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










Publication year range
1.
J Cell Sci ; 137(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38639717

ABSTRACT

Activation of the Wnt-ß-catenin signaling pathway by CHIR99021, a specific inhibitor of GSK3ß, induces Tcf7l1 protein degradation, which facilitates the maintenance of an undifferentiated state in mouse embryonic stem cells (mESCs); however, the precise mechanism is still unclear. Here, we showed that the overexpression of transducin-ß-like protein 1 (Tbl1, also known as Tbl1x) or its family member Tblr1 (also known as Tbl1xr1) can decrease Tcf7l1 protein levels, whereas knockdown of each gene increases Tcf7l1 levels without affecting Tcf7l1 transcription. Interestingly, only Tbl1, and not Tblr1, interacts with Tcf7l1. Mechanistically, Tbl1 translocates from the cytoplasm into the nucleus in association with ß-catenin (CTNNB1) after the addition of CHIR99021 and functions as an adaptor to promote ubiquitylation of the Tcf7l1 protein. Functional assays further revealed that enforced expression of Tbl1 is capable of delaying mESC differentiation. In contrast, knockdown of Tbl1 attenuates the effect of CHIR99021 on Tcf7l1 protein stability and mESC self-renewal. Our results provide insight into the regulatory network of the Wnt-ß-catenin signaling pathway involved in promoting the maintenance of naïve pluripotency.


Subject(s)
Mouse Embryonic Stem Cells , Proteolysis , Transcription Factor 7-Like 1 Protein , Ubiquitination , Wnt Signaling Pathway , beta Catenin , Animals , Mice , Mouse Embryonic Stem Cells/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factor 7-Like 1 Protein/genetics , beta Catenin/metabolism , Proteolysis/drug effects , Cell Differentiation/drug effects , Pyridines/pharmacology , beta-Transducin Repeat-Containing Proteins/metabolism , beta-Transducin Repeat-Containing Proteins/genetics , Pyrimidines/pharmacology , Humans
2.
Biochem Biophys Res Commun ; 621: 88-93, 2022 09 17.
Article in English | MEDLINE | ID: mdl-35810596

ABSTRACT

The basic helix-loop-helix (bHLH) family is one of the most conserved transcription factor families that plays an important role in regulating cell growth, differentiation and tissue development. Typically, members of this family form homo- or heterodimers to recognize specific motifs and activate transcription. MyoD is a vital transcription factor that regulates muscle cell differentiation. However, it is necessary for MyoD to form a heterodimer with E-proteins to activate transcription. Even though the crystal structure of the MyoD homodimer has been determined, the structure of the MyoD heterodimer in complex with the E-box protein remains unclear. In this study, we determined the crystal structure of the bHLH domain of the MyoD-E47 heterodimer at 2.05 Å. Our structural analysis revealed that MyoD interacts with E47 through a hydrophobic interface. Moreover, we confirmed that heterodimerization could enhance the binding affinity of MyoD to E-box sequences. Our results provide new structural insights into the heterodimer of MyoD and E-box protein, suggesting the molecular mechanism of transcription activation of MyoD upon binding to E-box protein.


Subject(s)
DNA-Binding Proteins , MyoD Protein , DNA-Binding Proteins/metabolism , Helix-Loop-Helix Motifs , MyoD Protein/metabolism , Protein Binding , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factors/metabolism
3.
Cell Rep ; 36(11): 109705, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525377

ABSTRACT

Mouse embryonic stem cells (mESCs) can be maintained in vitro in defined N2B27 medium supplemented with two chemical inhibitors for GSK3 and MEK (2i) and the cytokine leukemia inhibitory factor (LIF), which act synergistically to promote self-renewal and pluripotency. Here, we find that genetic deletion of the four genes encoding the TCF/LEF transcription factors confers mESCs with the ability to self-renew in N2B27 medium alone. TCF/LEF quadruple knockout (qKO) mESCs display dysregulation of several genes, including Aire, Dnmt3l, and IcosL, located adjacent to each other within a topologically associated domain (TAD). Aire, Dnmt3l, and IcosL appear to be regulated by TCF/LEF in a ß-catenin independent manner. Moreover, downregulation of Aire and Dnmt3l in wild-type mESCs mimics the loss of TCF/LEF and increases mESC survival in the absence of 2iL. Hence, this study identifies TCF/LEF effectors that mediate exit from the pluripotent state.


