Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
J Adv Res ; 33: 81-98, 2021 11.
Article in English | MEDLINE | ID: mdl-34603780

ABSTRACT

Introduction: Fate determination of germline stem cells remains poorly understood at the chromatin structure level. Objectives: Our research hopes to develop successful offspring production of ovarian organoids derived from spermatogonial stem cells (SSCs) by defined factors. Methods: The offspring production from oocytes transdifferentiated from mouse SSCs with tracking of transplanted SSCs in vivo, single cell whole exome sequencing, and in 3D cell culture reconstitution of the process of oogenesis derived from SSCs. The defined factors were screened with ovarian organoids. We uncovered extensive chromatin reorganization during SSC conversion into induced germline stem cells (iGSCs) using high throughput chromosome conformation. Results: We demonstrate successful production of offspring from oocytes transdifferentiated from mouse spermatogonial stem cells (SSCs). Furthermore, we demonstrate direct induction of germline stem cells (iGSCs) differentiated into functional oocytes by transduction of H19, Stella, and Zfp57 and inactivation of Plzf in SSCs after screening with ovarian organoids. We uncovered extensive chromatin reorganization during SSC conversion into iGSCs, which was highly similar to female germline stem cells. We observed that although topologically associating domains were stable during SSC conversion, chromatin interactions changed in a striking manner, altering 35% of inactive and active chromosomal compartments throughout the genome. Conclusion: We demonstrate successful offspring production of ovarian organoids derived from SSCs by defined factors with chromatin reorganization. These findings have important implications in various areas including mammalian gametogenesis, genetic and epigenetic reprogramming, biotechnology, and medicine.


Subject(s)
Adult Germline Stem Cells , Spermatogonia , Animals , Cell Culture Techniques, Three Dimensional , Chromatin/genetics , Female , Male , Mice , Organoids
2.
Stem Cell Res ; 31: 27-30, 2018 08.
Article in English | MEDLINE | ID: mdl-30007220

ABSTRACT

A skin biopsy was obtained from a 14-year-old female patient with a history of Myelomeningocele. Dermal fibroblasts were isolated and reprogrammed with Sendai virus (SeV) vectors encoding OCT3/4, SOX2, KLF4, and c-MYC. The generated induced Pluripotent Stem Cell (iPSC) clones NTDi4_09A were free of genomically integrated reprogramming genes, had a stable normal karyotype and expressed pluripotency markers. The iPSCs formed teratomas in mice, which were differentiated towards derivatives of the three germ layers in vivo. This iPSC line offers a useful resource to study a genetic profile of a patient with spina bifida.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Spinal Dysraphism/metabolism , Female , Humans , Infant , Kruppel-Like Factor 4 , Spinal Dysraphism/pathology
3.
Cell Reprogram ; 20(1): 27-37, 2018 02.
Article in English | MEDLINE | ID: mdl-29412738

ABSTRACT

p53 is a barrier to somatic cell reprogramming. Deletion or transient suppression of p53 increases the efficiency of reprogramming of somatic cells into induced pluripotent stem cells. Whether p53 represents an obstacle to a similar process transdifferentiation of somatic cells is unknown. However, it is predicted that inhibition of p53 would promote transdifferentiation of fibroblasts into cardiomyocytes. In this study, the effect of p53 on the capacity of cardiogenic transdifferentiation is evaluated using p53 wild-type (p53+/+), p53 heterozygous mutant (p53+/-), and p53 homozygous mutant (p53-/-) mouse embryonic fibroblasts (MEFs). Repression of p53 in MEFs increases the expression level of mesoderm transcription factors Brachyury (T) and MESP1. The cardiac-specific markers, Myh6 (Myosin, Heavy Chain 6), Myh7 (Myosin, Heavy Chain 7), and cTnI (cardiac muscle troponin I), show elevated expression in p53+/- and p53-/- MEFs compared with wild-type MEFs, but cardiac muscle troponin T (cTnT) showed a lower expression level when p53 was inhibited. After induction to cardiac differentiation, cTnT expression increased and markers of endoderm and ectoderm decreased in p53+/- and p53-/- MEFs. The effect of an important reprogramming factor Oct4 on cardiac transdifferentiation was also evaluated in the allelic series of p53 MEFs. We found that overexpression of Oct4 significantly enhanced Mesp1, Tbx5, and Isl1 expression in p53+/+ and p53+/- MEFs. Oct4 also enhanced cTnT expression in all three cell lines, especially in p53+/- MEFs. Thus, inhibition of p53 expression and viral expression of Oct4 both promote transdifferentiation of MEFs into cardiomyocytes, establishing reciprocity of action in the process.


