1
|
Yu G, Chen Y, Hu Y, Zhou Y, Ding X, Zhou X. Roles of transducin-like enhancer of split (TLE) family proteins in tumorigenesis and immune regulation. Front Cell Dev Biol 2022; 10:1010639. [PMID: 36438567 PMCID: PMC9692235 DOI: 10.3389/fcell.2022.1010639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/31/2022] [Indexed: 08/16/2023] Open
Abstract
Mammalian transducin-like enhancer of split family proteins (TLEs) are homologous to Drosophila Groucho (Gro) and are essential transcriptional repressors. Seven TLE family members, TLE1-7, have been identified to date. These proteins do not bind DNA directly; instead, they bind a set of transcription factors and thereby inhibit target gene expression. Loss of TLEs in mice usually leads to defective early development; however, TLE functions in developmentally mature cells are unclear. Recent studies have revealed that TLEs are dysregulated in certain human cancer types and may function as oncogenes or tumor suppressors in different contexts. TLE levels also affect the efficacy of cancer treatments and the development of drug resistance. In addition, TLEs play critical roles in the development and function of immune cells, including macrophages and lymphocytes. In this review, we provide updates on the expression, function, and mechanism of TLEs; discuss the roles played by TLEs in tumorigenesis and the inflammatory response; and elaborate on several TLE-associated signaling pathways, including the Notch, Wnt, and MAPK pathways. Finally, we discuss potential strategies for targeting TLEs in cancer therapy.
Collapse
Affiliation(s)
- Guiping Yu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, China
| | - Yiqi Chen
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Yuwen Hu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Yan Zhou
- Department of Periodontology, The Affiliated Nantong Stomatological Hospital of Nantong University, Nantong, China
| | - Xiaoling Ding
- Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaorong Zhou
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| |
Collapse
|
2
|
The Role of Protein Kinase CK2 in Development and Disease Progression: A Critical Review. J Dev Biol 2022; 10:jdb10030031. [PMID: 35997395 PMCID: PMC9397010 DOI: 10.3390/jdb10030031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Protein kinase CK2 (CK2) is a ubiquitous holoenzyme involved in a wide array of developmental processes. The involvement of CK2 in events such as neurogenesis, cardiogenesis, skeletogenesis, and spermatogenesis is essential for the viability of almost all organisms, and its role has been conserved throughout evolution. Further into adulthood, CK2 continues to function as a key regulator of pathways affecting crucial processes such as osteogenesis, adipogenesis, chondrogenesis, neuron differentiation, and the immune response. Due to its vast role in a multitude of pathways, aberrant functioning of this kinase leads to embryonic lethality and numerous diseases and disorders, including cancer and neurological disorders. As a result, CK2 is a popular target for interventions aiming to treat the aforementioned diseases. Specifically, two CK2 inhibitors, namely CX-4945 and CIBG-300, are in the early stages of clinical testing and exhibit promise for treating cancer and other disorders. Further, other researchers around the world are focusing on CK2 to treat bone disorders. This review summarizes the current understanding of CK2 in development, the structure of CK2, the targets and signaling pathways of CK2, the implication of CK2 in disease progression, and the recent therapeutics developed to inhibit the dysregulation of CK2 function in various diseases.
Collapse
|
3
|
Ma Q, Xiao F, Hao Y, Song Z, Zhang J, Si C, Liang C, Liu D. The prognostic role of the Transducin-like Enhancer of split protein family in lung adenocarcinoma. Transl Lung Cancer Res 2021; 10:3251-3263. [PMID: 34430362 PMCID: PMC8350083 DOI: 10.21037/tlcr-21-582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/26/2021] [Indexed: 02/01/2023]
Abstract
Background Lung cancer claims more lives than any other cancer worldwide. Lung adenocarcinoma (LUAD) accounts for approximately 40% of all lung cancers. Members of the Transducin-like Enhancer of split (TLE) protein family repress transcription through multiple mechanisms; however, their prognostic value in LUAD is still unclear. Methods A dataset from The Cancer Genome Atlas was used to analyze the relationship between the expression of TLE family members and outcomes of LUAD. The expression of TLE family members in 59 normal and 513 tumor samples in the TCGA dataset was selected. For paired analysis, 57 normal and 57 tumor paired tissues were selected. Gene Ontology (GO) term and Reactome pathway enrichment analyses of the TLE family members were performed. Progression-free survival (PFS) and overall survival (OS) served as endpoints in this study. All statistical analyses were performed with R 3.6.0. Results The expression levels of TLE family proteins differed between 59 normal and 513 tumor samples. High TLE1 and low TLE2 levels were associated with poor progression-free and OS (all P<0.050). Multivariate analysis demonstrated that high TLE1 expression and low TLE2 expression were independent risk factors for a poor outcome in LUAD. Moreover, the combined expression of these two proteins was a good tool for prognostication. Conclusions High TLE1 expression and low TLE2 are independent adverse prognostic factors in LUAD and can serve as prognostic biomarkers.
Collapse
Affiliation(s)
- Qianli Ma
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Fei Xiao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yang Hao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhiyi Song
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jin Zhang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Chaozeng Si
- Department of Information Management, China-Japan Friendship Hospital, Beijing, China
| | - Chaoyang Liang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Deruo Liu
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
4
|
Horvat L, Antica M, Matulić M. The Effect of Casein Kinase 2 Inhibition on three Leukemic Cell Lines. CURRENT DRUG THERAPY 2020. [DOI: 10.2174/1574885514666190724111509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background::
Casein Kinase 2 (CK2) is a Ser/Thr protein kinase that coregulates a great
number of signalling pathways in the cell. It is involved in cell cycle regulation and cell proliferation,
apoptosis, DNA damage response and gene transcription. Its substrates are numerous kinases
and transcription factors. It was found to be upregulated in different tumours, and certain types of
leukaemia are very sensitive to its inhibition.
Objective::
We analysed the effects of casein kinase 2 inhibition on three leukaemia cell lines of B
and T cell origin: Jurkat, a T cell line, CLL, a chronic B lymphocytic leukaemia cell line and 697, a
pre-B acute lymphocytic leukaemia cell line. Besides cell proliferation and cytotoxicity analysis, the
aim was to investigate the influence of CK2 inhibition on elements of the Notch signalling pathway.
Notch signalling has an important role in blood cell differentiation, and CK2 regulates Ikaros, a
tumour suppressor interfering with Notch signalling
Methods::
and T leukaemia cells were treated with different concentrations of the CK2 inhibitor,
CX-4945, for 6 days, and cell viability and proliferation were determined by Trypan Blue Exclusion
Method. Analysis of gene expression was performed by RT-qPCR.
Results::
All three cell lines were sensitive to CK2 inhibition and among them, 697 cells had two
times lower IC50. In Jurkat and CLL cells changes in c-Myc and Notch pathway gene expression
were found.
Conclusion::
As CK2 is involved in numerous signalling circuits, we concluded that each cell type
could have a cell-specific response in gene expression.
Collapse
Affiliation(s)
- Luka Horvat
- Department of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102A, 10000 Zagreb, Croatia
| | - Mariastefania Antica
- Division of Molecular Biology, Rudjer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - Maja Matulić
- Department of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102A, 10000 Zagreb, Croatia
| |
Collapse
|
5
|
Richard SA, Jia-Hao Z. Elucidating the pathogenic and biomarker potentials of FOXG1 in glioblastoma. Oncol Rev 2020; 14:444. [PMID: 32395201 PMCID: PMC7204822 DOI: 10.4081/oncol.2020.444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 03/20/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GB) is an extremely pugnacious brain cancer originating from neural stem (NS) cell-like cells. Forkhead box G1 (FOXG1; previously recognized as BF-1, qin, Chicken Brain Factor 1, or XBF-1 and renamed FOXG1 for mouse and human, and FoxG1 for other chordates) is an evolutionary preserved transcription factor driven from the forkhead box group of proteins FOXG1 modulates the speed of neurogenesis by maintaining progenitor cells in a proliferative mode as well as obstructing their differentiation into neurons during the initial periods of cortical formation. FOXG1 has been implicated in the formation of central nervous system (CNS) tumors and precisely GBs. Pathophysiologically, joint actions of FOXG1 and phosphatidylinositol- 3-kinases (PI3K) intermediate in intrinsic resistance of human GB cells to transforming growth factor-beta (TGF-β) stimulation of cyclin-dependent kinase inhibitor 1(p21Cip1) as well as growth inhibition. FOXG1 and NOTCH signaling pathways may functionally interrelate at different stages to facilitate gliomagenesis. Furthermore, FoxG1 actively contributed to the formation of transcription suppression complexes with corepressors of the Groucho/transducin-like Enhancer of split (Gro/TLEs). Also, FOXG1 was stimulated by Gro/TLE1 and abridged by Grg6. FOXG1 silencing in brain tumor-initiating cells (BTICs) also resulted in diminished secretion of markers characteristic undifferentiated natural neural stem/progenitor cells (NSPC) states, such as Oligodendrocyte transcription factor (OLIG2), (sex determining region Y)-box 2. (SOX2) and B lymphoma Mo-MLV insertion region 1 homolog (BMI1). This review therefore focuses on the pathogenic and biomarker potentials of FOXG1 in GB.
Collapse
Affiliation(s)
- Seidu A Richard
- Department of Neurosurgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, P.R. China.,Department of Medicine, Princefield University, Ho-Volta Region, Ghana, West Africa
| | - Zhou Jia-Hao
- Department of Neurosurgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, P.R. China
| |
Collapse
|
6
|
Chanoumidou K, Hadjimichael C, Athanasouli P, Ahlenius H, Klonizakis A, Nikolaou C, Drakos E, Kostouros A, Stratidaki I, Grigoriou M, Kretsovali A. Groucho related gene 5 (GRG5) is involved in embryonic and neural stem cell state decisions. Sci Rep 2018; 8:13790. [PMID: 30214018 PMCID: PMC6137157 DOI: 10.1038/s41598-018-31696-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 08/20/2018] [Indexed: 12/16/2022] Open
Abstract
Groucho related gene 5 (GRG5) is a multifunctional protein that has been implicated in late embryonic and postnatal mouse development. Here, we describe a previously unknown role of GRG5 in early developmental stages by analyzing its function in stem cell fate decisions. By both loss and gain of function approaches we demonstrate that ablation of GRG5 deregulates the Embryonic Stem Cell (ESC) pluripotent state whereas its overexpression leads to enhanced self-renewal and acquisition of cancer cell-like properties. The malignant characteristics of teratomas generated by ESCs that overexpress GRG5 reveal its pro-oncogenic potential. Furthermore, transcriptomic analysis and cell differentiation approaches underline GRG5 as a multifaceted signaling regulator that represses mesendodermal-related genes. When ESCs exit pluripotency, GRG5 promotes neuroectodermal specification via Wnt and BMP signaling suppression. Moreover, GRG5 promotes the neuronal reprogramming of fibroblasts and maintains the self-renewal of Neural Stem Cells (NSCs) by sustaining the activity of Notch/Hes and Stat3 signaling pathways. In summary, our results demonstrate that GRG5 has pleiotropic roles in stem cell biology functioning as a stemness factor and a neural fate specifier.