Subject(s)
Cell Self Renewal , Hepatocyte Nuclear Factor 1-alpha/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Transcription Factor 7-Like 1 Protein/genetics , Transcription Factor 7-Like 2 Protein/genetics , Animals , Benzamides/pharmacology , Cell Self Renewal/drug effects , Culture Media/chemistry , Culture Media/pharmacology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Down-Regulation/drug effects , Gene Editing , Hepatocyte Nuclear Factor 1-alpha/deficiency , Hepatocyte Nuclear Factor 1-alpha/metabolism , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Ligand/metabolism , Lymphoid Enhancer-Binding Factor 1/deficiency , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factor 7-Like 1 Protein/deficiency , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factor 7-Like 2 Protein/deficiency , Transcription Factor 7-Like 2 Protein/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/deficiency , beta Catenin/genetics , AIRE Protein
4.
Elife ; 102021 02 15.
Article in English | MEDLINE | ID: mdl-33587034

ABSTRACT

The canonical Wnt pathway transcriptional co-activator ß-catenin regulates self-renewal and differentiation of mammalian nephron progenitor cells (NPCs). We modulated ß-catenin levels in NPC cultures using the GSK3 inhibitor CHIR99021 (CHIR) to examine opposing developmental actions of ß-catenin. Low CHIR-mediated maintenance and expansion of NPCs are independent of direct engagement of TCF/LEF/ß-catenin transcriptional complexes at low CHIR-dependent cell-cycle targets. In contrast, in high CHIR, TCF7/LEF1/ß-catenin complexes replaced TCF7L1/TCF7L2 binding on enhancers of differentiation-promoting target genes. Chromosome confirmation studies showed pre-established promoter-enhancer connections to these target genes in NPCs. High CHIR-associated de novo looping was observed in positive transcriptional feedback regulation to the canonical Wnt pathway. Thus, ß-catenin's direct transcriptional role is restricted to the induction of NPCs, where rising ß-catenin levels switch inhibitory TCF7L1/TCF7L2 complexes to activating LEF1/TCF7 complexes at primed gene targets poised for rapid initiation of a nephrogenic program.


Subject(s)
Lymphoid Enhancer-Binding Factor 1/metabolism , Nephrons/metabolism , Stem Cells/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism , Animals , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Lymphoid Enhancer-Binding Factor 1/genetics , Mice , Nephrons/cytology , Nephrons/embryology , Promoter Regions, Genetic , Protein Binding , Stem Cells/cytology , Transcription Factor 7-Like 1 Protein/genetics , Transcription Factors/genetics
5.
PLoS One ; 15(10): e0236612, 2020.
Article in English | MEDLINE | ID: mdl-33104714

ABSTRACT

A precisely balanced activity of canonical Wnt signaling is essential for a number of biological processes and its perturbation leads to developmental defects or diseases. Here, we demonstrate that alternative isoforms of the KDM2A and KDM2B lysine demethylases have the ability to negatively regulate canonical Wnt signaling. These KDM2A and KDM2B isoforms (KDM2A-SF and KDM2B-SF) lack the N-terminal demethylase domain, but they still have the ability to bind to CpG islands in promoters and to interact with their protein partners via their other functional domains. We have observed that KDM2A-SF and KDM2B-SF bind to the promoters of axin 2 and cyclin D1, two canonical Wnt signaling target genes, and repress their activity. Moreover, KDM2A-SF and KDM2B-SF are both able to strongly repress a Wnt-responsive luciferase reporter. The transcriptional repression mediated by KDM2A-SF and KDM2B-SF, but also by KDM2A-LF, is dependent on their DNA binding domain, while the N-terminal demethylase domain is dispensable for this process. Surprisingly, KDM2B-LF is unable to repress both the endogenous promoters and the luciferase reporter. Finally, we show that both KDM2A-SF and KDM2B-SF are able to interact with TCF7L1, one of the transcriptional mediators of canonical Wnt signaling. KDM2A-SF and KDM2B-SF are thus likely to negatively affect the transcription of canonical Wnt signaling target genes by binding to their promoters and by interacting with TCF7L1 and other co-repressors.


