1
|
CHEN JIA, JIANG FEI, NIU KAIYI, ZHAO HAODONG, LI LI, YU HONGZHU. A novel Wnt/β-catenin signaling gene signature for progression and metastasis of gastric cancer. Oncol Res 2025; 33:1199-1215. [PMID: 40296906 PMCID: PMC12035655 DOI: 10.32604/or.2024.054366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 11/13/2024] [Indexed: 04/30/2025] Open
Abstract
Backgrounds As cancer progresses through various stages of malignancy, metastasis, and drug resistance, the Wnt/-catenin signaling is frequently dysregulated. Despite advancements in medical technology and therapeutic strategies, the prognosis for numerous gastric cancer patients remains unfavorable. Methods For the analysis of prognostic signature genes associated with Wnt signaling in GC, we used LASSO (least absolute shrinkage and selection operator) regression. To explore the function, cell specificity, and transcriptional regulation of the signature gene Carboxypeptidase Z (CPZ), we conducted co-expression analysis, single-cell RNA sequencing data analysis, transcription factor prediction, and dual luciferase reporter assay. The knockdown and overexpression experiments were also performed to observe the changes in the downstream gene expression, as well as the influence on the biological functions of GC cells. Results We identified a five-gene signature, including CPZ, Collagen Triple Helix Repeat Containing-1 (CTHRC1), Dickkopf-1 (DKK1), Epidermal Growth Factor (EGF), and Glypican Proteoglycan-3 (GPC3), with risk scores predictive of the prognosis of GC patients. We found that the adipocyte enhancer binding protein 1 (AEBP1) and transcription factor 3 (TCF3) could interact in the nucleus and synergistically enhance the expression of Wnt signaling-associated genes, including WNT2/FZD2 (Wnt family member 2/frizzled class receptor 2) and VIM (vimentin), thus promoting the invasion, migration, and malignant metastasis of GC. Conclusions Our study offers a precise gene-signature prediction method for the prognosis of GC. We discovered the synergistic effect of AEBP1 and TCF3 in the nucleus on GC metastasis. GC may benefit from the identification of this potential therapeutic target.
Collapse
Affiliation(s)
- JIA CHEN
- Department of General Surgery, Fuyang Hospital Affiliated of Anhui Medical University, Fuyang, 236000, China
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - FEI JIANG
- Department of General Surgery, Fuyang Hospital Affiliated of Anhui Medical University, Fuyang, 236000, China
| | - KAIYI NIU
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - HAODONG ZHAO
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - LI LI
- Department of General Surgery, Fuyang Hospital Affiliated of Anhui Medical University, Fuyang, 236000, China
| | - HONGZHU YU
- Department of General Surgery, Fuyang Hospital Affiliated of Anhui Medical University, Fuyang, 236000, China
| |
Collapse
|
2
|
Bae H, Jeon H, Lee C. Genetic regulation of B cell receptor signaling pathway: Insights from expression quantitative trait locus analysis using a mixed model. Comput Biol Chem 2024; 113:108188. [PMID: 39236423 DOI: 10.1016/j.compbiolchem.2024.108188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/07/2024]
Abstract
The B cell receptor (BCR) signaling pathway regulates non-immune cellular response through various pathways like MAPK, NF-kB, and PI3K-Akt. This study aimed to identify expression quantitative trait loci (eQTL) and their regulatory functions on BCR signaling pathway genes. A mixed model was employed to analyze eQTL using RNA expression levels in lymphoblastoid from 376 Europeans in the GEUVADIS dataset. In total, 266 SNPs, including 115 cis-acting SNPs, were found for association with transcription of 13 genes (P < 5 × 10-8), revealing 19 independent signals for five genes through linkage disequilibrium analysis. Functional analysis, aligning them with DNase sensitive sites, transcription factor binding sites, histone modification, promoters/enhancers, CpG islands, and ChIA-PET, identified regulatory variants targeting SYK, VAV2, and PLCG2. Notably, rs2562397 was validated as a SYK promoter variant, and rs694505, rs636667, and rs4889409 were confirmed as enhancer variants for VAV2 and PLCG2. Their allelic differences in gene expression were also confirmed using ENCODE ChIP-seq and Sei neural network prediction. Persistent differential expression of these genes by alleles might impact the adaptive immune system, vascular development, and/or relevant diseases that have been previously associated with other variants of the genes. Comprehensive genetic architecture studies of the BCR signaling pathway, along with experiments demonstrating related mechanisms, will greatly contribute to understanding the underlying mechanisms of relevant disease development and implementing precision medicine.
Collapse
Affiliation(s)
- Hojin Bae
- Department of Bioinformatics and Life Science, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, Republic of Korea
| | - Hyowon Jeon
- Department of Bioinformatics and Life Science, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, Republic of Korea
| | - Chaeyoung Lee
- Department of Bioinformatics and Life Science, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, Republic of Korea.
| |
Collapse
|
3
|
Vuong LM, Pan S, Sierra RA, Waterman ML, Gershon PD, Donovan PJ. Characterization of a chromatin-associated TCF7L1 complex in human embryonic stem cells. Proteomics 2024; 24:e2300641. [PMID: 38629187 DOI: 10.1002/pmic.202300641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 10/11/2024]
Abstract
Human embryonic stem cells (hESCs) resemble the pluripotent epiblast cells found in the early postimplantation human embryo and represent the "primed" state of pluripotency. One factor that helps primed pluripotent cells retain pluripotency and prepare genes for differentiation is the transcription factor TCF7L1, a member of a small family of proteins known as T cell factors/Lymphoid enhancer factors (TCF/LEF) that act as downstream components of the WNT signaling pathway. Transcriptional output of the WNT pathway is regulated, in part, by the activity of TCF/LEFs in conjunction with another component of the WNT pathway, β-CATENIN. Because TCF7L1 plays an important role in regulating pluripotency, we began to characterize the protein complex associated with TCF7L1 when bound to chromatin in hESCs using rapid immunoprecipitation of endogenous proteins (RIME). Data are available via ProteomeXchange with identifier PXD047582. These data identify known and new partners of TCF7L1 on chromatin and provide novel insights into how TCF7L1 and pluripotency itself might be regulated.
Collapse
Affiliation(s)
- Linh M Vuong
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
- Department of Biological Chemistry, University of California, Irvine, California, USA
- Sue and Bill Gross Stem Cell Research Center: A CIRM Institute, University of California, Irvine, California, USA
| | - Songqin Pan
- W.M. Keck Proteomics Laboratory, Institute of Integrated Genome Biology, Department of Botany and Plant Sciences, University of California, Riverside, California, USA
| | - Robert A Sierra
- Department of Biological Chemistry, University of California, Irvine, California, USA
- Sue and Bill Gross Stem Cell Research Center: A CIRM Institute, University of California, Irvine, California, USA
| | - Marian L Waterman
- Department of Microbiology and Molecular Genetics, University of California, Irvine, California, USA
| | - Paul D Gershon
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Peter J Donovan
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
- Department of Biological Chemistry, University of California, Irvine, California, USA
- Sue and Bill Gross Stem Cell Research Center: A CIRM Institute, University of California, Irvine, California, USA
| |
Collapse
|
4
|
Shi DL. Canonical and Non-Canonical Wnt Signaling Generates Molecular and Cellular Asymmetries to Establish Embryonic Axes. J Dev Biol 2024; 12:20. [PMID: 39189260 PMCID: PMC11348223 DOI: 10.3390/jdb12030020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
The formation of embryonic axes is a critical step during animal development, which contributes to establishing the basic body plan in each particular organism. Wnt signaling pathways play pivotal roles in this fundamental process. Canonical Wnt signaling that is dependent on β-catenin regulates the patterning of dorsoventral, anteroposterior, and left-right axes. Non-canonical Wnt signaling that is independent of β-catenin modulates cytoskeletal organization to coordinate cell polarity changes and asymmetric cell movements. It is now well documented that components of these Wnt pathways biochemically and functionally interact to mediate cell-cell communications and instruct cellular polarization in breaking the embryonic symmetry. The dysfunction of Wnt signaling disrupts embryonic axis specification and proper tissue morphogenesis, and mutations of Wnt pathway genes are associated with birth defects in humans. This review discusses the regulatory roles of Wnt pathway components in embryonic axis formation by focusing on vertebrate models. It highlights current progress in decoding conserved mechanisms underlying the establishment of asymmetry along the three primary body axes. By providing an in-depth analysis of canonical and non-canonical pathways in regulating cell fates and cellular behaviors, this work offers insights into the intricate processes that contribute to setting up the basic body plan in vertebrate embryos.
Collapse
Affiliation(s)
- De-Li Shi
- Department of Medical Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China;
- Laboratory of Developmental Biology, Centre National de la Recherche Scientifique (CNRS), UMR7622, Institut de Biologie Paris-Seine (IBPS), Sorbonne University, 75005 Paris, France
| |
Collapse
|
5
|
Bou-Rouphael J, Doulazmi M, Eschstruth A, Abdou A, Durand BC. Cerebellar granular neuron progenitors exit their germinative niche via BarH-like1 activity mediated partly by inhibition of T-cell factor. Development 2024; 151:dev202234. [PMID: 38860486 DOI: 10.1242/dev.202234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Cerebellar granule neuron progenitors (GNPs) originate from the upper rhombic lip (URL), a germinative niche in which developmental defects produce human diseases. T-cell factor (TCF) responsiveness and Notch dependence are hallmarks of self-renewal in neural stem cells. TCF activity, together with transcripts encoding proneural gene repressors hairy and enhancer of split (Hes/Hey), are detected in the URL; however, their functions and regulatory modes are undeciphered. Here, we established amphibian as a pertinent model for studying vertebrate URL development. The amphibian long-lived URL is TCF active, whereas the external granular layer (EGL) is non-proliferative and expresses hes4 and hes5 genes. Using functional and transcriptomic approaches, we show that TCF activity is necessary for URL emergence and maintenance. We establish that the transcription factor Barhl1 controls GNP exit from the URL, acting partly through direct TCF inhibition. Identification of Barhl1 target genes suggests that, besides TCF, Barhl1 inhibits transcription of hes5 genes independently of Notch signaling. Observations in amniotes suggest a conserved role for Barhl in maintenance of the URL and/or EGL via co-regulation of TCF, Hes and Hey genes.
