Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
Mol Cells ; 44(10): 723-735, 2021 Oct 31.
Article in English | MEDLINE | ID: mdl-34711690

ABSTRACT

Spemann organizer is a center of dorsal mesoderm and itself retains the mesoderm character, but it has a stimulatory role for neighboring ectoderm cells in becoming neuroectoderm in gastrula embryos. Goosecoid (Gsc) overexpression in ventral region promotes secondary axis formation including neural tissues, but the role of gsc in neural specification could be indirect. We examined the neural inhibitory and stimulatory roles of gsc in the same cell and neighboring cells contexts. In the animal cap explant system, Gsc overexpression inhibited expression of neural specific genes including foxd4l1.1, zic3, ncam, and neurod. Genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) and promoter analysis of early neural genes of foxd4l1.1 and zic3 were performed to show that the neural inhibitory mode of gsc was direct. Site-directed mutagenesis and serially deleted construct studies of foxd4l1.1 promoter revealed that Gsc directly binds within the foxd4l1.1 promoter to repress its expression. Conjugation assay of animal cap explants was also performed to demonstrate an indirect neural stimulatory role for gsc. The genes for secretory molecules, Chordin and Noggin, were up-regulated in gsc injected cells with the neural fate only achieved in gsc uninjected neighboring cells. These experiments suggested that gsc regulates neuroectoderm formation negatively when expressed in the same cell and positively in neighboring cells via soluble factors. One is a direct suppressive circuit of neural genes in gsc expressing mesoderm cells and the other is an indirect stimulatory circuit for neurogenesis in neighboring ectoderm cells via secreted BMP antagonizers.


Subject(s)
Goosecoid Protein/metabolism , Neural Plate/embryology , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals
4.
Eur J Med Chem ; 210: 113068, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33310292

ABSTRACT

Glioblastoma multiform (GBM) is the highly aggressive brain tumor with poor prognosis. Glioma stem cells (GSCs), small population of cancer cells that exist in GBM tissues, resistant to chemotherapy and radiotherapy and usually driving GBM recurrence, have been developed as effective therapeutic target. Steroidal saponins are one of important resources for anti-tumor agent and may be benefited to selectively clear GSCs. In this report, total of 97 natural steroidal saponins were investigated the relationship among structures/cytotoxicity/selectivity against GSCs, glioma cell lines and human untransformed cells, and revealed that tribulosaponin A was the most potent compound. Further investigation suggested that tribulosaponin A up-regulated the expression of NCF1 and NOX1 to accumulate ROS for triggering apoptosis in GSCs, but not in untransformed cells, and it was further supported by the assay that N-acetyl-l-cysteine (NAC) clearing ROS delayed GSCs apoptosis. Besides, tribulosaponin A damaged GSCs recapturing tumor spheres formation.


Subject(s)
Antineoplastic Agents/pharmacology , Biological Products/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Goosecoid Protein/antagonists & inhibitors , Saponins/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biological Products/chemical synthesis , Biological Products/chemistry , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioblastoma/metabolism , Glioblastoma/pathology , Goosecoid Protein/metabolism , Humans , Molecular Structure , Saponins/chemical synthesis , Saponins/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Elife ; 92020 05 26.
Article in English | MEDLINE | ID: mdl-32452768

ABSTRACT

Deciphering the mechanisms of axis formation in amphioxus is a key step to understanding the evolution of chordate body plan. The current view is that Nodal signaling is the only factor promoting the dorsal axis specification in the amphioxus, whereas Wnt/ß-catenin signaling plays no role in this process. Here, we re-examined the role of Wnt/ßcatenin signaling in the dorsal/ventral patterning of amphioxus embryo. We demonstrated that the spatial activity of Wnt/ß-catenin signaling is located in presumptive dorsal cells from cleavage to gastrula stage, and provided functional evidence that Wnt/ß-catenin signaling is necessary for the specification of dorsal cell fate in a stage-dependent manner. Microinjection of Wnt8 and Wnt11 mRNA induced ectopic dorsal axis in neurulae and larvae. Finally, we demonstrated that Nodal and Wnt/ß-catenin signaling cooperate to promote the dorsal-specific gene expression in amphioxus gastrula. Our study reveals high evolutionary conservation of dorsal organizer formation in the chordate lineage.


