Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Cell ; 147(7): 1511-24, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22196728

ABSTRACT

Specific chromatin marks keep master regulators of differentiation silent yet poised for activation by extracellular signals. We report that nodal TGF-ß signals use the poised histone mark H3K9me3 to trigger differentiation of mammalian embryonic stem cells. Nodal receptors induce the formation of companion Smad4-Smad2/3 and TRIM33-Smad2/3 complexes. The PHD-Bromo cassette of TRIM33 facilitates binding of TRIM33-Smad2/3 to H3K9me3 and H3K18ac on the promoters of mesendoderm regulators Gsc and Mixl1. The crystal structure of this cassette, bound to histone H3 peptides, illustrates that PHD recognizes K9me3, and Bromo binds an adjacent K18ac. The interaction between TRIM33-Smad2/3 and H3K9me3 displaces the chromatin-compacting factor HP1γ, making nodal response elements accessible to Smad4-Smad2/3 for Pol II recruitment. In turn, Smad4 increases K18 acetylation to augment TRIM33-Smad2/3 binding. Thus, nodal effectors use the H3K9me3 mark as a platform to switch master regulators of stem cell differentiation from the poised to the active state.


Subject(s)
Chromatin Assembly and Disassembly , Embryonic Stem Cells/metabolism , Histones/metabolism , Smad Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , Goosecoid Protein/genetics , Homeodomain Proteins/genetics , Humans , Mice , Models, Molecular , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Alignment
2.
Stem Cells ; 37(11): 1401-1415, 2019 11.
Article in English | MEDLINE | ID: mdl-31348575

ABSTRACT

Histone methyltransferases play a critical role in early human development, whereas their roles and precise mechanisms are less understood. SET and MYND domain-containing protein 2 (SMYD2) is a histone lysine methyltransferase induced during early differentiation of human embryonic stem cells (hESCs), but little is known about its function in undifferentiated hESCs and in their early lineage fate decision as well as underlying mechanisms. Here, we explored the role of SMYD2 in the self-renewal and mesendodermal lineage commitment of hESCs. We demonstrated that the expression of SMYD2 was significantly enhanced during mesendodermal but not neuroectodermal differentiation of hESCs. SMYD2 knockout (SMYD2-/- ) did not affect self-renewal and early neuroectodermal differentiation of hESCs, whereas it blocked the mesendodermal lineage commitment. This phenotype was rescued by reintroduction of SMYD2 into the SMYD2-/- hESCs. Mechanistically, the bindings of SMYD2 at the promoter regions of critical mesendodermal transcription factor genes, namely, brachyury (T), eomesodermin (EOMES), mix paired-like homeobox (MIXL1), and goosecoid homeobox (GSC) were significantly enhanced during mesendodermal differentiation of SMYD2+/+ hESCs but totally suppressed in SMYD2-/- ones. Concomitantly, such a suppression was associated with the remarkable reduction of methylation at histone 3 lysine 4 and lysine 36 but not at histone 4 lysine 20 globally and specifically on the promoter regions of mesendodermal genes, namely, T, EOMES, MIXL1, and GSC. These results reveal that the histone methyltransferase SMYD2 is dispensable in the undifferentiated hESCs and the early neuroectodermal differentiation, but it promotes the mesendodermal differentiation of hESCs through the epigenetic control of critical genes to mesendodermal lineage commitment. Stem Cells 2019;37:1401-1415.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Base Sequence , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Fetal Proteins/genetics , Fetal Proteins/metabolism , Flow Cytometry , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcriptional Activation/genetics , Transcriptional Activation/physiology
3.
Development ; 143(8): 1340-50, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26952988

ABSTRACT

Embryonic development relies on activating and repressing regulatory influences that are faithfully integrated at the core promoter of individual genes. In vertebrates, the basal machinery recognizing the core promoter includes TATA-binding protein (TBP) and two TBP-related factors. In Xenopus embryos, the three TBP family factors are all essential for development and are required for expression of distinct subsets of genes. Here, we report on a non-canonical TBP family-insensitive (TFI) mechanism of transcription initiation that involves mesoderm and organizer gene expression. Using TBP family single- and triple-knockdown experiments, α-amanitin treatment, transcriptome profiling and chromatin immunoprecipitation, we found that TFI gene expression cannot be explained by functional redundancy, is supported by active transcription and shows normal recruitment of the initiating form of RNA polymerase II to the promoter. Strikingly, recruitment of Gcn5 (also known as Kat2a), a co-activator that has been implicated in transcription initiation, to TFI gene promoters is increased upon depletion of TBP family factors. TFI genes are part of a densely connected TBP family-insensitive T-box-Otx2-Gsc interaction network. The results indicate that this network of genes bound by Vegt, Eomes, Otx2 and Gsc utilizes a novel, flexible and non-canonical mechanism of transcription that does not require TBP or TBP-related factors.