Subject(s)
Cell Transdifferentiation/genetics , Cell Transdifferentiation/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Ectoderm/cytology , Ectoderm/metabolism , Endoderm/cytology , Endoderm/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Genetic Markers , Heterozygote , Long-Term Synaptic Depression , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mutation , Tumor Suppressor Protein p53/deficiency , Up-Regulation
4.
Proc Natl Acad Sci U S A ; 113(34): 9551-6, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27512039

ABSTRACT

Understanding the mechanisms of early cardiac fate determination may lead to better approaches in promoting heart regeneration. We used a mesoderm posterior 1 (Mesp1)-Cre/Rosa26-EYFP reporter system to identify microRNAs (miRNAs) enriched in early cardiac progenitor cells. Most of these miRNA genes bear MESP1-binding sites and active histone signatures. In a calcium transient-based screening assay, we identified miRNAs that may promote the cardiomyocyte program. An X-chromosome miRNA cluster, miR-322/-503, is the most enriched in the Mesp1 lineage and is the most potent in the screening assay. It is specifically expressed in the looping heart. Ectopic miR-322/-503 mimicking the endogenous temporal patterns specifically drives a cardiomyocyte program while inhibiting neural lineages, likely by targeting the RNA-binding protein CUG-binding protein Elav-like family member 1 (Celf1). Thus, early miRNAs in lineage-committed cells may play powerful roles in cell-fate determination by cross-suppressing other lineages. miRNAs identified in this study, especially miR-322/-503, are potent regulators of early cardiac fate.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , CELF1 Protein/genetics , CELF1 Protein/metabolism , Cell Differentiation , Cell Lineage/genetics , Embryo, Mammalian , Gene Expression Profiling , Genes, Reporter , Integrases/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mesoderm/cytology , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Mice, Transgenic , MicroRNAs/metabolism , Morphogenesis/genetics , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Primary Cell Culture , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Signal Transduction
5.
Breast Cancer Res Treat ; 158(2): 263-76, 2016 07.
Article in English | MEDLINE | ID: mdl-27357812

ABSTRACT

Gametogenetin-binding protein 2 (GGNBP2) is encoded in human chromosome 17q12-q23, a region known as a breast and ovarian cancer susceptibility locus. GGNBP2, also referred to ZFP403, has a single C2H2 zinc finger and a consensus LxxLL nuclear receptor-binding motif. Here, we demonstrate that GGNBP2 expression is reduced in primary human breast tumors and in breast cancer cell lines, including T47D, MCF-7, LCC9, LY2, and MDA-MB-231 compared with normal, immortalized estrogen receptor α (ERα) negative MCF-10A and MCF10F breast epithelial cells. Overexpression of GGNBP2 inhibits the proliferation of T47D and MCF-7 ERα positive breast cancer cells without affecting MCF-10A and MCF10F. Stable GGNBP2 overexpression in T47D cells inhibits 17ß-estradiol (E2)-stimulated proliferation as well as migration, invasion, anchorage-independent growth in vitro, and xenograft tumor growth in mice. We further demonstrate that GGNBP2 protein physically interacts with ERα, inhibits E2-induced activation of estrogen response element-driven reporter activity, and attenuates ER target gene expression in T47D cells. In summary, our in vitro and in vivo findings suggest that GGNBP2 is a novel breast cancer tumor suppressor functioning as a nuclear receptor corepressor to inhibit ERα activity and tumorigenesis.