Collapse
Affiliation(s)
- Johnny Bou-Rouphael
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Mohamed Doulazmi
- Sorbonne Université, CNRS UMR8256, Institut de Biologie Paris-Seine (IBPS) - Laboratoire Adaptation Biologique et Vieillissement, 75005 Paris, France
| | - Alexis Eschstruth
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Asna Abdou
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Béatrice C Durand
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
- Sorbonne Université, CNRS UMR8256, Institut de Biologie Paris-Seine (IBPS) - Laboratoire Adaptation Biologique et Vieillissement, 75005 Paris, France
| |
Collapse
|
6
|
Yu Y, Liu L, Cao J, Huang R, Duan Q, Ye SD. Tbl1 promotes Wnt-β-catenin signaling-induced degradation of the Tcf7l1 protein in mouse embryonic stem cells. J Cell Sci 2024; 137:jcs261241. [PMID: 38639717 DOI: 10.1242/jcs.261241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Activation of the Wnt-β-catenin signaling pathway by CHIR99021, a specific inhibitor of GSK3β, 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-β-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 β-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-β-catenin signaling pathway involved in promoting the maintenance of naïve pluripotency.
Collapse
Affiliation(s)
- Yang Yu
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| | - Liwei Liu
- College of Medical Technology, Anhui Medical College, Hefei, Anhui, 230601, China
| | - Jianjian Cao
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| | - Ru Huang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| | - Quanchao Duan
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| | - Shou-Dong Ye
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| |
Collapse
|
7
|
Koo Y, Han W, Keum BR, Lutz L, Yun SH, Kim GH, Han JK. RNF2 regulates Wnt/ß-catenin signaling via TCF7L1 destabilization. Sci Rep 2023; 13:19750. [PMID: 37957244 PMCID: PMC10643375 DOI: 10.1038/s41598-023-47111-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/09/2023] [Indexed: 11/15/2023] Open
Abstract
The Wnt signaling pathway is a crucial regulator of various biological processes, such as development and cancer. The downstream transcription factors in this pathway play a vital role in determining the threshold for signaling induction and the length of the response, which vary depending on the biological context. Among the four transcription factors involved in canonical Wnt/ß-catenin signaling, TCF7L1 is known to possess an inhibitory function; however, the underlying regulatory mechanism remains unclear. In this study, we identified the E3 ligase, RNF2, as a novel positive regulator of the Wnt pathway. Here, we demonstrate that RNF2 promotes the degradation of TCF7L1 through its ubiquitination upon activation of Wnt signaling. Loss-of-function studies have shown that RNF2 consistently destabilizes nuclear TCF7L1 and is required for proper Wnt target gene transcription in response to Wnt activation. Furthermore, our results revealed that RNF2 controls the threshold, persistence, and termination of Wnt signaling by regulating TCF7L1. Overall, our study sheds light on the previously unknown degradation mechanism of TCF7L1 by a specific E3 ligase, RNF2, and provides new insights into the variability in cellular responses to Wnt activation.
Collapse
Affiliation(s)
- Youngmu Koo
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Wonhee Han
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Byeong-Rak Keum
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Leila Lutz
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Sung Ho Yun
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Gun-Hwa Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Jin-Kwan Han
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea.
| |
Collapse
|
8
|
Šopin T, Liška F, Kučera T, Cmarko D, Vacík T. Lysine Demethylase KDM2A Promotes Proteasomal Degradation of TCF/LEF Transcription Factors in a Neddylation-Dependent Manner. Cells 2023; 12:2620. [PMID: 37998355 PMCID: PMC10670284 DOI: 10.3390/cells12222620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Canonical Wnt signaling is essential for a plethora of biological processes ranging from early embryogenesis to aging. Malfunctions of this crucial signaling pathway are associated with various developmental defects and diseases, including cancer. Although TCF/LEF transcription factors (TCF/LEFs) are known to be essential for this pathway, the regulation of their intracellular levels is not completely understood. Here, we show that the lysine demethylase KDM2A promotes the proteasomal destabilization of TCF/LEFs independently of its demethylase domain. We found that the KDM2A-mediated destabilization of TCF/LEFs is dependent on the KDM2A zinc finger CXXC domain. Furthermore, we identified the C-terminal region of TCF7L2 and the CXXC domain of KDM2A as the domains responsible for the interaction between the two proteins. Our study is also the first to show that endogenous TCF/LEF proteins undergo KDM2A-mediated proteasomal degradation in a neddylation-dependent manner. Here, we reveal a completely new mechanism that affects canonical Wnt signaling by regulating the levels of TCF/LEF transcription factors through their KDM2A-promoted proteasomal degradation.
Collapse
Affiliation(s)
- Tijana Šopin
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, 128 01 Prague, Czech Republic; (F.L.); (T.Š.); (D.C.)
| | - František Liška
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, 128 01 Prague, Czech Republic; (F.L.); (T.Š.); (D.C.)
| | - Tomáš Kučera
- Institute of Histology and Embryology, First Faculty of Medicine, Charles University and General University Hospital in Prague, 128 01 Prague, Czech Republic;
| | - Dušan Cmarko
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, 128 01 Prague, Czech Republic; (F.L.); (T.Š.); (D.C.)
| | - Tomáš Vacík
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, 128 01 Prague, Czech Republic; (F.L.); (T.Š.); (D.C.)
| |
Collapse
|
9
|
Żak M, Støle TP, Plagnol V, Daudet N. Regulation of otic neurosensory specification by Notch and Wnt signalling: insights from RNA-seq screenings in the embryonic chicken inner ear. Front Cell Dev Biol 2023; 11:1245330. [PMID: 37900277 PMCID: PMC10600479 DOI: 10.3389/fcell.2023.1245330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
The Notch and Wnt signalling pathways play key roles in the formation of inner ear sensory organs, but little is known about their transcriptional effectors and targets in this context. Here, we perturbed Notch and Wnt activities in the embryonic chicken otic vesicle using pharmacological treatment or in ovo electroporation of plasmid DNA, and used RNA-Seq to analyse the resulting changes in gene expression. Compared to pharmacological treatments, in ovo electroporation changed the expression of fewer genes, a likely consequence of the variability and mosaicism of transfection. The pharmacological inhibition of Notch activity induced a rapid change in the expression of known effectors of this pathway and genes associated with neurogenesis, consistent with a switch towards an otic neurosensory fate. The Wnt datasets contained many genes associated with a neurosensory biological function, confirming the importance of this pathway for neurosensory specification in the otocyst. Finally, the results of a preliminary gain-of-function screening of selected transcription factors and Wnt signalling components suggest that the endogenous programs of otic neurosensory specification are very robust, and in general unaffected by the overexpression of a single factor. Altogether this work provides new insights into the effectors and candidate targets of the Notch and Wnt pathways in the early developing inner ear and could serve as a useful reference for future functional genomics experiments in the embryonic avian inner ear.
Collapse
Affiliation(s)
- Magdalena Żak
- UCL Ear Institute, University College London, London, United Kingdom
| | - Thea P. Støle
- UCL Ear Institute, University College London, London, United Kingdom
| | - Vincent Plagnol
- Genetics Institute, University College London, London, United Kingdom
| | - Nicolas Daudet
- UCL Ear Institute, University College London, London, United Kingdom
| |
Collapse
|
10
|
Yang S, Wang S, Chen L, Wang Z, Chen J, Ni Q, Guo X, Zhang L, Xue G. Neutrophil Extracellular Traps Delay Diabetic Wound Healing by Inducing Endothelial-to-Mesenchymal Transition via the Hippo pathway. Int J Biol Sci 2023; 19:347-361. [PMID: 36594092 PMCID: PMC9760440 DOI: 10.7150/ijbs.78046] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/05/2022] [Indexed: 12/23/2022] Open
Abstract
Diabetic foot ulcers (DFUs) are among the most frequent complications of diabetes with significant morbidity and mortality. Diabetes can trigger neutrophils to undergo histone citrullination by protein arginine deiminase 4 (encoded by Padi4 in mice) and release neutrophil extracellular traps (NETs). The specific mechanism of NETs-mediated wound healing impairment in diabetes remains unknown. In this study, we show neutrophils are more susceptible to NETosis in diabetic wound environments. Via in vitro experiments and in vivo models of wound healing using wide-type and Padi4 -/- mice, we demonstrate NETs can induce the activation of PAK2 via the membrane receptor TLR-9. Then PAK2 phosphorylates the intracellular protein Merlin/NF2 to inhibit the Hippo-YAP pathway. YAP binds to transcription factor SMAD2 and translocates from the cytoplasm into the nucleus to promote endothelial-to-mesenchymal transition (EndMT), which ultimately impedes angiogenesis and delays wound healing. Suppression of the Merlin/YAP/SMAD2 pathway can attenuate NET-induced EndMT. Inhibition of NETosis accelerates wound healing by reducing EndMT and promoting angiogenesis. Cumulatively, these data suggest NETosis delays diabetic wound healing by inducing EndMT via the Hippo-YAP pathway. Increased understanding of the molecular mechanism that regulates NETosis and EndMT will be of considerable value for providing cellular targets amenable to therapeutic intervention for DFUs.
Collapse
Affiliation(s)
- Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200127, China,✉ Corresponding authors: Shuofei Yang, , Telephone No. +86 13764227372; Lan Zhang, , Telephone No. +86 13764227372; Guanhua Xue, , Telephone No. +86 13310166810
| | - ShuangShuang Wang
- Songyuan Central Hospital, Songyuan Children's Hospital, Songyuan, China
| | - Liang Chen
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200127, China
| | - Zheyu Wang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200127, China
| | - Jiaquan Chen
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200127, China
| | - Qihong Ni
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200127, China
| | - Xiangjiang Guo
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200127, China
| | - Lan Zhang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200127, China,✉ Corresponding authors: Shuofei Yang, , Telephone No. +86 13764227372; Lan Zhang, , Telephone No. +86 13764227372; Guanhua Xue, , Telephone No. +86 13310166810
| | - Guanhua Xue
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200127, China,✉ Corresponding authors: Shuofei Yang, , Telephone No. +86 13764227372; Lan Zhang, , Telephone No. +86 13764227372; Guanhua Xue, , Telephone No. +86 13310166810
| |
Collapse
|
11
|
Rankin SA, Zorn AM. The homeodomain transcription factor Ventx2 regulates respiratory progenitor cell number and differentiation timing during
Xenopus
lung development. Dev Growth Differ 2022; 64:347-361. [PMID: 36053777 PMCID: PMC10088502 DOI: 10.1111/dgd.12807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/03/2022] [Accepted: 08/14/2022] [Indexed: 11/28/2022]
Abstract
Ventx2 is an Antennapedia superfamily/NK-like subclass homeodomain transcription factor best known for its roles in the regulation of early dorsoventral patterning during Xenopus gastrulation and in the maintenance of neural crest multipotency. In this work we characterize the spatiotemporal expression pattern of ventx2 in progenitor cells of the Xenopus respiratory system epithelium. We find that ventx2 is directly induced by BMP signaling in the ventral foregut prior to nkx2-1, the earliest epithelial marker of the respiratory lineage. Functional studies demonstrate that Ventx2 regulates the number of Nkx2-1/Sox9+ respiratory progenitor cells induced during foregut development, the timing and level of surfactant protein gene expression, and proper tracheal-esophageal separation. Our data suggest that Ventx2 regulates the balance of respiratory progenitor cell expansion and differentiation. While the ventx gene family has been lost from the mouse genome during evolution, humans have retained a ventx2-like gene (VENTX). Finally, we discuss how our findings might suggest a possible function of VENTX in human respiratory progenitor cells.
