Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Dev Biol ; 464(1): 11-23, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32450229

ABSTRACT

Development of the complex structure of the vertebrate limb requires carefully orchestrated interactions between multiple regulatory pathways and proteins. Among these, precise regulation of 5' Hox transcription factor expression is essential for proper limb bud patterning and elaboration of distinct limb skeletal elements. Here, we identified Geminin (Gmnn) as a novel regulator of this process. A conditional model of Gmnn deficiency resulted in loss or severe reduction of forelimb skeletal elements, while both the forelimb autopod and hindlimb were unaffected. 5' Hox gene expression expanded into more proximal and anterior regions of the embryonic forelimb buds in this Gmnn-deficient model. A second conditional model of Gmnn deficiency instead caused a similar but less severe reduction of hindlimb skeletal elements and hindlimb polydactyly, while not affecting the forelimb. An ectopic posterior SHH signaling center was evident in the anterior hindlimb bud of Gmnn-deficient embryos in this model. This center ectopically expressed Hoxd13, the HOXD13 target Shh, and the SHH target Ptch1, while these mutant hindlimb buds also had reduced levels of the cleaved, repressor form of GLI3, a SHH pathway antagonist. Together, this work delineates a new role for Gmnn in modulating Hox expression to pattern the vertebrate limb.


Subject(s)
Embryo, Mammalian/embryology , Geminin/metabolism , Gene Expression Regulation, Developmental , Hindlimb/embryology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Embryo, Mammalian/cytology , Geminin/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hindlimb/cytology , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Patched-1 Receptor/genetics , Patched-1 Receptor/metabolism , Transcription Factors/genetics
2.
Dev Biol ; 393(1): 44-56, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24995796

ABSTRACT

Geminin is a nucleoprotein that can directly bind chromatin regulatory complexes to modulate gene expression during development. Geminin knockout mouse embryos are preimplantation lethal by the 32-cell stage, precluding in vivo study of Geminin's role in neural development. Therefore, here we used a conditional Geminin allele in combination with several Cre-driver lines to define an essential role for Geminin during mammalian neural tube (NT) formation and patterning. Geminin was required in the NT within a critical developmental time window (embryonic day 8.5-10.5), when NT patterning and closure occurs. Geminin excision at these stages resulted in strongly diminished expression of genes that mark and promote dorsal NT identities and decreased differentiation of ventral motor neurons, resulting in completely penetrant NT defects, while excision after embryonic day 10.5 did not result in NT defects. When Geminin was deleted specifically in the spinal NT, both NT defects and axial skeleton defects were observed, but neither defect occurred when Geminin was excised in paraxial mesenchyme, indicating a tissue autonomous requirement for Geminin in developing neuroectoderm. Despite a potential role for Geminin in cell cycle control, we found no evidence of proliferation defects or altered apoptosis. Comparisons of gene expression in the NT of Geminin mutant versus wild-type siblings at embryonic day 10.5 revealed decreased expression of key regulators of neurogenesis, including neurogenic bHLH transcription factors and dorsal interneuron progenitor markers. Together, these data demonstrate a requirement for Geminin for NT patterning and neuronal differentiation during mammalian neurulation in vivo.


Subject(s)
Geminin/genetics , Neural Plate/embryology , Neural Tube Defects/genetics , Neural Tube/embryology , Neurogenesis/genetics , Animals , Apoptosis/genetics , Cell Proliferation , Chromatin , Gene Expression , Gene Expression Regulation, Developmental , Genotype , Mesoderm/embryology , Mice , Mice, Knockout , Neural Tube/abnormalities , Neurulation/genetics
3.
Stem Cells ; 31(8): 1477-87, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23630199

ABSTRACT

Embryonic cells use both growth factor signaling and cell intrinsic transcriptional and epigenetic regulation to acquire early cell fates. Underlying mechanisms that integrate these cues are poorly understood. Here, we investigated the role of Geminin, a nucleoprotein that interacts with both transcription factors and epigenetic regulatory complexes, during fate acquisition of mouse embryonic stem cells. In order to determine Geminin's role in mesendoderm formation, a process which occurs during embryonic gastrulation, we selectively over-expressed or knocked down Geminin in an in vitro model of differentiating mouse embryonic stem cells. We found that Geminin antagonizes mesendodermal fate acquisition, while these cells instead maintain elevated expression of genes associated with pluripotency of embryonic stem cells. During mesendodermal fate acquisition, Geminin knockdown promotes Wnt signaling, while Bmp, Fgf, and Nodal signaling are not affected. Moreover, we showed that Geminin facilitates the repression of mesendodermal genes that are regulated by the Polycomb repressor complex. Geminin directly binds several of these genes, while Geminin knockdown in mesendodermal cells reduces Polycomb repressor complex occupancy at these loci and increases trimethylation of histone H3 lysine 4, which correlates with active gene expression. Together, these results indicate that Geminin is required to restrain mesendodermal fate acquisition of early embryonic cells and that this is associated with both decreased Wnt signaling and enhanced Polycomb repressor complex retention at mesendodermal genes.