Collapse
Affiliation(s)
- Konstantina Chanoumidou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100, Alexandroupoli, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), 70013, Heraklion, Crete, Greece.,Lund Stem Cell Center, University Hospital, SE-221 84, Lund, Sweden
| | - Christiana Hadjimichael
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), 70013, Heraklion, Crete, Greece
| | - Paraskevi Athanasouli
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), 70013, Heraklion, Crete, Greece.,Department of Biology, University of Crete, 71409, Heraklion, Crete, Greece
| | - Henrik Ahlenius
- Lund Stem Cell Center, University Hospital, SE-221 84, Lund, Sweden
| | - Antonis Klonizakis
- Department of Biology, University of Crete, 71409, Heraklion, Crete, Greece
| | | | - Elias Drakos
- School of Medicine, University of Crete, 71003, Heraklion, Crete, Greece
| | - Antonis Kostouros
- School of Medicine, University of Crete, 71003, Heraklion, Crete, Greece
| | - Irene Stratidaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), 70013, Heraklion, Crete, Greece
| | - Maria Grigoriou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100, Alexandroupoli, Greece
| | - Androniki Kretsovali
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), 70013, Heraklion, Crete, Greece.
| |
Collapse
|
7
|
Dali R, Verginelli F, Pramatarova A, Sladek R, Stifani S. Characterization of a FOXG1:TLE1 transcriptional network in glioblastoma-initiating cells. Mol Oncol 2018; 12:775-787. [PMID: 29316219 PMCID: PMC5983107 DOI: 10.1002/1878-0261.12168] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 12/12/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common and deadly malignant brain cancer of glial cell origin, with a median patient survival of less than 20 months. Transcription factors FOXG1 and TLE1 promote GBM propagation by supporting maintenance of brain tumour-initiating cells (BTICs) with stem-like properties. Here, we characterize FOXG1 and TLE1 target genes in GBM patient-derived BTICs using ChIP-Seq and RNA-Seq approaches. These studies identify 150 direct FOXG1 targets, several of which are also TLE1 targets, involved in cell proliferation, differentiation, survival, chemotaxis and angiogenesis. Negative regulators of NOTCH signalling, including CHAC1, are among the transcriptional repression targets of FOXG1:TLE1 complexes, suggesting a crosstalk between FOXG1:TLE1 and NOTCH-mediated pathways in GBM. These results provide previously unavailable insight into the transcriptional programs underlying the tumour-promoting functions of FOXG1:TLE1 in GBM.
Collapse
Affiliation(s)
- Rola Dali
- Department of Neurology and NeurosurgeryMontreal Neurological InstituteMcGill UniversityMontrealCanada
- McGill Center for BioinformaticsMcGill UniversityMontrealCanada
| | - Federica Verginelli
- Department of Neurology and NeurosurgeryMontreal Neurological InstituteMcGill UniversityMontrealCanada
- Present address:
Laboratory of Cancer Stem Cell ResearchCandiolo Cancer InstituteFPO‐IRCCSCandioloItaly
| | - Albena Pramatarova
- Departments of Human Genetics and MedicineMcGill UniversityMontrealCanada
| | - Robert Sladek
- Departments of Human Genetics and MedicineMcGill UniversityMontrealCanada
| | - Stefano Stifani
- Department of Neurology and NeurosurgeryMontreal Neurological InstituteMcGill UniversityMontrealCanada
| |
Collapse
|
8
|
Sengupta D, Kar S. Alteration in MicroRNA Expression Governs the Nature and Timing of Cellular Fate Commitment. ACS Chem Neurosci 2018; 9:725-737. [PMID: 29181975 DOI: 10.1021/acschemneuro.7b00423] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In the central nervous system, the expression level of transcriptional repressor Hes1 (hairy and enhancer of split-1) tightly controls the alternative cell fate commitment during differentiation as well as the time required for such cellular transitions. A microRNA, miR-9, that interacts with Hes1 in a mutually antagonistic manner, influences both the process of lineage specification and timing of differentiation significantly, but the impact of the miR-9 in guiding these events still remains poorly understood. Here, we proposed a stochastic mathematical model of the miR-9/Hes1 double-negative feedback interaction network that at the outset shows how alternative cell fate such as quiescence, progenitor, and neuronal states can be accomplished through fine-tuning the Hes1 dynamics by altering the expression level of miR-9. The model simulations further foretell a correlated variation of the period of oscillation of Hes1, and the time delay observed between Hes1 mRNA and protein as the transcription rate of miR-9 increases during the neural progenitor state attainment. Importantly, the model simulations aided by the systematic sensitivity analysis predict that the timing of differentiation to the neuronal state crucially depends on the negative regulators (miR-9 and Hes6) of the Hes1. Our results indicate that miR-9/Hes1 interaction network can be effectively exploited for an efficient and well-timed neuronal transformation.
Collapse
Affiliation(s)
- Dola Sengupta
- Department of Chemistry, IIT Bombay, Powai, Mumbai 400076, India
| | - Sandip Kar
- Department of Chemistry, IIT Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
9
|
Yao X, Pham T, Temple B, Gray S, Cannon C, Hardy C, Fletcher K, Ireland SK, Hossain A, Chen R, Abdel-Mageed AB, Biliran H. TLE1 inhibits anoikis and promotes tumorigenicity in human lung cancer cells through ZEB1-mediated E-cadherin repression. Oncotarget 2017; 8:72235-72249. [PMID: 29069783 PMCID: PMC5641126 DOI: 10.18632/oncotarget.19703] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/26/2017] [Indexed: 01/06/2023] Open
Abstract
The Transducin-like enhancer of split 1 (TLE1) corepressor protein is overexpressed in human lung tumors and is a putative lung-specific oncogene. However, the molecular mechanism underlying its oncogenic function remains to be delineated. Here, we report an important role of TLE1 in promoting lung tumorigenesis by a mechanism involving induction of anoikis resistance. Using the human lung adenocarcinoma A549 and immortalized bronchial epithelial BEAS-2B cell lines, we observed that TLE1 inhibits anoikis through transcriptional repression of E-cadherin gene. In support of E-cadherin as a downstream target of TLE1 to block anoikis, forced expression of E-cadherin attenuated TLE1-induced anoikis resistance while E-cadherin downregulation decreased the anoikis sensitivity of TLE1 knockdown cells. Furthermore, we determined that E-cadherin expression is transcriptionally induced upon loss of cell attachment and functions as an effector of anoikis. Loss of E-cadherin via the siRNA strategy or exogenous TLE1 expression was sufficient to attenuate anoikis in A549 and BEAS-2B cells. Importantly, we demonstrated that the ZEB1 transcriptional factor is required for TLE1-mediated E-cadherin repression and anoikis resistance. ZEB1 interacted with and recruited the TLE1 to the E-cadherin promoter to impose histone deacetylation and gene silencing. In vivo, TLE1 strongly promoted tumorigenicity of A549 cells in a ZEB1-dependent manner. Underscoring its role in anoikis insensitivity of lung cancer cells, the TLE1-mediated E-cadherin repression was negatively regulated by the tumor suppressor Bcl-2 inhibitor of transcription 1 (Bit1) to effect anoikis. These findings identify the ZEB1/TLE1/E-cadherin transcriptional mechanism as a novel pathway that promotes anoikis resistance and oncogenicity of lung cancer cells.
Collapse
Affiliation(s)
- Xin Yao
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Tri Pham
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Brandi Temple
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Selena Gray
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Cornita Cannon
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Camry Hardy
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Kamari Fletcher
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Shubha Kale Ireland
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Ahamed Hossain
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Renwei Chen
- Center for Bioengineering, University of California, Santa Barbara, CA 93106, USA
| | - Asim B Abdel-Mageed
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hector Biliran
- Department of Biological and Public Health Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA
| |
Collapse
|
10
|
Epigenetic modification of TLE1 induce abnormal differentiation in diabetic mice intestinal epithelium. Mol Cell Biochem 2017; 438:85-96. [PMID: 28744818 DOI: 10.1007/s11010-017-3116-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/15/2017] [Indexed: 12/20/2022]
Abstract
The intestinal epithelium cells (IECs) in diabetes mellitus (DM) patients have been proven to be abnormally differentiated. During the differentiation of IECs, epigenetic modification acts as an important regulator. In this study, we aimed to examine the epigenetic alteration of Transducin-like Enhancer of Split 1 (TLE1), a multitask transcriptional co-repressor, contributing to the differentiation homeostasis in IECs of DM mice. The IECs of type 2 diabetic mice model were isolated and collected. Methylation states of whole genomic DNA promoter regions were investigated by microarray. Methylated-specific PCR was used to detect the methylation state of TLE1 promoter in DM mice IECs. The expression of TLE1, Hes1, and differentiated cell markers were measured through real-time PCR, Western blots, and immunohistochemistry; by transfection assay, TLE1 or Hes1 was independently down-regulated in intestinal epithelium cell line, IEC-6. Subsequent modulation on TLE1, Hes1, and differentiated intestinal cell markers were detected. Global gene promoter regions in DM intestinal epithelium were less methylated comparing to normal control. The expression of TLE1 was significantly increased via hypomethylated activation in DM mice IECs. Hes1 was significantly suppressed and the terminal cell markers abnormally expressed in DM mice IECs (P < 0.05). Inhibition or induction on the abundance of TLE1 in IEC-6 cell line resulted in the corresponding dysregulation of Hes1 and intestinal epithelium differentiation (P < 0.05). Demethylation of TLE1 promoter region activates the self-expression in diabetic mice IECs. Subsequently, TLE1, through the transcriptional suppression on expression of Hes1, contributes to the aberrant differentiation of IECs in DM mice.
Collapse
|
11
|
Götz C, Montenarh M. Protein kinase CK2 in development and differentiation. Biomed Rep 2016; 6:127-133. [PMID: 28357063 DOI: 10.3892/br.2016.829] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/06/2016] [Indexed: 12/15/2022] Open
Abstract
Among the human kinomes, protein kinase CK2 (formerly termed casein kinase II) is considered to be essential, as it is implicated in the regulation of various cellular processes. Experiments with pharmacological inhibitors of the kinase activity of CK2 provide evidence that CK2 is essential for development and differentiation. Therefore, the present review addresses the role of CK2 during embryogenesis, neuronal, adipogenic, osteogenic and myogenic differentiation in established model cell lines, and in embryonic, neural and mesenchymal stem cells. CK2 kinase activity appears to be essential in the early stages of differentiation, as CK2 inhibition at early time points generally prevents differentiation. In addition, the present review reports on target proteins of CK2 in embryogenesis and differentiation.