Collapse
Affiliation(s)
- Scott A. Rankin
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology Perinatal Institute, Cincinnati Children’s Hospital Medical Center Cincinnati OH
| | - Aaron M. Zorn
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology Perinatal Institute, Cincinnati Children’s Hospital Medical Center Cincinnati OH
- University of Cincinnati, College of Medicine, Department of Pediatrics Cincinnati OH
| |
Collapse
|
12
|
García-Corzo L, Calatayud-Baselga I, Casares-Crespo L, Mora-Martínez C, Julián Escribano-Saiz J, Hortigüela R, Asenjo-Martínez A, Jordán-Pla A, Ercoli S, Flames N, López-Alonso V, Vilar M, Mira H. The transcription factor LEF1 interacts with NFIX and switches isoforms during adult hippocampal neural stem cell quiescence. Front Cell Dev Biol 2022; 10:912319. [PMID: 35938168 PMCID: PMC9355129 DOI: 10.3389/fcell.2022.912319] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
Stem cells in adult mammalian tissues are held in a reversible resting state, known as quiescence, for prolonged periods of time. Recent studies have greatly increased our understanding of the epigenetic and transcriptional landscapes that underlie stem cell quiescence. However, the transcription factor code that actively maintains the quiescence program remains poorly defined. Similarly, alternative splicing events affecting transcription factors in stem cell quiescence have been overlooked. Here we show that the transcription factor T-cell factor/lymphoid enhancer factor LEF1, a central player in canonical β-catenin-dependent Wnt signalling, undergoes alternative splicing and switches isoforms in quiescent neural stem cells. We found that active β-catenin and its partner LEF1 accumulated in quiescent hippocampal neural stem and progenitor cell (Q-NSPC) cultures. Accordingly, Q-NSPCs showed enhanced TCF/LEF1-driven transcription and a basal Wnt activity that conferred a functional advantage to the cultured cells in a Wnt-dependent assay. At a mechanistic level, we found a fine regulation of Lef1 gene expression. The coordinate upregulation of Lef1 transcription and retention of alternative spliced exon 6 (E6) led to the accumulation of a full-length protein isoform (LEF1-FL) that displayed increased stability in the quiescent state. Prospectively isolated GLAST + cells from the postnatal hippocampus also underwent E6 retention at the time quiescence is established in vivo. Interestingly, LEF1 motif was enriched in quiescence-associated enhancers of genes upregulated in Q-NSPCs and quiescence-related NFIX transcription factor motifs flanked the LEF1 binding sites. We further show that LEF1 interacts with NFIX and identify putative LEF1/NFIX targets. Together, our results uncover an unexpected role for LEF1 in gene regulation in quiescent NSPCs, and highlight alternative splicing as a post-transcriptional regulatory mechanism in the transition from stem cell activation to quiescence.
Collapse
Affiliation(s)
- Laura García-Corzo
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
| | - Isabel Calatayud-Baselga
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
| | - Lucía Casares-Crespo
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
| | - Carlos Mora-Martínez
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
- Evo-devo Helsinki Community, Centre of Excellence in Experimental and Computational Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Juan Julián Escribano-Saiz
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
| | | | | | - Antonio Jordán-Pla
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
| | - Stefano Ercoli
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
| | - Nuria Flames
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
| | | | - Marçal Vilar
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
| | - Helena Mira
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), València, Spain
- *Correspondence: Helena Mira,
| |
Collapse
|
13
|
Torres-Aguila NP, Salonna M, Hoppler S, Ferrier DEK. Evolutionary diversification of the canonical Wnt signaling effector TCF/LEF in chordates. Dev Growth Differ 2022; 64:120-137. [PMID: 35048372 PMCID: PMC9303524 DOI: 10.1111/dgd.12771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 12/29/2022]
Abstract
Wnt signaling is essential during animal development and regeneration, but also plays an important role in diseases such as cancer and diabetes. The canonical Wnt signaling pathway is one of the most conserved signaling cascades in the animal kingdom, with the T‐cell factor/lymphoid enhancer factor (TCF/LEF) proteins being the major mediators of Wnt/β‐catenin‐regulated gene expression. In comparison with invertebrates, vertebrates possess a high diversity of TCF/LEF family genes, implicating this as a possible key change to Wnt signaling at the evolutionary origin of vertebrates. However, the precise nature of this diversification is only poorly understood. The aim of this study is to clarify orthology, paralogy, and isoform relationships within the TCF/LEF gene family within chordates via in silico comparative study of TCF/LEF gene structure, molecular phylogeny, and gene synteny. Our results support the notion that the four TCF/LEF paralog subfamilies in jawed vertebrates (gnathostomes) evolved via the two rounds of whole‐genome duplications that occurred during early vertebrate evolution. Importantly, gene structure comparisons and synteny analysis of jawless vertebrate (cyclostome) TCFs suggest that a TCF7L2‐like form of gene structure is a close proxy for the ancestral vertebrate structure. In conclusion, we propose a detailed evolutionary path based on a new pre‐whole‐genome duplication vertebrate TCF gene model. This ancestor gene model highlights the chordate and vertebrate innovations of TCF/LEF gene structure, providing the foundation for understanding the role of Wnt/β‐catenin signaling in vertebrate evolution.
Collapse
Affiliation(s)
- Nuria P Torres-Aguila
- Gatty Marine Laboratory, The Scottish Oceans Institute, School of Biology, University of St Andrews, St Andrews, UK
| | - Marika Salonna
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Stefan Hoppler
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - David E K Ferrier
- Gatty Marine Laboratory, The Scottish Oceans Institute, School of Biology, University of St Andrews, St Andrews, UK
| |
Collapse
|
14
|
Canonical Wnt Signaling in the Pathology of Iron Overload-Induced Oxidative Stress and Age-Related Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7163326. [PMID: 35116092 PMCID: PMC8807048 DOI: 10.1155/2022/7163326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/04/2022] [Indexed: 12/26/2022]
Abstract
Iron accumulates in the vital organs with aging. This is associated with oxidative stress, inflammation, and mitochondrial dysfunction leading to age-related disorders. Abnormal iron levels are linked to neurodegenerative diseases, liver injury, cancer, and ocular diseases. Canonical Wnt signaling is an evolutionarily conserved signaling pathway that regulates many cellular functions including cell proliferation, apoptosis, cell migration, and stem cell renewal. Recent evidences indicate that iron regulates Wnt signaling, and iron chelators like deferoxamine and deferasirox can inhibit Wnt signaling and cell growth. Canonical Wnt signaling is implicated in the pathogenesis of many diseases, and there are significant efforts ongoing to develop innovative therapies targeting the aberrant Wnt signaling. This review examines how intracellular iron accumulation regulates Wnt signaling in various tissues and their potential contribution in the progression of age-related diseases.
Collapse
|
15
|
Disoma C, Zhou Y, Li S, Peng J, Xia Z. Wnt/β-catenin signaling in colorectal cancer: Is therapeutic targeting even possible? Biochimie 2022; 195:39-53. [DOI: 10.1016/j.biochi.2022.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/03/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
16
|
Bou-Rouphael J, Durand BC. T-Cell Factors as Transcriptional Inhibitors: Activities and Regulations in Vertebrate Head Development. Front Cell Dev Biol 2021; 9:784998. [PMID: 34901027 PMCID: PMC8651982 DOI: 10.3389/fcell.2021.784998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022] Open
Abstract
Since its first discovery in the late 90s, Wnt canonical signaling has been demonstrated to affect a large variety of neural developmental processes, including, but not limited to, embryonic axis formation, neural proliferation, fate determination, and maintenance of neural stem cells. For decades, studies have focused on the mechanisms controlling the activity of β-catenin, the sole mediator of Wnt transcriptional response. More recently, the spotlight of research is directed towards the last cascade component, the T-cell factor (TCF)/Lymphoid-Enhancer binding Factor (LEF), and more specifically, the TCF/LEF-mediated switch from transcriptional activation to repression, which in both embryonic blastomeres and mouse embryonic stem cells pushes the balance from pluri/multipotency towards differentiation. It has been long known that Groucho/Transducin-Like Enhancer of split (Gro/TLE) is the main co-repressor partner of TCF/LEF. More recently, other TCF/LEF-interacting partners have been identified, including the pro-neural BarH-Like 2 (BARHL2), which belongs to the evolutionary highly conserved family of homeodomain-containing transcription factors. This review describes the activities and regulatory modes of TCF/LEF as transcriptional repressors, with a specific focus on the functions of Barhl2 in vertebrate brain development. Specific attention is given to the transcriptional events leading to formation of the Organizer, as well as the roles and regulations of Wnt/β-catenin pathway in growth of the caudal forebrain. We present TCF/LEF activities in both embryonic and neural stem cells and discuss how alterations of this pathway could lead to tumors.
Collapse
Affiliation(s)
| | - Béatrice C. Durand
- Sorbonne Université, CNRS UMR7622, IBPS Developmental Biology Laboratory, Campus Pierre et Marie Curie, Paris, France
| |
Collapse
|
17
|
Luo M, Liu Q, Ye S, Liu S, Hu Y, Lv D, Wang G, Li M, Jian C, Huang B. RNA-seq of buffalo fibroblasts over-expressed pluripotent-related genes to investigate characteristics of its preliminarily reprogrammed stage. Res Vet Sci 2021; 144:164-174. [PMID: 34839950 DOI: 10.1016/j.rvsc.2021.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/19/2021] [Accepted: 11/16/2021] [Indexed: 01/10/2023]
Abstract
Induced pluripotent stem cells (iPSCs) can enhance the efficiency of buffalo genetic improvements because of their differentiation potential and proliferation ability, which are similar to those of embryonic stem cells. However, very few studies have focussed on buffalo iPSCs, and a stable induction system has not been established for buffalo somatic cell reprogramming. In this study, we constructed a PiggyBac transposon vector co-expressing buffalo OCT4, C-MYC, KLF4 and SOX2 genes (PB_OMKS) separated by the nucleotide sequence of three 2A peptides and established the buffalo foetal skin fibroblast (BFSF) cell line BFSF_OMKS. RNA-seq technology and bioinformatics analysis methods were mainly employed to perform a transcriptome analysis between BFSF and BFSF_OMKS. The results revealed that over-expression of OCT4, C-MYC, KLF4 and SOX2 in BFSFs led to the activation of reprogramming-related LIF, activin, BMP4, SMAD1/5/9 and Wnt signals. These results increased our understanding of buffalo somatic cell reprogramming mechanisms and could provide a possible theory for the selection of small-molecule cocktails to promote reprogramming.