Subject(s)
Breast Neoplasms/metabolism , Down-Regulation , Estrogen Receptor alpha/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Estradiol/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Neoplasm Transplantation , Response Elements/drug effects
6.
Stem Cell Res ; 16(2): 522-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26972055

ABSTRACT

Induced pluripotent stem (iPS) cells can efficiently differentiate into the three germ layers similar to those formed by differentiated embryonic stem (ES) cells. This provides a new source of cells in which to establish preclinical allogeneic transplantation models. Our iPS cells were generated from mouse embryonic fibroblasts (MEFs) transfected with the Yamanaka factors, the four transcription factors (Oct4, Sox2, Klf4 and c-Myc), without antibiotic selection or MEF feeders. After the formation of embryoid bodies (EBs), iPS cells spontaneously differentiated into Flk1-positive cardiac progenitors and cardiomyocytes expressing cardiac-specific markers such as alpha sarcomeric actinin (α-actinin), cardiac alpha myosin heavy chain (α-MHC), cardiac troponin T (cTnT), and connexin 43 (CX43), as well as cardiac transcription factors Nk2 homebox 5 (Nkx2.5) and gata binding protein 4 (gata4). The electrophysiological activity of iPS cell-derived cardiomyocytes (iPS-CMs) was detected in beating cell clusters with optical mapping and RH237 a voltage-sensitive dye, and in single contracting cells with patch-clamp technology. Incompletely differentiated iPS cells formed teratomas when transplanted into a severe combined immunodeficiency (SCID) mouse model of myocardial infarction. Our results show that somatic cells can be reprogrammed into pluripotent stem cells, which in turn spontaneously differentiate into electrophysiologically functional mature cardiomyocytes expressing cardiac-specific makers, and that these cells can potentially be used to repair myocardial infarction (MI) in the future.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Cell Differentiation , Cell Line , Cellular Reprogramming , Connexin 43/metabolism , Disease Models, Animal , Fibroblasts/cytology , Flow Cytometry , Kruppel-Like Factor 4 , Mice , Mice, Inbred C57BL , Mice, SCID , Microscopy, Fluorescence , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/transplantation , Myosin Heavy Chains/metabolism , Patch-Clamp Techniques , Teratoma/metabolism , Teratoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Homologous , Troponin T/metabolism
7.
J Biol Chem ; 291(16): 8644-52, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26769970

ABSTRACT

Oct4 is considered a key transcription factor for pluripotent stem cell self-renewal. It binds to specific regions within target genes to regulate their expression and is downregulated upon induction of differentiation of pluripotent stem cells; however, the mechanisms that regulate the levels of human Oct4 expression remain poorly understood. Here we show that expression of human Oct4 is directly repressed by germ cell nuclear factor (GCNF), an orphan nuclear receptor, in hES cells. Knockdown of GCNF by siRNA resulted in maintenance of Oct4 expression during RA-induced hES cell differentiation. While overexpression of GCNF promoted repression of Oct4 expression in both undifferentiated and differentiated hES cells. The level of Oct4 repression was dependent on the level of GCNF expression in a dose-dependent manner. mRNA microarray analysis demonstrated that overexpression of GCNF globally regulates gene expression in undifferentiated and differentiated hES cells. Within the group of altered genes, GCNF down-regulated 36% of the genes, and up-regulated 64% in undifferentiated hES cells. In addition, GCNF also showed a regulatory gene pattern that is different from RA treatment during hES cell differentiation. These findings increase our understanding of the mechanisms that maintain hES cell pluripotency and regulate gene expression during the differentiation process.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation/physiology , Human Embryonic Stem Cells/metabolism , Nuclear Receptor Subfamily 6, Group A, Member 1/metabolism , Octamer Transcription Factor-3/biosynthesis , Repressor Proteins/metabolism , Cell Line , Human Embryonic Stem Cells/cytology , Humans , Nuclear Receptor Subfamily 6, Group A, Member 1/genetics , Octamer Transcription Factor-3/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Repressor Proteins/genetics
8.
Biol Reprod ; 94(2): 41, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26764350