Collapse
Affiliation(s)
- Claudia Götz
- Department of Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Germany
| | - Mathias Montenarh
- Department of Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Germany
| |
Collapse
|
12
|
Sengupta D, Kar S. Unraveling the differential dynamics of developmental fate in central and peripheral nervous systems. Sci Rep 2016; 6:36397. [PMID: 27805068 PMCID: PMC5090986 DOI: 10.1038/srep36397] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/14/2016] [Indexed: 12/01/2022] Open
Abstract
Bone morphogenetic protein 2 (BMP2), differentially regulates the developmental lineage commitment of neural stem cells (NSC’s) in central and peripheral nervous systems. However, the precise mechanism beneath such observations still remains illusive. To decipher the intricacies of this mechanism, we propose a generic mathematical model of BMP2 driven differentiation regulation of NSC’s. The model efficiently captures the dynamics of the wild-type as well as various mutant and over-expression phenotypes for NSC’s in central nervous system. Our model predicts that the differential developmental dynamics of the NSC’s in peripheral nervous system can be reconciled by altering the relative positions of the two mutually interconnected bi-unstable switches inherently present in the steady state dynamics of the crucial developmental fate regulatory proteins as a function of BMP2 dose. This model thus provides a novel mechanistic insight and has the potential to deliver exciting therapeutic strategies for neuronal regeneration from NSC’s of different origin.
Collapse
Affiliation(s)
- Dola Sengupta
- Department of Chemistry, IIT Bombay, Powai, Mumbai - 400076, India
| | - Sandip Kar
- Department of Chemistry, IIT Bombay, Powai, Mumbai - 400076, India
| |
Collapse
|
13
|
Wang WS, Zhu J, Zhang KX, Lü YT, Xu HH. A mutation of casein kinase 2 α4 subunit affects multiple developmental processes in Arabidopsis. PLANT CELL REPORTS 2016; 35:1071-1080. [PMID: 26883224 DOI: 10.1007/s00299-016-1939-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/20/2016] [Indexed: 06/05/2023]
Abstract
Arabidopsis CK2 α4 subunit regulates the primary root and hypocotyl elongation, lateral root formation, cotyledon expansion, rosette leaf initiation and growth, flowering, and anthocyanin biosynthesis. Casein kinase 2 (CK2) is a conserved tetrameric kinase composed of two α and two β subunits. The inhibition of CK2 activity usually results in severe developmental deficiency. Four genes (CKA1-CKA4) encode CK2 α subunit in Arabidopsis. Single mutations of CKA1, CKA2, and CKA3 do not affect the normal growth of Arabidopsis, while the cka1 cka2 cka3 triple mutants are defective in cotyledon and hypocotyl growth, lateral root development, and flowering. The inhibition of CKA4 expression in cka1 cka2 cka3 background further reduces the number of lateral roots and delays the flowering time. Here, we report the characterization of a novel knockout mutant of CKA4, which exhibits various developmental defects including reduced primary root and hypocotyl elongation, increased lateral root density, delayed cotyledon expansion, retarded rosette leaf initiation and growth, and late flowering. The examination of the cellular basis for abnormal root development of this mutant revealed reduced root meristem cells with enhanced RETINOBLASTOMA-RELATED (RBR) expression that promotes cell differentiation in root meristem. Moreover, this cka4-2 mutant accumulates higher anthocyanin in the aerial part and shows an increased expression of anthocyanin biosynthetic genes, suggesting a novel role of CK2 in modulating anthocyanin biosynthesis. In addition, the complementation test using primary root elongation assay as a sample confirms that the changed phenotypes of this cka4-2 mutant are due to the lack of CKA4. Taken together, this study reveals an essential role of CK2 α4 subunit in multiple developmental processes in Arabidopsis.
Collapse
Affiliation(s)
- Wen-Shu Wang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Huaihai Institute of Technology, Lianyungang, 222005, China
| | - Jiang Zhu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Huaihai Institute of Technology, Lianyungang, 222005, China
| | - Kun-Xiao Zhang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Huaihai Institute of Technology, Lianyungang, 222005, China
| | - Ying-Tang Lü
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Huaihai Institute of Technology, Lianyungang, 222005, China
| | - Heng-Hao Xu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Huaihai Institute of Technology, Lianyungang, 222005, China.
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Lianyungang, 222005, China.
| |
Collapse
|
14
|
Kwong PN, Chambers M, Vashisht AA, Turki-Judeh W, Yau TY, Wohlschlegel JA, Courey AJ. The Central Region of the Drosophila Co-repressor Groucho as a Regulatory Hub. J Biol Chem 2015; 290:30119-30. [PMID: 26483546 DOI: 10.1074/jbc.m115.681171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Indexed: 12/23/2022] Open
Abstract
Groucho (Gro) is a Drosophila co-repressor that regulates the expression of a large number of genes, many of which are involved in developmental control. Previous studies have shown that its central region is essential for function even though its three domains are poorly conserved and intrinsically disordered. Using these disordered domains as affinity reagents, we have now identified multiple embryonic Gro-interacting proteins. The interactors include protein complexes involved in chromosome organization, mRNA processing, and signaling. Further investigation of the interacting proteins using a reporter assay showed that many of them modulate Gro-mediated repression either positively or negatively. The positive regulators include components of the spliceosomal subcomplex U1 small nuclear ribonucleoprotein (U1 snRNP). A co-immunoprecipitation experiment confirms this finding and suggests that a sizable fraction of nuclear U1 snRNP is associated with Gro. The use of RNA-seq to analyze the gene expression profile of cells subjected to knockdown of Gro or snRNP-U1-C (a component of U1 snRNP) showed a significant overlap between genes regulated by these two factors. Furthermore, comparison of our RNA-seq data with Gro and RNA polymerase II ChIP data led to a number of insights, including the finding that Gro-repressed genes are enriched for promoter-proximal RNA polymerase II. We conclude that the Gro central domains mediate multiple interactions required for repression, thus functioning as a regulatory hub. Furthermore, interactions with the spliceosome may contribute to repression by Gro.
Collapse
Affiliation(s)
- Pak N Kwong
- From the Departments of Chemistry and Biochemistry and
| | | | | | - Wiam Turki-Judeh
- From the Departments of Chemistry and Biochemistry and Molecular Biology Institute, UCLA, Los Angeles, California 90095
| | - Tak Yu Yau
- From the Departments of Chemistry and Biochemistry and
| | - James A Wohlschlegel
- Biological Chemistry and Molecular Biology Institute, UCLA, Los Angeles, California 90095
| | - Albert J Courey
- From the Departments of Chemistry and Biochemistry and Molecular Biology Institute, UCLA, Los Angeles, California 90095
| |
Collapse
|
15
|
Itatani Y, Sonoshita M, Kakizaki F, Okawa K, Stifani S, Itoh H, Sakai Y, Taketo MM. Characterization of Aes nuclear foci in colorectal cancer cells. J Biochem 2015; 159:133-40. [PMID: 26229111 DOI: 10.1093/jb/mvv077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/20/2015] [Indexed: 11/14/2022] Open
Abstract
Amino-terminal enhancer of split (Aes) is a member of Groucho/Transducin-like enhancer (TLE) family. Aes is a recently found metastasis suppressor of colorectal cancer (CRC) that inhibits Notch signalling, and forms nuclear foci together with TLE1. Although some Notch-associated proteins are known to form subnuclear bodies, little is known regarding the dynamics or functions of these structures. Here, we show that Aes nuclear foci in CRC observed under an electron microscope are in a rather amorphous structure, lacking surrounding membrane. Investigation of their behaviour during the cell cycle by time-lapse cinematography showed that Aes nuclear foci dissolve during mitosis and reassemble after completion of cytokinesis. We have also found that heat shock cognate 70 (HSC70) is an essential component of Aes foci. Pharmacological inhibition of the HSC70 ATPase activity with VER155008 reduces Aes focus formation. These results provide insight into the understanding of Aes-mediated inhibition of Notch signalling.
Collapse
Affiliation(s)
- Yoshiro Itatani
- Department of Pharmacology and Department of Surgery, Graduate School of Medicine, Kyoto University, Yoshida Konoé-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | - Katsuya Okawa
- Drug Discovery Research Laboratories, Kyowa Hakko Kirin Co., Ltd, 1188 Shimotogari Nagaizumi-cho, Sunto-gun, Shizuoka 411-8731, Japan
| | - Stefano Stifani
- Montreal Neurological Institute, McGill University, 3801 rue University, Montreal, Quebec H3A 2B4, Canada; and
| | - Hideaki Itoh
- Department of Life Science, Faculty of Engineering and Resource Science, Akita University, 1-1 Tegata Gakuen Town, Akita, 010-0852 Akita, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Yoshida Konoé-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - M Mark Taketo
- Department of Pharmacology and Department of Surgery, Graduate School of Medicine, Kyoto University, Yoshida Konoé-cho, Sakyo-ku, Kyoto 606-8501, Japan;
| |
Collapse
|
16
|
Agarwal M, Kumar P, Mathew SJ. The Groucho/Transducin-like enhancer of split protein family in animal development. IUBMB Life 2015; 67:472-81. [PMID: 26172616 DOI: 10.1002/iub.1395] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 01/30/2023]
Abstract
Corepressors are proteins that cannot bind DNA directly but repress transcription by interacting with partner proteins. The Groucho/Transducin-Like Enhancer of Split (TLE) are a conserved family of corepressor proteins present in animals ranging from invertebrates such as Drosophila to vertebrates such as mice and humans. Groucho/TLE proteins perform important functions throughout the life span of animals, interacting with several pathways and regulating fundamental processes such as metabolism. However, these proteins have especially crucial functions in animal development, where they are required in multiple tissues in a temporally regulated manner. In this review, we summarize the functions of the Groucho/TLE proteins during animal development, emphasizing on specific tissues where they play essential roles.
Collapse
Affiliation(s)
- Megha Agarwal
- Regional Centre for Biotechnology, NCR Bio-Science Cluster, Faridabad, Haryana, India
| | - Pankaj Kumar
- Regional Centre for Biotechnology, NCR Bio-Science Cluster, Faridabad, Haryana, India
| | - Sam J Mathew
- Regional Centre for Biotechnology, NCR Bio-Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
17
|
Transcription factors FOXG1 and Groucho/TLE promote glioblastoma growth. Nat Commun 2014; 4:2956. [PMID: 24356439 PMCID: PMC3984242 DOI: 10.1038/ncomms3956] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/18/2013] [Indexed: 01/24/2023] Open
Abstract
Glioblastoma (GBM) is the most common and deadly malignant brain cancer, with a median survival of less than two years. GBM displays a cellular complexity that includes brain tumour-initiating cells (BTICs), which are considered as potential key targets for GBM therapies. Here we show that the transcription factors FOXG1 and Groucho/TLE are expressed in poorly differentiated astroglial cells in human GBM specimens and in primary cultures of GBM-derived BTICs, where they form a complex. FOXG1 knockdown in BTICs causes downregulation of neural stem/progenitor and proliferation markers, increased replicative senescence, upregulation of astroglial differentiation genes, and decreased BTIC-initiated tumour growth upon intracranial transplantation into host mice. These effects are phenocopied by Groucho/TLE knockdown or dominant-inhibition of the FOXG1:Groucho/TLE complex. These results provide evidence that transcriptional programs regulated by FOXG1 and Groucho/TLE are important for BTIC-initiated brain tumour growth, implicating FOXG1 and Groucho/TLE in GBM tumorigenesis.