Subject(s)
Lancelets/embryology , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Biological Evolution , Goosecoid Protein/metabolism , HEK293 Cells , Humans , Lancelets/metabolism , Nodal Protein/metabolism , Smad2 Protein/metabolism
6.
Stem Cells ; 38(8): 921-935, 2020 08.
Article in English | MEDLINE | ID: mdl-32346916

ABSTRACT

Spontaneous necrosis is a defining feature of glioblastomas (GBMs), the most malignant glioma. Despite its strong correlations with poor prognosis, it remains unclear whether necrosis could be a possible cause or mere consequence of glioma progression. Here we isolated a particular fraction of necrotic products spontaneously arising from glioma cells, morphologically and biochemically defined as autoschizis-like products (ALPs). When administered to granulocyte macrophage colony-stimulating factor (GM-CSF)-primed bone marrow-derived macrophage/dendritic cells (Mφ/DCs), ALPs were found to be specifically engulfed by Mφs expressing a tumor-associated macrophage (TAM) marker CD204. ALPs from glioma stem cells (GSCs) had higher activity for the TAM development than those from non-GSCs. Of note, expression of the Il12b gene encoding a common subunit of IL-12/23 was upregulated in ALPs-educated Mφs. Furthermore, IL-12 protein evidently enhanced the sphere-forming activity of GBM patient-derived cells, although interestingly IL-12 is generally recognized as an antitumoral M1-Mφ marker. Finally, in silico analysis of The Cancer Genome Atlas (TCGA) transcriptome data of primary and recurrent GBMs revealed that higher expression of these IL-12 family genes was well correlated with more infiltration of M1-type TAMs and closely associated with poorer prognosis in recurrent GBMs. Our results highlight a role of necrosis in GSC-driven self-beneficial niche construction and glioma progression, providing important clues for developing new therapeutic strategies against gliomas.


Subject(s)
Glioma/genetics , Goosecoid Protein/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Tumor-Associated Macrophages/metabolism , Animals , Female , Humans , Mice , Signal Transduction
7.
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
8.
Nature ; 558(7708): 132-135, 2018 06.
Article in English | MEDLINE | ID: mdl-29795348

ABSTRACT

In amniotes, the development of the primitive streak and its accompanying 'organizer' define the first stages of gastrulation. Although these structures have been characterized in detail in model organisms, the human primitive streak and organizer remain a mystery. When stimulated with BMP4, micropatterned colonies of human embryonic stem cells self-organize to generate early embryonic germ layers 1 . Here we show that, in the same type of colonies, Wnt signalling is sufficient to induce a primitive streak, and stimulation with Wnt and Activin is sufficient to induce an organizer, as characterized by embryo-like sharp boundary formation, markers of epithelial-to-mesenchymal transition and expression of the organizer-specific transcription factor GSC. Moreover, when grafted into chick embryos, human stem cell colonies treated with Wnt and Activin induce and contribute autonomously to a secondary axis while inducing a neural fate in the host. This fulfils the most stringent functional criteria for an organizer, and its discovery represents a milestone in human embryology.


Subject(s)
Nodal Protein/metabolism , Organizers, Embryonic/embryology , Organizers, Embryonic/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Activins/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Cell Line , Chick Embryo , Epithelial-Mesenchymal Transition , Goosecoid Protein/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Mice , Nerve Tissue/cytology , Nerve Tissue/embryology , Nerve Tissue/metabolism , Organizers, Embryonic/cytology , Primitive Streak/cytology , Primitive Streak/metabolism
9.
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
10.
Sci Rep ; 7: 43010, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28220837

ABSTRACT

Goosecoid (Gsc) expression marks the primary embryonic organizer in vertebrates and beyond. While functions have been assigned during later embryogenesis, the role of Gsc in the organizer has remained enigmatic. Using conditional gain-of-function approaches in Xenopus and mouse to maintain Gsc expression in the organizer and along the axial midline, neural tube closure defects (NTDs) arose and dorsal extension was compromised. Both phenotypes represent convergent extension (CE) defects, arising from impaired Wnt/planar cell polarity (PCP) signaling. Dvl2 recruitment to the cell membrane was inhibited by Gsc in Xenopus animal cap assays and key Wnt/PCP factors (RhoA, Vangl2, Prickle, Wnt11) rescued Gsc-mediated NTDs. Re-evaluation of endogenous Gsc functions in MO-mediated gene knockdown frog and knockout mouse embryos unearthed PCP/CE-related phenotypes as well, including cartilage defects in Xenopus and misalignment of inner ear hair cells in mouse. Our results assign a novel function to Gsc as an inhibitor of Wnt/PCP-mediated CE. We propose that in the organizer Gsc represses CE as well: Gsc-expressing prechordal cells, which leave the organizer first, migrate and do not undergo CE like the Gsc-negative notochordal cells, which subsequently emerge from the organizer. In this model, Gsc provides a switch between cell migration and CE, i.e. cell intercalation.