ABSTRACT

The Ggnbp2 null mutant embryos died in utero between Embryonic Days 13.5 to 15.5 with dysmorphic placentae, characterized by excessive nonvascular cell nests consisting of proliferative trophoblastic tissue and abundant trophoblast stem cells (TSCs) in the labyrinth. Lethality of Ggnbp2 null embryos was caused by insufficient placental perfusion as a result of remarkable decreases in both fetal and maternal blood vessels in the labyrinth. These defects were accompanied by a significant elevation of c-Met expression and phosphorylation and its downstream effector Stat3 activation. Knockdown of Ggnbp2 in wild-type TSCs in vitro provoked the proliferation but delayed the differentiation with an upregulation of c-Met expression and an enhanced phosphorylation of c-Met and Stat3. In contrast, overexpression of Ggnbp2 in wild-type TSCs exhibited completely opposite effects compared to knockdown TSCs. These results suggest that loss of GGNBP2 in the placenta aberrantly overactivates c-Met-Stat3 signaling, alters TSC proliferation and differentiation, and ultimately compromises the structure of placental vascular labyrinth. Our studies for the first time demonstrate that GGNBP2 is an essential factor for pregnancy success acting through the maintenance of a balance of TSC proliferation and differentiation during placental development.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Placentation/genetics , Stem Cells/cytology , Trophoblasts/cytology , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Female , Mice , Mice, Transgenic , Phosphorylation , Pregnancy , Proto-Oncogene Proteins c-met/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Stem Cells/metabolism , Trophoblasts/metabolism
9.
Stem Cells ; 33(11): 3254-65, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26205879

ABSTRACT

MESP1 is considered the first sign of the nascent cardiac mesoderm and plays a critical role in the appearance of cardiac progenitors, while exhibiting a transient expression in the developing embryo. We profiled the transcriptome of a pure population of differentiating MESP1-marked cells and found that they chiefly contribute to the mesendoderm lineage. High-throughput sequencing of endogenous MESP1-bound DNA revealed that MESP1 preferentially binds to two variants of E-box sequences and activates critical mesendoderm modulators, including Eomes, Gata4, Wnt5a, Wnt5b, Mixl1, T, Gsc, and Wnt3. These mesendoderm markers were enriched in the MESP1 marked population before the appearance of cardiac progenitors and myocytes. Further, MESP1-binding is globally associated with H(3)K(27) acetylation, supporting a novel pivotal role of it in regulating target gene epigenetics. Therefore, MESP1, the pioneer cardiac factor, primarily directs the appearance of mesendoderm, the intermediary of the earliest progenitors of mesoderm and endoderm organogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Endoderm/physiology , Gene Targeting , Genome-Wide Association Study/methods , Mesoderm/physiology , Transcriptional Activation/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/physiology , Cell Line, Transformed , Cell Lineage/physiology , Embryonic Stem Cells/physiology , Endoderm/embryology , Gene Targeting/methods , Male , Mesoderm/embryology , Mice , Myocytes, Cardiac/physiology , Protein Binding/physiology
11.
Genes Dev ; 29(8): 803-16, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25877919

ABSTRACT

Embryonic stem cells are maintained in a self-renewing and pluripotent state by multiple regulatory pathways. Pluripotent-specific transcriptional networks are sequentially reactivated as somatic cells reprogram to achieve pluripotency. How epigenetic regulators modulate this process and contribute to somatic cell reprogramming is not clear. Here we performed a functional RNAi screen to identify the earliest epigenetic regulators required for reprogramming. We identified components of the SAGA histone acetyltransferase complex, in particular Gcn5, as critical regulators of reprogramming initiation. Furthermore, we showed in mouse pluripotent stem cells that Gcn5 strongly associates with Myc and that, upon initiation of somatic reprogramming, Gcn5 and Myc form a positive feed-forward loop that activates a distinct alternative splicing network and the early acquisition of pluripotency-associated splicing events. These studies expose a Myc-SAGA pathway that drives expression of an essential alternative splicing regulatory network during somatic cell reprogramming.