Collapse
Affiliation(s)
- Man Luo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 530004, China; College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Quanhui Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 530004, China; College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Sheng Ye
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 530004, China; College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Shulin Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 530004, China; College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Yanan Hu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 530004, China; College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Danwei Lv
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 530004, China; College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Guodong Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 530004, China; College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Mengmei Li
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China.
| | - Chongdong Jian
- Department of Neurology, The Affiliated Hospital of Youjiang Medical for Nationalities, Baise, Guangxi 533000, China.
| | - Ben Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 530004, China; College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China.
| |
Collapse
|
18
|
Zhou W, Ma T, Ding S. Non-viral approaches for somatic cell reprogramming into cardiomyocytes. Semin Cell Dev Biol 2021; 122:28-36. [PMID: 34238675 DOI: 10.1016/j.semcdb.2021.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/04/2021] [Accepted: 06/23/2021] [Indexed: 11/27/2022]
Abstract
Heart disease is the leading cause of human deaths worldwide. Due to lacking cardiomyocytes with replicative capacity and cardiac progenitor cells with differentiation potential in adult hearts, massive loss of cardiomyocytes after ischemic events produces permanent damage, ultimately leading to heart failure. Cellular reprogramming is a promising strategy to regenerate heart by induction of cardiomyocytes from other cell types, such as cardiac fibroblasts. In contrast to conventional virus-based cardiac reprogramming, non-viral approaches greatly reduce the potential risk that includes disruption of genome integrity by integration of foreign DNAs, expression of exogenous genes with oncogenic potential, and appearance of partially reprogrammed cells harmful for the physiological functions of tissues/organs, which impedes their in-vivo applications. Here, we review the recent progress in development of non-viral approaches to directly reprogram somatic cells towards cardiomyocytes and their therapeutic application for heart regeneration.
Collapse
Affiliation(s)
- Wei Zhou
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Tianhua Ma
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Sheng Ding
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
19
|
Reis AH, Sokol SY. Rspo2 inhibits TCF3 phosphorylation to antagonize Wnt signaling during vertebrate anteroposterior axis specification. Sci Rep 2021; 11:13433. [PMID: 34183732 PMCID: PMC8239024 DOI: 10.1038/s41598-021-92824-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/10/2021] [Indexed: 01/20/2023] Open
Abstract
The Wnt pathway activates target genes by controlling the β-catenin-T-cell factor (TCF) transcriptional complex during embryonic development and cancer. This pathway can be potentiated by R-spondins, a family of proteins that bind RNF43/ZNRF3 E3 ubiquitin ligases and LGR4/5 receptors to prevent Frizzled degradation. Here we demonstrate that, during Xenopus anteroposterior axis specification, Rspo2 functions as a Wnt antagonist, both morphologically and at the level of gene targets and pathway mediators. Unexpectedly, the binding to RNF43/ZNRF3 and LGR4/5 was not required for the Wnt inhibitory activity. Moreover, Rspo2 did not influence Dishevelled phosphorylation in response to Wnt ligands, suggesting that Frizzled activity is not affected. Further analysis indicated that the Wnt antagonism is due to the inhibitory effect of Rspo2 on TCF3/TCF7L1 phosphorylation that normally leads to target gene activation. Consistent with this mechanism, Rspo2 anteriorizing activity has been rescued in TCF3-depleted embryos. These observations suggest that Rspo2 is a context-specific regulator of TCF3 phosphorylation and Wnt signaling.
Collapse
Affiliation(s)
- Alice H Reis
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA.
| | - Sergei Y Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
20
|
Cheng J, Tsuda M, Okolotowicz K, Dwyer M, Bushway PJ, Colas AR, Lancman JJ, Schade D, Perea-Gil I, Bruyneel AAN, Lee J, Vadgama N, Quach J, McKeithan WL, Biechele TL, Wu JC, Moon RT, Si Dong PD, Karakikes I, Cashman JR, Mercola M. Small-molecule probe reveals a kinase cascade that links stress signaling to TCF/LEF and Wnt responsiveness. Cell Chem Biol 2021; 28:625-635.e5. [PMID: 33503403 PMCID: PMC8140986 DOI: 10.1016/j.chembiol.2021.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/02/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022]
Abstract
Wnt signaling plays a central role in tissue maintenance and cancer. Wnt activates downstream genes through β-catenin, which interacts with TCF/LEF transcription factors. A major question is how this signaling is coordinated relative to tissue organization and renewal. We used a recently described class of small molecules that binds tubulin to reveal a molecular cascade linking stress signaling through ATM, HIPK2, and p53 to the regulation of TCF/LEF transcriptional activity. These data suggest a mechanism by which mitotic and genotoxic stress can indirectly modulate Wnt responsiveness to exert coherent control over cell shape and renewal. These findings have implications for understanding tissue morphogenesis and small-molecule anticancer therapeutics.
Collapse
Affiliation(s)
- Jiongjia Cheng
- Human BioMolecular Research Institute, 5310 Eastgate Mall, San Diego, CA 92121, USA
| | - Masanao Tsuda
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Karl Okolotowicz
- Human BioMolecular Research Institute, 5310 Eastgate Mall, San Diego, CA 92121, USA
| | - Mary Dwyer
- Human BioMolecular Research Institute, 5310 Eastgate Mall, San Diego, CA 92121, USA
| | - Paul J Bushway
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; University of California, San Diego, San Diego, CA 92093, USA
| | - Alexandre R Colas
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Joseph J Lancman
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Dennis Schade
- Human BioMolecular Research Institute, 5310 Eastgate Mall, San Diego, CA 92121, USA; Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstrasse 76, Kiel, Germany
| | - Isaac Perea-Gil
- Cardiovascular Institute, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA
| | - Arne A N Bruyneel
- Cardiovascular Institute, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA
| | - Jaechol Lee
- Cardiovascular Institute, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA
| | - Nirmal Vadgama
- Cardiovascular Institute, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA
| | - Justine Quach
- Human BioMolecular Research Institute, 5310 Eastgate Mall, San Diego, CA 92121, USA
| | - Wesley L McKeithan
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Cardiovascular Institute, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA
| | - Travis L Biechele
- Department of Pharmacology, University of Washington, Seattle, WA 98105, USA
| | - Joseph C Wu
- Cardiovascular Institute, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA
| | - Randall T Moon
- Department of Pharmacology, University of Washington, Seattle, WA 98105, USA
| | - P Duc Si Dong
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ioannis Karakikes
- Cardiovascular Institute, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA; Department of Cardiothoracic Surgery, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA
| | - John R Cashman
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mark Mercola
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; University of California, San Diego, San Diego, CA 92093, USA; Cardiovascular Institute, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, 240 Pasteur Drive, Palo Alto, CA 94305, USA.
| |
Collapse
|
21
|
Gupta A, Rarick KR, Ramchandran R. Established, New and Emerging Concepts in Brain Vascular Development. Front Physiol 2021; 12:636736. [PMID: 33643074 PMCID: PMC7907611 DOI: 10.3389/fphys.2021.636736] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/15/2021] [Indexed: 12/20/2022] Open
Abstract
In this review, we discuss the state of our knowledge as it relates to embryonic brain vascular patterning in model systems zebrafish and mouse. We focus on the origins of endothelial cell and the distinguishing features of brain endothelial cells compared to non-brain endothelial cells, which is revealed by single cell RNA-sequencing methodologies. We also discuss the cross talk between brain endothelial cells and neural stem cells, and their effect on each other. In terms of mechanisms, we focus exclusively on Wnt signaling and the recent developments associated with this signaling network in brain vascular patterning, and the benefits and challenges associated with strategies for targeting the brain vasculature. We end the review with a discussion on the emerging areas of meningeal lymphatics, endothelial cilia biology and novel cerebrovascular structures identified in vertebrates.
Collapse
Affiliation(s)
- Ankan Gupta
- Department of Pediatrics, Division of Neonatology, Developmental Vascular Biology Program, Children’s Research Institute (CRI), Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kevin R. Rarick
- Department of Pediatrics, Division of Critical Care, Children’s Research Institute (CRI), Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ramani Ramchandran
- Department of Pediatrics, Division of Neonatology, Developmental Vascular Biology Program, Children’s Research Institute (CRI), Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
22
|
Kim MJ, Huang Y, Park JI. Targeting Wnt Signaling for Gastrointestinal Cancer Therapy: Present and Evolving Views. Cancers (Basel) 2020; 12:E3638. [PMID: 33291655 PMCID: PMC7761926 DOI: 10.3390/cancers12123638] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/19/2022] Open
Abstract
Wnt signaling governs tissue development, homeostasis, and regeneration. However, aberrant activation of Wnt promotes tumorigenesis. Despite the ongoing efforts to manipulate Wnt signaling, therapeutic targeting of Wnt signaling remains challenging. In this review, we provide an overview of current clinical trials to target Wnt signaling, with a major focus on gastrointestinal cancers. In addition, we discuss the caveats and alternative strategies for therapeutically targeting Wnt signaling for cancer treatment.
Collapse
Affiliation(s)
- Moon Jong Kim
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.J.K.); (Y.H.)
| | - Yuanjian Huang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.J.K.); (Y.H.)
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.J.K.); (Y.H.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and Health Science Center, Houston, TX 77030, USA
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
23
|
Zhang H, Rong X, Wang C, Liu Y, Lu L, Li Y, Zhao C, Zhou J. VBP1 modulates Wnt/β-catenin signaling by mediating the stability of the transcription factors TCF/LEFs. J Biol Chem 2020; 295:16826-16839. [PMID: 32989053 PMCID: PMC7864075 DOI: 10.1074/jbc.ra120.015282] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/23/2020] [Indexed: 12/29/2022] Open
Abstract
The Wnt/β-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 β-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/β-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/β-catenin signaling pathway by controlling the stability of TCF/LEFs. Surprisingly, we found that either overexpression or knockdown of VBP1 decreased Wnt/β-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/β-catenin signaling pathway activity.
Collapse
Affiliation(s)
- Haifeng Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiaozhi Rong
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Caixia Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Ling Lu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yun Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Chengtian Zhao
- Institute of Evolution and Marine Biodiversity and College of Marine Biology, Ocean University of China, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jianfeng Zhou
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
24
|
Kobayashi H, Nishimura H, Kudo N, Osada H, Yoshida M. A novel GSK3 inhibitor that promotes self-renewal in mouse embryonic stem cells. Biosci Biotechnol Biochem 2020; 84:2113-2120. [PMID: 32640867 DOI: 10.1080/09168451.2020.1789445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Small molecules that regulate cell stemness have the potential to make a major contribution to regenerative medicine. In the course of screening for small molecules that affect stemness in mouse embryonic stem cells (mESCs), we discovered that NPD13432, an aurone derivative, promoted self-renewal of mESCs. Normally, mESCs start to differentiate upon withdrawal of 2i/LIF. However, cells treated with the compound continued to express endogenous Nanog, a pluripotency marker protein essential for sustaining the undifferentiated state, even in the absence of 2i/LIF. Biochemical characterization revealed that NPD13432 inhibited GSK3α and GSK3β with IC50 values of 92 nM and 310 nM, respectively, suggesting that the compound promotes self-renewal in mESCs by inhibiting GSK3. The chemical structure of the compound is unique among known molecules with this activity, providing an opportunity to develop new inhibitors of GSK3, as well as chemical tools for investigating cell stemness.