Collapse
|
18
|
Wu J, Bowe DB, Sadlonova A, Whisenhunt TR, Hu Y, Rustgi AK, Nie Y, Paterson AJ, Yang X. O-GlcNAc transferase is critical for transducin-like enhancer of split (TLE)-mediated repression of canonical Wnt signaling. J Biol Chem 2014; 289:12168-12176. [PMID: 24616106 DOI: 10.1074/jbc.m114.553859] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Drosophila Groucho protein and its mammalian orthologues the transducin-like enhancers of split (TLEs) are critical transcriptional corepressors that repress Wnt and other signaling pathways. Although it is known that Groucho/TLEs are recruited to target genes by pathway-specific transcription factors, molecular events after the corepressor recruitment are largely unclear. We report that association of TLEs with O-GlcNAc transferase, an enzyme that catalyzes posttranslational modification of proteins by O-linked N-acetylglucosamine, is essential for TLE-mediated transcriptional repression. Removal of O-GlcNAc from Wnt-responsive gene promoters is critical for gene activation from Wnt-responsive promoters. Thus, these studies identify a molecular mechanism by which Groucho/TLEs repress gene transcription and provide a model whereby O-GlcNAc may control distinct intracellular signaling pathways.
Collapse
Affiliation(s)
- Jing Wu
- Center for Mitochondrial Biology and Medicine, the Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China; Section of Comparative Medicine and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06519
| | - Damon B Bowe
- Division of Endocrinology and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Andrea Sadlonova
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia 30322
| | - Thomas R Whisenhunt
- Division of Endocrinology and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Yong Hu
- Division of Endocrinology and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Anil K Rustgi
- Department of Medicine (GI) and Genetics, Abramson Cancer Center, 600 CRB, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Andrew J Paterson
- Division of Endocrinology and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Xiaoyong Yang
- Section of Comparative Medicine and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06519.
| |
Collapse
|
19
|
Murthy M, Bocking S, Verginelli F, Stifani S. Transcription factor Runx1 inhibits proliferation and promotes developmental maturation in a selected population of inner olfactory nerve layer olfactory ensheathing cells. Gene 2014; 540:191-200. [PMID: 24582971 DOI: 10.1016/j.gene.2014.02.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 01/30/2014] [Accepted: 02/21/2014] [Indexed: 11/18/2022]
Abstract
The olfactory system undergoes persistent regeneration throughout life. Olfactory ensheathing cells (OECs) are a specialized class of glia found exclusively in the olfactory system. OECs wrap olfactory sensory neuron axons and support their growth from the olfactory epithelium, and targeting to the olfactory bulb, during development and life-long regeneration. Because of this function and their ability to cross the boundary between central and peripheral nervous systems, OECs are attractive candidates for cell-based regenerative therapies to promote axonal repair in the injured nervous system. OECs are a molecularly, topologically and functionally heterogeneous group of cells and the mechanisms underlying the development and function of specific OEC subpopulations are poorly defined. This situation has affected the outcome and interpretation of OEC-based regenerative strategies. Here we show that the transcription factor Runx1 is selectively expressed in OECs of the inner olfactory nerve layer of the mouse olfactory bulb and in their precursors in the OEC migratory mass. Furthermore, we provide evidence that in vivo knockdown of mouse Runx1 increases the proliferation of the OECs in which Runx1 is expressed. Conversely, Runx1 overexpression in primary cultures of OECs reduces cell proliferation in vitro. Decreased Runx1 activity also leads to an increase in Runx1-expressing OEC precursors, with a parallel decrease in the number of more developmentally mature OECs. These results identify Runx1 as a useful new marker of a distinct OEC subpopulation and suggest that Runx1 is important for the development of this group of OECs. These observations provide an avenue for further exploration into the molecular mechanisms underlying the development and function of specific OEC subpopulations.
Collapse
Affiliation(s)
- Manjari Murthy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Sarah Bocking
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Federica Verginelli
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
20
|
Ciarapica R, Methot L, Tang Y, Lo R, Dali R, Buscarlet M, Locatelli F, del Sal G, Rota R, Stifani S. Prolyl isomerase Pin1 and protein kinase HIPK2 cooperate to promote cortical neurogenesis by suppressing Groucho/TLE:Hes1-mediated inhibition of neuronal differentiation. Cell Death Differ 2013; 21:321-32. [PMID: 24270405 DOI: 10.1038/cdd.2013.160] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 09/10/2013] [Accepted: 10/09/2013] [Indexed: 01/15/2023] Open
Abstract
The Groucho/transducin-like Enhancer of split 1 (Gro/TLE1):Hes1 transcriptional repression complex acts in cerebral cortical neural progenitor cells to inhibit neuronal differentiation. The molecular mechanisms that regulate the anti-neurogenic function of the Gro/TLE1:Hes1 complex during cortical neurogenesis remain to be defined. Here we show that prolyl isomerase Pin1 (peptidyl-prolyl cis-trans isomerase NIMA-interacting 1) and homeodomain-interacting protein kinase 2 (HIPK2) are expressed in cortical neural progenitor cells and form a complex that interacts with the Gro/TLE1:Hes1 complex. This association depends on the enzymatic activities of both HIPK2 and Pin1, as well as on the association of Gro/TLE1 with Hes1, but is independent of the previously described Hes1-activated phosphorylation of Gro/TLE1. Interaction with the Pin1:HIPK2 complex results in Gro/TLE1 hyperphosphorylation and weakens both the transcriptional repression activity and the anti-neurogenic function of the Gro/TLE1:Hes1 complex. These results provide evidence that HIPK2 and Pin1 work together to promote cortical neurogenesis, at least in part, by suppressing Gro/TLE1:Hes1-mediated inhibition of neuronal differentiation.
Collapse
Affiliation(s)
- R Ciarapica
- 1] Montreal Neurological Institute, McGill University, 3801 rue University, Montreal, Quebec H3A2B4, Canada [2] Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - L Methot
- Montreal Neurological Institute, McGill University, 3801 rue University, Montreal, Quebec H3A2B4, Canada
| | - Y Tang
- Montreal Neurological Institute, McGill University, 3801 rue University, Montreal, Quebec H3A2B4, Canada
| | - R Lo
- Montreal Neurological Institute, McGill University, 3801 rue University, Montreal, Quebec H3A2B4, Canada
| | - R Dali
- Montreal Neurological Institute, McGill University, 3801 rue University, Montreal, Quebec H3A2B4, Canada
| | - M Buscarlet
- Montreal Neurological Institute, McGill University, 3801 rue University, Montreal, Quebec H3A2B4, Canada
| | - F Locatelli
- 1] Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy [2] University of Pavia, Pavia, Italy
| | - G del Sal
- 1] Laboratorio Nazionale CIB, Area Science Park, Trieste, Italy [2] Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| | - R Rota
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - S Stifani
- Montreal Neurological Institute, McGill University, 3801 rue University, Montreal, Quebec H3A2B4, Canada
| |
Collapse
|
21
|
Interaction and antagonistic roles of NF-κB and Hes6 in the regulation of cortical neurogenesis. Mol Cell Biol 2013; 33:2797-808. [PMID: 23689134 DOI: 10.1128/mcb.01610-12] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The involvement of nuclear factor kappa B (NF-κB) in several processes in the postnatal and adult brain, ranging from neuronal survival to synaptogenesis and plasticity, has been documented. In contrast, little is known about the functions of NF-κB during embryonic brain development. It is shown here that NF-κB is selectively activated in neocortical neural progenitor cells in the developing mouse telencephalon. Blockade of NF-κB activity leads to premature cortical neuronal differentiation and depletion of the progenitor cell pool. Conversely, NF-κB activation causes decreased cortical neurogenesis and expansion of the progenitor cell compartment. These effects are antagonized by the proneuronal transcription factor Hes6, which physically and functionally interacts with RelA-containing NF-κB complexes in cortical progenitor cells. In turn, NF-κB exerts an inhibitory effect on the ability of Hes6 to promote cortical neuronal differentiation. These results reveal previously uncharacterized functions and modes of regulation for NF-κB and Hes6 during cortical neurogenesis.
Collapse
|
22
|
Dastidar SG, Narayanan S, Stifani S, D'Mello SR. Transducin-like enhancer of Split-1 (TLE1) combines with Forkhead box protein G1 (FoxG1) to promote neuronal survival. J Biol Chem 2012; 287:14749-59. [PMID: 22354967 DOI: 10.1074/jbc.m111.328336] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Transducin-like enhancer of split-1 (TLE1) plays a critical role in the regulation of neurogenesis by inhibiting the differentiation of neural progenitor cells into neurons. Although TLE1 is also expressed highly in the postnatal brain and through adulthood, its role in postmitotic neurons is not clear. Using cultures of cerebellar granule neurons, we show that expression of TLE1 is reduced in neurons primed to die. Reestablishment of elevated TLE1 levels by ectopic expression protects neurons from death, whereas suppression of TLE1 expression in otherwise healthy neurons induces cell death. These results show that TLE1 is necessary for the maintenance of neuronal survival. Experiments using pharmacological inhibitors as well as expression of point mutants indicate that phosphorylation of TLE1 by casein kinase-2 (CK2) at Ser-239 and Ser-253 is necessary for its survival-promoting activity. TLE1-mediated survival is also inhibited by pharmacological inhibition of PI3K-Akt signaling but not by inhibitors of Raf-MEK-ERK signaling or other molecules, including histone deacetylases, calcium calmodulin kinase, or CK1. The survival-promoting activity of TLE1 depends critically on interaction with FoxG1, another protein involved in the regulation of neurogenesis and shown previously to promote survival of postmitotic neurons. Likewise, the ability of FoxG1 to promote neuronal survival depends on TLE1. Taken together, our study demonstrates that TLE1 cooperates with FoxG1 to promote neuronal survival in a CK2- and PI3K-Akt-dependent manner.