Subject(s)
Alternative Splicing , Cellular Reprogramming/genetics , Epigenomics , Histone Acetyltransferases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Differentiation , Cell Movement/genetics , Cells, Cultured , Embryonic Stem Cells , Gene Expression Regulation, Developmental , Histone Acetyltransferases/genetics , Mice , Pluripotent Stem Cells , RNA Interference , RNA Processing, Post-Transcriptional/genetics
12.
J Biol Chem ; 290(17): 10599-609, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25737450

ABSTRACT

All-trans retinoic acid (ATRA) is a differentiation agent that revolutionized the treatment of acute promyelocytic leukemia. However, it has not been useful for other types of acute myeloid leukemia (AML). Here we explored the effect of SALL4, a stem cell factor, on ATRA-induced AML differentiation in both ATRA-sensitive and ATRA-resistant AML cells. Aberrant SALL4 expression has been found in nearly all human AML cases, whereas, in normal bone marrow and peripheral blood cells, its expression is only restricted to hematopoietic stem/progenitor cells. We reason that, in AMLs, SALL4 activation may prevent cell differentiation and/or protect self-renewal that is seen in normal hematopoietic stem/progenitor cells. Indeed, our studies show that ATRA-mediated myeloid differentiation can be largely blocked by exogenous expression of SALL4, whereas ATRA plus SALL4 knockdown causes significantly increased AML differentiation and cell death. Mechanistic studies indicate that SALL4 directly associates with retinoic acid receptor α and modulates ATRA target gene expression. SALL4 is shown to recruit lysine-specific histone demethylase 1 (LSD1) to target genes and alter the histone methylation status. Furthermore, coinhibition of LSD1 and SALL4 plus ATRA treatment exhibited the strongest anti-AML effect. These findings suggest that SALL4 plays an unfavorable role in ATRA-based regimes, highlighting an important aspect of leukemia therapy.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Transcription Factors/antagonists & inhibitors , Tretinoin/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line, Tumor , Female , Gene Knockdown Techniques , HL-60 Cells , Hematopoietic Stem Cells/metabolism , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/genetics , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Small Interfering/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
13.
Dev Dyn ; 244(4): 619-27, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25619399

ABSTRACT

BACKGROUND: Mammalian first lineage segregation generates trophectoderm (TE) and pluripotent inner cell mass (ICM), which provides an ideal model for studying the mechanisms of maintenance and loss of pluripotency. In mouse, the transcription factor OCT4 restricts to ICM and plays a key role in TE/ICM specification and pluripotent regulatory networks. However, in pig, OCT4 does not restrict to ICM cells, suggesting a different molecular basis in TE/ICM specification and pluripotent regulatory networks. RESULTS: To explore molecular basis of porcine TE/ICM specification and pluripotent regulatory networks, we examined expression pattern of pluripotency factors, including SOX2, REX1, SALL4, ESG1, NANOG, TBX3, LIN28, KLF2, and KLF5, in porcine blastocysts. We found that SOX2 is a faithful pluripotent marker that anchored to the pluripotent cells including embryonic part cells, ICM cells and newly EPI cells along with developmental progress, whereas OCT4 expressed in almost all the cells at the same time. Consistently, analysis of spatiotemporal distribution of SOX2 and the TE marker CDX2 revealed an exclusive expression pattern in D6 blastocysts, whereas no correlation was observed between OCT4 and CDX2 at the same stage. CONCLUSIONS: Our results provide a molecular basis in porcine embryonic patterning and a clue for further studying porcine pluripotent regulatory networks.


Subject(s)
Gene Expression Regulation, Developmental , SOXB1 Transcription Factors/metabolism , Swine/embryology , Animals , Blastocyst/cytology , Blastocyst Inner Cell Mass/metabolism , Cell Differentiation , Cell Lineage , Gene Expression Profiling , Gene Regulatory Networks , Octamer Transcription Factor-3/metabolism , Oocytes/cytology , Pluripotent Stem Cells/metabolism
14.
PLoS One ; 9(8): e103985, 2014.
Article in English | MEDLINE | ID: mdl-25140725