Subject(s)
Gene Expression Regulation, Developmental , Gene Regulatory Networks , Goosecoid Protein/genetics , Otx Transcription Factors/genetics , TATA-Box Binding Protein/metabolism , Transcription Initiation, Genetic , Xenopus Proteins/genetics , Animals , Gastrulation , Gene Knockdown Techniques , Histone Acetyltransferases/metabolism , Otx Transcription Factors/metabolism , Protein Binding , TATA Box Binding Protein-Like Proteins/genetics , Xenopus , Xenopus Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 113(44): E6840-E6848, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27791112

ABSTRACT

Neurons of the Statoacoustic Ganglion (SAG), which innervate the inner ear, originate as neuroblasts in the floor of the otic vesicle and subsequently delaminate and migrate toward the hindbrain before completing differentiation. In all vertebrates, locally expressed Fgf initiates SAG development by inducing expression of Neurogenin1 (Ngn1) in the floor of the otic vesicle. However, not all Ngn1-positive cells undergo delamination, nor has the mechanism controlling SAG delamination been elucidated. Here we report that Goosecoid (Gsc), best known for regulating cellular dynamics in the Spemann organizer, regulates delamination of neuroblasts in the otic vesicle. In zebrafish, Fgf coregulates expression of Gsc and Ngn1 in partially overlapping domains, with delamination occurring primarily in the zone of overlap. Loss of Gsc severely inhibits delamination, whereas overexpression of Gsc greatly increases delamination. Comisexpression of Ngn1 and Gsc induces ectopic delamination of some cells from the medial wall of the otic vesicle but with a low incidence, suggesting the action of a local inhibitor. The medial marker Pax2a is required to restrict the domain of gsc expression, and misexpression of Pax2a is sufficient to block delamination and fully suppress the effects of Gsc The opposing activities of Gsc and Pax2a correlate with repression or up-regulation, respectively, of E-cadherin (cdh1). These data resolve a genetic mechanism controlling delamination of otic neuroblasts. The data also elucidate a developmental role for Gsc consistent with a general function in promoting epithelial-to-mesenchymal transition (EMT).


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Ganglia, Parasympathetic/growth & development , Ganglia, Parasympathetic/metabolism , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Organizers, Embryonic , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Animals , Cadherins/metabolism , Cell Differentiation/genetics , Ear, Inner/metabolism , Epithelial-Mesenchymal Transition/physiology , Ganglia, Parasympathetic/pathology , Gastrulation , Gene Expression Regulation, Developmental , Genes, Overlapping , Immunohistochemistry , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/genetics , Organizers, Embryonic/pathology , PAX2 Transcription Factor/metabolism , Signal Transduction , Up-Regulation , Vestibulocochlear Nerve/growth & development , Vestibulocochlear Nerve/metabolism , Zebrafish/genetics , Zebrafish/metabolism
5.
J Biol Chem ; 292(27): 11178-11188, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28500134

ABSTRACT

Poly-ubiquitination-mediated RUNX2 degradation is an important cause of age- and inflammation-related bone loss. NEDD4 family E3 ubiquitin protein ligases are thought to be the major regulators of RUNX2 poly-ubiquitination. However, we observed a mono-ubiquitination of RUNX2 that was catalyzed by WWP2, a member of the NEDD4 family of E3 ubiquitin ligases. WWP2 has been reported to catalyze the mono-ubiquitination of Goosecoid in chondrocytes, facilitating craniofacial skeleton development. In this study, we found that osteogenic differentiation of mesenchymal stem cells promoted WWP2 expression and nuclear accumulation. Knockdown of Wwp2 in mesenchymal stem cells and osteoblasts led to significant deficiencies of osteogenesis, including decreased mineral deposition and down-regulation of osteogenic marker genes. Co-immunoprecipitation experiments showed the interaction of WWP2 with RUNX2 in vitro and in vivo Mono-ubiquitination by WWP2 leads to RUNX2 transactivation, as evidenced by the wild type of WWP2, but not its ubiquitin ligase-dead mutant, augmenting RUNX2-reponsive reporter activity. Moreover, deletion of WWP2-dependent mono-ubiquitination resulted in striking defects of RUNX2 osteoblastic activity. In addition, ectopic expression of the constitutively active type 1A bone morphogenetic protein receptor enhanced WWP2-dependent RUNX2 ubiquitination and transactivation, demonstrating a regulatory role of bone morphogenetic protein signaling in the WWP2-RUNX2 axis. Taken together, our results provide evidence that WWP2 serves as a positive regulator of osteogenesis by augmenting RUNX2 transactivation in a non-proteolytic mono-ubiquitination manner.