Subject(s)
Goosecoid Protein/metabolism , Organizers, Embryonic/metabolism , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/metabolism , Animals , Cell Polarity , Dishevelled Proteins/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Embryonic Development , Genes, Reporter , Goosecoid Protein/deficiency , Goosecoid Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Scanning , Signal Transduction , Xenopus Proteins/genetics
11.
Oncotarget ; 8(5): 8785-8790, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28061432

ABSTRACT

Congenital absence of the uterus and vagina (CAUV) is the most extreme female Müllerian duct abnormality. Several researches proposed that genetic factors contributed to this disorder, whereas the precise genetic mechanism is far from full elucidation. Here, utilizing whole-exome sequencing (WES), we identified one novel missense mutation in LHX1 (NM_005568: c.G1108A, p.A370T) in one of ten unrelated patients diagnosed with CAUV. This mutation was absent from public databases and our internal database. Through the luciferase reporter analysis, we found that the mutation could change the transcriptional activity of LHX1 and its effect on the regulation of the downstream target gene GSC, which might be associated with urogenital system development. In short, we concluded that the LHX1 may be a pathogenic gene of CAUV. Our results demonstrate the power of whole exome sequencing and gene prioritization approach as diagnostic tools in clinical practice that help make genetic diagnosis of CAUV.


Subject(s)
46, XX Disorders of Sex Development/genetics , Congenital Abnormalities/genetics , LIM-Homeodomain Proteins/genetics , Mullerian Ducts/abnormalities , Mutation, Missense , Transcription Factors/genetics , 46, XX Disorders of Sex Development/diagnosis , 46, XX Disorders of Sex Development/metabolism , Congenital Abnormalities/diagnosis , Congenital Abnormalities/metabolism , DNA Mutational Analysis , Female , Gene Expression Regulation , Genetic Association Studies , Genetic Markers , Genetic Predisposition to Disease , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , HEK293 Cells , Humans , LIM-Homeodomain Proteins/metabolism , Mullerian Ducts/metabolism , Phenotype , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Exome Sequencing
12.
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
13.
Cell Res ; 26(6): 699-712, 2016 06.
Article in English | MEDLINE | ID: mdl-27126000

ABSTRACT

Craniofacial anomalies (CFAs) characterized by birth defects of skull and facial bones are the most frequent congenital disease. Genomic analysis has identified multiple genes responsible for CFAs; however, the underlying genetic mechanisms for the majority of CFAs remain largely unclear. Our previous study revealed that the Wwp2 E3 ubiquitin ligase facilitates craniofacial development in part through inducing monoubiquitination and activation of the paired-like homeobox transcription factor, Goosecoid (Gsc). Here we report that Gsc is also ubiquitinated and activated by the APC(Cdh1) E3 ubiquitin ligase, leading to transcriptional activation of various Gsc target genes crucial for craniofacial development. Consistenly, neural crest-specific Cdh1-knockout mice display similar bone malformation as Wwp2-deficient mice in the craniofacial region, characterized by a domed skull, a short snout and a twisted nasal bone. Mechanistically, like Wwp2-deficient mice, mice with Cdh1 deficiency in neural crest cells exhibit reduced Gsc/Sox6 transcriptional activities. Simultaneous deletion of Cdh1 and Wwp2 results in a more severe craniofacial defect compared with single gene deletion, suggesting a synergistic augmentation of Gsc activity by these two E3 ubiquitin ligases. Hence, our study reveals a novel role for Cdh1 in craniofacial development through promoting APC-dependent non-proteolytic ubiquitination and activation of Gsc.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cdh1 Proteins/metabolism , Face/embryology , Goosecoid Protein/metabolism , Skull/embryology , Ubiquitination , Amino Acid Sequence , Animals , Goosecoid Protein/chemistry , HeLa Cells , Heterozygote , Humans , Male , Mice, Knockout , Protein Binding , SOXD Transcription Factors/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism
14.
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
15.
Nat Commun ; 6: 6853, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25902370

ABSTRACT

The amniote middle ear is a classical example of the evolutionary novelty. Although paleontological evidence supports the view that mammals and diapsids (modern reptiles and birds) independently acquired the middle ear after divergence from their common ancestor, the developmental bases of these transformations remain unknown. Here we show that lower-to-upper jaw transformation induced by inactivation of the Endothelin1-Dlx5/6 cascade involving Goosecoid results in loss of the tympanic membrane in mouse, but causes duplication of the tympanic membrane in chicken. Detailed anatomical analysis indicates that the relative positions of the primary jaw joint and first pharyngeal pouch led to the coupling of tympanic membrane formation with the lower jaw in mammals, but with the upper jaw in diapsids. We propose that differences in connection and release by various pharyngeal skeletal elements resulted in structural diversity, leading to the acquisition of the tympanic membrane in two distinct manners during amniote evolution.