ABSTRACT

Expression of germ cell nuclear factor (GCNF; Nr6a1), an orphan member of the nuclear receptor gene family of transcription factors, during gastrulation and neurulation is critical for normal embryogenesis in mice. Gcnf represses the expression of the POU-domain transcription factor Oct4 (Pou5f1) during mouse post-implantation development. Although Gcnf expression is not critical for the embryonic segregation of the germ cell lineage, we found that sexually dimorphic expression of Gcnf in germ cells correlates with the expression of pluripotency-associated genes, such as Oct4, Sox2, and Nanog, as well as the early meiotic marker gene Stra8. To elucidate the role of Gcnf during mouse germ cell differentiation, we generated an ex vivo Gcnf-knockdown model in combination with a regulated CreLox mutation of Gcnf. Lack of Gcnf impairs normal spermatogenesis and oogenesis in vivo, as well as the derivation of germ cells from embryonic stem cells (ESCs) in vitro. Inactivation of the Gcnf gene in vivo leads to loss of repression of Oct4 expression in both male and female gonads.


Subject(s)
Gametogenesis/physiology , Gonads/growth & development , Nuclear Receptor Subfamily 6, Group A, Member 1/metabolism , Animals , Female , Gene Expression Regulation, Developmental , Gonads/metabolism , Male , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 6, Group A, Member 1/genetics , Octamer Transcription Factor-3/metabolism
15.
Stem Cells ; 32(6): 1527-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24578347

ABSTRACT

Cyclin D1 plays an important role in the regulation of cellular proliferation and its expression is activated during gastrulation in the mouse; however, it remains unknown how cyclin D1 expression is regulated during early embryonic development. Here, we define the role of germ cell nuclear factor (GCNF) in the activation of cyclin D1 expression during embryonic stem cell (ESC) differentiation as a model of early development. During our study of GCNF knockout (GCNF(-) (/) (-) ) ESC, we discovered that loss of GCNF leads to the repression of cyclin D1 activation during ESC differentiation. This was determined to be an indirect effect of deregulation Mir302a, which is a cyclin D1 suppressor via binding to the 3'UTR of cyclin D1 mRNA. Moreover, we showed that Mir302 is a target gene of GCNF that inhibits Mir302 expression by binding to a DR0 element within its promoter. Inhibition of Mir302a using Mir302 inhibitor during differentiation of GCNF(-) (/) (-) ESCs restored cyclin D1 expression. Similarly over-expression of GCNF during differentiation of GCNF(-) (/) (-) ESCs rescued the inhibition of Mir302a expression and the activation of cyclin D1. These results reveal that GCNF plays a key role in regulating activation of cyclin D1 expression via inhibition of Mir302a.


Subject(s)
Cell Differentiation/genetics , Cyclin D1/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Nuclear Receptor Subfamily 6, Group A, Member 1/metabolism , Repressor Proteins/metabolism , Animals , Cell Proliferation , Cell Shape , Colony-Forming Units Assay , Cyclin D1/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Nuclear Receptor Subfamily 6, Group A, Member 1/deficiency , Promoter Regions, Genetic/genetics , Protein Binding/genetics
16.
Proc Natl Acad Sci U S A ; 111(5): E572-81, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449888

ABSTRACT

The roles of microRNAs (miRNAs) and the miRNA processing machinery in the regulation of stem cell biology are not well understood. Here, we show that the p53 family member and p63 isoform, ΔNp63, is a transcriptional activator of a cofactor critical for miRNA processing (DGCR8). This regulation gives rise to a unique miRNA signature resulting in reprogramming cells to multipotency. Strikingly, ΔNp63(-/-) epidermal cells display profound defects in terminal differentiation and express a subset of markers and miRNAs present in embryonic stem cells and fibroblasts induced to pluripotency using Yamanaka factors. Moreover, ΔNp63(-/-) epidermal cells transduced with an inducible DGCR8 plasmid can differentiate into multiple cell fates in vitro and in vivo. We found that human primary keratinocytes depleted of ΔNp63 or DGCR8 can be reprogrammed in 6 d and express a unique miRNA and gene expression signature that is similar but not identical to human induced pluripotent stem cells. Our data reveal a role for ΔNp63 in the transcriptional regulation of DGCR8 to reprogram adult somatic cells into multipotent stem cells.


