigs through the direct injection of busulfan into the cavum vaginale of the scrotums of Landrace-Large bi-crossbreeding male pigs and Seghers male pigs, two economically-important types of pigs, to eliminate endogenous spermatogonia. No severe diseases or weight loss was observed in either pig type after the injection with busulfan. Histologic analysis showed an advanced and dose-dependent germ cell loss, with complete germ cell loss observed in the highest dose group, 3.0 mg/kg in the Landrace-Large bi-crossbreeding pigs and 2.0 mg/kg in the Seghers pigs. A smaller seminiferous tubule diameter, a vacuolized seminiferous epithelium and the overproliferation interstitial cells, frequently observed in mouse germ cell deficiency models, were present in the most of the high-dose busulfan-treated groups. Molecular markers detected in Seghers pigs further confirmed the depletion of endogenous germ cells, providing an accessible niche for exogenous SSCs. This study provides a basis to prepare the transplantation recipients of SSCs in pigs.


Subject(s)
Busulfan/pharmacology , Spermatogonia/drug effects , Stem Cell Transplantation/veterinary , Sterilization, Reproductive/veterinary , Swine , Animals , Male , Seminiferous Epithelium/drug effects , Seminiferous Epithelium/pathology , Seminiferous Tubules/drug effects , Seminiferous Tubules/pathology , Spermatogenesis/drug effects , Spermatogonia/cytology , Spermatogonia/transplantation , Stem Cell Transplantation/methods , Sterilization, Reproductive/methods
7.
Protein Cell ; 6(5): 363-372, 2015 May.
Article in English | MEDLINE | ID: mdl-25894090

ABSTRACT

Genome editing tools such as the clustered regularly interspaced short palindromic repeat (CRISPR)-associated system (Cas) have been widely used to modify genes in model systems including animal zygotes and human cells, and hold tremendous promise for both basic research and clinical applications. To date, a serious knowledge gap remains in our understanding of DNA repair mechanisms in human early embryos, and in the efficiency and potential off-target effects of using technologies such as CRISPR/Cas9 in human pre-implantation embryos. In this report, we used tripronuclear (3PN) zygotes to further investigate CRISPR/Cas9-mediated gene editing in human cells. We found that CRISPR/Cas9 could effectively cleave the endogenous ß-globin gene (HBB). However, the efficiency of homologous recombination directed repair (HDR) of HBB was low and the edited embryos were mosaic. Off-target cleavage was also apparent in these 3PN zygotes as revealed by the T7E1 assay and whole-exome sequencing. Furthermore, the endogenous delta-globin gene (HBD), which is homologous to HBB, competed with exogenous donor oligos to act as the repair template, leading to untoward mutations. Our data also indicated that repair of the HBB locus in these embryos occurred preferentially through the non-crossover HDR pathway. Taken together, our work highlights the pressing need to further improve the fidelity and specificity of the CRISPR/Cas9 platform, a prerequisite for any clinical applications of CRSIPR/Cas9-mediated editing.


Subject(s)
Blastocyst , CRISPR-Cas Systems , Hemoglobins, Abnormal/genetics , Hemoglobins, Abnormal/metabolism , Zygote , Humans
8.
Stem Cells ; 33(6): 1782-93, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25802002

ABSTRACT

Actl6a (actin-like protein 6A, also known as Baf53a or Arp4) is a subunit shared by multiple complexes including esBAF, INO80, and Tip60-p400, whose main components (Brg1, Ino80, and p400, respectively) are crucial for the maintenance of embryonic stem cells (ESCs). However, whether and how Actl6a functions in ESCs has not been investigated. ESCs originate from the epiblast (EPI) that is derived from the inner cell mass (ICM) in blastocysts, which also give rise to primitive endoderm (PrE). The molecular mechanisms for EPI/PrE specification remain unclear. In this study, we provide the first evidence that Actl6a can protect mouse ESCs (mESCs) from differentiating into PrE. While RNAi knockdown of Actl6a, which appeared highly expressed in mESCs and downregulated during differentiation, induced mESCs to differentiate towards the PrE lineage, ectopic expression of Actl6a was able to repress PrE differentiation. Our work also revealed that Actl6a could interact with Nanog and Sox2 and promote Nanog binding to pluripotency genes such as Oct4 and Sox2. Interestingly, cells depleted of p400, but not of Brg1 or Ino80, displayed similar PrE differentiation patterns. Mutant Actl6a with impaired ability to bind Tip60 and p400 failed to block PrE differentiation induced by Actl6a dysfunction. Finally, we showed that Actl6a could target to the promoters of key PrE regulators (e.g., Sall4 and Fgf4), repressing their expression and inhibiting PrE differentiation. Our findings uncover a novel function of Actl6a in mESCs, where it acts as a gatekeeper to prevent mESCs from entering into the PrE lineage through a Yin/Yang regulating pattern.