Subject(s)
Cyclin D1/metabolism , F-Box Proteins/metabolism , Gene Expression Regulation , Jumonji Domain-Containing Histone Demethylases/metabolism , Promoter Regions, Genetic , Transcription Factor 7-Like 1 Protein/metabolism , Wnt Signaling Pathway , CpG Islands , Cyclin D1/genetics , F-Box Proteins/genetics , HEK293 Cells , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Lysine/genetics , Lysine/metabolism , Protein Isoforms , Transcription Factor 7-Like 1 Protein/genetics
6.
J Biol Chem ; 295(49): 16826-16839, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32989053

ABSTRACT

The Wnt/ß-catenin pathway is one of the major pathways that regulates embryonic development, adult homeostasis, and stem cell self-renewal. In this pathway, transcription factors T-cell factor and lymphoid enhancer factor (TCF/LEF) serve as a key switch to repress or activate Wnt target gene transcription by recruiting repressor molecules or interacting with the ß-catenin effector, respectively. It has become evident that the protein stability of the TCF/LEF family members may play a critical role in controlling the activity of the Wnt/ß-catenin signaling pathway. However, factors that regulate the stability of TCF/LEFs remain largely unknown. Here, we report that pVHL binding protein 1 (VBP1) regulates the Wnt/ß-catenin signaling pathway by controlling the stability of TCF/LEFs. Surprisingly, we found that either overexpression or knockdown of VBP1 decreased Wnt/ß-catenin signaling activity in both cultured cells and zebrafish embryos. Mechanistically, VBP1 directly binds to all four TCF/LEF family members and von Hippel-Lindau tumor-suppressor protein (pVHL). Either overexpression or knockdown of VBP1 increases the association between TCF/LEFs and pVHL and then decreases the protein levels of TCF/LEFs via proteasomal degradation. Together, our results provide mechanistic insights into the roles of VBP1 in controlling TCF/LEFs protein stability and regulating Wnt/ß-catenin signaling pathway activity.


Subject(s)
Cytoskeletal Proteins/metabolism , Molecular Chaperones/metabolism , TCF Transcription Factors/metabolism , Wnt Signaling Pathway , Animals , Cell Line , Cell Proliferation , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Embryo, Nonmammalian/metabolism , Humans , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , TCF Transcription Factors/genetics , Transcription Factor 7-Like 1 Protein/genetics , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factor 7-Like 2 Protein/metabolism , Transcriptional Activation , Wnt Proteins/genetics , Wnt Proteins/metabolism , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
7.
Biochem Biophys Res Commun ; 524(2): 280-287, 2020 04 02.
Article in English | MEDLINE | ID: mdl-31987502

ABSTRACT

Activation of the Wnt/ß-catenin signaling pathway by the inhibition of glycogen synthase kinase-3 (GSK-3) will induce Tcf7l1 protein degradation to effectively promote embryonic stem cell (ESC) self-renewal. However, the exact mechanism remains unclear. Here, we found that inhibition of casein kinase 2 (Csnk2) by TBB or DMAT was sufficient to block the reduction of the Tcf7l1 protein induced by CHIR99021, a specific inhibitor of GSK-3. Similarly, downregulation of Csnk2 increased the Tcf7l1 level. In contrast, overexpression of Csnk2 significantly decreased Tcf7l1 protein stability in mouse ESCs. Notably, Csnk2α1 controls Tcf7l1 turnover to a greater degree than the other two isoforms of Csnk2, Csnk2α2 and Csnk2ß, as Csnk2α1-overexpressing mouse ESCs exhibited the lowest level of Tcf7l1. Csnk2α1 interacted with and phosphorylated Tcf7l1. In addition, the association of Csnk2α1 and Tcf7l1 was enhanced by CHIR99021. Our study demonstrated, for the first time, that Csnk2 is involved in Tcf7l1 turnover mediated by the Wnt/ß-catenin signaling pathway. These results expand our understanding of the function and circuit of Wnt/ß-catenin signaling pathway in ESCs.