Collapse
Affiliation(s)
- Hiroki Kobayashi
- Drug Discovery Seed Compounds Exploratory Unit, RIKEN Center for Sustainable Resource Science (CSRS) , Saitama, Japan
| | - Haruna Nishimura
- Drug Discovery Seed Compounds Exploratory Unit, RIKEN Center for Sustainable Resource Science (CSRS) , Saitama, Japan
| | - Norio Kudo
- Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science (CSRS) , Saitama, Japan
| | - Hiroyuki Osada
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science (CSRS) , Saitama, Japan
| | - Minoru Yoshida
- Drug Discovery Seed Compounds Exploratory Unit, RIKEN Center for Sustainable Resource Science (CSRS) , Saitama, Japan.,Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science (CSRS) , Saitama, Japan.,Department of Biotechnology, The University of Tokyo , Tokyo, Japan.,Collaborative Research Institute for Innovative Microbiology, The University of Tokyo , Tokyo, Japan
| |
Collapse
|
25
|
Zhang Y, Zhu Z, Ding H, Wan S, Zhang X, Li Y, Ji J, Wang X, Zhang M, Ye SD. β-catenin stimulates Tcf7l1 degradation through recruitment of casein kinase 2 in mouse embryonic stem cells. Biochem Biophys Res Commun 2020; 524:280-287. [PMID: 31987502 DOI: 10.1016/j.bbrc.2020.01.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/13/2020] [Indexed: 11/18/2022]
Abstract
Activation of the Wnt/β-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 Csnk2β, 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/β-catenin signaling pathway. These results expand our understanding of the function and circuit of Wnt/β-catenin signaling pathway in ESCs.
Collapse
Affiliation(s)
- Yan Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Zhenhua Zhu
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Huiwen Ding
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Shengpeng Wan
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Xinbao Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Yuting Li
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Junxiang Ji
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Xin Wang
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Meng Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Shou-Dong Ye
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China.
| |
Collapse
|
26
|
Kumar S, Umair Z, Kumar V, Lee U, Choi SC, Kim J. Ventx1.1 competes with a transcriptional activator Xcad2 to regulate negatively its own expression. BMB Rep 2020. [PMID: 31068250 PMCID: PMC6605524 DOI: 10.5483/bmbrep.2019.52.6.085] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Dorsoventral patterning of body axis in vertebrate embryo is tightly controlled by a complex regulatory network of transcription factors. Ventx1.1 is known as a transcriptional repressor to inhibit dorsal mesoderm formation and neural differentiation in Xenopus. In an attempt to identify, using chromatin immunoprecipitation (ChIP)-Seq, genome-wide binding pattern of Ventx1.1 in Xenopus gastrulae, we observed that Ventx1.1 associates with its own 5′-flanking sequence. In this study, we present evidence that Ventx1.1 binds a cis-acting Ventx1.1 response element (VRE) in its own promoter, leading to repression of its own transcription. Site-directed mutagenesis of the VRE in the Ventx1.1 promoter significantly abrogated this inhibitory autoregulation of Ventx1.1 transcription. Notably, Ventx1.1 and Xcad2, an activator of Ventx1.1 transcription, competitively co-occupied the VRE in the Ventx1.1 promoter. In support of this, mutation of the VRE down-regulated basal and Xcad2-induced levels of Ventx1.1 promoter activity. In addition, overexpression of Ventx1.1 prevented Xcad2 from binding to the Ventx1.1 promoter, and vice versa. Taken together, these results suggest that Ventx1.1 negatively regulates its own transcription in competition with Xcad2, thereby fine-tuning its own expression levels during dorsoventral patterning of Xenopus early embryo.
Collapse
Affiliation(s)
- Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Zobia Umair
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Chuncheon 24252, Korea
| | - Sun-Cheol Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
27
|
Young RM, Ewan KB, Ferrer VP, Allende ML, Godovac-Zimmermann J, Dale TC, Wilson SW. Developmentally regulated Tcf7l2 splice variants mediate transcriptional repressor functions during eye formation. eLife 2019; 8:e51447. [PMID: 31829936 PMCID: PMC6908431 DOI: 10.7554/elife.51447] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/24/2019] [Indexed: 12/11/2022] Open
Abstract
Tcf7l2 mediates Wnt/β-Catenin signalling during development and is implicated in cancer and type-2 diabetes. The mechanisms by which Tcf7l2 and Wnt/β-Catenin signalling elicit such a diversity of biological outcomes are poorly understood. Here, we study the function of zebrafish tcf7l2alternative splice variants and show that only variants that include exon five or an analogous human tcf7l2 variant can effectively provide compensatory repressor function to restore eye formation in embryos lacking tcf7l1a/tcf7l1b function. Knockdown of exon five specific tcf7l2 variants in tcf7l1a mutants also compromises eye formation, and these variants can effectively repress Wnt pathway activity in reporter assays using Wnt target gene promoters. We show that the repressive activities of exon5-coded variants are likely explained by their interaction with Tle co-repressors. Furthermore, phosphorylated residues in Tcf7l2 coded exon5 facilitate repressor activity. Our studies suggest that developmentally regulated splicing of tcf7l2 can influence the transcriptional output of the Wnt pathway.
Collapse
Affiliation(s)
- Rodrigo M Young
- Department of Cell and Developmental BiologyUCLLondonUnited Kingdom
| | - Kenneth B Ewan
- School of Bioscience, Cardiff UniversityCardiffUnited Kingdom
| | | | - Miguel L Allende
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de ChileSantiagoChile
| | | | - Trevor C Dale
- School of Bioscience, Cardiff UniversityCardiffUnited Kingdom
| | - Stephen W Wilson
- Department of Cell and Developmental BiologyUCLLondonUnited Kingdom
| |
Collapse
|
28
|
Shan J, Shen J, Wu M, Zhou H, Feng J, Yao C, Yang Z, Ma Q, Luo Y, Wang Y, Qian C. Tcf7l1 Acts as a Suppressor for the Self-Renewal of Liver Cancer Stem Cells and Is Regulated by IGF/MEK/ERK Signaling Independent of β-Catenin. Stem Cells 2019; 37:1389-1400. [PMID: 31322782 DOI: 10.1002/stem.3063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/26/2019] [Indexed: 12/19/2022]
Abstract
Tcf7l1, which is a key effector molecule of the Wnt/β-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 β-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.
Collapse
Affiliation(s)
- Juanjuan Shan
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Research Center of Bioinspired Materials Science and Engineering, College of Bioengineering, Chongqing University, Chongqing, People's Republic of China
| | - Junjie Shen
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Min Wu
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Haijun Zhou
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Juan Feng
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Chao Yao
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Zhi Yang
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Qinghua Ma
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yanfeng Luo
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Research Center of Bioinspired Materials Science and Engineering, College of Bioengineering, Chongqing University, Chongqing, People's Republic of China
| | - Yuanliang Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Research Center of Bioinspired Materials Science and Engineering, College of Bioengineering, Chongqing University, Chongqing, People's Republic of China
| | - Cheng Qian
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China.,Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| |
Collapse
|
29
|
Osteil P, Studdert JB, Goh HN, Wilkie EE, Fan X, Khoo PL, Peng G, Salehin N, Knowles H, Han JDJ, Jing N, Fossat N, Tam PPL. Dynamics of Wnt activity on the acquisition of ectoderm potency in epiblast stem cells. Development 2019; 146:dev.172858. [PMID: 30890572 DOI: 10.1242/dev.172858] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/11/2019] [Indexed: 01/12/2023]
Abstract
During embryogenesis, the stringent regulation of Wnt activity is crucial for the morphogenesis of the head and brain. The loss of function of the Wnt inhibitor Dkk1 results in elevated Wnt activity, loss of ectoderm lineage attributes from the anterior epiblast, and the posteriorisation of anterior germ layer tissue towards the mesendoderm. The modulation of Wnt signalling may therefore be crucial for the allocation of epiblast cells to ectoderm progenitors during gastrulation. To test this hypothesis, we examined the lineage characteristics of epiblast stem cells (EpiSCs) that were derived and maintained under different signalling conditions. We showed that suppression of Wnt activity enhanced the ectoderm propensity of the EpiSCs. Neuroectoderm differentiation of these EpiSCs was further empowered by the robust re-activation of Wnt activity. Therefore, during gastrulation, the tuning of the signalling activities that mediate mesendoderm differentiation is instrumental for the acquisition of ectoderm potency in the epiblast.
Collapse
Affiliation(s)
- Pierre Osteil
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia .,School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Josh B Studdert
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Hwee Ngee Goh
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Emilie E Wilkie
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia.,Bioinformatics Group, Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Xiaochen Fan
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Poh-Lynn Khoo
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Guangdun Peng
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nazmus Salehin
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Hilary Knowles
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Jing-Dong J Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nicolas Fossat
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia.,School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Patrick P L Tam
- Embryology Unit, Children's Medical Research Institute, Westmead, NSW 2145, Australia.,School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
30
|
Sena E, Rocques N, Borday C, Amin HSM, Parain K, Sitbon D, Chesneau A, Durand BC. Barhl2 maintains T-cell factors as repressors, and thereby switches off the Wnt/β-Catenin response driving Spemann organizer formation. Development 2019; 146:dev.173112. [DOI: 10.1242/dev.173112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/11/2019] [Indexed: 12/19/2022]
Abstract
A hallmark of Wnt/β-Catenin signaling is the extreme diversity of its transcriptional response, which varies depending on cell and developmental context. What controls this diversity is poorly understood. In all cases, the switch from transcriptional repression to activation depends on a nuclear increase in β-Catenin, which detaches the transcription factor T-cell Factor-7 like 1 (Tcf7l1) bound to Groucho (Gro) transcriptional co-repressors from its DNA binding sites and transiently converts Tcf7/Lymphoid enhancer binding factor 1 (Lef1) into a transcriptional activator. One of the earliest and evolutionarily conserved functions of Wnt/β-Catenin signaling is the induction of the blastopore lip organizer. Here, we demonstrate that the evolutionarily conserved BarH-like homeobox-2 (Barhl2) protein stabilizes the Tcf7l1-Gro complex and maintains repressed expression of Tcf target genes by a mechanism that depends on histone deacetylase 1 (Hdac-1) activity. In this way, Barhl2 switches off the Wnt/β-Catenin-dependent early transcriptional response, thereby limiting the formation of the organizer in time and/or space. This study reveals a novel nuclear inhibitory mechanism of Wnt/Tcf signaling that switches off organizer fate determination.