Subject(s)
Actins/metabolism , Blastocyst/cytology , Cell Differentiation/physiology , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Endoderm/cytology , Germ Layers/cytology , Mouse Embryonic Stem Cells/cytology , Animals , Cell Lineage/genetics , Gene Expression Regulation, Developmental/physiology , Mice , Octamer Transcription Factor-3/metabolism
9.
Cell Biochem Funct ; 31(5): 365-73, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23657870

ABSTRACT

Recent studies have demonstrated that germ-like cells could be differentiated from human umbilical cord mesenchymal stem cells (hUC-MSCs) in vitro. Whether the sexuality of hUC-MSCs affects the formation efficiency of germ-like cells derived from hUC-MSCs is still unclear. To clearly test the formation efficiency of oocyte-like cells from male and female hUC-MSCs, obtained hUC-MSCs were induced by 20% follicular fluid (FF) according to the method that has been proved by our previous studies. Results showed that hUC-MSCs differentiated into oocyte-like structures and expressed germ cell makers. It was noted that the presence of advanced oocyte-like cells in male hUC-MSCs (m-hUC-MSCs) was similar as that in female hUC-MSCs (f-hUC-MSCs); however, the expression of germ cell's specific markers in m-hUC-MSCs was delayed compared with that in f-hUC-MSCs. In addition, immunofluorescence analysis demonstrated that germ cell-specific markers, Oct4, Vasa, Dazl, ZP2, ZP3 and Stra8, were expressed on the 14th day after induction in both f-hUC-MSCs and m-hUC-MSCs. However, the size of oocyte-like cells from f-hUC-MSCs was larger than that in m-hUC-MSCs. The level of secreted oestradiol was significantly higher in f-hUC-MSCs than m-hUC-MSCs. We sought to determine whether critical germ cell's transcription factor-Figlα will promote the development of oocyte-like cells. Some germ cell-specific markers were increased when exogenous Figlα was transfected into hUC-MSCs. This process implied that germ-like cells might be produced by over-expression of exogenous germ cell-specific gene, and this process was similar as that in production of germ cells in induced pluripotent stem cells (iPSCs). Finally, to verify the feasibility that hUC-MSCs differentiate into germ cells, hUC-MSCs were transplanted into seminiferous tubules and kidney capsule of mouse, respectively, and we found the transplanted cells differentiated into germ-like cells in recipient's seminiferous tubules and kidney capsule. This study will provide a simple model to study mammalian germ cell specification using hUC-MSCs in vitro.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Oocytes/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/metabolism , Cell Differentiation , Cell Size , Cells, Cultured , Estradiol/metabolism , Female , Fetal Blood/metabolism , Gene Expression , Humans , Kidney/cytology , Kidney/metabolism , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred ICR , Oocytes/metabolism , Seminiferous Tubules/cytology , Seminiferous Tubules/metabolism , Sex Factors , Transfection , Transplantation, Heterologous
10.
Cell Biochem Funct ; 31(2): 159-65, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22961649

ABSTRACT

This study was designed to investigate the effect of platelet-derived growth factor (PDGF) on the proliferation of human umbilical cord mesenchymal stem cells (UC-MSCs) and further explore the mechanism of PDGF in promoting the proliferation of UC-MSCs. The human UC-MSCs were treated with different concentrations of PDGF, and the effects were evaluated by counting the cell number, the cell viability, the expression of PDGF receptors analyzed by RT-PCR, and the detection of the gene expression of cell proliferation, cell cycle and pluripotency, and Brdu assay by immunofluorescent staining and Quantitative real-time (QRT-PCR). The results showed that PDGF could promote the proliferation of UC-MSCs in vitro in a dose-dependent way, and 10 to 50 ng/ml PDGF had a significant proliferation effect on UC-MSCs; the most obvious concentration was 50 ng/ml. Significant inhibition on the proliferation of UC-MSCs was observed when the concentration of PDGF was higher than 100 ng/ml, and all cells died when the concentration reached 200 ng/ml PDGF. The PDGF-treated cells had stronger proliferation and antiapoptotic capacity than the control group by Brdu staining. The expression of the proliferation-related genes C-MYC, PCNA and TERT and cell cycle-related genes cyclin A, cyclin 1 and CDK2 were up-regulated in PDGF medium compared with control. However, pluripotent gene OCT4 was not significantly different between cells cultured in PDGF and cells analyzed by immunofluorescence and QRT-PCR. The PDGF could promote the proliferation of human UC-MSCs in vitro.