Subject(s)
Ambystoma mexicanum/embryology , Endothelin-1/genetics , Lizards/embryology , Mice/embryology , Receptor, Endothelin A/genetics , Sharks/embryology , Tympanic Membrane/embryology , Ambystoma mexicanum/genetics , Animals , Base Sequence , Embryo, Mammalian , Embryo, Nonmammalian , Endothelin-1/metabolism , Evolution, Molecular , Gene Expression Regulation, Developmental , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lizards/genetics , Mandible/embryology , Maxilla/embryology , Mice/genetics , Molecular Sequence Data , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Receptor, Endothelin A/metabolism , Sharks/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Dev Biol ; 399(2): 337-47, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25601451

ABSTRACT

Organizer activity, once thought to be restricted to vertebrates, has ancient origins. However, among non-bilaterians, it has only been subjected to detailed investigation during embryonic development of the sea anemone, Nematostella vectensis. As a step toward establishing the extent to which findings in Nematostella can be generalized across the large and diverse phylum Cnidaria, we examined the expression of some key organizer and gastrulation genes during the embryonic development of the coral Acropora millepora. Although anemones and corals both belong to the cnidarian class Anthozoa, the two lineages diverged during the Cambrian and the morphological development of Acropora differs in several important respects from that of Nematostella. While the expression patterns of the key genes brachyury, bmp2/4, chordin, goosecoid and forkhead are broadly similar, developmental differences between the two species enable novel observations, and new interpretations of their significance. Specifically, brachyury expression during the flattened prawnchip stage before gastrulation, a developmental peculiarity of Acropora, leads us to suggest that it is the key gene demarcating ectoderm from endoderm in Acropora, and by implication in other cnidarians, whereas previous studies in Nematostella proposed that forkhead plays this role. Other novel observations include the transient expression of Acropora forkhead in scattered ectodermal cells shortly after gastrulation, and in the developing mesenterial filaments, with no corresponding expression reported in Nematostella. In addition, the expression patterns of goosecoid and bmp2/4 confirm the fundamental bilaterality of the Anthozoa.


Subject(s)
Anthozoa/embryology , Biological Evolution , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Organizers, Embryonic/metabolism , T-Box Domain Proteins/metabolism , Animals , Anthozoa/metabolism , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , DNA Primers/genetics , DNA, Complementary/genetics , Ectoderm/embryology , Ectoderm/metabolism , Endoderm/embryology , Endoderm/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental/genetics , Goosecoid Protein/metabolism , Image Processing, Computer-Assisted , In Situ Hybridization , Species Specificity
17.
Oncol Rep ; 32(1): 189-98, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24858567

ABSTRACT

Ovarian carcinoma is the most lethal cancer among all gynecological malignancies due to recurrence through chemoresistance. The aim of the present study was to identify new biomarkers to predict chemoresistance and prognosis in ovarian carcinomas. The mRNA expression by qRT-PCR was examined in 60 ovarian serous carcinomas for selected genes from the screening by PCR array focusing on apoptosis, epithelial-to-mesenchymal transition and cancer pathways. The clinical impact was assessed by analyzing the correlation between gene expression and clinicopathological variables. Further validation with immunohistochemistry was performed with 75 cases of serous carcinomas. The chemoresistance was significantly associated with high expression of FOS, GSC, SNAI1, TERT and TNFRSF10D, and low expression of CDKN1A, TNFRSF10A, TNFRSF10C and TRAF1 (p<0.05, t-test). Low expression of TRAF1 and high expression of E2F1, FOS, TERT and GSC were significantly associated with advanced clinical stage (p<0.05, χ2-test). Lymph node metastasis was significantly associated with high expression of GSC. The upregulation group of TERT, GSC, NOTCH1 and SNAI1, and downregulation group of TRAF1 were significantly associated with poor overall survival (p<0.05, log-rank test). On further validation with immunohistochemistry, overexpression of goosecoid homeobox (GSC) was associated with poor overall survival. The results suggest that GSC is the most potential biomarker of drug response and poor prognosis in ovarian serous carcinomas.