Collapse
Affiliation(s)
- Elena Sena
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
| | - Nathalie Rocques
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
| | - Caroline Borday
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - Harem Sabr Muhamad Amin
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, S1.7 CNRS 8197, INSERM U1024 46 rue d'Ulm 75005, Paris F-75005, France
| | - Karine Parain
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - David Sitbon
- Institut Curie, PSL Research University, CNRS, UMR3664, Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Albert Chesneau
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - Béatrice C. Durand
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, S1.7 CNRS 8197, INSERM U1024 46 rue d'Ulm 75005, Paris F-75005, France
| |
Collapse
|
31
|
Watanabe T, Yamamoto T, Tsukano K, Hirano S, Horikawa A, Michiue T. Fam46a regulates BMP-dependent pre-placodal ectoderm differentiation in Xenopus. Development 2018; 145:dev.166710. [PMID: 30291163 DOI: 10.1242/dev.166710] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 09/06/2018] [Indexed: 12/30/2022]
Abstract
The pre-placodal ectoderm (PPE) is a specialized ectodermal region which gives rise to the sensory organs and other systems. The PPE is induced from the neural plate border during neurulation, but the molecular mechanism of PPE formation is not fully understood. In this study, we examined the role of a newly identified PPE gene, Fam46a, during embryogenesis. Fam46a contains a nucleoside triphosphate transferase domain, but its function in early development was previously unclear. We show that Fam46a is expressed in the PPE in Xenopus embryos, and Fam46a knockdown induces abnormalities in the eye formation and the body color. At the neurula stage, Fam46a upregulates the expression of PPE genes and inhibits neural crest formation. We also show that Fam46a physically interacts with Smad1/Smad4 and positively regulates BMP signaling. From these results, we conclude that Fam46a is required for PPE formation via the positive regulation of BMP signaling. Our study provides a new mechanism of ectodermal patterning via cell-autonomous regulation of BMP signaling in the PPE.
Collapse
Affiliation(s)
- Tomoko Watanabe
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Takayoshi Yamamoto
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Kohei Tsukano
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Sayuki Hirano
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Ayumi Horikawa
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Tatsuo Michiue
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| |
Collapse
|
32
|
Wingless Signaling: A Genetic Journey from Morphogenesis to Metastasis. Genetics 2018; 208:1311-1336. [PMID: 29618590 DOI: 10.1534/genetics.117.300157] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 12/13/2017] [Indexed: 12/15/2022] Open
Abstract
This FlyBook chapter summarizes the history and the current state of our understanding of the Wingless signaling pathway. Wingless, the fly homolog of the mammalian Wnt oncoproteins, plays a central role in pattern generation during development. Much of what we know about the pathway was learned from genetic and molecular experiments in Drosophila melanogaster, and the core pathway works the same way in vertebrates. Like most growth factor pathways, extracellular Wingless/Wnt binds to a cell surface complex to transduce signal across the plasma membrane, triggering a series of intracellular events that lead to transcriptional changes in the nucleus. Unlike most growth factor pathways, the intracellular events regulate the protein stability of a key effector molecule, in this case Armadillo/β-catenin. A number of mysteries remain about how the "destruction complex" destabilizes β-catenin and how this process is inactivated by the ligand-bound receptor complex, so this review of the field can only serve as a snapshot of the work in progress.
Collapse
|
33
|
Grainger S, Willert K. Mechanisms of Wnt signaling and control. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2018; 10:e1422. [PMID: 29600540 PMCID: PMC6165711 DOI: 10.1002/wsbm.1422] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/14/2018] [Accepted: 02/22/2018] [Indexed: 01/17/2023]
Abstract
The Wnt signaling pathway is a highly conserved system that regulates complex biological processes across all metazoan species. At the cellular level, secreted Wnt proteins serve to break symmetry and provide cells with positional information that is critical to the patterning of the entire body plan. At the organismal level, Wnt signals are employed to orchestrate fundamental developmental processes, including the specification of the anterior-posterior body axis, induction of the primitive streak and ensuing gastrulation movements, and the generation of cell and tissue diversity. Wnt functions extend into adulthood where they regulate stem cell behavior, tissue homeostasis, and damage repair. Disruption of Wnt signaling activity during embryonic development or in adults results in a spectrum of abnormalities and diseases, including cancer. The molecular mechanisms that underlie the myriad of Wnt-regulated biological effects have been the subject of intense research for over three decades. This review is intended to summarize our current understanding of how Wnt signals are generated and interpreted. This article is categorized under: Biological Mechanisms > Cell Signaling Developmental Biology > Stem Cell Biology and Regeneration.
Collapse
Affiliation(s)
- Stephanie Grainger
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla California
| | - Karl Willert
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla California
| |
Collapse
|
34
|
Moorer MC, Riddle RC. Regulation of Osteoblast Metabolism by Wnt Signaling. Endocrinol Metab (Seoul) 2018; 33:318-330. [PMID: 30112869 PMCID: PMC6145954 DOI: 10.3803/enm.2018.33.3.318] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 07/01/2018] [Accepted: 07/08/2018] [Indexed: 12/13/2022] Open
Abstract
Wnt/β-catenin signaling plays a critical role in the achievement of peak bone mass, affecting the commitment of mesenchymal progenitors to the osteoblast lineage and the anabolic capacity of osteoblasts depositing bone matrix. Recent studies suggest that this evolutionarily-conserved, developmental pathway exerts its anabolic effects in part by coordinating osteoblast activity with intermediary metabolism. These findings are compatible with the cloning of the gene encoding the low-density lipoprotein related receptor-5 (LRP5) Wnt co-receptor from a diabetes-susceptibility locus and the now well-established linkage between Wnt signaling and metabolism. In this article, we provide an overview of the role of Wnt signaling in whole-body metabolism and review the literature regarding the impact of Wnt signaling on the osteoblast's utilization of three different energy sources: fatty acids, glucose, and glutamine. Special attention is devoted to the net effect of nutrient utilization and the mode of regulation by Wnt signaling. Mechanistic studies indicate that the utilization of each substrate is governed by a unique mechanism of control with β-catenin-dependent signaling regulating fatty acid β-oxidation, while glucose and glutamine utilization are β-catenin-independent and downstream of mammalian target of rapamycin complex 2 (mTORC2) and mammalian target of rapamycin complex 1 (mTORC1) activation, respectively. The emergence of these data has provided a new context for the mechanisms by which Wnt signaling influences bone development.
Collapse
Affiliation(s)
- Megan C Moorer
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Baltimore Veterans Administration Medical Center, Baltimore, MD, USA.
| |
Collapse
|
35
|
Aloysius A, DasGupta R, Dhawan J. The transcription factor Lef1 switches partners from β-catenin to Smad3 during muscle stem cell quiescence. Sci Signal 2018; 11:11/540/eaan3000. [PMID: 30042129 DOI: 10.1126/scisignal.aan3000] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skeletal muscle stem cells (MuSCs), also known as satellite cells, persist in adult mammals by entering a state of quiescence (G0) during the early postnatal period. Quiescence is reversed during damage-induced regeneration and re-established after regeneration. Entry of cultured myoblasts into G0 is associated with a specific, reversible induction of Wnt target genes, thus implicating members of the Tcf and Lef1 (Tcf/Lef) transcription factor family, which mediate transcriptional responses to Wnt signaling, in the initiation of quiescence. We found that the canonical Wnt effector β-catenin, which cooperates with Tcf/Lef, was dispensable for myoblasts to enter quiescence. Using pharmacological and genetic approaches in cultured C2C12 myoblasts and in MuSCs, we demonstrated that Tcf/Lef activity during quiescence depended not on β-catenin but on the transforming growth factor-β (TGF-β) effector and transcriptional coactivator Smad3, which colocalized with Lef1 at canonical Wnt-responsive elements and directly interacted with Lef1 specifically in G0 Depletion of Smad3, but not β-catenin, reduced Lef1 occupancy at target promoters, Tcf/Lef target gene expression, and self-renewal of myoblasts. In vivo, MuSCs underwent a switch from β-catenin-Lef1 to Smad3-Lef1 interactions during the postnatal switch from proliferation to quiescence, with β-catenin-Lef1 interactions recurring during damage-induced reactivation. Our findings suggest that the interplay of Wnt-Tcf/Lef and TGF-β-Smad3 signaling activates canonical Wnt target promoters in a manner that depends on β-catenin during myoblast proliferation but is independent of β-catenin during MuSC quiescence.
Collapse
Affiliation(s)
- Ajoy Aloysius
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India.,Centre for Cellular and Molecular Biology, Hyderabad 500007, India.,Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India
| | | | - Jyotsna Dhawan
- Centre for Cellular and Molecular Biology, Hyderabad 500007, India. .,Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India
| |
Collapse
|
36
|
Abstract
TGF-β family ligands function in inducing and patterning many tissues of the early vertebrate embryonic body plan. Nodal signaling is essential for the specification of mesendodermal tissues and the concurrent cellular movements of gastrulation. Bone morphogenetic protein (BMP) signaling patterns tissues along the dorsal-ventral axis and simultaneously directs the cell movements of convergence and extension. After gastrulation, a second wave of Nodal signaling breaks the symmetry between the left and right sides of the embryo. During these processes, elaborate regulatory feedback between TGF-β ligands and their antagonists direct the proper specification and patterning of embryonic tissues. In this review, we summarize the current knowledge of the function and regulation of TGF-β family signaling in these processes. Although we cover principles that are involved in the development of all vertebrate embryos, we focus specifically on three popular model organisms: the mouse Mus musculus, the African clawed frog of the genus Xenopus, and the zebrafish Danio rerio, highlighting the similarities and differences between these species.