Subject(s)
Down-Regulation/genetics , Keratinocytes/metabolism , Multipotent Stem Cells/cytology , Phosphoproteins/genetics , Proteins/genetics , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Adult , Animals , Cell Differentiation , Cell Line , Cell Lineage , Cell Proliferation , Chimera , Embryo, Mammalian/cytology , Epidermal Cells , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Keratinocytes/cytology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Multipotent Stem Cells/metabolism , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Phosphoproteins/deficiency , Phosphoproteins/metabolism , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Trans-Activators/deficiency , Trans-Activators/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism
17.
Nucleic Acids Res ; 42(1): 205-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24078252

ABSTRACT

How tumor suppressor p53 selectively responds to specific signals, especially in normal cells, is poorly understood. We performed genome-wide profiling of p53 chromatin interactions and target gene expression in human embryonic stem cells (hESCs) in response to early differentiation, induced by retinoic acid, versus DNA damage, caused by adriamycin. Most p53-binding sites are unique to each state and define stimulus-specific p53 responses in hESCs. Differentiation-activated p53 targets include many developmental transcription factors and, in pluripotent hESCs, are bound by OCT4 and NANOG at chromatin enriched in both H3K27me3 and H3K4me3. Activation of these genes occurs with recruitment of p53 and H3K27me3-specific demethylases, UTX and JMJD3, to chromatin. In contrast, genes associated with cell migration and motility are bound by p53 specifically after DNA damage. Surveillance functions of p53 in cell death and cell cycle regulation are conserved in both DNA damage and differentiation. Comparative genomic analysis of p53-targets in mouse and human ESCs supports an inter-species divergence in p53 regulatory functions during evolution. Our findings expand the registry of p53-regulated genes to define p53-regulated opposition to pluripotency during early differentiation, a process highly distinct from stress-induced p53 response in hESCs.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Animals , Binding Sites , Cell Line , DNA Damage , Embryonic Stem Cells/cytology , Genome, Human , Histone Demethylases/metabolism , Histones/metabolism , Humans , Mice , Transcription Factors/metabolism
18.
PLoS One ; 8(9): e74556, 2013.
Article in English | MEDLINE | ID: mdl-24040281

ABSTRACT

Loss of Dicer, an enzyme critical for microRNA biogenesis, results in lethality due to a block in mouse embryonic stem cell (mES) differentiation. Using ChIP-Seq we found increased H3K9me2 at over 900 CpG islands in the Dicer(-/-)ES epigenome. Gene ontology analysis revealed that promoters of chromatin regulators to be among the most impacted by increased CpG island H3K9me2 in ES (Dicer(-/-)). We therefore, extended the study to include H3K4me3 and H3K27me3 marks for selected genes. We found that the ES (Dicer(-/-)) mutant epigenome was characterized by a shift in the overall balance between transcriptionally favorable (H3K4me3) and unfavorable (H3K27me3) marks at key genes regulating ES cell differentiation. Pluripotency genes Oct4, Sox2 and Nanog were not impacted in relation to patterns of H3K27me3 and H3K4me3 and showed no changes in the rates of transcript down-regulation in response to RA. The most striking changes were observed in regards to genes regulating differentiation and the transition from self-renewal to differentiation. An increase in H3K4me3 at the promoter of Lin28b was associated with the down-regulation of this gene at a lower rate in Dicer(-/-)ES as compared to wild type ES. An increase in H3K27me3 in the promoters of differentiation genes Hoxa1 and Cdx2 in Dicer(-/-)ES cells was coincident with an inability to up-regulate these genes at the same rate as ES upon retinoic acid (RA)-induced differentiation. We found that siRNAs Ezh2 and post-transcriptional silencing of Ezh2 by let-7 g rescued this effect suggesting that Ezh2 up-regulation is in part responsible for increased H3K27me3 and decreased rates of up-regulation of differentiation genes in Dicer(-/-)ES.