Collapse
|
23
|
Establishment of motor neuron-V3 interneuron progenitor domain boundary in ventral spinal cord requires Groucho-mediated transcriptional corepression. PLoS One 2012; 7:e31176. [PMID: 22363571 PMCID: PMC3281934 DOI: 10.1371/journal.pone.0031176] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 01/03/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Dorsoventral patterning of the developing spinal cord is important for the correct generation of spinal neuronal types. This process relies in part on cross-repressive interactions between specific transcription factors whose expression is regulated by Sonic hedgehog. Groucho/transducin-like Enhancer of split (TLE) proteins are transcriptional corepressors suggested to be recruited by at least certain Sonic hedgehog-controlled transcription factors to mediate the formation of spatially distinct progenitor domains within the ventral spinal cord. The aim of this study was to characterize the involvement of TLE in mechanisms regulating the establishment of the boundary between the most ventral spinal cord progenitor domains, termed pMN and p3. Because the pMN domain gives rise to somatic motor neurons while the p3 domain generates V3 interneurons, we also examined the involvement of TLE in the acquisition of these neuronal fates. METHODOLOGY AND PRINCIPAL FINDINGS A combination of in vivo loss- and gain-of-function studies in the developing chick spinal cord was performed to characterize the role of TLE in ventral progenitor domain formation. It is shown here that TLE overexpression causes increased numbers of p3 progenitors and promotes the V3 interneuron fate while suppressing the motor neuron fate. Conversely, dominant-inhibition of TLE increases the numbers of pMN progenitors and postmitotic motor neurons. CONCLUSION Based on these results, we propose that TLE is important to promote the formation of the p3 domain and subsequent generation of V3 interneurons.
Collapse
|
24
|
Turki-Judeh W, Courey AJ. The unconserved groucho central region is essential for viability and modulates target gene specificity. PLoS One 2012; 7:e30610. [PMID: 22319573 PMCID: PMC3272004 DOI: 10.1371/journal.pone.0030610] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 12/26/2011] [Indexed: 12/31/2022] Open
Abstract
Groucho (Gro) is a Drosophila corepressor required by numerous DNA-binding repressors, many of which are distributed in gradients and provide positional information during development. Gro contains well-conserved domains at its N- and C-termini, and a poorly conserved central region that includes the GP, CcN, and SP domains. All lethal point mutations in gro map to the conserved regions, leading to speculation that the unconserved central domains are dispensable. However, our sequence analysis suggests that the central domains are disordered leading us to suspect that the lack of lethal mutations in this region reflects a lack of order rather than an absence of essential functions. In support of this conclusion, genomic rescue experiments with Gro deletion variants demonstrate that the GP and CcN domains are required for viability. Misexpression assays using these same deletion variants show that the SP domain prevents unrestrained and promiscuous repression by Gro, while the GP and CcN domains are indispensable for repression. Deletion of the GP domain leads to loss of nuclear import, while deletion of the CcN domain leads to complete loss of repression. Changes in Gro activity levels reset the threshold concentrations at which graded repressors silence target gene expression. We conclude that co-regulators such as Gro are not simply permissive components of the repression machinery, but cooperate with graded DNA-binding factors in setting borders of gene expression. We suspect that disorder in the Gro central domains may provide the flexibility that allows this region to mediate multiple interactions required for repression.
Collapse
Affiliation(s)
- Wiam Turki-Judeh
- Department of Chemistry and Biochemistry and Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Albert J. Courey
- Department of Chemistry and Biochemistry and Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
25
|
Indulekha CL, Divya TS, Divya MS, Sanalkumar R, Rasheed VA, Dhanesh SB, Sebin A, George A, James J. Hes-1 regulates the excitatory fate of neural progenitors through modulation of Tlx3 (HOX11L2) expression. Cell Mol Life Sci 2012; 69:611-27. [PMID: 21744064 PMCID: PMC11114997 DOI: 10.1007/s00018-011-0765-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Revised: 06/22/2011] [Accepted: 06/23/2011] [Indexed: 10/18/2022]
Abstract
Tlx3 (HOX11L2) is regarded as one of the selector genes in excitatory versus inhibitory fate specification of neurons in distinct regions of the nervous system. Expression of Tlx3 in a post-mitotic immature neuron favors a glutamatergic over GABAergic fate. The factors that regulate Tlx3 have immense importance in the fate specification of glutamatergic neurons. Here, we have shown that Notch target gene, Hes-1, negatively regulates Tlx3 expression, resulting in decreased generation of glutamatergic neurons. Down-regulation of Hes-1 removed the inhibition on Tlx3 promoter, thus promoting glutamatergic differentiation. Promoter-protein interaction studies with truncated/mutated Hes-1 protein suggested that the co-repressor recruitment mediated through WRPW domain of Hes-1 has contributed to the repressive effect. Our results clearly demonstrate a new and unique role for canonical Notch signaling through Hes-1, in neurotransmitter/subtype fate specification of neurons in addition to its known functional role in proliferation/maintenance of neural progenitors.
Collapse
Affiliation(s)
- Chandrasekharan Lalitha Indulekha
- Neuro Stem Cell Biology Laboratory, Department of Neurobiology, Rajiv Gandhi Center for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695 014 India
| | - Thulasi Sheela Divya
- Neuro Stem Cell Biology Laboratory, Department of Neurobiology, Rajiv Gandhi Center for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695 014 India
| | - Mundackal Sivaraman Divya
- Neuro Stem Cell Biology Laboratory, Department of Neurobiology, Rajiv Gandhi Center for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695 014 India
| | - Rajendran Sanalkumar
- Neuro Stem Cell Biology Laboratory, Department of Neurobiology, Rajiv Gandhi Center for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695 014 India
| | - Vazhanthodi Abdul Rasheed
- Neuro Stem Cell Biology Laboratory, Department of Neurobiology, Rajiv Gandhi Center for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695 014 India
| | - Sivadasan Bindu Dhanesh
- Neuro Stem Cell Biology Laboratory, Department of Neurobiology, Rajiv Gandhi Center for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695 014 India
| | - Anu Sebin
- Neuro Stem Cell Biology Laboratory, Department of Neurobiology, Rajiv Gandhi Center for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695 014 India
| | - Amitha George
- Neuro Stem Cell Biology Laboratory, Department of Neurobiology, Rajiv Gandhi Center for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695 014 India
| | - Jackson James
- Neuro Stem Cell Biology Laboratory, Department of Neurobiology, Rajiv Gandhi Center for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695 014 India
| |
Collapse
|
26
|
Abstract
Drosophila Groucho (Gro) is the founding member of a family of metazoan corepressors. Gro mediates repression through interactions with a myriad of DNA-binding repressor proteins to direct the silencing of genes involved in many developmental processes, including neurogenesis and patterning of the main body axis, as well as receptor tyrosine kinase/Ras/MAPK, Notch, Wingless (Wg)/Wnt, and Decapentaplegic (Dpp) signaling. Gro mediates repression by multiple molecular mechanisms, depending on the regulatory context. Because Gro is a broadly expressed nuclear factor, whereas its repressor partners display restricted temporal and spatial distribution, it was presumed that this corepressor played permissive rather than instructive roles in development. However, a wide range of studies demonstrates that this is not the case. Gro can sense and integrate many cellular inputs to modulate the expression of variety of genes, making it a versatile corepressor with crucial instructive roles in development and signaling.
Collapse
Affiliation(s)
- Wiam Turki-Judeh
- Department of Chemistry & Biochemistry and Molecular Biology Institute, University of California, Los Angeles, California, USA
| | | |
Collapse
|
27
|
Lee JE, Golz JF. Diverse roles of Groucho/Tup1 co-repressors in plant growth and development. PLANT SIGNALING & BEHAVIOR 2012; 7:86-92. [PMID: 22301974 PMCID: PMC3357377 DOI: 10.4161/psb.7.1.18377] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Transcriptional regulation involves coordinated and often complex interactions between activators and repressors that together dictate the temporal and spatial activity of target genes. While the study of developmental regulation has often focused on positively acting transcription factors, it is becoming increasingly clear that transcriptional repression is a key regulatory mechanism underpinning many developmental processes in both plants and animals. In this review, we focus on the plant Groucho (Gro)/Tup1-like co-repressors and discuss their roles in establishing the apical-basal axis of the developing embryo, maintaining the stem cell population in the shoot apex and determining floral organ identity. As well as being developmental regulators, recent studies have shown that these co-repressors play a central role in regulating auxin and jasmonate signalling pathways and are also linked to the regulation of pectin structure in the seed coat. These latest findings point to the Gro/Tup1-like co-repressors playing a much broad role in plant growth and development than previously thought; an observation that underlines the central importance of transcriptional repression in plant gene regulation.
Collapse
|
28
|
Kamata T, Bong YS, Mood K, Park MJ, Nishanian TG, Lee HS. EphrinB1 interacts with the transcriptional co-repressor Groucho/xTLE4. BMB Rep 2011; 44:199-204. [PMID: 21429299 DOI: 10.5483/bmbrep.2011.44.3.199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ephrin signaling is involved in various morphogenetic events, such as axon guidance, hindbrain segmentation, and angiogenesis. We conducted a yeast two-hybrid screen using the intracellular domain (ICD) of EphrinB1 to gain biochemical insightinto the function of the EphrinB1 ICD. We identified the transcriptional co-repressor xTLE1/Groucho as an EphrinB1 interacting protein. Whole-mount in situ hybridization of Xenopus embryos confirmed the co-localization of EphrinB1 and a Xenopus counterpart to TLE1, xTLE4, during various stages of development. The EphrinB1/xTLE4 interaction was confirmed by co-immunoprecipitation experiments. Further characterization of the interaction revealed that the carboxy-terminal PDZ binding motif of EphrinB1 and the SP domain of xTLE4 are required for binding. Additionally, phosphorylation of EphrinB1 by a constitutively activated fibroblast growth factor receptor resulted in loss of the interaction, suggesting that the interaction is modulated by tyrosine phosphorylation of the EphrinB1 ICD.
Collapse
Affiliation(s)
- Teddy Kamata
- Laboratory of Cell and Developmental Signaling, National Cancer Institute-Frederick, Maryland 21702, USA
| | | | | | | | | | | |
Collapse
|
29
|
Kagale S, Rozwadowski K. EAR motif-mediated transcriptional repression in plants: an underlying mechanism for epigenetic regulation of gene expression. Epigenetics 2011; 6:141-6. [PMID: 20935498 DOI: 10.4161/epi.6.2.13627] [Citation(s) in RCA: 344] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Ethylene-responsive element binding factor-associated Amphiphilic Repression (EAR) motif-mediated transcriptional repression is emerging as one of the principal mechanisms of plant gene regulation. The EAR motif, defined by the consensus sequence patterns of either LxLxL or DLNxxP, is the most predominant form of transcriptional repression motif so far identified in plants. Additionally, this active repression motif is highly conserved in transcriptional regulators known to function as negative regulators in a broad range of developmental and physiological processes across evolutionarily diverse plant species. Recent discoveries of co-repressors interacting with EAR motifs, such as TOPLESS (TPL) and AtSAP18, have begun to unravel the mechanisms of EAR motif-mediated repression. The demonstration of genetic interaction between mutants of TPL and AtHDA19, co-complex formation between TPL-related 1 (TPR1) and AtHDA19, as well as direct physical interaction between AtSAP18 and AtHDA19 support a model where EAR repressors, via recruitment of chromatin remodeling factors, facilitate epigenetic regulation of gene expression. Here, we discuss the biological significance of EAR-mediated gene regulation in the broader context of plant biology and present literature evidence in support of a model for EAR motif-mediated repression via the recruitment and action of chromatin modifiers. Additionally, we discuss the possible influences of phosphorylation and ubiquitination on the function and turnover of EAR repressors.