Subject(s)
Casein Kinase II/metabolism , Mouse Embryonic Stem Cells/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , beta Catenin/metabolism , Animals , Cell Line , Mice , Protein Interaction Maps , Proteolysis
8.
Exp Mol Pathol ; 110: 104293, 2019 10.
Article in English | MEDLINE | ID: mdl-31381875

ABSTRACT

The accurate classification and proper identification of testicular germ cell tumors is imperative for treatment selection and clinical prognosis. Although such distinction can often be achieved by microscopic morphology alone, ancillary tests may at times be needed. T-cell factor 7 L1 (TCF7L1, also known as TCF3), a component of the Wnt signaling pathway, plays important roles in embryonic stem cell self-renewal and lineage specification. Here we examined the immunohistochemical expression and diagnostic utility of TCF7L1 in testicular germ cell tumors. Fifty cases of testicular germ cell tumors were collected, including 23 seminomas, 6 embryonal carcinomas, 1 teratoma, 1 choriocarcinoma, and 19 mixed germ cell tumors. The components of the mixed germ cell tumors were seminoma (n = 3), embryonal carcinoma (n = 18), yolk sac tumor (n = 9), teratoma (n = 15), and choriocarcinoma (n = 4). On immunohistochemistry of TCF7L1, only nuclear staining was considered positive. Staining was graded as negative (<5% of tumor cells stained), minimal (5-25% positive), focal (26-50%), and diffuse (>50%). All non-seminomatous components (n = 54) exhibited distinct nuclear expression of TCF7L1 (54/54; 100%). In contrast, no TCF7L1 expression was detected in the majority of seminomatous tumor component (24/26; 92%). Two seminomas (2/26; 8%) exhibited minimal weak nuclear staining (5% and 10%, respectively) for TCF7L1. In conclusion, TCF7L1, highly expressed in non-seminomatous testicular germ cell tumors, might be used as a marker for diagnosis of testicular germ cell tumors, two therapeutically different entities, for better patient management.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms, Germ Cell and Embryonal/diagnosis , Seminoma/diagnosis , Testicular Neoplasms/diagnosis , Transcription Factor 7-Like 1 Protein/metabolism , Adult , Diagnosis, Differential , Humans , Immunohistochemistry , Male , Neoplasms, Germ Cell and Embryonal/metabolism , Patient Selection , Seminoma/metabolism , Testicular Neoplasms/metabolism
9.
Stem Cells ; 37(11): 1389-1400, 2019 11.
Article in English | MEDLINE | ID: mdl-31322782

ABSTRACT

Tcf7l1, which is a key effector molecule of the Wnt/ß-catenin signaling pathway, is highly expressed in various cancers, and it promotes tumor growth. In this study, we demonstrated that unlike its tumor-promoting effects in several other types of cancers, Tcf7l1 expression is downregulated in hepatocarcinoma compared with their adjacent nontumor counterparts. Underexpression of Tcf7l1 is correlated with poorer survival. In liver cancer stem cell (CSC) populations, Tcf7l1 expression is downregulated. Ectopic expression of Tcf7l1 attenuates the self-renewal abilities of liver CSCs. Mechanistically, Tcf7l1 regulates the self-renewal abilities of liver CSCs through transcriptional repression of the Nanog gene, and the effect is independent of ß-catenin. Moreover, we found that Tcf7l1 expression is controlled by extracellular insulin-like growth factor (IGF) signaling, and we demonstrated for the first time that IGF signaling stimulates Tcf7l1 phosphorylation and degradation through the mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway. Overall, our results provide some new insights into how extracellular signals modulate the self-renewal of liver CSCs and highlight the inhibitory roles of Tcf7l1 in cancer. Stem Cells 2019;37:1389-1400.