Subject(s)
Cell Differentiation/physiology , Core Binding Factor Alpha 1 Subunit/metabolism , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , Transcriptional Activation/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Animals , Cell Line , Core Binding Factor Alpha 1 Subunit/genetics , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mice , Osteoblasts/cytology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/genetics
6.
Breast Cancer Res ; 20(1): 2, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29291741

ABSTRACT

BACKGROUND: Disseminated tumor cells (DTCs) found in the bone marrow (BM) of patients with breast cancer portend a poor prognosis and are thought to be intermediaries in the metastatic process. To assess the clinical relevance of a mouse model for identifying possible prognostic and predictive biomarkers of these cells, we have employed patient-derived xenografts (PDX) for propagating and molecularly profiling human DTCs. METHODS: Previously developed mouse xenografts from five breast cancer patients were further passaged by implantation into NOD/SCID mouse mammary fat pads. BM was collected from long bones at early, serial passages and analyzed for human-specific gene expression by qRT-PCR as a surrogate biomarker for the detection of DTCs. Microarray-based gene expression analyses were performed to compare expression profiles between primary xenografts, solid metastasis, and populations of BM DTCs. Differential patterns of gene expression were then compared to previously generated microarray data from primary human BM aspirates from patients with breast cancer and healthy volunteers. RESULTS: Human-specific gene expression of SNAI1, GSC, FOXC2, KRT19, and STAM2, presumably originating from DTCs, was detected in the BM of all xenograft mice that also developed metastatic tumors. Human-specific gene expression was undetectable in the BM of those xenograft lines with no evidence of distant metastases and in non-transplanted control mice. Comparative gene expression analysis of BM DTCs versus the primary tumor of one mouse line identified multiple gene transcripts associated with epithelial-mesenchymal transition, aggressive clinical phenotype, and metastatic disease development. Sixteen of the PDX BM associated genes also demonstrated a statistically significant difference in expression in the BM of healthy volunteers versus the BM of breast cancer patients with distant metastatic disease. CONCLUSION: Unique and reproducible patterns of differential gene expression can be identified that presumably originate from BM DTCs in mouse PDX lines. Several of these identified genes are also detected in the BM of patients with breast cancer who develop early metastases, which suggests that they may be clinically relevant biomarkers. The PDX model may also provide a clinically relevant system for analyzing and targeting these intermediaries of metastases.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Neoplasm Metastasis/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Cells/pathology , Breast Neoplasms/pathology , Disease Models, Animal , Endosomal Sorting Complexes Required for Transport/genetics , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , Goosecoid Protein/genetics , Humans , Keratin-19/genetics , Mice , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , Snail Family Transcription Factors/genetics , Xenograft Model Antitumor Assays
7.
J Biol Chem ; 291(7): 3333-45, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26694615

ABSTRACT

Inscuteable (Insc) regulates cell fate decisions in several types of stem cells. Although it is recognized that the expression levels of mouse INSC govern the balance between symmetric and asymmetric stem cell division, regulation of mouse Insc gene expression remains poorly understood. Here, we showed that mouse Insc expression transiently increases at an early stage of differentiation, when mouse embryonic stem (mES) cells differentiate into bipotent mesendoderm capable of producing both endoderm and mesoderm in defined culture conditions. We identified the minimum transcriptional regulatory element (354 bases) that drives mouse Insc transcription in mES cells within a region >5 kb upstream of the mouse Insc transcription start site. We found that the transcription factor reticuloendotheliosis oncogene (c-Rel) bound to the minimum element and promoted mouse Insc expression in mES cells. In addition, short interfering RNA-mediated knockdown of either mouse INSC or c-Rel protein decreased mesodermal cell populations without affecting differentiation into the mesendoderm or endoderm. Furthermore, overexpression of mouse INSC rescued the mesoderm-reduced phenotype induced by knockdown of c-Rel. We propose that regulation of mouse Insc expression by c-Rel modulates cell fate decisions during mES cell differentiation.


Subject(s)
Cell Cycle Proteins/agonists , Cell Differentiation , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chromatin Immunoprecipitation , Endoderm/cytology , Endoderm/metabolism , Genes, Reporter , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-rel/antagonists & inhibitors , Proto-Oncogene Proteins c-rel/genetics , RNA Interference , RNA, Small Interfering , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Regulatory Elements, Transcriptional , Transcription Initiation Site
8.
Am J Hum Genet ; 93(6): 1135-42, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24290375