Collapse
Affiliation(s)
- Sateesh Kagale
- Saskatoon Research Centre, Agriculture and Agri-Food Canada, 107 Science Place, Saskatoon, Saskatchewan, Canada
| | | |
Collapse
|
30
|
Roth M, Bonev B, Lindsay J, Lea R, Panagiotaki N, Houart C, Papalopulu N. FoxG1 and TLE2 act cooperatively to regulate ventral telencephalon formation. Development 2010; 137:1553-62. [PMID: 20356955 PMCID: PMC2853852 DOI: 10.1242/dev.044909] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2010] [Indexed: 12/22/2022]
Abstract
FoxG1 is a conserved transcriptional repressor that plays a key role in the specification, proliferation and differentiation of the telencephalon, and is expressed from the earliest stages of telencephalic development through to the adult. How the interaction with co-factors might influence the multiplicity and diversity of FoxG1 function is not known. Here, we show that interaction of FoxG1 with TLE2, a Xenopus tropicalis co-repressor of the Groucho/TLE family, is crucial for regulating the early activity of FoxG1. We show that TLE2 is co-expressed with FoxG1 in the ventral telencephalon from the early neural plate stage and functionally cooperates with FoxG1 in an ectopic neurogenesis assay. FoxG1 has two potential TLE binding sites: an N-terminal eh1 motif and a C-terminal YWPMSPF motif. Although direct binding seems to be mediated by the N-terminal motif, both motifs appear important for functional synergism. In the neurogenesis assay, mutation of either motif abolishes functional cooperation of TLE2 with FoxG1, whereas in the forebrain deletion of both motifs renders FoxG1 unable to induce the ventral telencephalic marker Nkx2.1. Knocking down either FoxG1 or TLE2 disrupts the development of the ventral telencephalon, supporting the idea that endogenous TLE2 and FoxG1 work together to specify the ventral telencephalon.
Collapse
Affiliation(s)
- Martin Roth
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Boyan Bonev
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Jennefer Lindsay
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Robert Lea
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Niki Panagiotaki
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Corinne Houart
- Medical Research Council Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK
| | - Nancy Papalopulu
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
31
|
Kang WH, Park YH, Park HM. The LAMMER kinase homolog, Lkh1, regulates Tup transcriptional repressors through phosphorylation in Schizosaccharomyces pombe. J Biol Chem 2010; 285:13797-806. [PMID: 20200159 PMCID: PMC2859543 DOI: 10.1074/jbc.m110.113555] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Disruption of the fission yeast LAMMER kinase, Lkh1, gene resulted in diverse phenotypes, including adhesive filamentous growth and oxidative stress sensitivity, but an exact cellular function had not been assigned to Lkh1. Through an in vitro pull-down approach, a transcriptional repressor, Tup12, was identified as an Lkh1 binding partner. Interactions between Lkh1 and Tup11 or Tup12 were confirmed by in vitro and in vivo binding assays. Tup proteins were phosphorylated by Lkh1 in a LAMMER motif-dependent manner. The LAMMER motif was also necessary for substrate recognition in vitro and cellular function in vivo. Transcriptional activity assays using promoters negatively regulated by Tup11 and Tup12 showed 6 or 2 times higher activity in the Δlkh1 mutant than the wild type, respectively. Northern analysis revealed derepressed expression of the fbp1+ mRNA in Δlkh1 and in Δtup11Δtup12 mutant cells under repressed conditions. Δlkh1 and Δtup11Δtup12 mutant cells showed flocculation, which was reversed by co-expression of Tup11 and -12 with Ssn6. Here, we presented a new aspect of the LAMMER kinase by demonstrating that the activities of global transcriptional repressors, Tup11 and Tup12, were positively regulated by Lkh1-mediated phosphorylation.
Collapse
Affiliation(s)
- Won-Hwa Kang
- Department of Microbiology, School of Bioscience and Biotechnology, Chungnam National University, Gung-dong 220, Yuseong-gu, Daejeon 305-764, Korea
| | | | | |
Collapse
|
32
|
Carvalho LR, Brinkmeier ML, Castinetti F, Ellsworth BS, Camper SA. Corepressors TLE1 and TLE3 interact with HESX1 and PROP1. Mol Endocrinol 2010; 24:754-65. [PMID: 20181723 DOI: 10.1210/me.2008-0359] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Pituitary hormone deficiency causes short stature in one in 4000 children born and can be caused by mutations in transcription factor genes, including HESX1, PROP1, and POU1F1. HESX1 interacts with a member of the groucho-related gene family, TLE1, through an engrailed homology domain and represses PROP1 activity. Mice with Prop1 deficiency exhibit failed differentiation of the POU1F1 lineage, resulting in lack of TSH, GH, and prolactin. In addition, these mutants exhibit profound pituitary dysmorphology and excess Hesx1 and Tle3 expression. The ability of HESX1 to interact with TLE3 has not been explored previously. We tested the ability of TLE3 to enhance HESX1-mediated repression of PROP1 in cell culture. Both TLE3 and TLE1 repress PROP1 in conjunction with HESX1 with similar efficiencies. TLE1 and TLE3 can each repress PROP1 in the absence of HESX1 via a protein-protein interaction. We tested the functional consequences of ectopic TLE3 and HESX1 expression in transgenic mice by driving constitutive expression in pituitary thyrotrophs and gonadotrophs. Terminal differentiation of these cells was suppressed by HESX1 alone and by TLE3 and HESX1 together but not by TLE3 alone. In summary, we present evidence that HESX1 is a strong repressor that can be augmented by the corepressors TLE1 and TLE3. Our in vitro studies suggest that TLE1 and TLE3 might also play roles independent of HESX1 by interacting with other transcription factors like PROP1.
Collapse
Affiliation(s)
- Luciani R Carvalho
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|
33
|
Cofactor-activated phosphorylation is required for inhibition of cortical neuron differentiation by Groucho/TLE1. PLoS One 2009; 4:e8107. [PMID: 19956621 PMCID: PMC2779591 DOI: 10.1371/journal.pone.0008107] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 11/07/2009] [Indexed: 12/03/2022] Open
Abstract
Background Transcriptional co-repressors of the Groucho/transducin-like Enhancer of split (Gro/TLE) family regulate the expression of a variety of genes and are involved in numerous developmental processes in both invertebrate and vertebrate species. More specifically, Gro/TLE1 participates in mechanisms that inhibit/delay the differentiation of cerebral cortex neural progenitor cells into neurons during mammalian forebrain development. The anti-neurogenic function of Gro/TLE1 depends on the formation of protein complexes with specific DNA-binding transcription factors that engage Gro/TLE1 through WRP(W/Y) sequences. Interaction with those transcription partners results in Gro/TLE1 recruitment to selected DNA sites and causes increased Gro/TLE1 phosphorylation. The physiological significance of the latter event, termed “cofactor-activated phosphorylation,” had not been determined. Therefore, this study aimed at clarifying the role of cofactor-activated phosphorylation in the anti-neurogenic function of Gro/TLE1. Methods and Principal Findings A combination of site-directed mutagenesis, mass spectrometry, biochemistry, primary cell culture, and immunocytochemical assays was utilized to characterize point mutations of Ser-286, a residue that is phosphorylated in vivo and is located within the serine/proline-rich (SP) domain of Gro/TLE1. Mutation of Ser-286 to alanine or glutamic acid does not perturb the interaction of Gro/TLE1 with DNA-binding partners, including the basic helix-loop-helix transcription factor Hes1, a prototypical anti-neurogenic WRP(W/Y) motif protein. Ser-286 mutations do not prevent the recruitment of Gro/TLE1 to DNA, but they impair cofactor-activated phosphorylation and weaken the interaction of Gro/TLE1 with chromatin. These effects are correlated with an impairment of the anti-neurogenic activity of Gro/TLE1. Similar results were obtained when mutations of Ser-289 and Ser-298, which are also located within the SP domain of Gro/TLE1, were analyzed. Conclusion Based on the positive correlation between Gro/TLE1 cofactor-activated phosphorylation and ability to inhibit cortical neuron differentiation, we propose that hyperphosphorylation induced by cofactor binding plays a positive role in the regulation of Gro/TLE1 anti-neurogenic activity.
Collapse
|
34
|
Larder R, Mellon PL. Otx2 induction of the gonadotropin-releasing hormone promoter is modulated by direct interactions with Grg co-repressors. J Biol Chem 2009; 284:16966-16978. [PMID: 19401468 PMCID: PMC2719334 DOI: 10.1074/jbc.m109.002485] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Indexed: 12/12/2022] Open
Abstract
Hormonal communication between the hypothalamus, pituitary, and gonads orchestrates the development and regulation of mammalian reproductive function. In mice, gonadotropin-releasing hormone (GnRH) expression is limited to approximately 1000 neurons that originate in the olfactory placode then migrate to specific positions scattered throughout the hypothalamus. Coordination of the hypothalamic-pituitary-gonadal axis is dependent upon correct migration of GnRH neurons into the hypothalamus followed by the appropriate synthesis and pulsatile secretion of GnRH. Defects in any one of these processes can cause infertility. Recently, substantial progress has been made in identifying transcription factors, and their cofactors, that regulate not only adult expression of GnRH, but also the maturation of GnRH neurons. Here, we show that expression of Otx2, a homeodomain protein required for the formation of the forebrain, is dramatically up-regulated during GnRH neuronal maturation and that overexpression of Otx2 increases GnRH promoter activity in GnRH neuronal cell lines. Furthermore, Otx2 transcriptional activity is modulated by Grg4, a member of the Groucho-related-gene (Grg) family. Using mutational analysis, we show that a WRPW peptide motif within the Otx2 protein is required for physical interaction between Otx2 and Grg4. Without this physical interaction, Grg4 cannot repress Otx2-dependent activation of GnRH gene transcription. Taken together, these data show that Otx2 is important for GnRH expression and that direct interaction between Otx2 and Grg co-repressors regulates GnRH gene expression in hypothalamic neurons.
Collapse
Affiliation(s)
- Rachel Larder
- From the Department of Reproductive Medicine and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674
| | - Pamela L Mellon
- From the Department of Reproductive Medicine and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674.
| |
Collapse
|
35
|
de-Leon SBT, Davidson EH. Modeling the dynamics of transcriptional gene regulatory networks for animal development. Dev Biol 2009; 325:317-28. [PMID: 19028486 PMCID: PMC4100934 DOI: 10.1016/j.ydbio.2008.10.043] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2008] [Revised: 10/14/2008] [Accepted: 10/21/2008] [Indexed: 01/04/2023]
Abstract
The dynamic process of cell fate specification is regulated by networks of regulatory genes. The architecture of the network defines the temporal order of specification events. To understand the dynamic control of the developmental process, the kinetics of mRNA and protein synthesis and the response of the cis-regulatory modules to transcription factor concentration must be considered. Here we review mathematical models for mRNA and protein synthesis kinetics which are based on experimental measurements of the rates of the relevant processes. The model comprises the response functions of cis-regulatory modules to their transcription factor inputs, by incorporating binding site occupancy and its dependence on biologically measurable quantities. We use this model to simulate gene expression, to distinguish between cis-regulatory execution of "AND" and "OR" logic functions, rationalize the oscillatory behavior of certain transcriptional auto-repressors and to show how linked subcircuits can be dealt with. Model simulations display the effects of mutation of binding sites, or perturbation of upstream gene expression. The model is a generally useful tool for understanding gene regulation and the dynamics of cell fate specification.