Subject(s)
Cell Survival/physiology , Liver/cytology , Liver/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Somatomedins/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , beta Catenin/metabolism , Cell Line , Cell Survival/genetics , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Immunoassay , Immunohistochemistry , Immunoprecipitation , In Vitro Techniques , Lentivirus , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Phosphorylation , Plasmids/genetics , Real-Time Polymerase Chain Reaction , Somatomedins/genetics , Transcription Factor 7-Like 1 Protein/genetics , beta Catenin/genetics
10.
Cell Stem Cell ; 25(2): 210-224.e6, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31104942

ABSTRACT

Progression through states of pluripotency is required for cells in early mammalian embryos to transition away from heightened self-renewal and toward competency for lineage specification. Here, we use a CRISPR mutagenesis screen in mouse embryonic stem cells (ESCs) to identify unexpected roles for nuclear export and intracellular Ca2+ homeostasis during the exit out of the naive state of pluripotency. Mutation of a plasma membrane Ca2+ pump encoded by Atp2b1 increased intracellular Ca2+ such that it overcame effects of intracellular Ca2+ reduction, which is required for naive exit. Persistent self-renewal of ESCs was supported both in Atp2b1-/-Tcf7l1-/- double-knockout ESCs passaged in defined media alone (no LIF or inhibitors) and in wild-type cells passaged in media containing only calcitonin and a GSK3 inhibitor. These new findings suggest a central role for intracellular Ca2+ in safeguarding naive pluripotency.


Subject(s)
Calcium Signaling/physiology , Intracellular Space/metabolism , Mouse Embryonic Stem Cells/physiology , Plasma Membrane Calcium-Transporting ATPases/metabolism , Pluripotent Stem Cells/physiology , Transcription Factor 7-Like 1 Protein/metabolism , Active Transport, Cell Nucleus , Animals , Cell Differentiation , Cell Lineage , Cell Self Renewal/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Glycogen Synthase Kinase 3/metabolism , Homeostasis , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , Transcription Factor 7-Like 1 Protein/genetics
11.
Elife ; 82019 02 19.
Article in English | MEDLINE | ID: mdl-30777146

ABSTRACT

The vertebrate eye originates from the eye field, a domain of cells specified by a small number of transcription factors. In this study, we show that Tcf7l1a is one such transcription factor that acts cell-autonomously to specify the eye field in zebrafish. Despite the much-reduced eye field in tcf7l1a mutants, these fish develop normal eyes revealing a striking ability of the eye to recover from a severe early phenotype. This robustness is not mediated through genetic compensation at neural plate stage; instead, the smaller optic vesicle of tcf7l1a mutants shows delayed neurogenesis and continues to grow until it achieves approximately normal size. Although the developing eye is robust to the lack of Tcf7l1a function, it is sensitised to the effects of additional mutations. In support of this, a forward genetic screen identified mutations in hesx1, cct5 and gdf6a, which give synthetically enhanced eye specification or growth phenotypes when in combination with the tcf7l1a mutation.


Subject(s)
Eye/growth & development , Morphogenesis , Transcription Factor 7-Like 1 Protein/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Animals , Cell Proliferation , Embryo, Nonmammalian/metabolism , Eye/pathology , Female , Gene Expression Regulation, Developmental , Genetic Loci , Kinetics , Male , Mutation/genetics , Neural Plate/embryology , Neurogenesis , Penetrance , Phenotype , Prosencephalon/embryology , Transcription Factor 7-Like 1 Protein/genetics , Up-Regulation/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zygote/metabolism
12.
Acta Biochim Biophys Sin (Shanghai) ; 51(4): 375-385, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30811526

ABSTRACT

Gastric cancer is one of the most common cancers worldwide and is the third leading cause of cancer-related deaths globally. Although significant progress has been made in the diagnosis and treatment for the cancer, less improvement has been made in overall survival rate. Thus, there is an urgent need for a better understanding of the biological aspects of the cancer. The transcription factor transcription factor 7-like 1 (TCF7L1) is an embryonic stem cell signature gene that is upregulated in multiple aggressive cancer types, but its role in gastric cancer has seldom been discussed. In the present study, by using the Cancer Genome Atlas dataset analysis, we demonstrated that patients with higher expression of TCF7L1 could be used to reflect prognosis. An examination of the mechanisms demonstrated that TCF7L1 could positively regulate antioxidant response in gastric cancer cells by positively regulating Keap1/NRF2 [Kelch-like ECH-associated protein 1/nuclear factor (erythroid-derived 2)-like 2] pathway. Collectively, our data demonstrated that TCF7L1 is a novel marker for predicting overall survival of gastric cancer and provided the possible underlying molecular mechanism.