Collapse
Affiliation(s)
- Joseph Zinski
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Benjamin Tajer
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Mary C Mullins
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| |
Collapse
|
37
|
Teegala S, Chauhan R, Lei E, Weinstein DC. Tbx2 is required for the suppression of mesendoderm during early Xenopus development. Dev Dyn 2018; 247:903-913. [PMID: 29633424 DOI: 10.1002/dvdy.24633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/14/2018] [Accepted: 03/31/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND T-box family proteins are DNA-binding transcriptional regulators that play crucial roles during germ layer formation in the early vertebrate embryo. Well-characterized members of this family, including the transcriptional activators Brachyury and VegT, are essential for the proper formation of mesoderm and endoderm, respectively. To date, T-box proteins have not been shown to play a role in the promotion of the third primary germ layer, ectoderm. RESULTS Here, we report that the T-box factor Tbx2 is both sufficient and necessary for ectodermal differentiation in the frog Xenopus laevis. Tbx2 is expressed zygotically in the presumptive ectoderm, during blastula and gastrula stages. Ectopic expression of Tbx2 represses mesoderm and endoderm, while loss of Tbx2 leads to inappropriate expression of mesoderm- and endoderm-specific genes in the region fated to give rise to ectoderm. Misexpression of Tbx2 also promotes neural tissue in animal cap explants, suggesting that Tbx2 plays a role in both the establishment of ectodermal fate and its dorsoventral patterning. CONCLUSIONS Our studies demonstrate that Tbx2 functions as a transcriptional repressor during germ layer formation, and suggest that this activity is mediated in part through repression of target genes that are stimulated, in the mesendoderm, by transactivating T-box proteins. Taken together, our results point to a critical role for Tbx2 in limiting the potency of blastula-stage progenitor cells during vertebrate germ layer differentiation. Developmental Dynamics 247:903-913, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sushma Teegala
- Department of Biology, The Graduate Center, City University of New York, New York.,Department of Biology, Queens College, City University of New York, Flushing, New York
| | - Riddhi Chauhan
- Department of Biology, Queens College, City University of New York, Flushing, New York
| | - Emily Lei
- Department of Biology, Queens College, City University of New York, Flushing, New York
| | - Daniel C Weinstein
- Department of Biology, Queens College, City University of New York, Flushing, New York
| |
Collapse
|
38
|
Xu X, Guo M, Zhang N, Ye S. Telomeric noncoding RNA promotes mouse embryonic stem cell self-renewal through inhibition of TCF3 activity. Am J Physiol Cell Physiol 2018. [PMID: 29513567 DOI: 10.1152/ajpcell.00292.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
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/β-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.
Collapse
Affiliation(s)
- Xiaojuan Xu
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences , Hefei , People's Republic of China.,University of Science and Technology of China , Hefei , People's Republic of China
| | - Mengmeng Guo
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University , Hefei , People's Republic of China
| | - Na Zhang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences , Hefei , People's Republic of China
| | - Shoudong Ye
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University , Hefei , People's Republic of China
| |
Collapse
|
39
|
Blaquiere JA, Wong KKL, Kinsey SD, Wu J, Verheyen EM. Homeodomain-interacting protein kinase promotes tumorigenesis and metastatic cell behavior. Dis Model Mech 2018; 11:dmm.031146. [PMID: 29208636 PMCID: PMC5818076 DOI: 10.1242/dmm.031146] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/08/2017] [Indexed: 12/21/2022] Open
Abstract
Aberrations in signaling pathways that regulate tissue growth often lead to tumorigenesis. Homeodomain-interacting protein kinase (Hipk) family members are reported to have distinct and contradictory effects on cell proliferation and tissue growth. From these studies, it is clear that much remains to be learned about the roles of Hipk family protein kinases in proliferation and cell behavior. Previous work has shown that Drosophila Hipk is a potent growth regulator, thus we predicted that it could have a role in tumorigenesis. In our study of Hipk-induced phenotypes, we observed the formation of tumor-like structures in multiple cell types in larvae and adults. Furthermore, elevated Hipk in epithelial cells induces cell spreading, invasion and epithelial-to-mesenchymal transition (EMT) in the imaginal disc. Further evidence comes from cell culture studies, in which we expressed Drosophila Hipk in human breast cancer cells and showed that it enhances proliferation and migration. Past studies have shown that Hipk can promote the action of conserved pathways implicated in cancer and EMT, such as Wnt/Wingless, Hippo, Notch and JNK. We show that Hipk phenotypes are not likely to arise from activation of a single target, but rather through a cumulative effect on numerous target pathways. Most Drosophila tumor models involve mutations in multiple genes, such as the well-known RasV12 model, in which EMT and invasiveness occur after the additional loss of the tumor suppressor gene scribble. Our study reveals that elevated levels of Hipk on their own can promote both hyperproliferation and invasive cell behavior, suggesting that Hipk family members could be potent oncogenes and drivers of EMT. Summary: The protein kinase Hipk can promote proliferation and invasive behaviors, and can synergize with known cancer pathways, in a new Drosophila model for tumorigenesis.
Collapse
Affiliation(s)
- Jessica A Blaquiere
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Kenneth Kin Lam Wong
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Stephen D Kinsey
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Jin Wu
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Esther M Verheyen
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| |
Collapse
|
40
|
RNAi-mediated TCF-3 gene silencing inhibits proliferation of Eca-109 esophageal cancer cells by inducing apoptosis. Biosci Rep 2017; 37:BSR20170799. [PMID: 28864779 PMCID: PMC5678029 DOI: 10.1042/bsr20170799] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/24/2017] [Accepted: 08/30/2017] [Indexed: 01/24/2023] Open
Abstract
Esophageal cancer (EC) remains an important health problem in China. In the present study, through the use of siRNA, specific gene knockdown of transcription factor 3 gene (TCF-3) was achieved in vitro and the effect of TCF-3 gene on human EC Eca-109 cell proliferation and apoptosis. Eca-109 cells were treated using negative control (NC) of siRNA against TCF-3 (siTCF-3) and siTCF-3 group. Colony formation assay was used to detect the colony formation ability in Eca-109 cells. MTT assay was used to measure the cell growth and viability, whereas BrDU assay was used to evaluate cell proliferation, and flow cytometry (FCM) to assess cell apoptosis. Reverse-transcription quantitative PCR (RT-qPCR) was applied to measure TCF-3 gene expression. Protein expressions of TCF-3, apoptosis-related proteins, Bcl-2, Bax, and caspase-3 were determined using Western blotting. Transfection of siTCF-3 successfully down-regulated TCF-3 gene expression. In addition, siTCF-3, reduced Eca-109 cell viability and proliferation, in a time-dependent manner, and inhibited progression of cell cycle from G0/G1 to S-stage. When treated with siTCF-3, the Eca-109 cells exhibited increased apoptosis, with up-regulated cleaved caspase and Bax expressions, whereas Bcl-2 expression was down-regulated. The present study shows that TCF-3 gene silencing inhibits Eca-109 cell growth and proliferation, suppresses cell cycle progression, and promotes apoptosis, which might serve as a new objective for EC treatment.
Collapse
|
41
|
Cao Q, Shen Y, Zheng W, Liu H, Liu C. Tcf7l1 promotes transcription of Kruppel-likefactor 4 during Xenopus embryogenesis. J Biomed Res 2017; 32:215. [PMID: 29336356 PMCID: PMC6265397 DOI: 10.7555/jbr.32.20170056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/27/2017] [Indexed: 11/30/2022] Open
Abstract
Kruppel-like factor 4 (Klf4) is a zinc finger transcriptionfactor and plays crucial roles in Xenopus embryogenesis. However, its regulation during embryogenesis is stillunclear. Here, we report that Tcf7l1, a key downstream transducerof the Wnt signaling pathway, could promote Klf4 transcription and stimulate Klf4 promoter activity in early Xenopus embryos. Furthermore, cycloheximide treatmentshowed a direct effect on Klf4 transcriptionfacilitated by Tcf7l1. Moreover, the dominant negative form of Tcf7l1(dnTcf7l1), which lacks N-terminusof the β-catenin binding motif, could still activate Klf4 transcription, suggesting that thisregulation is Wnt/β-catenin independent. Taken together, ourresults demonstrate that Tcf7l1 lies upstream of Klf4 to maintainits expression level during Xenopus embryogenesis.
Collapse
Affiliation(s)
- Qing Cao
- . College of Medicine, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Yan Shen
- . College of Medicine, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Wei Zheng
- . College of Medicine, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Hao Liu
- . College of Medicine, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Chen Liu
- . Department of Developmental Genetics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
42
|
Su H, Sureda-Gomez M, Rabaneda-Lombarte N, Gelabert M, Xie J, Wu W, Adell T. A C-terminally truncated form of β-catenin acts as a novel regulator of Wnt/β-catenin signaling in planarians. PLoS Genet 2017; 13:e1007030. [PMID: 28976975 PMCID: PMC5643146 DOI: 10.1371/journal.pgen.1007030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 10/16/2017] [Accepted: 09/17/2017] [Indexed: 12/12/2022] Open
Abstract
β-Catenin, the core element of the Wnt/β-catenin pathway, is a multifunctional and evolutionarily conserved protein which performs essential roles in a variety of developmental and homeostatic processes. Despite its crucial roles, the mechanisms that control its context-specific functions in time and space remain largely unknown. The Wnt/β-catenin pathway has been extensively studied in planarians, flatworms with the ability to regenerate and remodel the whole body, providing a ‘whole animal’ developmental framework to approach this question. Here we identify a C-terminally truncated β-catenin (β-catenin4), generated by gene duplication, that is required for planarian photoreceptor cell specification. Our results indicate that the role of β-catenin4 is to modulate the activity of β-catenin1, the planarian β-catenin involved in Wnt signal transduction in the nucleus, mediated by the transcription factor TCF-2. This inhibitory form of β-catenin, expressed in specific cell types, would provide a novel mechanism to modulate nuclear β-catenin signaling levels. Genomic searches and in vitro analysis suggest that the existence of a C-terminally truncated form of β-catenin could be an evolutionarily conserved mechanism to achieve a fine-tuned regulation of Wnt/β-catenin signaling in specific cellular contexts. The Wnt signaling pathway is essential for proper intercellular communication in every developmental process since it controls basic cellular events as cell fate or proliferation. The key element of the Wnt signaling is β-catenin, which controls the transcription of multiple genes in the Wnt receiving cell. A main level of regulation of the Wnt/β-catenin signaling occurs in the cytoplasm, where β-catenin protein levels depend on the activity of the β-catenin destruction complex. However, once it reaches the nucleus, β-catenin transcriptional activity requires a fine-tuned regulation to enable the multiple context-specific responses that it performs. These nuclear mechanisms that regulate the Wnt/β-catenin signaling remain poorly understood. Here we report the existence of C-terminal truncated forms of β-catenin in planarians (β-cat3 and 4), which, in vitro, do not show transactivation activity and compete with the canonical planarian β-catenin (β-cat1), thus acting as competitor inhibitors. Functional analyses in planarians indicate that β-cat4 acts as a negative regulator of β-cat1 during planarian eye photoreceptor specification. We provide evidence to suggest that this novel mechanism for the regulation of nuclear β-catenin activity could be conserved across animal evolution.