ABSTRACT

Short stature, auditory canal atresia, mandibular hypoplasia, and skeletal abnormalities (SAMS) has been reported previously to be a rare, autosomal-recessive developmental disorder with other, unique rhizomelic skeletal anomalies. These include bilateral humeral hypoplasia, humeroscapular synostosis, pelvic abnormalities, and proximal defects of the femora. To identify the genetic basis of SAMS, we used molecular karyotyping and whole-exome sequencing (WES) to study small, unrelated families. Filtering of variants from the WES data included segregation analysis followed by comparison of in-house exomes. We identified a homozygous 306 kb microdeletion and homozygous predicted null mutations of GSC, encoding Goosecoid homeobox protein, a paired-like homeodomain transcription factor. This confirms that SAMS is a human malformation syndrome resulting from GSC mutations. Previously, Goosecoid has been shown to be a determinant at the Xenopus gastrula organizer region and a segment-polarity determinant in Drosophila. In the present report, we present data on Goosecoid protein localization in staged mouse embryos. These data and the SAMS clinical phenotype both suggest that Goosecoid is a downstream effector of the regulatory networks that define neural-crest cell-fate specification and subsequent mesoderm cell lineages in mammals, particularly during shoulder and hip formation. Our findings confirm that Goosecoid has an essential role in human craniofacial and joint development and suggest that Goosecoid is an essential regulator of mesodermal patterning in mammals and that it has specific functions in neural crest cell derivatives.


Subject(s)
Abnormalities, Multiple/genetics , Bone and Bones/abnormalities , Dwarfism/genetics , Ear Canal/abnormalities , Goosecoid Protein/genetics , Mandible/abnormalities , Mutation , Abnormalities, Multiple/diagnosis , Adult , Animals , Child , DNA Mutational Analysis , Female , Genetic Association Studies , Homozygote , Humans , Male , Mice , Pedigree , Phenotype , Syndrome , Young Adult
9.
J Craniofac Surg ; 27(6): e583-6, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27526242

ABSTRACT

Treacher Collins syndrome is an autosomal dominant craniofacial malformation mainly caused by mutations in the TCOF1 gene. Few cases have been observed in the Chinese population. Herein, the authors report the mutational analysis of TCOF1, GSC, and HOXA2 to determine the mutational features of the 3 genes in Chinese patients with Treacher Collins syndrome. Genomic DNA of the patients and their parents was extracted from peripheral blood following a standard protocol. DNA sequencing analysis was performed on all exons and the exon-intron borders of TCOF1, GSC, and HOXA2 in addition to the 1200-bp upstream of TCOF1. Four novel single nucleotide polymorphisms were detected in TCOF1, one of which was in the promoter region. Mutations in GSC and HOXA2 were not found in the 3 patients. Our results suggest the possibility of genetic heterogeneity or different mechanisms leading to the disease. Further functional study of the alteration is necessary to obtain more definitive information.


Subject(s)
DNA Mutational Analysis/methods , Goosecoid Protein/genetics , Homeodomain Proteins/genetics , Mandibulofacial Dysostosis/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Humans , Polymorphism, Single Nucleotide/genetics
10.
Biochim Biophys Acta ; 1839(7): 559-70, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24844180

ABSTRACT

In mammals, an array of MEF2C proteins is generated by alternative splicing (AS), yet specific functions have not been ascribed to each isoform. Teleost fish possess two MEF2C paralogues, mef2ca and mef2cb. In zebrafish, the Mef2cs function to promote cardiomyogenic differentiation and myofibrillogenesis in nascent skeletal myofibers. We found that zebrafish mef2ca and mef2cb are alternatively spliced in the coding exons 4-6 region and these splice variants differ in their biological activity. Of the two, mef2ca is more abundantly expressed in developing skeletal muscle, its activity is tuned through zebrafish development by AS. By 24hpf, we found the prevalent expression of the highly active full length protein in differentiated muscle in the somites. The splicing isoform of mef2ca that lacks exon 5 (mef2ca 4-6), encodes a protein that has 50% lower transcriptional activity, and is found mainly earlier in development, before muscle differentiation. mef2ca transcripts including exon 5 (mef2ca 4-5-6) are present early in the embryo. Over-expression of this isoform alters the expression of genes involved in early dorso-ventral patterning of the embryo such as chordin, nodal related 1 and goosecoid, and induces severe developmental defects. AS of mef2cb generates a long splicing isoform in the exon 5 region (Mef2cbL) that predominates during somitogenesis. Mef2cbL contains an evolutionarily conserved domain derived from exonization of a fragment of intron 5, which confers the ability to induce ectopic muscle in mesoderm upon over-expression of the protein. Taken together, the data show that AS is a significant regulator of Mef2c activity.