Subject(s)
Embryonic Stem Cells/physiology , Geminin/physiology , Mesoderm/physiology , Polycomb-Group Proteins/physiology , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Geminin/genetics , Geminin/metabolism , Gene Expression Regulation, Developmental , Mesoderm/cytology , Mesoderm/metabolism , Mice , Microarray Analysis , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Wnt Signaling Pathway
4.
Mol Cell Biol ; 32(22): 4549-60, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22949506

ABSTRACT

Regulating the transition from lineage-restricted progenitors to terminally differentiated cells is a central aspect of nervous system development. Here, we investigated the role of the nucleoprotein geminin in regulating neurogenesis at a mechanistic level during both Xenopus primary neurogenesis and mammalian neuronal differentiation in vitro. The latter work utilized neural cells derived from embryonic stem and embryonal carcinoma cells in vitro and neural stem cells from mouse forebrain. In all of these contexts, geminin antagonized the ability of neural basic helix-loop-helix (bHLH) transcription factors to activate transcriptional programs promoting neurogenesis. Furthermore, geminin promoted a bivalent chromatin state, characterized by the presence of both activating and repressive histone modifications, at genes encoding transcription factors that promote neurogenesis. This epigenetic state restrains the expression of genes that regulate commitment of undifferentiated stem and neuronal precursor cells to neuronal lineages. However, maintaining geminin at high levels was not sufficient to prevent terminal neuronal differentiation. Therefore, these data support a model whereby geminin promotes the neuronal precursor cell state by modulating both the epigenetic status and expression of genes encoding neurogenesis-promoting factors. Additional developmental signals acting in these cells can then control their transition toward terminal neuronal or glial differentiation during mammalian neurogenesis.


Subject(s)
Cell Cycle Proteins/genetics , Epigenesis, Genetic , Neurogenesis/genetics , Nuclear Proteins/genetics , Prosencephalon/metabolism , Xenopus laevis/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation , Chromatin/genetics , Chromatin/metabolism , Embryo, Nonmammalian , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Geminin , Gene Expression Regulation, Developmental , Histones/genetics , Histones/metabolism , Mice , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/metabolism , Prosencephalon/cytology , Prosencephalon/embryology , Transcriptional Activation , Xenopus Proteins , Xenopus laevis/embryology , Xenopus laevis/metabolism
5.
Proc Natl Acad Sci U S A ; 108(8): 3294-9, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21300881

ABSTRACT

Formation of the complex vertebrate nervous system begins when pluripotent cells of the early embryo are directed to acquire a neural fate. Although cell intrinsic controls play an important role in this process, the molecular nature of this regulation is not well defined. Here we assessed the role for Geminin, a nuclear protein expressed in embryonic cells, during neural fate acquisition from mouse embryonic stem (ES) cells. Whereas Geminin knockdown does not affect the ability of ES cells to maintain or exit pluripotency, we found that it significantly impairs their ability to acquire a neural fate. Conversely, Geminin overexpression promotes neural gene expression, even in the presence of growth factor signaling that antagonizes neural transcriptional responses. These data demonstrate that Geminin's activity contributes to mammalian neural cell fate acquisition. We investigated the mechanistic basis of this phenomenon and found that Geminin maintains a hyperacetylated and open chromatin conformation at neural genes. Interestingly, recombinant Geminin protein also rapidly alters chromatin acetylation and accessibility even when Geminin is combined with nuclear extract and chromatin in vitro. Together, these data support a role for Geminin as a cell intrinsic regulator of neural fate acquisition that promotes expression of neural genes by regulating chromatin accessibility and histone acetylation.


Subject(s)
Cell Cycle Proteins/physiology , Embryonic Stem Cells/cytology , Nervous System/growth & development , Nuclear Proteins/physiology , Acetylation , Animals , Chromatin/chemistry , Geminin , Histones/metabolism , Mice , Nervous System/cytology , Pluripotent Stem Cells/cytology
6.
J Cell Biochem ; 111(2): 391-401, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20506506

ABSTRACT

OCT3/4 is a POU domain transcription factor that is critical for maintenance of pluripotency and self-renewal by embryonic stem (ES) cells and cells of the early mammalian embryo. It has been demonstrated to bind and regulate a number of genes, often in conjunction with the transcription factors SOX2 and NANOG. In an effort to further understand this regulatory network, chromatin immunoprecipitation was used to prepare a library of DNA segments specifically bound by OCT3/4 in undifferentiated mouse ES (mES) cell chromatin. One segment corresponds to a region within the first intron of the gene encoding histone deacetylase 4 (Hdac4), a Class II histone deacetylase. This region acts as a transcriptional repressor and contains at least two functional sites that are specifically bound by OCT3/4. HDAC4 is not expressed in the nuclei of OCT3/4+ mES cells and is upregulated upon differentiation. These findings demonstrate the participation of OCT3/4 in the repression of Hdac4 in ES cells.