Subject(s)
Cell Differentiation/drug effects , Chromatin/metabolism , DEAD-box RNA Helicases/genetics , Embryonic Stem Cells/drug effects , Gene Expression Regulation, Developmental/drug effects , Ribonuclease III/genetics , Tretinoin/pharmacology , Animals , CDX2 Transcription Factor , Cells, Cultured , CpG Islands , DEAD-box RNA Helicases/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Histones/genetics , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Methylation , Mice , Mice, Knockout , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins , Ribonuclease III/deficiency , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Semin Cell Dev Biol ; 24(10-12): 679-86, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24029702

ABSTRACT

GCNF (NR6A1) is essential for embryonic development. GCNF belongs to the nuclear receptor (NR) gene family, it is distantly related to other NRs and is the only member of subfamily 6. As the ligand for GCNF has not been identified, GCNF is designated an orphan nuclear receptor. GCNF has been found to be a transcriptional repressor, through specific binding to DR0 response elements, which is found in the Oct4 proximal promoter for example. GCNF is expressed widely in early mouse embryos, and later in the developing nervous system. GCNF knockout mouse embryos die around E10.5. GCNF is required for the restriction of Oct4 expression to primordial germ cells after gastrulation. GCNF is expressed in ES/EC cells and during their differentiation, and has been reported to be required for pluripotency gene repression during retinoic acid (RA)-induced mES cell differentiation. GCNF can interact with DNA methylation proteins, and is suggested to recruit DNA methylation complexes to repress and silence Oct4 expression. Nuclear receptor regulation in embryonic development is a complex process, as different nuclear receptors have overlapping and distinct functions. In-depth exploration of GCNF function and mechanism of action will help to comprehensively understand the nuclear receptor regulation in embryonic development.


Subject(s)
Embryonic Development , Nuclear Receptor Subfamily 6, Group A, Member 1/metabolism , Amino Acid Sequence , Animals , DNA/metabolism , Humans , Molecular Sequence Data , Nuclear Receptor Subfamily 6, Group A, Member 1/chemistry , Nuclear Receptor Subfamily 6, Group A, Member 1/genetics , Protein Multimerization , Repressor Proteins/metabolism , Response Elements/genetics
20.
Biol Reprod ; 89(4): 95, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24006280

ABSTRACT

Proper regulation of trophoblast proliferation, differentiation, and function are critical for placenta development and function. The RNA-binding protein, LIN28A, has been well characterized as a potent regulator of differentiation in embryonic stem cells; however, little is known about the function of LIN28A in the placenta. We assessed LIN28A in vitro using mouse trophoblast stem (mTS) cells and human trophoblast cells (ACH-3P). We observed that LIN28A decreased and let-7 miRNA increased when mTS cells were induced to differentiate into mouse trophoblast giant cells (mTGCs) upon the removal of FGF4, heparin and conditioned medium. Similarly, we observed that LIN28A decreased in ACH-3P cells induced to syncytialize with forskolin treatment. To assess LIN28A in vivo we examined Embryonic Day 11.5 mouse placenta and observed abundant LIN28A in the chorioallantoic interface and labyrinth layer, with little LIN28A staining in spongiotrophoblast or differentiated mTGCs. Additionally, shRNA-mediated LIN28A knockdown in ACH-3P cells resulted in increased spontaneous syncytialization, and increased levels of syncytiotrophoblast markers hCG, LGALS13, and ERVW-1 mRNA. Additionally, targeted degradation of LIN28A mRNA increased responsiveness to forskolin-induced differentiation. In contrast, targeted degradation of Lin28a mRNA in mTS cells did not alter cell phenotype when maintained under proliferative culture conditions. Together, these data establish that LIN28A has a functional role in regulating trophoblast differentiation and function, and that loss of LIN28A in human trophoblast is sufficient to induce differentiation, but does not induce differentiation in the mouse.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Models, Biological , RNA-Binding Proteins/metabolism , Trophoblasts/metabolism , Animals , Cell Line , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Humans , Mice , MicroRNAs/metabolism , Placenta/cytology , Placenta/embryology , Placenta/metabolism , Placentation , Pregnancy , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Species Specificity , Trophoblasts/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...