Subject(s)
Cell Proliferation/genetics , Kelch-Like ECH-Associated Protein 1/genetics , NF-E2-Related Factor 2/genetics , Stomach Neoplasms/genetics , Transcription Factor 7-Like 1 Protein/genetics , Antioxidants/metabolism , Cell Line, Tumor , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic , Glycolysis/genetics , Humans , Kaplan-Meier Estimate , Kelch-Like ECH-Associated Protein 1/metabolism , Male , Middle Aged , NF-E2-Related Factor 2/metabolism , Prognosis , Signal Transduction/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcription Factor 7-Like 1 Protein/metabolism
13.
Cell Rep ; 25(3): 571-584.e5, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332639

ABSTRACT

Wnt/ß-catenin signaling is necessary for lymphatic vascular development. Oscillatory shear stress (OSS) enhances Wnt/ß-catenin signaling in cultured lymphatic endothelial cells (LECs) to induce expression of the lymphedema-associated transcription factors GATA2 and FOXC2. However, the mechanisms by which OSS regulates Wnt/ß-catenin signaling and GATA2 and FOXC2 expression are unknown. We show that OSS activates autocrine Wnt/ß-catenin signaling in LECs in vitro. Tissue-specific deletion of Wntless, which is required for the secretion of Wnt ligands, reveals that LECs and vascular smooth muscle cells are complementary sources of Wnt ligands that regulate lymphatic vascular development in vivo. Further, the LEC master transcription factor PROX1 forms a complex with ß-catenin and the TCF/LEF transcription factor TCF7L1 to enhance Wnt/ß-catenin signaling and promote FOXC2 and GATA2 expression in LECs. Thus, our work defines Wnt sources, reveals that PROX1 directs cell fate by acting as a Wnt signaling component, and dissects the mechanisms of PROX1 and Wnt synergy.


Subject(s)
Endothelial Cells/cytology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Tumor Suppressor Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Female , Forkhead Transcription Factors/metabolism , GATA2 Transcription Factor/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Wnt Signaling Pathway
14.
Am J Physiol Cell Physiol ; 314(6): C712-C720, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29513567

ABSTRACT

Although long noncoding RNAs (lncRNAs) are emerging as new modulators in the fate decision of pluripotent stem cells, the functions of specific lncRNAs remain unclear. Here, we found that telomeric RNA (TERRA or TelRNA), one type of lncRNAs, is highly expressed in mouse embryonic stem cells (mESCs) but declines significantly upon differentiation. TERRA is induced by the Wnt/ß-catenin signaling pathway and can reproduce its self-renewal-promoting effect when overexpressed. Further studies revealed that T cell factor 3 ( TCF3) is a potential downstream target of TERRA and mediates the effect of TERRA in mESC maintenance. TERRA inhibits TCF3 transcription, while enforced TCF3 expression abrogates the undifferentiated state of mESCs supported by TERRA. Accordingly, the transcripts of the pluripotency genes Esrrb, Tfcp2l1, and Klf2, repressed by TCF3 in mESCs, are increased in TERRA-overexpressing cells. Our study therefore highlights the important role of TERRA in mESC maintenance and also uncovers a mechanism by which TERRA promotes self-renewal. These data will expand our understanding of the pluripotent regulatory network of ESCs.