Subject(s)
Cell Differentiation/genetics , MEF2 Transcription Factors/genetics , Muscle Development/genetics , Muscle Proteins/genetics , Zebrafish Proteins/genetics , Alternative Splicing/genetics , Animals , Gene Expression Regulation, Developmental , Glycoproteins/biosynthesis , Glycoproteins/genetics , Goosecoid Protein/biosynthesis , Goosecoid Protein/genetics , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Nodal Signaling Ligands/biosynthesis , Nodal Signaling Ligands/genetics , Protein Isoforms/genetics , RNA Splicing/genetics , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/biosynthesis
11.
PLoS Genet ; 8(10): e1002999, 2012.
Article in English | MEDLINE | ID: mdl-23071453

ABSTRACT

Holoprosencephaly (HPE) is a remarkably common congenital anomaly characterized by failure to define the midline of the forebrain and midface. HPE is associated with heterozygous mutations in Sonic hedgehog (SHH) pathway components, but clinical presentation is extremely variable and many mutation carriers are unaffected. It has been proposed that these observations are best explained by a multiple-hit model, in which the penetrance and expressivity of an HPE mutation is enhanced by a second mutation or the presence of cooperating, but otherwise silent, modifier genes. Non-genetic risk factors are also implicated in HPE, and gene-environment interactions may provide an alternative multiple-hit model to purely genetic multiple-hit models; however, there is little evidence for this contention. We report here a mouse model in which there is dramatic synergy between mutation of a bona fide HPE gene (Cdon, which encodes a SHH co-receptor) and a suspected HPE teratogen, ethanol. Loss of Cdon and in utero ethanol exposure in 129S6 mice give little or no phenotype individually, but together produce defects in early midline patterning, inhibition of SHH signaling in the developing forebrain, and a broad spectrum of HPE phenotypes. Our findings argue that ethanol is indeed a risk factor for HPE, but genetically predisposed individuals, such as those with SHH pathway mutations, may be particularly susceptible. Furthermore, gene-environment interactions are likely to be important in the multifactorial etiology of HPE.


Subject(s)
Cell Adhesion Molecules/genetics , Ethanol/adverse effects , Holoprosencephaly/chemically induced , Holoprosencephaly/genetics , Maternal Exposure/adverse effects , Mutation , Signal Transduction , Animals , Brain/abnormalities , Craniofacial Abnormalities/chemically induced , Craniofacial Abnormalities/genetics , Developmental Disabilities/chemically induced , Developmental Disabilities/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Goosecoid Protein/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Holoprosencephaly/embryology , Mice , Mice, 129 Strain , Mice, Knockout , Neural Tube Defects/chemically induced , Neural Tube Defects/embryology , Neural Tube Defects/genetics , Phenotype , Signal Transduction/drug effects
12.
J Appl Toxicol ; 34(1): 66-75, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23378141

ABSTRACT

Fetal alcohol spectrum disorder (FASD) is a set of developmental malformations caused by excess alcohol consumption during pregnancy. Using an in vitro system, we examined the role that chronic ethanol (EtOH) exposure plays in gene expression changes during the early stage of embryonic differentiation. We demonstrated that EtOH affected the cell morphology, cell cycle progression and also delayed the down-regulation of OCT4 and NANOG during differentiation. Gene expression profiling and pathway analysis demonstrated that EtOH deregulates many genes and pathways that are involved in early embryogenesis. Follow-up analyzes revealed that EtOH exposure to embryoid bodies (EBs) induced the expression of an organizer-specific gene, goosecoid (GSC), in comparison to controls. Moreover, EtOH treatment altered several important genes that are involved in embryonic structure formation, nervous system development, and placental and embryonic vascularization, which are all common processes that FASD can disrupt. Specifically, EtOH treatment let to a reduction in ALDOC, ENO2 and CDH1 expression, whereas EtOH treatment induced the expression of PTCH1, EGLN1, VEGFA and DEC2 in treated EBs. We also found that folic acid (FA) treatment was able to correct the expression of the majority of genes deregulated by EtOH exposure during early embryo development. Finally, the present study identified a gene set including GSC, which was deregulated by EtOH exposure that may contribute to the etiology of fetal alcohol syndrome (FAS). We also reported that EtOH-induced GSC expression is mediated by Nodal signaling, which may provide a new avenue for analyzing the molecular mechanisms behind EtOH teratogenicity in FASD individuals.


Subject(s)
Ethanol/adverse effects , Fetal Alcohol Spectrum Disorders/genetics , Goosecoid Protein/metabolism , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Cluster Analysis , Down-Regulation , Embryonic Development/drug effects , Female , Gene Expression/drug effects , Gene Expression Profiling , Goosecoid Protein/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Microarray Analysis , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Placenta/drug effects , Pregnancy , Reproducibility of Results , Signal Transduction
13.
Mol Cells ; 47(4): 100058, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38522664