Subject(s)
Mesenchymal Stem Cells/cytology , Platelet-Derived Growth Factor/pharmacology , Umbilical Cord/cytology , Biomarkers/metabolism , Bromodeoxyuridine/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Humans , Infant, Newborn , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Up-Regulation/drug effects
11.
Cell Biochem Funct ; 31(1): 12-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23086862

ABSTRACT

Historically, our understanding of molecular genetic aspects of germ cell development has been limited. Recently, results demonstrated that the derivation of pluripotent stem cells may provide the necessary genetic system to study germ cell development. Here, we characterized an induced pluripotent stem cell (iPSC) line, which can spontaneously differentiate into embryonic bodies (EBs) after 3 days of suspension culture, expressing specific markers of three germ layers. Then, we induced the iPSCs to differentiate into germ cells by culturing adherent EBs in retinoic acid (RA) and porcine follicular fluid (PFF) differentiation medium or seminiferous tubule transplantation. Our results indicated that RA and PFF were beneficial for the derivation of germ cells and oocyte-like cells from iPSCs, and iPSCs transplantation could make a contribution to repairing the testis of infertile mice. Our study offers an approach for further study on the development and the differentiation of germ cells derived from iPSCs.


Subject(s)
Germ Cells/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Busulfan/toxicity , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cells, Cultured/drug effects , Culture Media/pharmacology , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Female , Follicular Fluid/physiology , Gene Expression Profiling , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/transplantation , Infertility, Male/chemically induced , Infertility, Male/surgery , Male , Mice , Seminiferous Tubules , Suspensions , Swine , Teratoma/etiology , Transplantation, Heterotopic , Tretinoin/pharmacology
12.
Cell Biochem Funct ; 30(8): 623-32, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23097316

ABSTRACT

Embryonic stem cells (ESCs) have the capacity to differentiate into nearly all sorts of cell types, including germ cells, which were regarded as one type of highly specialized cells in mammals, taking the responsibility of transferring genetic materials to the next generation. Studies on induction differentiation of murine embryonic stem cells (mESCs) into male germ cells, but with a low efficiency, basic reason is that the regulation mechanism of germ cell development in mammals is still unclear. miRNA might play an important role in spermatogenesis in mammals. In this study, several miRNAs, which might be related to spermatogenesis, were initially selected and detected in the mouse tissues by semi-polymerase chain reaction (PCR) and quantitative real time (qRT)-PCR to find a testis-specific miRNA. To study its effect on mESCs differentiation into male germ cells, miR-34c mimics were synthesized and pri-miR-34c-GFP plasmid was constructed, transfected into mESCs and combined with retinoic acid induction. The effects of miR-34c were analysed by morphology, alkaline phosphatase staining, qRT-PCR_and immunofluorescent staining. The results showed that miR-34c promoted mESCs differentiation into male germ-like cells, to some extent. Then miR-34c targeted genes were predicted by bioinformatics; Retinoic acid receptor gamma (RARg) was selected, and two dual-luciferase reporter vectors contained the normal and mutated 3'untranslated region of RARg were constructed, respectively. By miRNA mimics and vector co-transfection experiment, the predicted target gene-RARg was confirmed. In conclusion, we found a mammalian male germ cell specific miRNA--miR-34c, and it might be pivotal in mESCs differentiation into male germ cells through its target--RARg.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Fluorescent Antibody Technique , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Male , Mice , Mutation , Receptors, Retinoic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spermatozoa/cytology , Spermatozoa/metabolism , Transfection , Retinoic Acid Receptor gamma
13.
J Cell Biochem ; 113(12): 3629-42, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22821411

ABSTRACT

A small molecular chemical-Reversine has been shown to promote cell reprogramming and induce dedifferentiation of multiple terminally differentiated mesodermal origin cells, and then differentiate into other cell types within mesodermal lineages as well as neuroectodermal. However, the possibilities of these cells to give rise to germ cell lineages have not been examined. The objective of the current study was to detect the effect of Reversine on PMDSCs differentiation into germ cells. PMDSCs from fetal porcine skeletal muscle and their potential of differentiation into germ cells in vitro were investigated. The phenotype, proliferation potential, characteristic markers of the first adhesion cells (pp1), and the purified 2 times cells (pp3) were analyzed by growth curve, FACS, and RT-PCR, respectively. Then, the purified cells were induced with 10% or 20% bovine follicular fluid (FF), the results showed that some of the induced pp3 cells were similar as porcine oocyte, and expressed germ cell and oocyte markers analyzed by semi-quantitative RT-PCR and immunofluorescent staining. Reversine clearly increased the potentiality of PMDSCs differentiation into large round germ-like cells in FF induction medium analyzed by morpholgogy, QRT-PCR and immunofluoresce. The BrdU labeled PMDSCs might differentiate into female germ-like cells in recipient's kidney capsule, which were positive for germ cell and meiotic markers (Dazl, Vasa, Figla, Stra8, Scp3) and oocyte markers (Zp2, Zp3). These findings provided an efficient model to study the mechanism of cell proliferation and germ cell differentiation in livestock promoted by Reversine.