Subject(s)
Embryonic Stem Cells/metabolism , Histone Deacetylases/genetics , Octamer Transcription Factor-3/physiology , Transcription, Genetic , Animals , Binding Sites , Chromatin , DNA/metabolism , Gene Expression Regulation/genetics , Gene Regulatory Networks , Mice , Octamer Transcription Factor-3/metabolism , Transcription Factors
7.
Physiol Genomics ; 34(3): 277-84, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18523156

ABSTRACT

SOX17 is a SRY-related high-mobility group (HMG) box transcription factor that is necessary for endoderm formation in multiple species. Despite its essential function during endoderm formation and differentiation, few direct targets of SOX17 are known. To identify targets of SOX17, we isolated SOX17 binding sites with a chromatin immunoprecipitation (ChIP)-cloning screen. SOX17-ChIP identified zinc finger protein 202 (Zfp202) as a direct target of SOX17 during endoderm differentiation of F9 embryonal carcinoma cells. A sequence in the first intron of Zfp202 activated transcription in differentiated F9 cells, and overexpression of Sox17 increased the transcriptional activity of this sequence. SOX17 binds to a site within this sequence in electrophoretic mobility shift assays, and mutation of this site decreases the transcriptional activation. Zfp202 is induced concomitantly with Sox17 during endoderm differentiation of F9 cells. We also show that ZFP202 represses Hnf4a, which has been reported for the human ortholog ZNF202. Identifying targets of SOX17 will help to elucidate the molecular basis of endoderm differentiation and may provide a better understanding of the role of endoderm in patterning the other germ layers.


Subject(s)
Cell Differentiation , Endoderm/cytology , Repressor Proteins/genetics , SOXF Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Clone Cells , Cloning, Molecular , Electrophoretic Mobility Shift Assay , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 4/genetics , Humans , Mice , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins/metabolism , SOXF Transcription Factors/genetics
8.
Bioessays ; 29(10): 949-52, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17876773

ABSTRACT

In many animals, early development of the embryo is characterized by synchronous, biphasic cell divisions. These cell divisions are controlled by maternally inherited proteins and RNAs. A critical question in developmental biology is how the embryo transitions to a later pattern of asynchronous cell divisions and transfers the prior maternal control of development to the zygotic genome. The most-common model regarding how this transition from maternal to zygotic control is regulated posits that this is a consequence of the limitation of maternal gene products, due to their titration during early cell divisions. Here we discuss a recent article by Crest et al.1 that instead proposes that the balance of Cyclin-dependent Kinase 1 and Cyclin B (Cdk1-CycB) activity relative to that of the Drosophila checkpoint kinase Chk1 determines when asynchronous divisions begin.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin B/metabolism , DNA Replication , Drosophila/embryology , Protein Kinases/metabolism , Animals , Checkpoint Kinase 1 , Cyclin B1 , Drosophila/genetics , Embryo, Nonmammalian , Female , Models, Biological , Time Factors
9.
Diabetes Metab Res Rev ; 18(6): 442-50, 2002.
Article in English | MEDLINE | ID: mdl-12469358

ABSTRACT

Recent success with immunosuppression following islet cell transplantation offers hope that a cell transplantation treatment for type 1 (juvenile) diabetes may be possible if sufficient quantities of safe and effective cells can be produced. For the treatment of type 1 diabetes, the two therapeutically essential functions are the ability to monitor blood glucose levels and the production of corresponding and sufficient levels of mature insulin to maintain glycemic control. Stem cells can replicate themselves and produce cells that take on more specialized functions. If a source of stem cells capable of yielding glucose-responsive insulin-producing (GRIP) cells can be identified, then transplantation-based treatment for type 1 diabetes may become widely available. Currently, stem cells from embryonic and adult sources are being investigated for their ability to proliferate and differentiate into cells with GRIP function. Human embryonic pluripotent stem cells, commonly referred to as embryonic stem (ES) cells and embryonic germ (EG) cells, have received significant attention owing to their broad capacity to differentiate and ability to proliferate well in culture. Their application to diabetes research is of particular promise, as it has been demonstrated that mouse ES cells are capable of producing cells able to normalize glucose levels of diabetic mice, and human ES cells can differentiate into cells capable of insulin production. Cells with GRIP function have also been derived from stem cells residing in adult organisms, here referred to as endogenous stem cell sources. Independent of source, stem cells capable of producing cells with GRIP function may provide a widely available cell transplantation treatment for type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation , Stem Cell Transplantation , Animals , Blood Glucose/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , Embryo, Mammalian , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...