Subject(s)
Cell Self Renewal , Mouse Embryonic Stem Cells/metabolism , RNA, Untranslated/metabolism , Telomere/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Animals , Cell Differentiation , Cell Line , Cell Lineage , Cell Self Renewal/drug effects , Gene Expression Regulation, Developmental , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, 129 Strain , Mouse Embryonic Stem Cells/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Untranslated/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Telomere/genetics , Transcription Factor 7-Like 1 Protein/genetics , Transcription, Genetic , Wnt Signaling Pathway
15.
Development ; 145(4)2018 02 23.
Article in English | MEDLINE | ID: mdl-29361574

ABSTRACT

Human embryonic stem cells (hESCs) are exquisitely sensitive to WNT ligands, which rapidly cause differentiation. Therefore, hESC self-renewal requires robust mechanisms to keep the cells in a WNT inactive but responsive state. How they achieve this is largely unknown. We explored the role of transcriptional regulators of WNT signaling, the TCF/LEFs. As in mouse ESCs, TCF7L1 is the predominant family member expressed in hESCs. Genome-wide, it binds a gene cohort involved in primitive streak formation at gastrulation, including NODAL, BMP4 and WNT3 Comparing TCF7L1-bound sites with those bound by the WNT signaling effector ß-catenin indicates that TCF7L1 acts largely on the WNT signaling pathway. TCF7L1 overlaps less with the pluripotency regulators OCT4 and NANOG than in mouse ESCs. Gain- and loss-of-function studies indicate that TCF7L1 suppresses gene cohorts expressed in the primitive streak. Interestingly, we find that BMP4, another driver of hESC differentiation, downregulates TCF7L1, providing a mechanism of BMP and WNT pathway intersection. Together, our studies indicate that TCF7L1 plays a major role in maintaining hESC pluripotency, which has implications for human development during gastrulation.


Subject(s)
Human Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Primitive Streak/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Wnt Signaling Pathway/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation , Cell Lineage , Electrophoresis, Polyacrylamide Gel , Gene Expression , Humans , Immunohistochemistry , Microarray Analysis , Polymerase Chain Reaction
16.
Cell Rep ; 20(10): 2424-2438, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28877475

ABSTRACT

Co-expression and cross-regulation of the four TCF/LEFs render their redundant and unique functions ambiguous. Here, we describe quadruple-knockout (QKO) mouse ESCs lacking all full-length TCF/LEFs and cell lines rescued with TCF7 or TCF7L1. QKO cells self-renew, despite gene expression patterns that differ significantly from WT, and display delayed, neurectoderm-biased, embryoid body (EB) differentiation. QKO EBs have no contracting cardiomyocytes and differentiate poorly into mesendoderm but readily generate neuronal cells. QKO cells and TCF7L1-rescued cells cannot efficiently activate TCF reporters, whereas TCF7-rescued cells exhibit significant reporter responsiveness. Surprisingly, despite dramatically different transactivation capacities, re-expression of TCF7L1 or TCF7 in QKO cells restores their tri-lineage differentiation ability, with similar lineage marker expression patterns and beating cardiomyocyte frequencies observed in EBs. Both factors also similarly affect the transcriptome of QKO cells. Our data reveal that a single TCF, regardless of its activation capacity, is sufficient for effective trilineage differentiation of ESCs.


Subject(s)
Cell Differentiation/physiology , TCF Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunohistochemistry , Male , Mice , TCF Transcription Factors/genetics , Transcription Factor 7-Like 1 Protein/genetics , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/genetics , beta Catenin/metabolism
17.
Mol Cell Biol ; 37(18)2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28674186

ABSTRACT

Transforming growth factor ß (TGF-ß)-induced migration of triple-negative breast cancer (TNBC) cells is dependent on nuclear export of the orphan receptor NR4A1, which plays a role in proteasome-dependent degradation of SMAD7. In this study, we show that TGF-ß induces p38α (mitogen-activated protein kinase 14 [MAPK14]), which in turn phosphorylates NR4A1, resulting in nuclear export of the receptor. TGF-ß/p38α and NR4A1 also play essential roles in the induction of epithelial-to-mesenchymal transition (EMT) and induction of ß-catenin in TNBC cells, and these TGF-ß-induced responses and nuclear export of NR4A1 are blocked by NR4A1 antagonists, the p38 inhibitor SB202190, and kinase-dead [p38(KD)] and dominant-negative [p38(DN)] forms of p38α. Inhibition of NR4A1 nuclear export results in nuclear export of TGF-ß-induced ß-catenin, which then undergoes proteasome-dependent degradation. TGF-ß-induced ß-catenin also regulates NR4A1 expression through formation of the ß-catenin-TCF-3/TCF-4/LEF-1 complex on the NR4A1 promoter. Thus, TGF-ß-induced nuclear export of NR4A1 in TNBC cells plays an essential role in cell migration, SMAD7 degradation, EMT, and induction of ß-catenin, and all of these pathways are inhibited by bis-indole-derived NR4A1 antagonists that inhibit nuclear export of the receptor and thereby block TGF-ß-induced migration and EMT.