ABSTRACT

A comprehensive regulatory network of transcription factors controls the dorsoventral patterning of the body axis in developing vertebrate embryos. Bone morphogenetic protein signaling is essential for activating the Ventx family of homeodomain transcription factors, which regulates embryonic patterning and germ layer identity during Xenopus gastrulation. Although Ventx1.1 and Ventx2.1 of the Xenopus Ventx family have been extensively investigated, Ventx3.2 remains largely understudied. Therefore, this study aimed to investigate the transcriptional regulation of ventx3.2 during the embryonic development of Xenopus. We used goosecoid (Gsc) genome-wide chromatin immunoprecipitation-sequencing data to isolate and replicate the promoter region of ventx3.2. Serial deletion and site-directed mutagenesis were used to identify the cis-acting elements for Gsc and caudal type homeobox 1 (Cdx1) within the ventx3.2 promoter. Cdx1 and Gsc differentially regulated ventx3.2 transcription in this study. Additionally, positive cis-acting and negative response elements were observed for Cdx1 and Gsc, respectively, within the 5' flanking region of the ventx3.2 promoter. This result was corroborated by mapping the active Cdx1 response element (CRE) and Gsc response element (GRE). Moreover, a point mutation within the CRE and GRE completely abolished the activator and repressive activities of Cdx1 and Gsc, respectively. Furthermore, the chromatin immunoprecipitation-polymerase chain reaction confirmed the direct binding of Cdx1 and Gsc to the CRE and GRE, respectively. Inhibition of Cdx1 and Gsc activities at their respective functional regions, namely, the ventral marginal zone and dorsal marginal zone, reversed their effects on ventx3.2 transcription. These results indicate that Cdx1 and Gsc modulate ventx3.2 transcription in the ventral marginal zone and dorsal marginal zone by directly binding to the promoter region during Xenopus gastrulation.


Subject(s)
Gastrula , Homeodomain Proteins , Promoter Regions, Genetic , Xenopus Proteins , Xenopus laevis , Animals , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Gastrula/metabolism , Gene Expression Regulation, Developmental , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription, Genetic , Xenopus laevis/genetics , Xenopus laevis/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
14.
Dev Biol ; 368(2): 231-41, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22627292

ABSTRACT

Signaling inputs from multiple pathways are essential for the establishment of distinct cell and tissue types in the embryo. Therefore, multiple signals must be integrated to activate gene expression and confer cell fate, but little is known about how this occurs at the level of target gene promoters. During early embryogenesis, Wnt and Nodal signals are required for formation of the Spemann organizer, which is essential for germ layer patterning and axis formation. Signaling by both Wnt and Nodal pathways is required for the expression of multiple organizer genes, suggesting that integration of these signals is required for organizer formation. Here, we demonstrate transcriptional cooperation between the Wnt and Nodal pathways in the activation of the organizer genes Goosecoid (Gsc), Cerberus (Cer), and Chordin (Chd). Combined Wnt and Nodal signaling synergistically activates transcription of these organizer genes. Effectors of both pathways occupy the Gsc, Cer and Chd promoters and effector occupancy is enhanced with active Wnt and Nodal signaling. This suggests that, at organizer gene promoters, a stable transcriptional complex containing effectors of both pathways forms in response to combined Wnt and Nodal signaling. Consistent with this idea, the histone acetyltransferase p300 is recruited to organizer promoters in a Wnt and Nodal effector-dependent manner. Taken together, these results offer a mechanism for spatial and temporal restriction of organizer gene transcription by the integration of two major signaling pathways, thus establishing the Spemann organizer domain.


Subject(s)
Nodal Protein/metabolism , Organizers, Embryonic/metabolism , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Body Patterning/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Goosecoid Protein/genetics , Immunohistochemistry , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Nodal Protein/genetics , Organizers, Embryonic/embryology , Promoter Regions, Genetic/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Time Factors , Transcriptional Activation , Wnt Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
15.
Dev Biol ; 362(1): 94-103, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22178155

ABSTRACT

TGFß signaling patterns the primitive streak, yet little is known about transcriptional effectors that mediate the cell fate choices during streak-like development in mammalian embryos and in embryonic stem (ES) cells. Here we demonstrate that cross-antagonistic actions of EVEN-SKIPPED HOMEOBOX 1 (EVX1) and GOOSECOID (GSC) regulate cell fate decisions in streak-like progenitors derived from human ES cells exposed to BMP4 and/or activin. We found that EVX1 repressed GSC expression and promoted formation of posterior streak-like progeny in response to BMP4, and conversely that GSC repressed EVX1 expression and was required for development of anterior streak-like progeny in response to activin. Chromatin immunoprecipitation assays showed that EVX1 bound to the GSC 5'-flanking region in BMP4 treated human ES cells, and band shift assays identified two EVX1 binding sites in the GSC 5'-region. Significantly, we found that intact EVX1 binding sites were required for BMP4-mediated repression of GSC reporter constructs. We conclude that BMP4-induced EVX1 repress GSC directly and the two genes form the core of a gene regulatory network (GRN) controlling cell fates in streak-like human ES cell progeny.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental/physiology , Gene Regulatory Networks/genetics , Goosecoid Protein/metabolism , Homeodomain Proteins/metabolism , Primitive Streak/embryology , Activins/metabolism , Analysis of Variance , Binding Sites/genetics , Blotting, Western , Bone Morphogenetic Protein 4/metabolism , Chromatin Immunoprecipitation , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Flow Cytometry , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Goosecoid Protein/genetics , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Luciferases , Primitive Streak/cytology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Proc Natl Acad Sci U S A ; 107(6): 2521-6, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20133783