Collapse
Affiliation(s)
| | - Eric H. Davidson
- Division of Biology 156-29, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
36
|
Ahn JW, Lee YA, Ahn JH, Choi CY. Covalent conjugation of Groucho with SUMO-1 modulates its corepressor activity. Biochem Biophys Res Commun 2009; 379:160-5. [DOI: 10.1016/j.bbrc.2008.12.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 12/09/2008] [Indexed: 10/21/2022]
|
37
|
Cinnamon E, Paroush Z. Context-dependent regulation of Groucho/TLE-mediated repression. Curr Opin Genet Dev 2008; 18:435-40. [PMID: 18721877 DOI: 10.1016/j.gde.2008.07.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 07/14/2008] [Accepted: 07/23/2008] [Indexed: 01/15/2023]
Abstract
Groucho/TLE proteins are global corepressors that are recruited to target promoters by different families of DNA-binding repressors. As these corepressors are widely expressed, the long-standing view had been that Groucho/TLE-mediated repression is regulated solely by the spatial and temporal distribution of partner repressors. It has recently emerged, however, that Groucho/TLE repressor activity is itself regulated, in a signal induced, context-dependent manner. Here we review the essential roles played by Groucho/TLE factors in different cell-signalling processes that illustrate different modes for regulating Groucho/TLE-mediated repression: (i) via the expression of partner repressors; (ii) by competition with coactivators and (iii) through post-translational modifications of Groucho/TLE. We also discuss how the intrinsic properties of repressors can result in differential responses to Groucho/TLE regulation.
Collapse
Affiliation(s)
- Einat Cinnamon
- Department of Biochemistry, Faculty of Medicine, The Hebrew University, PO Box 12272, Jerusalem 91120, Israel.
| | | |
Collapse
|
38
|
Buscarlet M, Perin A, Laing A, Brickman JM, Stifani S. Inhibition of cortical neuron differentiation by Groucho/TLE1 requires interaction with WRPW, but not Eh1, repressor peptides. J Biol Chem 2008; 283:24881-8. [PMID: 18611861 DOI: 10.1074/jbc.m800722200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In both invertebrates and vertebrates, transcriptional co-repressors of the Groucho/transducin-like Enhancer of split (Gro/TLE) family regulate a number of developmental mechanisms, including neuronal differentiation. The pleiotropic activity of Gro/TLE depends on context-specific interactions with a variety of DNA-binding proteins. Most of those factors engage Gro/TLE through two different types of short peptide motifs, the WRP(W/Y) tetrapeptide and the Engrailed homology 1 (Eh1) sequence (FXIXXIL). The aim of this study was to elucidate the contribution of WRP(W/Y) and Eh1 motifs to mammalian Gro/TLE anti-neurogenic activity. Here we describe point mutations within the C-terminal WD40 repeat domain of Gro/TLE1 that do not perturb protein folding but disrupt the ability of Gro/TLE1 to inhibit the differentiation of cerebral cortex neural progenitor cells into neurons. One of those mutations, L743F, selectively blocks binding to Hes1, an anti-neurogenic basic helix-loop-helix protein that harbors a WRPW motif. In contrast, the L743F mutation does not disrupt binding to Engrailed1 and FoxG1, which both contain Eh1 motifs, nor to Tcf3, which binds to the Gro/TLE N terminus. These results demonstrate that the recruitment of transcription factors harboring WRP(W/Y) tetrapeptides is essential to the anti-neurogenic function of Gro/TLE1.
Collapse
Affiliation(s)
- Manuel Buscarlet
- Centre for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | | | | | | | | |
Collapse
|
39
|
The FoxO3a gene is a key negative target of canonical Notch signalling in the keratinocyte UVB response. EMBO J 2008; 27:1243-54. [PMID: 18388864 DOI: 10.1038/emboj.2008.45] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Accepted: 12/17/2007] [Indexed: 11/08/2022] Open
Abstract
Notch signalling has an important role in skin homeostasis, promoting keratinocyte differentiation and suppressing tumorigenesis. Here we show that this pathway also has an essential anti-apoptotic function in the keratinocyte UVB response. Notch1 expression and activity are significantly induced, in a p53-dependent manner, by UVB exposure of primary keratinocytes as well as intact epidermis of both mouse and human origin. The apoptotic response to UVB is increased by deletion of the Notch1 gene or down-modulation of Notch signalling by pharmacological inhibition or genetic suppression of 'canonical' Notch/CSL/MAML1-dependent transcription. Conversely, Notch activation protects keratinocytes against apoptosis through a mechanism that is not linked to Notch-induced cell cycle withdrawal or NF-kappaB activation. Rather, transcription of FoxO3a, a key pro-apoptotic gene, is under direct negative control of Notch/HERP transcription in keratinocytes, and upregulation of this gene accounts for the increased susceptibility to UVB of cells with suppressed Notch signalling. Thus, the canonical Notch/HERP pathway functions as a protective anti-apoptotic mechanism in keratinocytes through negative control of FoxO3a expression.
Collapse
|
40
|
Jacobsen KX, Vanderluit JL, Slack RS, Albert PR. HES1 regulates 5-HT1A receptor gene transcription at a functional polymorphism: essential role in developmental expression. Mol Cell Neurosci 2008; 38:349-58. [PMID: 18499474 DOI: 10.1016/j.mcn.2008.03.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Revised: 03/06/2008] [Accepted: 03/17/2008] [Indexed: 11/30/2022] Open
Abstract
Mammalian HES1 and HES5 are abundant in developing CNS and inhibit neurogenesis, while HES6 promotes neurogenesis. An early serotonergic differentiation marker, the 5-HT1A receptor, is repressed by HES5 and DEAF1 which recognize the C(-1019), but not G(-1019) allele of a human 5-HT1A promoter polymorphism associated with mood disorders. We tested whether HES1 and HES6 regulate transcriptional activity at this element. HES1 strongly repressed 5-HT1A transcription in neuronal and non-neuronal cells, while HES6 reversed HES1- and HES5-mediated repression. Mutation of a putative HES consensus site blocked HES1 and HES5, but, unlike HES5, HES1 repressed at the G(-1019) allele. To address its role in vivo, the temporal expression of 5-HT1A receptor RNA and protein was examined in HES1-/- mice, and elevated levels in E12.5 hindbrain and midbrain were observed. Thus, HES1 and HES6 oppositely regulate 5-HT1A receptor transcription and HES1 is required for its correct developmental expression.
Collapse
Affiliation(s)
- Kirsten X Jacobsen
- Ottawa Health Research Institute (Neuroscience) and Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON Canada
| | | | | | | |
Collapse
|
41
|
Abstract
The Groucho family of co-repressor proteins are essential for development and may also have a role in some human cancers.
Collapse
Affiliation(s)
- Barbara H Jennings
- Developmental Genetics Laboratory, Cancer Research UK, Lincoln's Inn Fields, London, WC2A 3PX, UK
| | | |
Collapse
|
42
|
Cinnamon E, Helman A, Ben-Haroush Schyr R, Orian A, Jiménez G, Paroush Z. Multiple RTK pathways downregulate Groucho-mediated repression in Drosophila embryogenesis. Development 2008; 135:829-37. [PMID: 18216172 DOI: 10.1242/dev.015206] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
RTK pathways establish cell fates in a wide range of developmental processes. However, how the pathway effector MAPK coordinately regulates the expression of multiple target genes is not fully understood. We have previously shown that the EGFR RTK pathway causes phosphorylation and downregulation of Groucho, a global co-repressor that is widely used by many developmentally important repressors for silencing their various targets. Here, we use specific antibodies that reveal the dynamics of Groucho phosphorylation by MAPK, and show that Groucho is phosphorylated in response to several RTK pathways during Drosophila embryogenesis. Focusing on the regulation of terminal patterning by the Torso RTK pathway, we demonstrate that attenuation of Groucho's repressor function via phosphorylation is essential for the transcriptional output of the pathway and for terminal cell specification. Importantly, Groucho is phosphorylated by an efficient mechanism that does not alter its subcellular localisation or decrease its stability; rather, modified Groucho endures long after MAPK activation has terminated. We propose that phosphorylation of Groucho provides a widespread, long-term mechanism by which RTK signals control target gene expression.
Collapse
Affiliation(s)
- Einat Cinnamon
- Department of Biochemistry, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
The anticipated therapeutic uses of neural stem cells depend on their ability to retain a certain level of developmental plasticity. In particular, cells must respond to developmental manipulations designed to specify precise neural fates. Studies in vivo and in vitro have shown that the developmental potential of neural progenitor cells changes and becomes progressively restricted with time. For in vitro cultured neural progenitors, it is those derived from embryonic stem cells that exhibit the greatest developmental potential. It is clear that both extrinsic and intrinsic mechanisms determine the developmental potential of neural progenitors and that epigenetic, or chromatin structural, changes regulate and coordinate hierarchical changes in fate-determining gene expression. Here, we review the temporal changes in developmental plasticity of neural progenitor cells and discuss the epigenetic mechanisms that underpin these changes. We propose that understanding the processes of epigenetic programming within the neural lineage is likely to lead to the development of more rationale strategies for cell reprogramming that may be used to expand the developmental potential of otherwise restricted progenitor populations.
Collapse
Affiliation(s)
- Nicholas D Allen
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3US, UK.
| |
Collapse
|
44
|
Belanger-Jasmin S, Llamosas E, Tang Y, Joachim K, Osiceanu AM, Jhas S, Stifani S. Inhibition of cortical astrocyte differentiation by Hes6 requires amino- and carboxy-terminal motifs important for dimerization and phosphorylation. J Neurochem 2007; 103:2022-34. [PMID: 17868320 DOI: 10.1111/j.1471-4159.2007.04902.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hairy/Enhancer of split (Hes) 6 is a basic helix-loop-helix protein that interacts with the transcriptional co-repressor, Groucho, and antagonizes the neural functions of the Notch pathway. More specifically, mouse Hes6 regulates cerebral corticogenesis by promoting neurogenesis and suppressing astrocyte differentiation. The molecular mechanisms underlying the anti-astrogenic function of Hes6 are poorly defined. Here we describe studies aimed at testing whether Hes6 inhibits astrocyte differentiation by antagonizing the transcription repression activity of Notch-activated Hes family members like Hes1. It is reported that Hes6 preferentially forms homodimers. Heterodimerization with Hes1 is antagonized in part by a conserved N-terminal patch of negatively charged residues. Mutation of this motif enhances heterodimerization with Hes1 and increases Hes6 ability to antagonize Hes1-mediated transcriptional repression. However, this mutation does not increase, but instead decreases, the anti-astrogenic activity of Hes6. It is shown further that Hes6 harbors a second conserved sequence, a C-terminal SPXXSP motif. This sequence is phosphorylated by the mitogen activated protein kinase pathway and its mutation disrupts the anti-astrogenic activity of Hes6 without affecting its ability to suppress Hes1. Together, these observations suggest that Hes6 homodimers regulate astrocyte differentiation through mechanisms that depend on the phosphorylation of Hes6 C-terminal domain but are independent of its ability to suppress Hes1-mediated transcriptional repression.