Subject(s)
Active Transport, Cell Nucleus/physiology , Epithelial-Mesenchymal Transition/physiology , Mitogen-Activated Protein Kinase 14/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Transforming Growth Factor beta/genetics , Triple Negative Breast Neoplasms/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement , Female , Humans , Imidazoles/pharmacology , Lymphoid Enhancer-Binding Factor 1/metabolism , Mitogen-Activated Protein Kinase 14/biosynthesis , Multiprotein Complexes/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phosphorylation , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Smad7 Protein/metabolism , Transcription Factor 4 , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism
18.
Elife ; 62017 05 03.
Article in English | MEDLINE | ID: mdl-28467300

ABSTRACT

The transcription factor TCF7L1 is an embryonic stem cell signature gene that is upregulated in multiple aggressive cancer types, but its role in skin tumorigenesis has not yet been defined. Here we document TCF7L1 upregulation in skin squamous cell carcinoma (SCC) and demonstrate that TCF7L1 overexpression increases tumor incidence, tumor multiplicity, and malignant progression in the chemically induced mouse model of skin SCC. Additionally, we show that downregulation of TCF7L1 and its paralogue TCF7L2 reduces tumor growth in a xenograft model of human skin SCC. Using separation-of-function mutants, we show that TCF7L1 promotes tumor growth, enhances cell migration, and overrides oncogenic RAS-induced senescence independently of its interaction with ß-catenin. Through transcriptome profiling and combined gain- and loss-of-function studies, we identified LCN2 as a major downstream effector of TCF7L1 that drives tumor growth. Our findings establish a tumor-promoting role for TCF7L1 in skin and elucidate the mechanisms underlying its tumorigenic capacity.


Subject(s)
Carcinogenesis , Carcinoma, Squamous Cell/physiopathology , Lipocalin-2/metabolism , Skin Neoplasms/physiopathology , Transcription Factor 7-Like 1 Protein/metabolism , beta Catenin/metabolism , Animals , Disease Models, Animal , Gene Expression Profiling , Heterografts , Humans , Mice
19.
Biochem Biophys Res Commun ; 487(3): 716-722, 2017 Jun 03.
Article in English | MEDLINE | ID: mdl-28450117

ABSTRACT

The T-cell factor/Lymphoid enhancer factor (TCF/LEF; hereafter TCF) family of transcription factors are critical regulators of colorectal cancer (CRC) cell growth. Of the four TCF family members, TCF7L1 functions predominantly as a repressor of gene expression. Few studies have addressed the role of TCF7L1 in CRC and only a handful of target genes regulated by this repressor are known. By silencing TCF7L1 expression in HCT116 cells, we show that it promotes cell proliferation and tumorigenesis in vivo by driving cell cycle progression. Microarray analysis of transcripts differentially expressed in control and TCF7L1-silenced CRC cells identified genes that control cell cycle kinetics and cancer pathways. Among these, expression of the Wnt antagonist DICKKOPF4 (DKK4) was upregulated when TCF7L1 levels were reduced. We found that TCF7L1 recruits the C-terminal binding protein (CtBP) and histone deacetylase 1 (HDAC1) to the DKK4 promoter to repress DKK4 gene expression. In the absence of TCF7L1, TCF7L2 and ß-catenin occupancy at the DKK4 promoter is stimulated and DKK4 expression is increased. These findings uncover a critical role for TCF7L1 in repressing DKK4 gene expression to promote the oncogenic potential of CRCs.


Subject(s)
Alcohol Oxidoreductases/metabolism , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Carcinogenesis/metabolism , Cell Line, Tumor , Humans , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...