Subject(s)
Carcinoma, Acinar Cell/genetics , Drug Resistance, Neoplasm , Goosecoid Protein/genetics , Lymphatic Metastasis/genetics , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Acinar Cell/drug therapy , Carcinoma, Acinar Cell/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Goosecoid Protein/metabolism , Humans , Lymphatic Metastasis/diagnosis , Lymphatic Metastasis/pathology , Middle Aged , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Survival Analysis
18.
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
19.
PLoS One ; 8(2): e57698, 2013.
Article in English | MEDLINE | ID: mdl-23469052

ABSTRACT

The organizer is one of the earliest structures to be established during vertebrate development and is crucial to subsequent patterning of the embryo. We have previously shown that the SoxB1 transcription factor, Sox3, plays a central role as a transcriptional repressor of zebrafish organizer gene expression. Recent data suggest that Fgf signaling has a positive influence on organizer formation, but its role remains to be fully elucidated. In order to better understand how Fgf signaling fits into the complex regulatory network that determines when and where the organizer forms, the relationship between the positive effects of Fgf signaling and the repressive effects of the SoxB1 factors must be resolved. This study demonstrates that both fgf3 and fgf8 are required for expression of the organizer genes, gsc and chd, and that SoxB1 factors (Sox3, and the zebrafish specific factors, Sox19a and Sox19b) can repress the expression of both fgf3 and fgf8. However, we also find that these SoxB1 factors inhibit the expression of gsc and chd independently of their repression of fgf expression. We show that ectopic expression of organizer genes induced solely by the inhibition of SoxB1 function is dependent upon the activation of fgf expression. These data allow us to describe a comprehensive signaling network in which the SoxB1 factors restrict organizer formation by inhibiting Fgf, Nodal and Wnt signaling, as well as independently repressing the targets of that signaling. The organizer therefore forms only where Nodal-induced Fgf signaling overlaps with Wnt signaling and the SoxB1 proteins are absent.


Subject(s)
Fibroblast Growth Factors/metabolism , Organizers, Embryonic/cytology , Organizers, Embryonic/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Animals , Conserved Sequence , Evolution, Molecular , Fibroblast Growth Factor 3/genetics , Fibroblast Growth Factor 3/metabolism , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Goosecoid Protein/metabolism , Humans , Mesoderm/cytology , Mesoderm/metabolism , Mice , Promoter Regions, Genetic/genetics
20.
Mech Dev ; 129(9-12): 263-74, 2012.
Article in English | MEDLINE | ID: mdl-22820002

ABSTRACT

The high mobility group (HMG) proteins constitute a superfamily of nuclear proteins that regulate the expression of a wide range of genes through architectural remodeling of the chromatin structure, and the formation of multiple protein complexes on promoter/enhancer regions, but their function in germ layer specification during early development is not clear. Here we show that hmgb genes regulate mesoderm formation and dorsoventral patterning both in zebrafish and Xenopus early embryos. Overexpression of hmgb3 blocks the expression of the pan-mesoderm gene no tail/Xbra and other ventrolateral mesoderm genes, and results in embryos with shortened anteroposterior axis, while overexpression of hmgb3EnR, which contains the engrailed repressor domain, most potently repressed no tail expression and mesoderm formation. However, hmgb3VP16, which contains the transcriptional activation domain of VP16, had an opposite effect, indicating that hmgb3 may function as a repressor during mesoderm induction and patterning. In addition, we show that hmgb3 inhibits target gene expression downstream of mesoderm-inducing factors. Furthermore, using reporter gene assays in Xenopus whole embryos, we show that hmgb3 differentially regulates the activation of various mesendoderm reporter genes. In particular, it up-regulates the goosecoid, but inhibits the Xbra reporter gene activation. Therefore, our results suggest that hmgb genes may function to fine-tune the specification and/or dorsoventral patterning of mesoderm during zebrafish and Xenopus development.


Subject(s)
Body Patterning/physiology , Gene Expression Regulation, Developmental , HMGB Proteins/physiology , Xenopus/growth & development , Zebrafish/growth & development , Animals , Body Patterning/genetics , Fetal Proteins , Goosecoid Protein/genetics , Goosecoid Protein/metabolism , HMGB Proteins/genetics , HMGB Proteins/metabolism , HMGB3 Protein/genetics , HMGB3 Protein/metabolism , Mesoderm/growth & development , Mesoderm/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcriptional Activation/genetics , Up-Regulation , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...