ABSTRACT

Dorsal organizer formation is one of the most critical steps in early embryonic development. Several genes and signaling pathways that positively regulate the dorsal organizer development have been identified; however, little is known about the factor(s) that negatively regulates the organizer formation. Here, we show that Setdb2, a SET domain-containing protein possessing potential histone H3K9 methyltransferase activity, restricts dorsal organizer development and regulates left-right asymmetry by suppressing fibroblast growth factor 8 (fgf8) expression. Knockdown of Setdb2 results in a massive expansion of dorsal organizer markers floating head (flh), goosecoid (gsc), and chordin (chd), as well as a significant increase of fgf8, but not fgf4 mRNAs. Consequently, disrupted midline patterning and resultant randomization of left-right asymmetry are observed in Setdb2-deficient embryos. These characteristic changes induced by Setdb2 deficiency can be nearly corrected by either overexpression of a dominant-negative fgf receptor or knockdown of fgf8, suggesting an essential role for Setdb2-Fgf8 signaling in restricting dorsal organizer territory and regulating left-right asymmetry. These results provide unique evidence that a SET domain-containing protein potentially involved in the epigenetic control negatively regulates dorsal organizer formation during early embryonic development.


Subject(s)
Body Patterning/genetics , Embryo, Nonmammalian/metabolism , Fibroblast Growth Factor 8/genetics , Histone-Lysine N-Methyltransferase/genetics , Zebrafish Proteins/genetics , Amino Acid Sequence , Animals , Embryo, Nonmammalian/embryology , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Glycoproteins/genetics , Goosecoid Protein/genetics , Homeodomain Proteins/genetics , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Transcription Factors/genetics
17.
Proc Natl Acad Sci U S A ; 107(35): 15449-54, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20713713

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) produces cancer cells that are invasive, migratory, and exhibit stem cell characteristics, hallmarks of cells that have the potential to generate metastases. Inducers of the EMT include several transcription factors (TFs), such as Goosecoid, Snail, and Twist, as well as the secreted TGF-beta1. Each of these factors is capable, on its own, of inducing an EMT in the human mammary epithelial (HMLE) cell line. However, the interactions between these regulators are poorly understood. Overexpression of each of the above EMT inducers up-regulates a subset of other EMT-inducing TFs, with Twist, Zeb1, Zeb2, TGF-beta1, and FOXC2 being commonly induced. Up-regulation of Slug and FOXC2 by either Snail or Twist does not depend on TGF-beta1 signaling. Gene expression signatures (GESs) derived by overexpressing EMT-inducing TFs reveal that the Twist GES and Snail GES are the most similar, although the Goosecoid GES is the least similar to the others. An EMT core signature was derived from the changes in gene expression shared by up-regulation of Gsc, Snail, Twist, and TGF-beta1 and by down-regulation of E-cadherin, loss of which can also trigger an EMT in certain cell types. The EMT core signature associates closely with the claudin-low and metaplastic breast cancer subtypes and correlates negatively with pathological complete response. Additionally, the expression level of FOXC1, another EMT inducer, correlates strongly with poor survival of breast cancer patients.


Subject(s)
Breast Neoplasms/genetics , Claudins/genetics , Epithelial Cells/metabolism , Gene Expression Profiling , Mesoderm/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cluster Analysis , Down-Regulation , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Goosecoid Protein/genetics , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors/genetics , Transforming Growth Factor beta1/genetics , Twist-Related Protein 1/genetics
18.
Cells ; 12(6)2023 03 11.
Article in English | MEDLINE | ID: mdl-36980215