Collapse
Affiliation(s)
- Hanxia Su
- MOE Key Laboratory of Protein Science, School of Life Sciences, Tsinghua University, Beijing, China
| | - Miquel Sureda-Gomez
- Departament de Genètica, Microbiologia i Estadística,Facultat de Biologia, Universitat de Barcelona and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Catalunya, Spain
| | - Neus Rabaneda-Lombarte
- Departament de Genètica, Microbiologia i Estadística,Facultat de Biologia, Universitat de Barcelona and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Catalunya, Spain
| | - Maria Gelabert
- Departament de Genètica, Microbiologia i Estadística,Facultat de Biologia, Universitat de Barcelona and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Catalunya, Spain
| | - Jianlei Xie
- MOE Key Laboratory of Protein Science, School of Life Sciences, Tsinghua University, Beijing, China
| | - Wei Wu
- MOE Key Laboratory of Protein Science, School of Life Sciences, Tsinghua University, Beijing, China
| | - Teresa Adell
- Departament de Genètica, Microbiologia i Estadística,Facultat de Biologia, Universitat de Barcelona and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Catalunya, Spain
- * E-mail:
| |
Collapse
|
43
|
Abstract
Wnt/β-catenin signaling is highly conserved throughout metazoans, is required for numerous essential events in development, and serves as a stem cell niche signal in many contexts. Misregulation of the pathway is linked to several human pathologies, most notably cancer. Wnt stimulation results in stabilization and nuclear import of β-catenin, which then acts as a transcriptional co-activator. Transcription factors of the T-cell family (TCF) are the best-characterized nuclear binding partners of β-catenin and mediators of Wnt gene regulation. This review provides an update on what is known about the transcriptional activation of Wnt target genes, highlighting recent work that modifies the conventional model. Wnt/β-catenin signaling regulates genes in a highly context-dependent manner, and the role of other signaling pathways and TCF co-factors in this process will be discussed. Understanding Wnt gene regulation has served to elucidate many biological roles of the pathway, and we will use examples from stem cell biology, metabolism, and evolution to illustrate some of the rich Wnt biology that has been uncovered.
Collapse
Affiliation(s)
| | - Ken M Cadigan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
44
|
Ubiquitin C-terminal hydrolase37 regulates Tcf7 DNA binding for the activation of Wnt signalling. Sci Rep 2017; 7:42590. [PMID: 28198400 PMCID: PMC5309806 DOI: 10.1038/srep42590] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/11/2017] [Indexed: 12/18/2022] Open
Abstract
The Tcf/Lef family of transcription factors mediates the Wnt/β-catenin pathway that is involved in a wide range of biological processes, including vertebrate embryogenesis and diverse pathogenesis. Post-translational modifications, including phosphorylation, sumoylation and acetylation, are known to be important for the regulation of Tcf/Lef proteins. However, the importance of ubiquitination and ubiquitin-mediated regulatory mechanisms for Tcf/Lef activity are still unclear. Here, we newly show that ubiquitin C-terminal hydrolase 37 (Uch37), a deubiquitinase, interacts with Tcf7 (formerly named Tcf1) to activate Wnt signalling. Biochemical analyses demonstrated that deubiquitinating activity of Uch37 is not involved in Tcf7 protein stability but is required for the association of Tcf7 to target gene promoter in both Xenopus embryo and human liver cancer cells. In vivo analyses further revealed that Uch37 functions as a positive regulator of the Wnt/β-catenin pathway downstream of β-catenin stabilization that is required for the expression of ventrolateral mesoderm genes during Xenopus gastrulation. Our study provides a new mechanism for chromatin occupancy of Tcf7 and uncovers the physiological significance of Uch37 during early vertebrate development by regulating the Wnt/β-catenin pathway.
Collapse
|
45
|
Houston DW. Vertebrate Axial Patterning: From Egg to Asymmetry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 953:209-306. [PMID: 27975274 PMCID: PMC6550305 DOI: 10.1007/978-3-319-46095-6_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The emergence of the bilateral embryonic body axis from a symmetrical egg has been a long-standing question in developmental biology. Historical and modern experiments point to an initial symmetry-breaking event leading to localized Wnt and Nodal growth factor signaling and subsequent induction and formation of a self-regulating dorsal "organizer." This organizer forms at the site of notochord cell internalization and expresses primarily Bone Morphogenetic Protein (BMP) growth factor antagonists that establish a spatiotemporal gradient of BMP signaling across the embryo, directing initial cell differentiation and morphogenesis. Although the basics of this model have been known for some time, many of the molecular and cellular details have only recently been elucidated and the extent that these events remain conserved throughout vertebrate evolution remains unclear. This chapter summarizes historical perspectives as well as recent molecular and genetic advances regarding: (1) the mechanisms that regulate symmetry-breaking in the vertebrate egg and early embryo, (2) the pathways that are activated by these events, in particular the Wnt pathway, and the role of these pathways in the formation and function of the organizer, and (3) how these pathways also mediate anteroposterior patterning and axial morphogenesis. Emphasis is placed on comparative aspects of the egg-to-embryo transition across vertebrates and their evolution. The future prospects for work regarding self-organization and gene regulatory networks in the context of early axis formation are also discussed.
Collapse
Affiliation(s)
- Douglas W Houston
- Department of Biology, The University of Iowa, 257 BB, Iowa City, IA, 52242, USA.
| |
Collapse
|
46
|
Masuda T, Ishitani T. Context-dependent regulation of the β-catenin transcriptional complex supports diverse functions of Wnt/β-catenin signaling. J Biochem 2016; 161:9-17. [PMID: 28013224 DOI: 10.1093/jb/mvw072] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022] Open
Abstract
Wnt/β-catenin signaling is activated repeatedly during an animal's lifespan, and it controls gene expression through its essential nuclear effector, β-catenin, to regulate embryogenesis, organogenesis, and adult homeostasis. Although the β-catenin transcriptional complex has the ability to induce the expression of many genes to exert its diverse roles, it chooses and transactivates a specific gene set from among its numerous target genes depending on the context. For example, the β-catenin transcriptional complex stimulates the expression of cell cycle-related genes and consequent cell proliferation in neural progenitor cells, while it promotes the expression of neural differentiation-related genes in differentiating neurons. Recent studies using animal and cell culture models have gradually improved our understanding of the molecular basis underlying such context-dependent actions of the β-catenin transcriptional complex. Here, we describe eight mechanisms that support β-catenin-mediated context-dependent gene regulation, and their spatio-temporal regulation during vertebrate development. In addition, we discuss their contribution to the diverse functions of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Takamasa Masuda
- Division of Cell Regulation Systems, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Tohru Ishitani
- Division of Cell Regulation Systems, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| |
Collapse
|
47
|
Blaquiere JA, Verheyen EM. Homeodomain-Interacting Protein Kinases: Diverse and Complex Roles in Development and Disease. Curr Top Dev Biol 2016; 123:73-103. [PMID: 28236976 DOI: 10.1016/bs.ctdb.2016.10.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Homeodomain-interacting protein kinase (Hipk) family of proteins plays diverse, and at times conflicting, biological roles in normal development and disease. In this review we will highlight developmental and cellular roles for Hipk proteins, with an emphasis on the pleiotropic and essential physiological roles revealed through genetic studies. We discuss the myriad ways of regulating Hipk protein function, and how these may contribute to the diverse cellular roles. Furthermore we will describe the context-specific activities of Hipk family members in diseases such as cancer and fibrosis, including seemingly contradictory tumor-suppressive and oncogenic activities. Given the diverse signaling pathways regulated by Hipk proteins, it is likely that Hipks act to fine-tune signaling and may mediate cross talk in certain contexts. Such regulation is emerging as vital for development and in disease.
Collapse
Affiliation(s)
- Jessica A Blaquiere
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Esther M Verheyen
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
48
|
Lin H, Min Z, Tao Q. The MLL/Setd1b methyltransferase is required for the Spemann's organizer gene activation in Xenopus. Mech Dev 2016; 142:1-9. [DOI: 10.1016/j.mod.2016.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 08/07/2016] [Accepted: 08/08/2016] [Indexed: 01/22/2023]
|
49
|
Glycans define the stemness of naïve and primed pluripotent stem cells. Glycoconj J 2016; 34:737-747. [PMID: 27796614 DOI: 10.1007/s10719-016-9740-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 10/20/2022]
Abstract
Cell surface glycans are tissue-specific and developmentally regulated. They function as essential modulators in cell-cell interactions, cell-extracellular matrix interactions, and ligand-receptor interactions, binding to various ligands, including Wnt, fibroblast growth factors, and bone morphogenetic proteins. Embryonic stem (ES) cells, originally derived from the inner cell mass of blastocysts, have the essential characteristics of pluripotency and self-renewal. Recently, it has been proposed that mouse and human conventional ES cells are present in different developmental stages, namely pre-implantation blastocyst and post-implantation blastocyst stages, also called the naïve state and the primed state, respectively. They therefore require different extrinsic signals for the maintenance of self-renewal and pluripotency, and also appear to require different surface glycans. Understanding of molecular mechanisms involving glycans in self-renewal and pluripotency of ES cells is increasingly important for potential clinical applications, as well as for basic research. This review focuses on the roles of glycans in the two different states of pluripotent stem cells, namely the naïve state and the primed state, and the transition between these two states.
Collapse
|
50
|
Lu T, Bao Z, Wang Y, Yang L, Lu B, Yan K, Wang S, Wei H, Zhang Z, Cui G. Karyopherinβ1 regulates proliferation of human glioma cells via Wnt/β-catenin pathway. Biochem Biophys Res Commun 2016; 478:1189-97. [PMID: 27568288 DOI: 10.1016/j.bbrc.2016.08.093] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 08/14/2016] [Indexed: 11/28/2022]
Abstract
Karyopherinβ1 (KPNB1), one of the cytosolic factors involved in the selective protein transport across nucleus, docked at nuclear pore complex and transported through nuclear envelope in an ATP-dependent style, assisting proteins to be recognized as import substrates. It has been reported to be bound up with the origination and progress of lung cancer, cervical cancer, head and neck cancer and hepatocellular carcinoma. In current study, we demonstrated for the first time that the role of KPNB1 in human glioma. KPNB1 was over-expressed as the well-known trend of Ki-67(p < 0.01) and tightly closed to poor prognosis, as an independent prognostic factor. In vitro, up-regulation of KPNB1 was accompanied by certain rising levels of proliferation markers, employing U251 and U87MG cells as serum-starve models. Silencing KPNB1 in U251 and U87MG led to G1 phase arrested directly via flow cytometry analysis. In the nucleus of KPNB1-depletion cell models, the decreasing expression of KPNB1 and β-catenin was detected respectively, which indicated that KPNB1 functioned via β-catenin signal. Besides, the interaction between KPNB1 and β-catenin was proved clearly by immunoprecipitation. Taken together, it showed that KPNB1 might enhance human glioma proliferation via Wnt/β-Catenin Pathway.
Collapse
Affiliation(s)
- Ting Lu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Zhen Bao
- Department of Neurosurgery, The Affiliated Dushuhu Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Yunfeng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Lixiang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Bing Lu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Ke Yan
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Shaozhen Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - He Wei
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Zhe Zhang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Gang Cui
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China.
| |
Collapse
|