Collapse
Affiliation(s)
- Stephanie Belanger-Jasmin
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
45
|
Buscarlet M, Stifani S. The 'Marx' of Groucho on development and disease. Trends Cell Biol 2007; 17:353-61. [PMID: 17643306 DOI: 10.1016/j.tcb.2007.07.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 04/19/2007] [Accepted: 07/02/2007] [Indexed: 10/23/2022]
Abstract
Groucho proteins are abundant and broadly expressed nuclear factors that lack intrinsic DNA-binding activity but can interact with a variety of DNA-binding proteins. The recruitment of Groucho to specific gene regulatory sequences results in transcriptional repression. In both invertebrates and vertebrates, Groucho family members act as important regulators of several signaling mechanisms, including the Notch, Wingless/Wnt and Dpp/BMP/TGF-beta signaling pathways. Recent studies of embryonic development in several species point to an important role for Groucho in the regulation of multiple patterning and differentiation events. Moreover, a deregulated expression of human Groucho family members is correlated with several neoplastic conditions. Here we focus on the functions of Groucho proteins during body patterning and their implication in tumorigenesis.
Collapse
Affiliation(s)
- Manuel Buscarlet
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
46
|
McKenzie O, Ponte I, Mangelsdorf M, Finnis M, Colasante G, Shoubridge C, Stifani S, Gécz J, Broccoli V. Aristaless-related homeobox gene, the gene responsible for West syndrome and related disorders, is a Groucho/transducin-like enhancer of split dependent transcriptional repressor. Neuroscience 2007; 146:236-47. [PMID: 17331656 DOI: 10.1016/j.neuroscience.2007.01.038] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 12/30/2006] [Accepted: 01/17/2007] [Indexed: 11/17/2022]
Abstract
Aristaless-related homeobox gene (ARX) is an important paired-type homeobox gene involved in the development of human brain. The ARX gene mutations are a significant contributor to various forms of X-chromosome-linked mental retardation with and without additional features including epilepsy, lissencephaly with abnormal genitalia, hand dystonia or autism. Here we demonstrate that the human ARX protein is a potent transcriptional repressor, which binds to Groucho/transducin-like enhancer of split (TLE) co-factor proteins and the TLE1 in particular through its octapeptide (Engrailed homology repressor domain (eh-1) homology) domain. We show that the transcription repression activity of ARX is modulated by two strong repression domains, one located within the octapeptide domain and the second in the region of the polyalanine tract 4, and one activator domain, the aristaless domain. Importantly, we show that the transcription repression activity of ARX is affected by various naturally occurring mutations. The introduction of the c.98T>C (p.L33P) mutation results in the lack of binding to TLE1 protein and relaxed transcription repression. The introduction of the two most frequent ARX polyalanine tract expansion mutations increases the repression activity in a manner dependent on the number of extra alanines. Interestingly, deletions of alanine residues within polyalanine tracts 1 and 2 show low or no effect. In summary we demonstrate that the ARX protein is a strong transcription repressor, we identify novel ARX interacting proteins (TLE) and offer an explanation of a molecular pathogenesis of some ARX mutations, including the most frequent ARX mutations, the polyalanine tract expansion mutations, c.304ins(GCG)7 and c.428_451dup.
Collapse
Affiliation(s)
- O McKenzie
- Department of Genetic Medicine, Women's and Children's Hospital, and Department of Paediatrics, University of Adelaide, Adelaide, South Australia, 5006, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wang S, Jones KA. CK2 controls the recruitment of Wnt regulators to target genes in vivo. Curr Biol 2007; 16:2239-44. [PMID: 17113388 DOI: 10.1016/j.cub.2006.09.034] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Revised: 09/15/2006] [Accepted: 09/19/2006] [Indexed: 02/08/2023]
Abstract
Nuclear beta-catenin is a transcriptional coactivator of LEF-1/TCF DNA-binding proteins in the Wnt/Wg signaling pathway. Casein Kinase 2 (CK2), a positive regulator of Wnt signaling, is present in beta-catenin complexes and activated in Wnt-signaling cells. We show here that CK2 enhances beta-catenin:LEF-1 transactivation in vivo and in vitro and that beta-catenin and CK2 cycle on and off the DNA in an alternating manner with the TLE1 corepressor at Wnt target genes. Interestingly, CK2 phosphorylates hLEF-1 directly and stimulates binding and transactivation of beta-catenin:LEF-1 complexes on chromatin templates in vitro. In vitro, CK2 phosphorylation of hLEF-1 strongly enhances its affinity for beta-catenin and reduces its affinity for TLE1. MALDI-TOF mass spectrometry (MS) identified two CK2 phosphorylation sites (S42, S61) within the amino terminus of hLEF-1, and mutation of these sites reduced binding to beta-catenin in vitro and transactivation in vivo. Remarkably, treatment of cells with TBB, a pharmaceutical inhibitor of CK2, blocked the recruitment and cycling of beta-catenin and TLE1 at Wnt target genes in vivo. Taken together, these data indicate that CK2 is required for the assembly and cycling of Wnt-enhancer complexes in vivo.
Collapse
Affiliation(s)
- Song Wang
- Regulatory Biology Laboratory, The Salk Institute, 10010 N. Torrey Pines Road, La Jolla, California 92037, USA
| | | |
Collapse
|
48
|
Jhas S, Ciura S, Belanger-Jasmin S, Dong Z, Llamosas E, Theriault FM, Joachim K, Tang Y, Liu L, Liu J, Stifani S. Hes6 inhibits astrocyte differentiation and promotes neurogenesis through different mechanisms. J Neurosci 2006; 26:11061-71. [PMID: 17065448 PMCID: PMC6674651 DOI: 10.1523/jneurosci.1358-06.2006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The mechanisms regulating the generation of cell diversity in the mammalian cerebral cortex are beginning to be elucidated. In that regard, Hairy/Enhancer of split (Hes) 1 and 5 are basic helix-loop-helix (bHLH) factors that inhibit the differentiation of pluripotent cortical progenitors into neurons. In contrast, a related Hes family member termed Hes6 promotes neurogenesis. It is shown here that knockdown of endogenous Hes6 causes supernumerary cortical progenitors to differentiate into cells that exhibit an astrocytic morphology and express the astrocyte marker protein GFAP. Conversely, exogenous Hes6 expression in cortical progenitors inhibits astrocyte differentiation. The negative effect of Hes6 on astrocyte differentiation is independent of its ability to promote neuronal differentiation. We also show that neither its proneuronal nor its anti-gliogenic functions appear to depend on Hes6 ability to bind to DNA via the basic arm of its bHLH domain. Both of these activities require Hes6 to be localized to nuclei, but only its anti-gliogenic function depends on two short peptides, LNHLL and WRPW, that are conserved in all Hes6 proteins. These findings suggest that Hes6 is an important regulator of the neurogenic phase of cortical development by promoting the neuronal fate while suppressing astrocyte differentiation. They suggest further that separate molecular mechanisms underlie the proneuronal and anti-gliogenic activities of Hes6 in cortical progenitor cells.
Collapse
Affiliation(s)
- Sumit Jhas
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Sorana Ciura
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Stephanie Belanger-Jasmin
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Zhifeng Dong
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Estelle Llamosas
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Francesca M. Theriault
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Kerline Joachim
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Yeman Tang
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Lauren Liu
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Jisheng Liu
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Stefano Stifani
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada H3A 2B4
| |
Collapse
|
49
|
Abstract
The transcription factor DeltaFosB (also referred to as FosB2 or FosB[short form]) is an important mediator of the long-term plasticity induced in brain by chronic exposure to several types of psychoactive stimuli, including drugs of abuse, stress, and electroconvulsive seizures. A distinct feature of DeltaFosB is that, once induced, it persists in brain for relatively long periods of time in the absence of further stimulation. The mechanisms underlying this apparent stability, however, have remained unknown. Here, we demonstrate that DeltaFosB is a relatively stable transcription factor, with a half-life of approximately 10 h in cell culture. Furthermore, we show that DeltaFosB is a phosphoprotein in brain and that phosphorylation of a highly conserved serine residue (Ser27) in DeltaFosB protects it from proteasomal degradation. We provide several lines of evidence suggesting that this phosphorylation is mediated by casein kinase 2. These findings constitute the first evidence that DeltaFosB is phosphorylated and demonstrate that phosphorylation contributes to its stability, which is at the core of its ability to mediate long-lasting adaptations in brain.
Collapse
|
50
|
Marçal N, Patel H, Dong Z, Belanger-Jasmin S, Hoffman B, Helgason CD, Dang J, Stifani S. Antagonistic effects of Grg6 and Groucho/TLE on the transcription repression activity of brain factor 1/FoxG1 and cortical neuron differentiation. Mol Cell Biol 2006; 25:10916-29. [PMID: 16314515 PMCID: PMC1316978 DOI: 10.1128/mcb.25.24.10916-10929.2005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Groucho (Gro)/TLE transcriptional corepressors are involved in a variety of developmental mechanisms, including neuronal differentiation. They contain a conserved C-terminal WD40 repeat domain that mediates interactions with several DNA-binding proteins. In particular, Gro/TLE1 interacts with forkhead transcription factor brain factor 1 (BF-1; also termed FoxG1). BF-1 is an essential regulator of neuronal differentiation during cerebral cortex development and represses transcription together with Gro/TLE1. Gro/TLE-related gene product 6 (Grg6) shares with Gro/TLEs a conserved WD40 repeat domain but is more distantly related at its N-terminal half. We demonstrate that Grg6 is expressed in cortical neural progenitor cells and interacts with BF-1. In contrast to Gro/TLE1, however, Grg6 does not promote, but rather suppresses, BF-1-mediated transcriptional repression. Consistent with these observations, Grg6 interferes with the binding of Gro/TLE1 to BF-1 and does not repress transcription when targeted to DNA. Moreover, coexpression of Grg6 and BF-1 in cortical progenitor cells leads to a decrease in the number of proliferating cells and increased neuronal differentiation. Conversely, Grg6 knockdown by RNA interference causes decreased neurogenesis. These results identify a new role for Grg6 in cortical neuron development and establish a functional link between Grg6 and BF-1.
Collapse
Affiliation(s)
- Nathalie Marçal
- Center for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|