Subject(s)
Cell Differentiation/drug effects , Follicular Fluid/metabolism , Morpholines/pharmacology , Oocytes/cytology , Purines/pharmacology , Satellite Cells, Skeletal Muscle/cytology , Stem Cells/cytology , Animals , Biomarkers/metabolism , Bromodeoxyuridine , Cattle , Cell Adhesion , Cell Lineage , Cell Proliferation , Cell Shape , Culture Media/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Meiosis , Mice , Oocytes/metabolism , Ovary/cytology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Satellite Cells, Skeletal Muscle/drug effects , Stem Cell Transplantation , Stem Cells/metabolism , Swine
14.
Histochem Cell Biol ; 138(4): 593-603, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22684927

ABSTRACT

Umbilical cord (UC) has been suggested as a new source of mesenchymal stem cells (MSCs). In this report, we isolated MSCs from the fetal UC of goat and investigated their multipotency of differentiation into germ cells in vitro, in the presence of 0-20 % bovine follicular fluid (FF). The phenotypes, capacity of proliferation and expression of MSC markers were served as the indexes of multipotency of the isolated UC-MSCs, those were ascertained by growth curves, RT-PCR and immunofluorescent staining, respectively. Our results showed that the UC-MSCs shared a similar immunophenotype to those cells reported in mouse and human bone marrow MSCs, as well as some characteristics seen in embryonic stem cells (ESCs). In addition, our data also demonstrated that a dose-dependent function of FF on the states of differentiation of goat UC-MSCs. From 2 to 20 % of the FF can promote the proliferation of goat UC-MSC, especially the 5 % concentration of follicular fluid promote proliferation was significantly higher than 2 %. In contrast, higher concentration of follicular fluid (>10 %) induced goat UC-MSCs differentiation into oocyte-like cells. These findings provide an efficient model to study the mechanism on cell proliferation and germ cell differentiation in livestock using FF.


Subject(s)
Cell Differentiation , Follicular Fluid/metabolism , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Animals , Cattle , Cell Proliferation , Cells, Cultured , Dose-Response Relationship, Drug , Female , Flow Cytometry , Fluorescent Antibody Technique , Goats , Polymerase Chain Reaction
15.
J Cell Biochem ; 113(4): 1111-21, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22213070

ABSTRACT

Previous studies have demonstrated that germ cells can be derived from mouse embryonic stem cells (ESCs). However, there is still no efficient system, which can visualize the stage of germ cell specification in vitro, and further to identify and enrich germ cells derived from ESCs. Figla (factor in the germline, alpha) gene encodes a germ cell specific transcription factor that coordinates the expression of the oocyte-specific zona pellucida (Zp) genes and is essential for folliculogenesis in mouse. Here, we first constructed a pFigla-EGFP recombinant plasmid that expressed enhanced green fluorescent protein (EGFP) under the control of Figla promoter, and generated and characterized an ESC line stably carrying this pFigla-EGFP reporter construct. Then the ESCs were induced to differentiate into female germ-like cells by culturing adherent embryoid bodies (EBs) in retinoic acid (RA) induction medium or transplanting ESCs under the kidney capsule with ovarian cells. A population of differentiated ESCs expressed GFP, and these cells were analyzed by RT-PCR and immunofluorescence. The GFP positive cells showed the expression of germ cell markers Vasa, meiotic specific gene Stra8, Scp3, oocyte markers Gdf9, Zp3 and Figla, indicating that this method could be used for the purification and selection of female germ cells. Our study establishes a new selective system of female germ-like cell derivation and offers an approach for further research on the development and the differentiation of germ cells derived from stem cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Embryonic Stem Cells/cytology , Green Fluorescent Proteins/genetics , Ovum/metabolism , Promoter Regions, Genetic , Animals , Base Sequence , Cell Differentiation , Cell Line , Cell Proliferation , Cell Transplantation , DNA Primers , Embryonic Stem Cells/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...