ABSTRACT

The reciprocal inhibition between two signaling centers, the Spemann organizer (dorsal mesoderm) and ventral region (mesoderm and ectoderm), collectively regulate the overall development of vertebrate embryos. Each center expresses key homeobox transcription factors (TFs) that directly control target gene transcription. Goosecoid (Gsc) is an organizer (dorsal mesoderm)-specific TF known to induce dorsal fate and inhibit ventral/ectodermal specification. Ventx1.1 (downstream of Bmp signaling) induces the epidermal lineage and inhibits dorsal organizer-specific genes from the ventral region. Chordin (Chrd) is an organizer-specific secreted Bmp antagonist whose expression is primarily activated by Gsc. Alternatively, chrd expression is repressed by Bmp/Ventx1.1 in the ventral/epidermal region. However, the regulatory mechanisms underlying the transcription mediated by Gsc and Ventx1.1 remain elusive. Here, we found that the chrd promoter contained two cis-acting response elements that responded negatively to Ventx1.1 and positively to Gsc. In the ventral/ectodermal region, Ventx1.1 was directly bound to the Ventx1.1 response element (VRE) and inhibited chrd transcription. In the organizer region, Gsc was bound to the Gsc response elements (GRE) to activate chrd transcription. The Gsc-mediated positive response on the chrd promoter completely depended on another adjacent Wnt response cis-acting element (WRE), which was the TCF7 (also known as Tcf1) binding element. Site-directed mutagenesis of VRE, GRE, or WRE completely abolished the repressive or activator activity of Ventx1.1 and Gsc, respectively. The ChIP-PCR results confirmed the direct binding of Ventx1.1 and Gsc/Tcf7 to VRE and GRE/WRE, respectively. These results demonstrated that chrd expression is oppositely modulated by homeobox TFs, Ventx1.1, and Gsc/Tcf7 during the embryonic patterning of Xenopus gastrula.


Subject(s)
Gastrula , Glycoproteins , Goosecoid Protein , Transcription Factors , Xenopus Proteins , Xenopus laevis , Animals , Gastrula/metabolism , Genes, Homeobox , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Glycoproteins/metabolism
19.
Dev Biol ; 352(2): 367-81, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21295564

ABSTRACT

The Spemann organizer is an essential signaling center in Xenopus germ layer patterning and axis formation. Organizer formation occurs in dorsal blastomeres receiving both maternal Wnt and zygotic Nodal signals. In response to stabilized ßcatenin, dorsal blastomeres express the closely related transcriptional activators, Siamois (Sia) and Twin (Twn), members of the paired homeobox family. Sia and Twn induce organizer formation and expression of organizer-specific genes, including Goosecoid (Gsc). In spite of the similarity of Sia and Twn sequence and expression pattern, it is unclear whether these factors function equivalently in promoter binding and subsequent transcriptional activation, or if Sia and Twn are required for all aspects of organizer function. Here we report that Sia and Twn activate Gsc transcription by directly binding to a conserved P3 site within the Wnt-responsive proximal element of the Gsc promoter. Sia and Twn form homodimers and heterodimers by direct homeodomain interaction and dimer forms are indistinguishable in both DNA-binding and activation functions. Sequential chromatin immunoprecipitation reveals that the endogenous Gsc promoter can be occupied by either Sia or Twn homodimers or Sia-Twn heterodimers. Knockdown of Sia and Twn together, but not individually, results in a failure of organizer gene expression and a disruption of axis formation, consistent with a redundant role for Sia and Twn in organizer formation. Furthermore, simultaneous knockdown of Sia and Twn blocks axis induction in response to ectopic Wnt signaling, demonstrating an essential role for Sia and Twn in mediating the transcriptional response to the maternal Wnt pathway. The results demonstrate the functional redundancy of Sia and Twn and their essential role in direct transcriptional responses necessary for Spemann organizer formation.


Subject(s)
Homeodomain Proteins/metabolism , Organizers, Embryonic/embryology , Organizers, Embryonic/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , Animals , Base Sequence , Binding Sites/genetics , Body Patterning , Conserved Sequence , DNA/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Protein Multimerization , Sequence Homology, Nucleic Acid , Signal Transduction , Wnt Proteins/genetics , Wnt Proteins/metabolism , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus laevis/genetics
20.
Dev Dyn ; 240(6): 1422-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21538683

ABSTRACT

We have previously shown that the Gsx family homeobox gene Gsh2 is part of the regulatory network specifying dorsoventral pattern of primary neurons in the developing amphibian embryo. Here, we investigate the role of Gsx transcription factors in regulating the transcription of Iroquois family homeobox genes in the amphibian neural plate. Iroquois genes are key regulators of neural patterning and their expression is coincident with that of the Gsx genes during open neural plate stages. We show that Gsx proteins repress Iroquois expression in the embryo and conversely, inhibition of Gsx activity with either antisense morpholino oligos or an anti-morphic Gsx protein up-regulates Iroquois expression. These data indicate that Gsx factors act as negative regulators of Iroquois gene expression in the amphibian neural plate and support a model in which the Gsx proteins promote neuronal differentiation by repressing the expression of known inhibitors of neuronal differentiation such as Iro3.


Subject(s)
Amphibians/embryology , Amphibians/genetics , Gene Expression Regulation, Developmental , Goosecoid Protein/physiology , Homeodomain Proteins/genetics , Animals , Animals, Genetically Modified , Cells, Cultured , Down-Regulation/genetics , Embryo, Nonmammalian , Goosecoid Protein/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Models, Biological , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL