1
|
Chiu C, Küchler A, Depienne C, Preuße C, Marina AD, Reis A, Kaiser FJ, Nolte K, Hentschel A, Schara-Schmidt U, Kölbel H, Roos A. Skeletal muscle vulnerability in a child with Pitt-Hopkins syndrome. Skelet Muscle 2024; 14:15. [PMID: 39026379 PMCID: PMC11256580 DOI: 10.1186/s13395-024-00348-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 06/29/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND TCF4 acts as a transcription factor that binds to the immunoglobulin enhancer Mu-E5/KE5 motif. Dominant variants in TCF4 are associated with the manifestation of Pitt-Hopkins syndrome, a rare disease characterized by severe mental retardation, certain features of facial dysmorphism and, in many cases, with abnormalities in respiratory rhythm (episodes of paroxysmal tachypnea and hyperventilation, followed by apnea and cyanosis). Frequently, patients also develop epilepsy, microcephaly, and postnatal short stature. Although TCF4 is expressed in skeletal muscle and TCF4 seems to play a role in myogenesis as demonstrated in mice, potential myopathological findings taking place upon the presence of dominant TCF4 variants are thus far not described in human skeletal muscle. METHOD To address the pathological effect of a novel deletion affecting exons 15 and 16 of TCF4 on skeletal muscle, histological and immunofluorescence studies were carried out on a quadriceps biopsy in addition to targeted transcript studies and global proteomic profiling. RESULTS We report on muscle biopsy findings from a Pitt-Hopkins patient with a novel heterozygous deletion spanning exon 15 and 16 presenting with neuromuscular symptoms. Microscopic characterization of the muscle biopsy revealed moderate fiber type I predominance, imbalance in the proportion of fibroblasts co-expressing Vimentin and CD90, and indicate activation of the complement cascade in TCF4-mutant muscle. Protein dysregulations were unraveled by proteomic profiling. Transcript studies confirmed a mitochondrial vulnerability in muscle and confirmed reduced TCF4 expression. CONCLUSION Our combined findings, for the first time, unveil myopathological changes as phenotypical association of Pitt-Hopkins syndrome and thus expand the current clinical knowledge of the disease as well as support data obtained on skeletal muscle of a mouse model.
Collapse
Affiliation(s)
- Celine Chiu
- Centre for Neuromuscular Disorders, Department of Pediatric Neurology, Centre for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147, Essen, Germany
| | - Alma Küchler
- Center for Rare Diseases Essen, Institute for Human Genetics, University Hospital Essen, University Duisburg-Essen, 45147, Essen, Germany
| | - Christel Depienne
- Center for Rare Diseases Essen, Institute for Human Genetics, University Hospital Essen, University Duisburg-Essen, 45147, Essen, Germany
| | - Corinna Preuße
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Adela Della Marina
- Centre for Neuromuscular Disorders, Department of Pediatric Neurology, Centre for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147, Essen, Germany
| | - Andre Reis
- Institute for Human Genetics, University Hospital Erlangen, Friedrich-Alexander-University, 91054, Erlangen, Germany
| | - Frank J Kaiser
- Center for Rare Diseases Essen, Institute for Human Genetics, University Hospital Essen, University Duisburg-Essen, 45147, Essen, Germany
| | - Kay Nolte
- Department of Neuropathology, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | - Andreas Hentschel
- Leibniz-Institute for Analytical Science -ISAS- E.V, 44127, Dortmund, Germany
| | - Ulrike Schara-Schmidt
- Centre for Neuromuscular Disorders, Department of Pediatric Neurology, Centre for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147, Essen, Germany
| | - Heike Kölbel
- Centre for Neuromuscular Disorders, Department of Pediatric Neurology, Centre for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147, Essen, Germany
| | - Andreas Roos
- Centre for Neuromuscular Disorders, Department of Pediatric Neurology, Centre for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147, Essen, Germany.
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, K1H 5B2, Canada.
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225, Düsseldorf, Germany.
| |
Collapse
|
2
|
Chen L, Lin W, Zhang H, Geng S, Le Z, Wan F, Huang Q, Chen H, Liu X, Lu JJ, Kong L. TRIB3 promotes malignancy of head and neck squamous cell carcinoma via inhibiting ferroptosis. Cell Death Dis 2024; 15:178. [PMID: 38429254 PMCID: PMC10907716 DOI: 10.1038/s41419-024-06472-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 03/03/2024]
Abstract
Tribbles pseudokinase 3 (TRIB3) has been identified recently as a novel oncogene in several cancers. Still, further extensive research is imperative to elucidate its function and the molecular mechanisms underlying its involvement in the progression of head and neck squamous cell carcinoma (HNSCC). In our study, we found that TRIB3 silencing significantly promoted cell death by inducing ferroptosis. The interaction of TRIB3 with Transcription Factor 4 (TCF4) and β-catenin created a heterotrimeric complex, which directly interacts with the ALOXE3 promoter, detrimentally impacting its activation. The consequential partial neutralization of ferroptosis induced by TRIB3 deficiency is observed through the implementation of ALOXE3 knockdown. Furthermore, the study demonstrated that the molecular inhibitor hesperidin, targeting TRIB3, not only reduced cell malignancy but also induced ferroptosis, thereby suppressing tumor growth. Overall, our findings unequivocally validate the proposition that TRIB3 deficiency precipitates the iron death mechanism, thereby indicating that the strategic targeting of TRIB3 could emerge as an innovative therapeutic strategy for HNSCC.
Collapse
Affiliation(s)
- Li Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Wanzun Lin
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Haojiong Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Shikai Geng
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Ziyu Le
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Fangzhu Wan
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Qingting Huang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Huaiyuan Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Xingyu Liu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Jiade J Lu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Lin Kong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China.
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China.
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China.
| |
Collapse
|
3
|
Isoform-Specific Reduction of the Basic Helix-Loop-Helix Transcription Factor TCF4 Levels in Huntington's Disease. eNeuro 2021; 8:ENEURO.0197-21.2021. [PMID: 34518368 PMCID: PMC8519306 DOI: 10.1523/eneuro.0197-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/19/2021] [Accepted: 08/22/2021] [Indexed: 11/21/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder with onset of characteristic motor symptoms at midlife, preceded by subtle cognitive and behavioral disturbances. Transcriptional dysregulation emerges early in the disease course and is considered central to HD pathogenesis. Using wild-type (wt) and HD knock-in mouse striatal cell lines we observed a HD genotype-dependent reduction in the protein levels of transcription factor 4 (TCF4), a member of the basic helix-loop-helix (bHLH) family with critical roles in brain development and function. We characterized mouse Tcf4 gene structure and expression of alternative mRNAs and protein isoforms in cell-based models of HD, and in four different brain regions of male transgenic HD mice (R6/1) from young to mature adulthood. The largest decrease in the levels of TCF4 at mRNA and specific protein isoforms were detected in the R6/1 mouse hippocampus. Translating this finding to human disease, we found reduced expression of long TCF4 isoforms in the postmortem hippocampal CA1 area and in the cerebral cortex of HD patients. Additionally, TCF4 protein isoforms showed differential synergism with the proneural transcription factor ASCL1 in activating reporter gene transcription in hippocampal and cortical cultured neurons. Induction of neuronal activity increased these synergistic effects in hippocampal but not in cortical neurons, suggesting brain region-dependent differences in TCF4 functions. Collectively, this study demonstrates isoform-specific changes in TCF4 expression in HD that could contribute to the progressive impairment of transcriptional regulation and neuronal function in this disease.
Collapse
|
4
|
Ong Tone S, Kocaba V, Böhm M, Wylegala A, White TL, Jurkunas UV. Fuchs endothelial corneal dystrophy: The vicious cycle of Fuchs pathogenesis. Prog Retin Eye Res 2021; 80:100863. [PMID: 32438095 PMCID: PMC7648733 DOI: 10.1016/j.preteyeres.2020.100863] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/05/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022]
Abstract
Fuchs endothelial corneal dystrophy (FECD) is the most common primary corneal endothelial dystrophy and the leading indication for corneal transplantation worldwide. FECD is characterized by the progressive decline of corneal endothelial cells (CECs) and the formation of extracellular matrix (ECM) excrescences in Descemet's membrane (DM), called guttae, that lead to corneal edema and loss of vision. FECD typically manifests in the fifth decades of life and has a greater incidence in women. FECD is a complex and heterogeneous genetic disease where interaction between genetic and environmental factors results in cellular apoptosis and aberrant ECM deposition. In this review, we will discuss a complex interplay of genetic, epigenetic, and exogenous factors in inciting oxidative stress, auto(mito)phagy, unfolded protein response, and mitochondrial dysfunction during CEC degeneration. Specifically, we explore the factors that influence cellular fate to undergo apoptosis, senescence, and endothelial-to-mesenchymal transition. These findings will highlight the importance of abnormal CEC-DM interactions in triggering the vicious cycle of FECD pathogenesis. We will also review clinical characteristics, diagnostic tools, and current medical and surgical management options for FECD patients. These new paradigms in FECD pathogenesis present an opportunity to develop novel therapeutics for the treatment of FECD.
Collapse
Affiliation(s)
- Stephan Ong Tone
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Viridiana Kocaba
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Myriam Böhm
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Adam Wylegala
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Tomas L White
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Ula V Jurkunas
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
5
|
Fautsch MP, Wieben ED, Baratz KH, Bhattacharyya N, Sadan AN, Hafford-Tear NJ, Tuft SJ, Davidson AE. TCF4-mediated Fuchs endothelial corneal dystrophy: Insights into a common trinucleotide repeat-associated disease. Prog Retin Eye Res 2020; 81:100883. [PMID: 32735996 PMCID: PMC7988464 DOI: 10.1016/j.preteyeres.2020.100883] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/24/2020] [Accepted: 07/04/2020] [Indexed: 12/13/2022]
Abstract
Fuchs endothelial corneal dystrophy (FECD) is a common cause for heritable visual loss in the elderly. Since the first description of an association between FECD and common polymorphisms situated within the transcription factor 4 (TCF4) gene, genetic and molecular studies have implicated an intronic CTG trinucleotide repeat (CTG18.1) expansion as a causal variant in the majority of FECD patients. To date, several non-mutually exclusive mechanisms have been proposed that drive and/or exacerbate the onset of disease. These mechanisms include (i) TCF4 dysregulation; (ii) toxic gain-of-function from TCF4 repeat-containing RNA; (iii) toxic gain-of-function from repeat-associated non-AUG dependent (RAN) translation; and (iv) somatic instability of CTG18.1. However, the relative contribution of these proposed mechanisms in disease pathogenesis is currently unknown. In this review, we summarise research implicating the repeat expansion in disease pathogenesis, define the phenotype-genotype correlations between FECD and CTG18.1 expansion, and provide an update on research tools that are available to study FECD as a trinucleotide repeat expansion disease. Furthermore, ongoing international research efforts to develop novel CTG18.1 expansion-mediated FECD therapeutics are highlighted and we provide a forward-thinking perspective on key unanswered questions that remain in the field. FECD is a common, age-related corneal dystrophy. The majority of cases are associated with expansion of a CTG repeat (CTG18.1). FECD is the most common trinucleotide repeat expansion disease in humans. Evidence supports multiple molecular mechanisms underlying the pathophysiology. Novel CTG18.1-targeted therapeutics are in development.
Collapse
Affiliation(s)
- Michael P Fautsch
- Department of Ophthalmology, 200 1st St SW, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Eric D Wieben
- Department of Biochemistry and Molecular Biology, 200 1st St SW, Mayo Clinic, Rochester, MN, USA.
| | - Keith H Baratz
- Department of Ophthalmology, 200 1st St SW, Mayo Clinic, Rochester, MN, 55905, USA.
| | | | - Amanda N Sadan
- University College London Institute of Ophthalmology, London, ECIV 9EL, UK.
| | | | - Stephen J Tuft
- University College London Institute of Ophthalmology, London, ECIV 9EL, UK; Moorfields Eye Hospital, London, EC1V 2PD, UK.
| | - Alice E Davidson
- University College London Institute of Ophthalmology, London, ECIV 9EL, UK.
| |
Collapse
|
6
|
Hellwig M, Lauffer MC, Bockmayr M, Spohn M, Merk DJ, Harrison L, Ahlfeld J, Kitowski A, Neumann JE, Ohli J, Holdhof D, Niesen J, Schoof M, Kool M, Kraus C, Zweier C, Holmberg D, Schüller U. TCF4 (E2-2) harbors tumor suppressive functions in SHH medulloblastoma. Acta Neuropathol 2019; 137:657-673. [PMID: 30830316 DOI: 10.1007/s00401-019-01982-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 12/31/2022]
Abstract
The TCF4 gene encodes for the basic helix-loop-helix transcription factor 4 (TCF4), which plays an important role in the development of the central nervous system (CNS). Haploinsufficiency of TCF4 was found to cause Pitt-Hopkins syndrome (PTHS), a severe neurodevelopmental disorder. Recently, the screening of a large cohort of medulloblastoma (MB), a highly aggressive embryonal brain tumor, revealed almost 20% of adult patients with MB of the Sonic hedgehog (SHH) subtype carrying somatic TCF4 mutations. Interestingly, many of these mutations have previously been detected as germline mutations in patients with PTHS. We show here that overexpression of wild-type TCF4 in vitro significantly suppresses cell proliferation in MB cells, whereas mutant TCF4 proteins do not to the same extent. Furthermore, RNA sequencing revealed significant upregulation of multiple well-known tumor suppressors upon expression of wild-type TCF4. In vivo, a prenatal knockout of Tcf4 in mice caused a significant increase in apoptosis accompanied by a decreased proliferation and failed migration of cerebellar granule neuron precursor cells (CGNP), which are thought to be the cells of origin for SHH MB. In contrast, postnatal in vitro and in vivo knockouts of Tcf4 with and without an additional constitutive activation of the SHH pathway led to significantly increased proliferation of CGNP or MB cells. Finally, publicly available data from human MB show that relatively low expression levels of TCF4 significantly correlate with a worse clinical outcome. These results not only point to time-specific roles of Tcf4 during cerebellar development but also suggest a functional linkage between TCF4 mutations and the formation of SHH MB, proposing that TCF4 acts as a tumor suppressor during postnatal stages of cerebellar development.
Collapse
Affiliation(s)
- Malte Hellwig
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Martinistrasse 52, N63 (HPI), 20251, Hamburg, Germany
| | - Marlen C Lauffer
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michael Bockmayr
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Martinistrasse 52, N63 (HPI), 20251, Hamburg, Germany
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Spohn
- Research Institute Children's Cancer Center Hamburg, Martinistrasse 52, N63 (HPI), 20251, Hamburg, Germany
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel J Merk
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany
- Hertie Institute for Clinical Brain Research, University Hospital Tübingen, Tübingen, Germany
| | - Luke Harrison
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany
- Research Unit Neurobiology of Diabetes, Helmholtz Center Munich, Neuherberg, Germany
| | - Julia Ahlfeld
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Annabel Kitowski
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Julia E Neumann
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Ohli
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Dörthe Holdhof
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Martinistrasse 52, N63 (HPI), 20251, Hamburg, Germany
| | - Judith Niesen
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Martinistrasse 52, N63 (HPI), 20251, Hamburg, Germany
| | - Melanie Schoof
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Martinistrasse 52, N63 (HPI), 20251, Hamburg, Germany
| | - Marcel Kool
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cornelia Kraus
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christiane Zweier
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Dan Holmberg
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Research Institute Children's Cancer Center Hamburg, Martinistrasse 52, N63 (HPI), 20251, Hamburg, Germany.
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany.
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
7
|
Mohamed ZI, Tee SF, Chow TJ, Loh SY, Yong HS, Bakar AKA, Tang PY. Functional characterization of two variants in the 3'-untranslated region (UTR) of transcription factor 4 gene and their association with schizophrenia in sib-pairs from multiplex families. Asian J Psychiatr 2019; 40:76-81. [PMID: 30771755 DOI: 10.1016/j.ajp.2019.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/30/2019] [Accepted: 02/07/2019] [Indexed: 12/12/2022]
Abstract
Transcription factor 4 (TCF4) gene plays an important role in nervous system development and it always associated with the risk of schizophrenia. Since miRNAs regulate targetgenes by binding to 3'UTRs of target mRNAs, the functional variants located in 3'UTR of TCF4 are highly suggested to affect the gene expressions in schizophrenia. To test the hypothesis regarding the effects of the variants located in 3'UTR of TCF4, we conducted an in silico analysis to identify the functional variants and their predicted functions. In this study, we sequenced the 3'UTR of TCF4 in 13 multiplex schizophrenia families and 14 control families. We found two functional variants carried by three unrelated patients. We determined that the C allele of rs1272363 and the TC insert of rs373174214 might suppress post- transcriptional expression. Secondly, we cloned the region that flanked these two variants into a dual luciferase reporter system and compared the luciferase activities between the pmirGLO-TCF4 (control), pmirGLO-TCF4-rs373174214 and pmirGLO-TCF4-rs1273263. Both pmirGLO-TCF4-rs373174214 and pmirGLO-TCF4-rs1273263 caused lower reporter gene activities, as compared to the control. However, only the C allele of rs1272363 reduced the luciferase activity significantly (p = 0.0231). Our results suggested that rs1273263 is a potential regulator of TCF4 expression, and might be associated with schizophrenia.
Collapse
Affiliation(s)
- Zahra Isnaini Mohamed
- Department of Mechatronics and Biomedical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Bandar Sungai Long, Cheras 43000 Kajang, Malaysia
| | - Shiau Foon Tee
- Department of Chemical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Bandar Sungai Long, Cheras 43000 Kajang, Malaysia
| | - Tze Jen Chow
- Department of Mechatronics and Biomedical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Bandar Sungai Long, Cheras 43000 Kajang, Malaysia
| | - Siew Yim Loh
- Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Hoi Sen Yong
- Institute of Biological Sciences, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Pek Yee Tang
- Department of Mechatronics and Biomedical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Bandar Sungai Long, Cheras 43000 Kajang, Malaysia.
| |
Collapse
|
8
|
Grajkowska LT, Ceribelli M, Lau CM, Warren ME, Tiniakou I, Nakandakari Higa S, Bunin A, Haecker H, Mirny LA, Staudt LM, Reizis B. Isoform-Specific Expression and Feedback Regulation of E Protein TCF4 Control Dendritic Cell Lineage Specification. Immunity 2017; 46:65-77. [PMID: 27986456 PMCID: PMC5243153 DOI: 10.1016/j.immuni.2016.11.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 09/22/2016] [Accepted: 10/24/2016] [Indexed: 12/24/2022]
Abstract
The cell fate decision between interferon-producing plasmacytoid DC (pDC) and antigen-presenting classical DC (cDC) is controlled by the E protein transcription factor TCF4 (E2-2). We report that TCF4 comprises two transcriptional isoforms, both of which are required for optimal pDC development in vitro. The long Tcf4 isoform is expressed specifically in pDCs, and its deletion in mice impaired pDCs development and led to the expansion of non-canonical CD8+ cDCs. The expression of Tcf4 commenced in progenitors and was further upregulated in pDCs, correlating with stage-specific activity of multiple enhancer elements. A conserved enhancer downstream of Tcf4 was required for its upregulation during pDC differentiation, revealing a positive feedback loop. The expression of Tcf4 and the resulting pDC differentiation were selectively sensitive to the inhibition of enhancer-binding BET protein activity. Thus, lineage-specifying function of E proteins is facilitated by lineage-specific isoform expression and by BET-dependent feedback regulation through distal regulatory elements.
Collapse
Affiliation(s)
- Lucja T Grajkowska
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Michele Ceribelli
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA; Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Colleen M Lau
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Margaret E Warren
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Graduate Program in Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Ioanna Tiniakou
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | | | - Anna Bunin
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Hans Haecker
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Leonid A Mirny
- Department of Physics and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Louis M Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Boris Reizis
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
9
|
Schrankel CS, Solek CM, Buckley KM, Anderson MK, Rast JP. A conserved alternative form of the purple sea urchin HEB/E2-2/E2A transcription factor mediates a switch in E-protein regulatory state in differentiating immune cells. Dev Biol 2016; 416:149-161. [PMID: 27265865 DOI: 10.1016/j.ydbio.2016.05.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/26/2016] [Accepted: 05/27/2016] [Indexed: 12/11/2022]
Abstract
E-proteins are basic helix-loop-helix (bHLH) transcription factors with essential roles in animal development. In mammals, these are encoded by three loci: E2-2 (ITF-2/ME2/SEF2/TCF4), E2A (TCF3), and HEB (ME1/REB/TCF12). The HEB and E2-2 paralogs are expressed as alternative (Alt) isoforms with distinct N-terminal sequences encoded by unique exons under separate regulatory control. Expression of these alternative transcripts is restricted relative to the longer (Can) forms, suggesting distinct regulatory roles, although the functions of the Alt proteins remain poorly understood. Here, we characterize the single sea urchin E-protein ortholog (SpE-protein). The organization of the SpE-protein gene closely resembles that of the extended HEB/E2-2 vertebrate loci, including a transcript that initiates at a homologous alternative transcription start site (SpE-Alt). The existence of an Alt form in the sea urchin indicates that this feature predates the emergence of the vertebrates. We present additional evidence indicating that this transcript was present in the common bilaterian ancestor. In contrast to the widely expressed canonical form (SpE-Can), SpE-Alt expression is tightly restricted. SpE-Alt is expressed in two phases: first in aboral non-skeletogenic mesenchyme (NSM) cells and then in oral NSM cells preceding their differentiation and ingression into the blastocoel. Derivatives of these cells mediate immune response in the larval stage. Inhibition of SpE-Alt activity interferes with these events. Notably, although the two isoforms are initially co-expressed, as these cells differentiate, SpE-Can is excluded from the SpE-Alt(+) cell population. This mutually exclusive expression is dependent on SpE-Alt function, which reveals a previously undescribed negative regulatory linkage between the two E-protein forms. Collectively, these findings reorient our understanding of the evolution of this transcription factor family and highlight fundamental properties of E-protein biology.
Collapse
Affiliation(s)
- Catherine S Schrankel
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Cynthia M Solek
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Katherine M Buckley
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Michele K Anderson
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Jonathan P Rast
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
10
|
Hill MJ, Forrest MP, Martin-Rendon E, Blake DJ. Association of Transcription Factor 4 (TCF4) variants with schizophrenia and intellectual disability. Curr Behav Neurosci Rep 2014. [DOI: 10.1007/s40473-014-0027-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
11
|
in 't Hout FEM, van der Reijden BA, Monteferrario D, Jansen JH, Huls G. High expression of transcription factor 4 (TCF4) is an independent adverse prognostic factor in acute myeloid leukemia that could guide treatment decisions. Haematologica 2014; 99:e257-9. [PMID: 25150259 DOI: 10.3324/haematol.2014.110437] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Florentien E M in 't Hout
- Department of Laboratory medicine, Laboratory of Hematology, RadboudUMC; and Department of Hematology, RadboudUMC, Nijmegen, The Netherlands
| | | | - Davide Monteferrario
- Department of Laboratory medicine, Laboratory of Hematology, RadboudUMC; and Department of Hematology, RadboudUMC, Nijmegen, The Netherlands
| | - Joop H Jansen
- Department of Laboratory medicine, Laboratory of Hematology, RadboudUMC; and
| | - Gerwin Huls
- Department of Laboratory medicine, Laboratory of Hematology, RadboudUMC; and Department of Hematology, RadboudUMC, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Shin HW, Choi H, So D, Kim YI, Cho K, Chung HJ, Lee KH, Chun YS, Cho CH, Kang GH, Kim WH, Park JW. ITF2 prevents activation of the β-catenin-TCF4 complex in colon cancer cells and levels decrease with tumor progression. Gastroenterology 2014; 147:430-442.e8. [PMID: 24846398 DOI: 10.1053/j.gastro.2014.04.047] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 04/07/2014] [Accepted: 04/28/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Immunoglobulin transcription factor 2 (ITF2) was believed to promote neoplastic transformation via activation of β-catenin. However, ITF2 recently was reported to suppress colon carcinogenesis. We investigated the roles of ITF2 in colorectal cancer cell lines and tumor formation and growth in mice. METHODS Levels of ITF2, β-catenin, and c-Myc were measured in 12 human colorectal tumor samples and by immunohistochemistry. ITF2 regulation of β-catenin and T-cell factor (TCF) were analyzed using luciferase reporter, reverse-transcription quantitative polymerase chain reaction, flow cytometry, and immunoblot analyses. Mice were given subcutaneous injections of human colorectal cancer cell lines that stably express ITF2, small hairpin RNAs to reduce levels of ITF2, or control plasmids; xenograft tumor growth was assessed. Human colorectal carcinoma tissue arrays were used to associate levels of ITF2 expression and clinical outcomes. RESULTS Levels of β-catenin, cMyc, and ITF2 were increased in areas of human colon adenomas and carcinomas, compared with nontumor areas of the same tissues. ITF2 levels were reduced and cMyc levels were increased in areas of carcinoma, compared with adenoma. In human colorectal cancer cell lines, activation of the β-catenin-TCF4 complex and expression of its target genes were regulated negatively by ITF2. ITF2 inhibited formation of the β-catenin-TCF4 complex by competing with TCF4 for β-catenin binding. Stable transgenic expression of ITF2 in human colorectal cancer cell lines reduced their proliferation and tumorigenic potential in mice, whereas small hairpin RNA knockdown of ITF2 promoted growth of xenograft tumors in mice. In an analysis of colorectal tumor tissue arrays, loss of ITF2 from colorectal tumor tissues was associated with poor outcomes of patients. A gene set enrichment analysis supported the negative correlation between the level of ITF2 and activity of the β-catenin-TCF4 complex. CONCLUSIONS In human colorectal cancer cell lines and tissue samples, ITF2 appears to prevent activation of the β-catenin-TCF4 complex and transcription of its gene targets. Loss of ITF2 promotes the ability of colorectal cancer cells to form xenograft tumors, and is associated with tumor progression and shorter survival times of patients.
Collapse
Affiliation(s)
- Hyun-Woo Shin
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyunsung Choi
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea
| | - Daeho So
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea; Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Im Kim
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea; Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Kumsun Cho
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Hee-Joon Chung
- Seoul National University Biomedical Informatics, Seoul National University College of Medicine, Seoul, Korea
| | - Kyoung-Hwa Lee
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea; Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
| | - Yang-Sook Chun
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea; Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea; Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
| | - Chung-Hyun Cho
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Gyeong Hoon Kang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Wan Park
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea; Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
13
|
The emerging roles of TCF4 in disease and development. Trends Mol Med 2014; 20:322-31. [PMID: 24594265 DOI: 10.1016/j.molmed.2014.01.010] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 01/28/2014] [Accepted: 01/30/2014] [Indexed: 10/25/2022]
Abstract
Genome-wide association studies have identified common variants in transcription factor 4 (TCF4) as susceptibility loci for schizophrenia, Fuchs' endothelial corneal dystrophy, and primary sclerosing cholangitis. By contrast, rare TCF4 mutations cause Pitt-Hopkins syndrome, a disorder characterized by intellectual disability and developmental delay, and have also been described in patients with other neurodevelopmental disorders. TCF4 therefore sits at the nexus between common and rare disorders. TCF4 interacts with other basic helix-loop-helix proteins, forming transcriptional networks that regulate the differentiation of several distinct cell types. Here, we review the role of TCF4 in these seemingly diverse disorders and discuss recent data implicating TCF4 as an important regulator of neurodevelopment and epithelial-mesenchymal transition.
Collapse
|
14
|
Gow CH, Guo C, Wang D, Hu Q, Zhang J. Differential involvement of E2A-corepressor interactions in distinct leukemogenic pathways. Nucleic Acids Res 2013; 42:137-52. [PMID: 24064250 PMCID: PMC3874172 DOI: 10.1093/nar/gkt855] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
E2A is a member of the E-protein family of transcription factors. Previous studies have reported context-dependent regulation of E2A-dependent transcription. For example, whereas the E2A portion of the E2A-Pbx1 leukemia fusion protein mediates robust transcriptional activation in t(1;19) acute lymphoblastic leukemia, the transcriptional activity of wild-type E2A is silenced by high levels of corepressors, such as the AML1-ETO fusion protein in t(8;21) acute myeloid leukemia and ETO-2 in hematopoietic cells. Here, we show that, unlike the HEB E-protein, the activation domain 1 (AD1) of E2A has specifically reduced corepressor interaction due to E2A-specific amino acid changes in the p300/CBP and ETO target motif. Replacing E2A-AD1 with HEB-AD1 abolished the ability of E2A-Pbx1 to activate target genes and to induce cell transformation. On the other hand, the weak E2A-AD1-corepressor interaction imposes a critical importance on another ETO-interacting domain, downstream ETO-interacting sequence (DES), for corepressor-mediated repression. Deletion of DES abrogates silencing of E2A activity by AML1-ETO in t(8;21) leukemia cells or by ETO-2 in normal hematopoietic cells. Our results reveal an E2A-specific mechanism important for its context-dependent activation and repression function, and provide the first evidence for the differential involvement of E2A-corepressor interactions in distinct leukemogenic pathways.
Collapse
Affiliation(s)
- Chien-Hung Gow
- Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | | | | | | | | |
Collapse
|
15
|
Navarrete K, Pedroso I, De Jong S, Stefansson H, Steinberg S, Stefansson K, Ophoff RA, Schalkwyk LC, Collier DA. TCF4 (e2-2; ITF2): a schizophrenia-associated gene with pleiotropic effects on human disease. Am J Med Genet B Neuropsychiatr Genet 2013; 162B:1-16. [PMID: 23129290 DOI: 10.1002/ajmg.b.32109] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 09/27/2012] [Indexed: 12/22/2022]
Abstract
Common SNPs in the transcription factor 4 (TCF4; ITF2, E2-2, SEF-2) gene, which encodes a basic Helix-Loop-Helix (bHLH) transcription factor, are associated with schizophrenia, conferring a small increase in risk. Other common SNPs in the gene are associated with the common eye disorder Fuch's corneal dystrophy, while rare, mostly de novo inactivating mutations cause Pitt-Hopkins syndrome. In this review, we present a systematic bioinformatics and literature review of the genomics, biological function and interactome of TCF4 in the context of schizophrenia. The TCF4 gene is present in all vertebrates, and although protein length varies, there is high conservation of primary sequence, including the DNA binding domain. Humans have a unique leucine-rich nuclear export signal. There are two main isoforms (A and B), as well as complex splicing generating many possible N-terminal amino acid sequences. TCF4 is highly expressed in the brain, where plays a role in neurodevelopment, interacting with class II bHLH transcription factors Math1, HASH1, and neuroD2. The Ca(2+) sensor protein calmodulin interacts with the DNA binding domain of TCF4, inhibiting transcriptional activation. It is also the target of microRNAs, including mir137, which is implicated in schizophrenia. The schizophrenia-associated SNPs are in linkage disequilibrium with common variants within putative DNA regulatory elements, suggesting that regulation of expression may underlie association with schizophrenia. Combined gene co-expression analyses and curated protein-protein interaction data provide a network involving TCF4 and other putative schizophrenia susceptibility genes. These findings suggest new opportunities for understanding the molecular basis of schizophrenia and other mental disorders.
Collapse
Affiliation(s)
- Katherinne Navarrete
- Social, Genetic and Developmental Psychiatry Centre, King's College London, Institute of Psychiatry, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Muthu K, Panneerselvam M, Jayaraman M, Topno NS, Das AA, Ramadas K. Structural insights into interacting mechanism of ID1 protein with an antagonist ID1/3-PA7 and agonist ETS-1 in treatment of ovarian cancer: molecular docking and dynamics studies. J Mol Model 2012; 18:4865-84. [DOI: 10.1007/s00894-012-1489-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 05/28/2012] [Indexed: 11/30/2022]
|
17
|
HEB in the spotlight: Transcriptional regulation of T-cell specification, commitment, and developmental plasticity. Clin Dev Immunol 2012; 2012:678705. [PMID: 22577461 PMCID: PMC3346973 DOI: 10.1155/2012/678705] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 12/12/2011] [Indexed: 12/02/2022]
Abstract
The development of T cells from multipotent progenitors in the thymus occurs by cascades of interactions between signaling molecules and transcription factors, resulting in the loss of alternative lineage potential and the acquisition of the T-cell functional identity. These processes require Notch signaling and the activity of GATA3, TCF1, Bcl11b, and the E-proteins HEB and E2A. We have shown that HEB factors are required to inhibit the thymic NK cell fate and that HEBAlt allows the passage of T-cell precursors from the DN to DP stage but is insufficient for suppression of the NK cell lineage choice. HEB factors are also required to enforce the death of cells that have not rearranged their TCR genes. The synergistic interactions between Notch1, HEBAlt, HEBCan, GATA3, and TCF1 are presented in a gene network model, and the influence of thymic stromal architecture on lineage choice in the thymus is discussed.
Collapse
|
18
|
Sepp M, Kannike K, Eesmaa A, Urb M, Timmusk T. Functional diversity of human basic helix-loop-helix transcription factor TCF4 isoforms generated by alternative 5' exon usage and splicing. PLoS One 2011; 6:e22138. [PMID: 21789225 PMCID: PMC3137626 DOI: 10.1371/journal.pone.0022138] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 06/16/2011] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Transcription factor 4 (TCF4 alias ITF2, E2-2, ME2 or SEF2) is a ubiquitous class A basic helix-loop-helix protein that binds to E-box DNA sequences (CANNTG). While involved in the development and functioning of many different cell types, recent studies point to important roles for TCF4 in the nervous system. Specifically, human TCF4 gene is implicated in susceptibility to schizophrenia and TCF4 haploinsufficiency is the cause of the Pitt-Hopkins mental retardation syndrome. However, the structure, expression and coding potential of the human TCF4 gene have not been described in detail. PRINCIPAL FINDINGS In the present study we used human tissue samples to characterize human TCF4 gene structure and TCF4 expression at mRNA and protein level. We report that although widely expressed, human TCF4 mRNA expression is particularly high in the brain. We demonstrate that usage of numerous 5' exons of the human TCF4 gene potentially yields in TCF4 protein isoforms with 18 different N-termini. In addition, the diversity of isoforms is increased by alternative splicing of several internal exons. For functional characterization of TCF4 isoforms, we overexpressed individual isoforms in cultured human cells. Our analysis revealed that subcellular distribution of TCF4 isoforms is differentially regulated: Some isoforms contain a bipartite nuclear localization signal and are exclusively nuclear, whereas distribution of other isoforms relies on heterodimerization partners. Furthermore, the ability of different TCF4 isoforms to regulate E-box controlled reporter gene transcription is varied depending on whether one or both of the two TCF4 transcription activation domains are present in the protein. Both TCF4 activation domains are able to activate transcription independently, but act synergistically in combination. CONCLUSIONS Altogether, in this study we have described the inter-tissue variability of TCF4 expression in human and provided evidence about the functional diversity of the alternative TCF4 protein isoforms.
Collapse
Affiliation(s)
- Mari Sepp
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Kaja Kannike
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Ave Eesmaa
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Mari Urb
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Tõnis Timmusk
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
- * E-mail:
| |
Collapse
|
19
|
Mologni L, Dekhil H, Ceccon M, Purgante S, Lan C, Cleris L, Magistroni V, Formelli F, Gambacorti-Passerini CB. Colorectal tumors are effectively eradicated by combined inhibition of {beta}-catenin, KRAS, and the oncogenic transcription factor ITF2. Cancer Res 2010; 70:7253-63. [PMID: 20823162 DOI: 10.1158/0008-5472.can-10-1108] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Colorectal carcinomas (CRC) harbor well-defined genetic abnormalities, including aberrant activation of β-catenin (β-cat) and KRAS, but independent targeting of these molecules seems to have limited therapeutic effect. In this study, we report therapeutic effects of combined targeting of different oncogenes in CRC. Inducible short hairpin RNA (shRNA)-mediated silencing of β-cat, ITF2, or KRAS decreased proliferation by 88%, 72%, and 45%, respectively, with no significant apoptosis in any case. In contrast, combined blockade of β-cat and ITF2 inhibited proliferation by 99% with massive apoptosis. Similar effects occurred after combined shRNA against β-cat and KRAS. In vivo, single oncogene blockade inhibited the growth of established tumors by up to 30%, whereas dual β-cat and ITF2 targeting caused 93% inhibition. Similar tumor growth suppression was achieved by double β-cat/KRAS shRNA in vivo. Our findings illustrate an effective therapeutic principle in CRC based on a combination targeting strategy that includes the ITF2 oncogene, which represents a novel therapeutic target.
Collapse
Affiliation(s)
- Luca Mologni
- University of Milano Bicocca, Monza, Italy; McGill University, Montreal, Canada; and National Cancer Institute, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Baratz KH, Tosakulwong N, Ryu E, Brown WL, Branham K, Chen W, Tran KD, Schmid-Kubista KE, Heckenlively JR, Swaroop A, Abecasis G, Bailey KR, Edwards AO. E2-2 protein and Fuchs's corneal dystrophy. N Engl J Med 2010; 363:1016-24. [PMID: 20825314 DOI: 10.1056/nejmoa1007064] [Citation(s) in RCA: 211] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Fuchs's corneal dystrophy (FCD) is a leading cause of corneal transplantation and affects 5% of persons in the United States who are over the age of 40 years. Clinically visible deposits called guttae develop under the corneal endothelium in patients with FCD. A loss of endothelial cells and deposition of an abnormal extracellular matrix are observed microscopically. In advanced disease, the cornea swells and becomes cloudy because the remaining endothelial cells are not sufficient to keep the cornea dehydrated and clear. Although rare genetic variation that contributes to both early-onset and typical late-onset forms of FCD has been identified, to our knowledge, no common variants have been reported. METHODS We performed a genomewide association study and replicated the most significant observations in a second, independent group of subjects. RESULTS Alleles in the transcription factor 4 gene (TCF4), encoding a member of the E-protein family (E2-2), were associated with typical FCD (P=2.3x10(-26)). The association increased the odds of having FCD by a factor of 30 for persons with two copies of the disease variants (homozygotes) and discriminated between case subjects and control subjects with about 76% accuracy. At least two regions of the TCF4 locus were associated independently with FCD. Alleles in the gene encoding protein tyrosine phosphatase receptor type G (PTPRG) were associated with FCD (P=4.0x10(-7)), but the association did not reach genomewide significance. CONCLUSIONS Genetic variation in TCF4 contributes to the development of FCD. (Funded by the National Eye Institute and others.)
Collapse
Affiliation(s)
- Keith H Baratz
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wang D, Claus CL, Rajkumar P, Braunstein M, Moore AJ, Sigvardsson M, Anderson MK. Context-dependent regulation of hematopoietic lineage choice by HEBAlt. THE JOURNAL OF IMMUNOLOGY 2010; 185:4109-17. [PMID: 20826759 DOI: 10.4049/jimmunol.0901783] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hematopoietic development is controlled by combinatorial interactions between E-protein transcription factors and other lineage regulators that operate in the context of gene-regulatory networks. The E-proteins HEB and E2A are critical for T cell and B cell development, but the mechanisms by which their activities are directed to different genes in each lineage are unclear. We found that a short form of HEB, HEBAlt, acts downstream of Delta-like (DL)-Notch signaling to promote T cell development. In this paper, we show that forced expression of HEBAlt in mouse hematopoietic progenitors inhibited B cell development, but it allowed them to adopt a myeloid fate. HEBAlt interfered with the activity of E2A homodimers and with the expression of the transcription factor Pax5, both of which are critical for B cell development. However, when combined with DL-Notch signaling, HEBAlt enhanced the generation of T cell progenitors at the expense of myeloid cells. The longer form of HEB, HEBCan, also inhibited E47 activity and Pax5 expression, but it did not collaborate with DL-Notch signaling to suppress myeloid potential. Therefore, HEBAlt can suppress B cell or myeloid potential in a context-specific manner, which suggests a role for this factor in maintaining T lineage priming prior to commitment.
Collapse
Affiliation(s)
- Duncheng Wang
- Division of Molecular and Cellular Biology, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
22
|
Appaiah H, Bhat-Nakshatri P, Mehta R, Thorat M, Badve S, Nakshatri H. ITF2 is a target of CXCR4 in MDA-MB-231 breast cancer cells and is associated with reduced survival in estrogen receptor-negative breast cancer. Cancer Biol Ther 2010; 10:600-14. [PMID: 20603605 DOI: 10.4161/cbt.10.6.12586] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
CXCR4, a chemokine receptor, plays an important role in breast cancer growth, invasion, and metastasis. The transcriptional targets of CXCR4 signaling are not known. Microarray analysis of CXCR4-enriched and CXCR4-low subpopulations of the MDA-MB-231 breast cancer cell line, which has a constitutively active CXCR4 signaling network, revealed differential expression of ∼ 200 genes in the CXCR4-enriched subpopulation. ITF2, upregulated in CXCR4-enriched cells, was investigated further. Expression array datasets of primary breast tumors revealed higher ITF2 expression in estrogen receptor negative tumors, which correlated with reduced progression free and overall survival and suggested its relevance in breast cancer progression. CXCL12, a CXCR4 ligand, increased ITF2 expression in MDA-MB-231 cells. ITF2 is a basic helix-loop-helix transcription factor that controls the epithelial-to-mesenchymal transition and the function of the ID family (inhibitor-of-differentiation) of transcription factors, such as ID2. ID2 promotes differentiation of breast epithelial cells and its reduced expression in breast cancer is associated with an unfavorable prognosis. Both CXCR4 and ITF2 repressed ID2 expression. In xenograft studies, CXCR4-enriched cells formed large tumors and exhibited significantly elevated lung metastasis. Short interfering RNA against ITF2 reduced invasion of the CXCR4-enriched MDA-MB-231 subpopulation, whereas ITF2 overexpression restored the invasive capacity of MDA-MB-231 cells expressing CXCR4shRNA. Furthermore, overexpression of ITF2 in these cells enhanced tumor growth. We propose that ITF2 is one of the CXCR4 targets, which is involved in CXCR4-dependent tumor growth and invasion of breast cancer cells.
Collapse
Affiliation(s)
- Hitesh Appaiah
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | |
Collapse
|
23
|
Cano A, Portillo F. An emerging role for class I bHLH E2-2 proteins in EMT regulation and tumor progression. Cell Adh Migr 2010; 4:56-60. [PMID: 20023376 DOI: 10.4161/cam.4.1.9995] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
EMT is a complex process whereby cells lose cell-cell interactions and other epithelial properties whilst acquiring a migratory and mesenchymal phenotype. EMT is presently recognized as an important even for tumor invasion and metastasis. Functional E-cadherin loss is a hallmark of EMT and required for tumor invasion in the majority of carcinomas. Transcriptional downregulation is one of the major mechanisms for E-cadherin suppression in carcinomas. In the last decade several E-cadherin repressors, belonging to different transcriptional families, have been identified that, importantly, also act as potent EMT inducers. One of the last additions to EMT regulators are the class I bHLH factors E2-2 (also known as TCF4). However, the hierarchical and functional interrelations between the different EMT inducers are still poorly understood. Here, we comment on the new and so far unrecognized function of E2-2 factors in EMT and discuss on the potential interactions among various EMT inducers. Emerging evidence supporting the participation of TCF4 in human malignancies is also discussed. Thus, increasing understanding of EMT and its regulators is providing meaningful insights into the present knowledge on tumor progression.
Collapse
Affiliation(s)
- Amparo Cano
- Departmento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain.
| | | |
Collapse
|
24
|
The transcription factor ITF-2A induces cell cycle arrest via p21Cip1. Biochem Biophys Res Commun 2009; 387:736-40. [DOI: 10.1016/j.bbrc.2009.07.102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 07/22/2009] [Indexed: 11/21/2022]
|
25
|
Sobrado VR, Moreno-Bueno G, Cubillo E, Holt LJ, Nieto MA, Portillo F, Cano A. The class I bHLH factors E2-2A and E2-2B regulate EMT. J Cell Sci 2009; 122:1014-24. [DOI: 10.1242/jcs.028241] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Functional loss of the cell-cell adhesion molecule E-cadherin is an essential event for epithelial-mesenchymal transition (EMT), a process that allows cell migration during embryonic development and tumour invasion. In most carcinomas, transcriptional repression has emerged as the main mechanism responsible for E-cadherin downregulation. Here, we report the identification of class I bHLH factor E2-2 (TCF4/ITF2) as a new EMT regulator. Both isoforms of E2-2 (E2-2A and E2-2B) induce a full EMT when overexpressed in MDCK cells but without affecting the tumorigenic properties of parental cells, in contrast to other EMT inducers, such as Snail1 or class I bHLH E47. E-cadherin repression mediated by E2-2 is indirect and independent of proximal E-boxes of the promoter. Knockdown studies indicate that E2-2 expression is dispensable for maintenance of the EMT driven by Snail1 and E47. Comparative gene-profiling analysis reveals that E2-2 factors induce similar, yet distinct, genetic programs to that induced by E47 in MDCK cells. These results, together with the embryonic expression pattern of Tcf4 and E2A (which encodes E12/E47), support a distinct role for E2-2 and suggest an interesting interplay between E-cadherin repressors in the regulation of physiological and pathological EMT processes.
Collapse
Affiliation(s)
- Verónica R. Sobrado
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas `Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| | - Gema Moreno-Bueno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas `Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| | - Eva Cubillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas `Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| | - Liam J. Holt
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas `Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| | - M. Angela Nieto
- Instituto de Neurociencias de Alicante, CSIC-UMH, 03550 Sant Joan d'Alacant, Spain
| | - Francisco Portillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas `Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| | - Amparo Cano
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas `Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| |
Collapse
|
26
|
Itahana Y, Piens M, Fong S, Singh J, Sumida T, Desprez PY. Expression of Id and ITF-2 genes in the mammary gland during pregnancy. Biochem Biophys Res Commun 2008; 372:826-30. [PMID: 18519032 DOI: 10.1016/j.bbrc.2008.05.139] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Accepted: 05/22/2008] [Indexed: 10/22/2022]
Abstract
Id-1 is one of the four related helix-loop-helix Id proteins that act as inhibitors of the basic helix-loop-helix (bHLH) transcription factors that control cell differentiation, development and carcinogenesis. We previously found that Id-1 regulated the growth, differentiation, apoptosis and invasion of mouse mammary epithelial cells in culture. Using the techniques of immunohistochemistry and in situ hybridization, we now show that Id-1 gene expression is specifically detected in the epithelial cells of growing ductal structures during early pregnancy. Using adjacent sections, we determined that Id-1 was expressed in keratin 8/18 positive cells. We also demonstrated that the up-regulation of Id-2 during late pregnancy correlated with the down-regulation of Id-1. Using the yeast-two hybrid system, we identified the bHLH transcription factors, ITF-2A and ITF-2B, as the Id-interacting proteins. The levels of expression of these two splice variants did not change during the transition from growing ductal structures to differentiated alveoli. Therefore Id-1 and Id-2, but not the ubiquitous bHLH proteins, appear to represent the key factors whose expression is modulated during different stages of pregnancy in mouse mammary glands.
Collapse
Affiliation(s)
- Yoko Itahana
- California Pacific Medical Center, Cancer Research Institute, 475 Brannan Street, Suite 220, San Francisco, CA 94107, USA
| | | | | | | | | | | |
Collapse
|
27
|
Moreno-Bueno G, Cubillo E, Sarrió D, Peinado H, Rodríguez-Pinilla SM, Villa S, Bolós V, Jordá M, Fabra A, Portillo F, Palacios J, Cano A. Genetic profiling of epithelial cells expressing E-cadherin repressors reveals a distinct role for Snail, Slug, and E47 factors in epithelial-mesenchymal transition. Cancer Res 2007; 66:9543-56. [PMID: 17018611 DOI: 10.1158/0008-5472.can-06-0479] [Citation(s) in RCA: 248] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The transcription factors Snail, Slug, and bHLH E47 have been recently described as direct repressors of E-cadherin and inducers of epithelial-mesenchymal transition (EMT) and invasion when overexpressed in epithelial cells. Although a role of those factors in tumor progression and invasion has been proposed, whether the different repressors play distinct or redundant roles in the tumorigenic process has not been established. To further investigate this important issue, we have analyzed the gene expression profiling of Madin-Darby canine kidney (MDCK) epithelial cells expressing the different repressors (MDCK-Snail, MDCK-Slug, and MDCK-E47 cells) versus control MDCK cells by cDNA microarrays. A total of 243 clones (228 genes and 15 expressed sequence tags) were found to be differentially expressed between either of the three MDCK-derived cell lines and control MDCK cells. Twenty two of the candidate genes were validated by Northern blot, Western blot, immunofluorescence, and promoter analyses in cell lines and by immunohistochemistry in xenografted tumors. Gene clustering analysis indicated that about a third of the 243 candidate genes were common to MDCK cells expressing Snail, Slug, or E47 factors, whereas the rest of the genes were regulated in only one or two cell types. Differentially regulated genes include those related to EMT (45 genes), transcriptional regulation (18 genes), cell proliferation and signaling (54 genes), apoptosis (12 genes), and angiogenesis (9 genes). These results indicate that Snail, Slug, and E47 transcription factors induce common and specific genetic programs, supporting a differential role of the factors in tumor progression and invasion.
Collapse
Affiliation(s)
- Gema Moreno-Bueno
- Breast and Gynecological Cancer Group, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Laursen KB, Mielke E, Iannaccone P, Füchtbauer EM. Mechanism of transcriptional activation by the proto-oncogene Twist1. J Biol Chem 2007; 282:34623-33. [PMID: 17893140 DOI: 10.1074/jbc.m707085200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian Twist1, a master regulator in development and a key factor in tumorigenesis, is known to repress transcription by several mechanisms and is therefore considered to mediate its function mainly through inhibition. A role of Twist1 as transactivator has also been reported but, so far, without providing a mechanism for such an activity. Here we show that heterodimeric complexes of Twist1 and E12 mediate E-box-dependent transcriptional activation. We identify a novel Twist1 transactivation domain that coactivates together with the less potent E12 transactivation domain. We found three specific residues in the highly conserved WR domain to be essential for the transactivating function of murine Twist1 and suggest an alpha-helical structure of the transactivation domain.
Collapse
|
29
|
Muir T, Sadler-Riggleman I, Stevens JD, Skinner MK. Role of the basic helix-loop-helix protein ITF2 in the hormonal regulation of Sertoli cell differentiation. Mol Reprod Dev 2007; 73:491-500. [PMID: 16425294 DOI: 10.1002/mrd.20397] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Sertoli cells are a post-mitotic terminally differentiated cell population that forms the seminiferous tubules in the adult testis and provides the microenvironment and structural support for developing germ cells. During pubertal development, Sertoli cells are responsive to follicle-stimulating hormone (FSH) to promote the expression of differentiated gene products. The basic helix-loop-helix (bHLH) and inhibitors of differentiation (Id) transcription factors are involved in the differentiation of a variety of cell lineages during development. Both bHLH and Id transcription factors have been identified in Sertoli cells. A yeast two-hybrid screen was conducted using a rat Sertoli cell cDNA library to identify bHLH dimerization partners for the Id1 transcription factor. The ubiquitous bHLH protein ITF2 (i.e., E2-2) was identified as one of the interacting partners. The current study investigates the expression and function of ITF2 in Sertoli cells. ITF2 was found to be ubiquitously expressed in all testicular cell types including germ cells, peritubular myoid cells, and Sertoli cells. Stimulation of cultured Sertoli cells with FSH or dibutryl cAMP resulted in a transient decrease in expression of ITF2 mRNA levels followed by a rise in expression with FSH treatment. ITF2 expression was at its highest in mid-pubertal 20-day-old rat Sertoli cells. ITF2 was found to directly bind to negative acting Id HLH proteins and positive acting bHLH proteins such as scleraxis. Transient overexpression of ITF2 protein in cultured Sertoli cells stimulated transferrin promoter activity, which is a marker of Sertoli cell differentiation. Co-transfections of ITF2 and Id proteins sequestered the inhibitory effects of the Id family of proteins. Observations suggest ITF2 can enhance FSH actions through suppressing the inhibitory actions of the Id family of proteins and increasing the actions of stimulatory bHLH proteins (i.e., scleraxis) in Sertoli cells.
Collapse
Affiliation(s)
- Terla Muir
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-4231, USA
| | | | | | | |
Collapse
|
30
|
Wang D, Claus CL, Vaccarelli G, Braunstein M, Schmitt TM, Zúñiga-Pflücker JC, Rothenberg EV, Anderson MK. The basic helix-loop-helix transcription factor HEBAlt is expressed in pro-T cells and enhances the generation of T cell precursors. THE JOURNAL OF IMMUNOLOGY 2006; 177:109-19. [PMID: 16785505 DOI: 10.4049/jimmunol.177.1.109] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The basic helix-loop-helix (bHLH) transcription factors HEB and E2A are critical mediators of gene regulation during lymphocyte development. We have cloned a new transcription factor, called HEBAlt, from a pro-T cell cDNA library. HEBAlt is generated by alternative transcriptional initiation and splicing from the HEB gene locus, which also encodes the previously characterized E box protein HEBCan. HEBAlt contains a unique N-terminal coding exon (the Alt domain) that replaces the first transactivation domain of HEBCan. Downstream of the Alt domain, HEBAlt is identical to HEBCan, including the DNA binding domain. HEBAlt is induced in early thymocyte precursors and down-regulated permanently at the double negative to double positive (DP) transition, whereas HEBCan mRNA expression peaks at the DP stage of thymocyte development. HEBAlt mRNA is up-regulated synergistically by a combination of HEBCan activity and Delta-Notch signaling. Retroviral transduction of HEBAlt or HEBCan into hemopoietic stem cells followed by OP9-DL1 coculture revealed that HEBAlt-transduced precursors generated more early T lineage precursors and more DP pre-T cells than control transduced cells. By contrast, HEBCan-transduced cells that maintained high level expression of the HEBCan transgene were inhibited in expansion and progression through T cell development. HEB(-/-) fetal liver precursors transduced with HEBAlt were rescued from delayed T cell specification, but HEBCan-transduced HEB(-/-) precursors were not. Therefore, HEBAlt and HEBCan are functionally distinct transcription factors, and HEBAlt is specifically required for the efficient generation of early T cell precursors.
Collapse
Affiliation(s)
- Duncheng Wang
- Sunnybrook Research Institute, and Department of Immunology, University of Toronto, 2075 Bayview Avenue, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Lu Y, Sheng DQ, Mo ZC, Li HF, Wu NH, Shen YF. A negative regulatory element-dependent inhibitory role of ITF2B on IL-2 receptor alpha gene. Biochem Biophys Res Commun 2005; 336:142-9. [PMID: 16126178 DOI: 10.1016/j.bbrc.2005.08.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2005] [Accepted: 08/05/2005] [Indexed: 11/23/2022]
Abstract
Despite the fact that the negative regulatory element (NRE) within the upstream regulatory region of human IL-2 receptor alpha (IL-2Ralpha) gene has been identified two decades ago, mechanisms of the NRE function on the gene are hitherto unknown. In this paper, we report for the first time that the immunoglobulin transcription factor 2B (ITF2B) encoded by transcription factor 4 (TCF4) gene is a NRE binding protein. The full-length TCF4 cDNA clone was obtained from a HTLV-1 transformed human peripheral T cell MACHERMAKER cDNA library with NRE as the bait in yeast one-hybrid system. The NRE binding ability of ITF2B was further confirmed in chromatin-immunoprecipitation assay. Competitive RT-PCR-based promoter activity assay showed that over-expression of ITF2B protein inhibited the expression of IL-2Ralpha gene in Jurkat cells in an NRE-dependent manner. The function of ITF2B on the inhibition of both the IL-2Ralpha and the 5'LTR activity of HIV-1 shed light on the essence of NRE binding protein as a potential target for immune therapy and treatment in AIDS patients.
Collapse
Affiliation(s)
- Yu Lu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | | | | | | | | | | |
Collapse
|
32
|
Muir T, Sadler-Riggleman I, Skinner MK. Role of the basic helix-loop-helix transcription factor, scleraxis, in the regulation of Sertoli cell function and differentiation. Mol Endocrinol 2005; 19:2164-74. [PMID: 15831523 DOI: 10.1210/me.2004-0473] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Sertoli cells are a postmitotic terminally differentiated cell population in the adult testis that form the seminiferous tubules and provide the microenvironment and structural support for developing germ cells. The transcription factors that regulate Sertoli cell differentiation remain to be elucidated. The basic helix-loop-helix transcription factors are involved in the differentiation of a variety of cell lineages during development and are expressed in pubertal Sertoli cells. A yeast-two-hybrid procedure was used to screen a Sertoli cell library from 20-d-old pubertal rats to identify dimerization partners with the ubiquitous E47 basic helix-loop-helix transcription factor. Scleraxis was identified as one of the interacting partners. Among the cell types of the testis, scleraxis expression was found to be specific to Sertoli cells. Analysis of the expression pattern of scleraxis mRNA in developing Sertoli cells revealed an increase in scleraxis message at the onset of puberty. Sertoli cells respond to FSH to promote expression of differentiated gene products such as transferrin that aid in proper development of the germ cells. Analysis of the hormonal regulation of scleraxis expression revealed a 4-fold increase in scleraxis mRNA in response to the presence of FSH or dibutryl cAMP in cultured Sertoli cells. An antisense oligonucleotide procedure and overexpression analysis were used to determine whether scleraxis regulates the expression of Sertoli cell differentiated gene products. An antisense oligonucleotide to scleraxis down-regulated transferrin promoter activity in Sertoli cells. A transient overexpression of scleraxis in Sertoli cells stimulated transferrin and androgen binding protein promoter activities and the expression of a number of differentiated genes. Observations suggest scleraxis functions in a number of adult tissues and is involved in the regulation and maintenance of Sertoli cell function and differentiation. This is one of the first adult and nontendon/chondrocyte-associated functions described for scleraxis.
Collapse
Affiliation(s)
- Tera Muir
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-4231, USA
| | | | | |
Collapse
|
33
|
Hikima JI, Cioffi CC, Middleton DL, Wilson MR, Miller NW, Clem LW, Warr GW. Evolution of Transcriptional Control of theIgHLocus: Characterization, Expression, and Function of TF12/HEB Homologs of the Catfish. THE JOURNAL OF IMMUNOLOGY 2004; 173:5476-84. [PMID: 15494495 DOI: 10.4049/jimmunol.173.9.5476] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The transcriptional enhancer (Emu3') of the IgH locus of the channel catfish, Ictalurus punctatus, differs from enhancers of the mammalian IgH locus in terms of its position, structure, and function. Transcription factors binding to multiple octamer motifs and a single muE5 motif (an E-box site, consensus CANNTG) interact for its function. E-box binding transcription factors of the class I basic helix-loop-helix family were cloned from a catfish B cell cDNA library in this study, and homologs of TF12/HEB were identified as the most highly represented E-proteins. Two alternatively spliced forms of catfish TF12 (termed CFEB1 and -2) were identified and contained regions homologous to the basic helix-loop-helix and activation domains of other vertebrate E-proteins. CFEB message is widely expressed, with CFEB1 message predominating over that of CFEB2. Both CFEB1 and -2 strongly activated transcription from a muE5-dependent artificial promoter. In catfish B cells, CFEB1 and -2 also activated transcription from the core region of the catfish IgH enhancer (Emu3') in a manner dependent on the presence of the muE5 site. Both CFEB1 and -2 bound the muE5 motif, and formed both homo- and heterodimers. CFEB1 and -2 were weakly active or inactive (in a promoter-dependent fashion) in mammalian B-lineage cells. Although E-proteins have been highly conserved in vertebrate evolution, the present results indicate that, at the phylogenetic level of a teleost fish, the TF12/HEB homolog differs from that of mammals in terms of 1) its high level of expression and 2) the presence of isoforms generated by alternative RNA processing.
Collapse
Affiliation(s)
- Jun-Ichi Hikima
- Center for Marine Biomedicine and Environmental Sciences, and Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29407, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Kaput J, Klein KG, Reyes EJ, Kibbe WA, Cooney CA, Jovanovic B, Visek WJ, Wolff GL. Identification of genes contributing to the obese yellow Avy phenotype: caloric restriction, genotype, diet x genotype interactions. Physiol Genomics 2004; 18:316-24. [PMID: 15306695 DOI: 10.1152/physiolgenomics.00065.2003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The incidence and severity of obesity and type 2 diabetes are increasing in Western societies. The progression of obesity to type 2 diabetes is gradual with overlapping symptoms of insulin resistance, hyperinsulinemia, hyperglycemia, dyslipidemias, ion imbalance, and inflammation; this complex syndrome has been called diabesity. We describe here comparisons of gene expression in livers of A/a (agouti) vs. A(vy)/A (obese yellow) segregants (i.e., littermates) from BALB/cStCrlfC3H/Nctr x VYWffC3Hf/Nctr-A(vy)/a matings in response to 70% and 100% of ad libitum caloric intakes of a reproducible diet. Twenty-eight (28) genes regulated by diet, genotype, or diet x genotype interactions mapped to diabesity quantitative trait loci. A subset of the identified genes is linked to abnormal physiological signs observed in obesity and diabetes.
Collapse
Affiliation(s)
- Jim Kaput
- University of California at Davis, Davis, California 95616, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Liu YP, Burleigh D, Durning M, Hudson L, Chiu IM, Golos TG. Id2 is a primary partner for the E2-2 basic helix-loop-helix transcription factor in the human placenta. Mol Cell Endocrinol 2004; 222:83-91. [PMID: 15249128 DOI: 10.1016/j.mce.2004.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Accepted: 04/27/2004] [Indexed: 10/26/2022]
Abstract
We screened a term placental cDNA library by the yeast two-hybrid approach with Id2, a negative regulator of basic helix-loop-helix (bHLH) factors. Of the clones obtained, approximately one-third were the E2-2 bHLH transcription factor. Id2 and E2-2 were shown to interact in direct two-hybrid assays in yeast cells, as well as immunoprecipitation assays in mammalian cells. Immunohistochemical analysis demonstrated co-localization of both Id2 and E2-2 in placental trophoblasts. Co-transfection of JEG-3 cells with E2-2 and Id2, and a luciferase reporter construct under the control of the human chorionic gonadotropin alpha-subunit promoter revealed that E2-2 had a negative effect on CGalpha-subunit transcription, which could be relieved by overexpression of Id2. The library was in turn rescreened with E2-2, and Id2 and Id1 were essentially the only clones obtained. We conclude that Id2 is a primary binding partner for the bHLH transcription factor E2-2 in the human placenta.
Collapse
Affiliation(s)
- Yi-Ping Liu
- Wisconsin National Primate Research Center and the Department of Obstetrics and Gynecology, University of Wisconsin Medical School, Madison, WI 53715, USA
| | | | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- Xiao-Hong Sun
- Immunobiology and Cancer Program, Oklahoma Medical Research Foundation Oklahoma City, OK 73104, USA
| |
Collapse
|
37
|
Desprez PY, Sumida T, Coppé JP. Helix-loop-helix proteins in mammary gland development and breast cancer. J Mammary Gland Biol Neoplasia 2003; 8:225-39. [PMID: 14635797 DOI: 10.1023/a:1025957025773] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The basic helix-loop-helix (bHLH) family of transcription factors functions in the coordinated regulation of gene expression, cell lineage commitment, and cell differentiation in most mammalian tissues. Helix-loop-helix Id (Inhibitor of DNA binding) proteins are distinct from bHLH transcription factors in that they lack the basic domain necessary for DNA binding. Id proteins thus function as dominant negative regulators of bHLH transcription factors. The inhibition of bHLH factor activity by forced constitutive expression of Id proteins is closely associated with the inhibition of differentiation in a number of different cell types, including mammary epithelial cells. Moreover, recent literature suggests important roles of HLH proteins in many normal and transformed tissues, including mammary gland. Therefore, future directions for prognosis or therapeutic treatments of breast cancer may be able to exploit bHLH and Id genes as useful molecular targets. The purpose of this review is to summarize the evidence implicating HLH proteins in the regulation of normal and transformed mammary epithelial cell phenotypes.
Collapse
Affiliation(s)
- Pierre-Yves Desprez
- California Pacific Medical Center, Cancer Research Institute, San Francisco, California 94115, USA.
| | | | | |
Collapse
|
38
|
Wolff GL. Regulation of yellow pigment formation in mice: a historical perspective. PIGMENT CELL RESEARCH 2003; 16:2-15. [PMID: 12519120 DOI: 10.1034/j.1600-0749.2003.00012.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Pigment synthesis by hair follicle melanocytes is modulated by a large number of environmental and genetic factors, many of which are discussed in this review. Eumelanic (non-yellow) pigment is produced by hair follicle melanocytes following the binding of alpha-melanocyte stimulating hormone to melanocortin receptor 1. Binding of this hormone to the melanocyte membrane is blocked by agouti signaling protein (ASP) which is encoded by the agouti locus and results in the synthesis of yellow pigment, instead of non-yellow (black/brown) pigment. The cyclical release of ASP by hair follicle cells results in a black/brown hair with a subapical yellow band. This is the wild-type coat color pattern of many mammals and is called agouti. Several dominant mutations at the agouti locus in mice, induced by retrotransposon-like intracisternal A particles, result in ectopic over-expression of ASP and animals with much higher proportions of all-yellow hairs. This abnormal presence of ASP in essentially all body cells results in the 'yellow agouti obese mouse syndrome.' The obesity has been associated with binding of ASP to melanocortin receptor 4 inactivating the latter. The syndrome also includes hyperinsulinemia, increased somatic growth, and increased susceptibility to hyperplasia and carcinogenesis. The physiologic and molecular bases for these syndrome components have not yet been elucidated. This historically orientated review is subdivided, where applicable, into pre- and post-1992 subsections to emphasize the impact of the cloning of the agouti and extension loci and their protein products on the identification of the molecular and physiological pathways modulating the manifold aspects of pheomelanogenesis.
Collapse
Affiliation(s)
- George L Wolff
- Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration, Jefferson, AR 72079, USA.
| |
Collapse
|
39
|
Spinner DS, Liu S, Wang SW, Schmidt J. Interaction of the myogenic determination factor myogenin with E12 and a DNA target: mechanism and kinetics. J Mol Biol 2002; 317:431-45. [PMID: 11922675 DOI: 10.1006/jmbi.2002.5440] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The myogenic determination factors MyoD, myogenin, myf5, and MRF4 are members of the basic helix-loop-helix (bHLH) family of transcription factors and crucial agents of myogenesis. The bHLH regions of these proteins enable them to dimerize with E proteins, another class of the bHLH family, and to bind a specific DNA element known as an E box (CANNTG consensus sequence), which results in the activation of muscle-specific gene expression. As a model for such assembly of the myogenic determination factor/E protein-DNA ternary complex, we have studied the physiologically relevant association of myogenin, E12, and the 3' E box of the acetylcholine receptor (AChR) alpha-subunit gene enhancer. Using the technique of electrophoretic mobility shift assay combined with order-of-addition and time-course experiments, we find that heterodimerization of myogenin with E12 occurs prior to DNA-binding. In addition, we deduce the dissociation (Kd) and rate (k) constants for each step in the formation of the myogenin/E12-DNA ternary complex. Kinetic simulations indicate that at 37 degrees C myogenin and E12 heterodimerize with a Kd of 36 microM (k(on) of 573 M(-1) x s(-1) and k(off )of 0.0205 x s(-1)), and that subsequently the heterodimer binds the AChR alpha-subunit gene enhancer 3' E box with a Kd of 8.8 nM (with possible k(on) and k(off) values ranging from 1.0x10(8) to 14.1x10(8) M(-1) x s(-1), and 0.875 to 12.3 s(-1), respectively).
Collapse
Affiliation(s)
- Daryl S Spinner
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook 11794-5215, USA
| | | | | | | |
Collapse
|
40
|
Kolligs FT, Nieman MT, Winer I, Hu G, Van Mater D, Feng Y, Smith IM, Wu R, Zhai Y, Cho KR, Fearon ER. ITF-2, a downstream target of the Wnt/TCF pathway, is activated in human cancers with beta-catenin defects and promotes neoplastic transformation. Cancer Cell 2002; 1:145-55. [PMID: 12086873 DOI: 10.1016/s1535-6108(02)00035-1] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In many cancers, inactivation of the adenomatous polyposis coli (APC) or Axin tumor suppressor proteins or activating mutations in beta-catenin lead to elevated beta-catenin levels, enhanced binding of beta-catenin to T cell factor (TCF) proteins, and increased expression of TCF-regulated genes. We found that the gene for the basic helix-loop-helix transcription factor ITF-2 (immunoglobulin transcription factor-2) was activated in rat E1A-immortalized RK3E cells following neoplastic transformation by beta-catenin or ligand-induced activation of a beta-catenin-estrogen receptor fusion protein. Human cancers with beta-catenin regulatory defects had elevated ITF-2 expression, and ITF-2 was repressed by restoring wild-type APC function or inhibiting TCF activity. Of note, ITF-2 promoted neoplastic transformation of RK3E cells. We propose that ITF-2 is a TCF-regulated gene, which functions in concert with other TCF target genes to promote growth and/or survival of cancer cells with defects in beta-catenin regulation.
Collapse
Affiliation(s)
- Frank T Kolligs
- Department of Internal Medicine, Division of Medical Genetics and the Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Dhulipala PD, Lianos EA, Kotlikoff MI. Regulation of human P2X1 promoter activity by beta helix-loop-helix factors in smooth muscle cells. Gene 2001; 269:167-75. [PMID: 11376948 DOI: 10.1016/s0378-1119(01)00442-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We isolated and characterized genomic clones of the human P2X1 receptor (hP2X1) gene in an effort to understand its tissue specific expression. The hP2X1 gene contains 12 exons spanning 20 kb, with exon sizes ranging from 59 to 143 bp. A 385 bp upstream fragment promoted hP2X1 gene expression in smooth muscle (A7R5 and primary trachealis) and fibroblast (NIH3T3) cell lines, and mutation of a consensus E box sequence (CACCTG) within this fragment (-340 to -345) did not alter basal promoter activity. However, co-transfected bHLH factors regulated activity of the 385 bp minimal P2X1 promoter in a tissue-specific manner. E12 expression inhibited and ITF2b augmented activity in A7R5 cells, but had no effect in NIH3T3 cells. ITF2a, Myo-D, and Id1 proteins had no effect on either cell line, but co-expression of ITF2a blocked E12 inhibition in A7R5 cells, while ITF2b failed to reverse the inhibition. Northern analysis of A7R5 RNA identified high levels of E12 and ITF2b transcripts, and gel shift assays using A7R5 and NIH3T3 nuclear extracts indicated the formation of a protein-DNA complex with an oligonucleotide corresponding to -330 and -348, which was abolished by base substitutions within the E box motif. Our results identify a critical E box response element in the hP2X1 promoter that binds bHLH factors and demonstrate smooth muscle specific transcriptional regulation by E proteins.
Collapse
MESH Headings
- 3T3 Cells
- Amino Acid Sequence
- Animals
- Base Sequence
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors
- Binding Sites
- Cloning, Molecular
- DNA, Complementary
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Gene Expression Profiling
- Gene Expression Regulation
- Helix-Loop-Helix Motifs
- Humans
- Inhibitor of Differentiation Protein 1
- Mice
- Molecular Sequence Data
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- MyoD Protein/genetics
- MyoD Protein/metabolism
- Nerve Tissue Proteins
- Promoter Regions, Genetic
- Receptors, Purinergic P2/genetics
- Receptors, Purinergic P2X
- Repressor Proteins
- TCF Transcription Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factor 4
- Transcription Factor 7-Like 1 Protein
- Transcription Factor 7-Like 2 Protein
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- P D Dhulipala
- Department of Medicine, Division of Nephrology, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | | | | |
Collapse
|
42
|
Furumura M, Potterf SB, Toyofuku K, Matsunaga J, Muller J, Hearing VJ. Involvement of ITF2 in the transcriptional regulation of melanogenic genes. J Biol Chem 2001; 276:28147-54. [PMID: 11382753 DOI: 10.1074/jbc.m101626200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In response to agouti signal protein, melanocytes switch from producing eumelanin to pheomelanin concomitant with the down-regulation of melanogenic gene transcription. We previously reported that a ubiquitous basic helix-loop-helix transcription factor, known as ITF2, is up-regulated during this switch, and we now report that treatment of melanocytes with melanocyte-stimulating hormone down-regulates expression of ITF2. To more fully characterize the involvement of ITF2 in regulating melanogenic gene transcription, ITF2 sense or antisense constructs were introduced into melan-a melanocytes. Gene and protein expression analyses and luciferase reporter assays using promoters from melanogenic genes showed that up-regulation of ITF2 suppressed melanogenic gene expression as well as the expression of Mitf, a melanocyte-specific transcription factor. In addition, stable ITF2 sense transfectants had significant reductions in pigmentation and a less dendritic phenotype compared with mock transfectants. In contrast, ITF2 antisense-transfected melanocytes were more pigmented and more dendritic. These results demonstrate that up-regulation of ITF2 during the pheomelanin switch is functionally significant and reveal that differential expression of a ubiquitous basic helix-loop-helix transcription factor can modulate expression of melanogenic genes and the differentiation of melanocytes.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors
- Blotting, Northern
- Cell Differentiation
- Cyclic AMP/metabolism
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/physiology
- Dendritic Cells/metabolism
- Down-Regulation
- Genes, Reporter
- Helix-Loop-Helix Motifs
- Luciferases/metabolism
- MART-1 Antigen
- Melanins/metabolism
- Melanocytes/metabolism
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Microscopy, Electron
- Models, Biological
- Neoplasm Proteins/metabolism
- Nerve Tissue Proteins
- Oligonucleotides, Antisense/metabolism
- Phenotype
- Plasmids/metabolism
- Precipitin Tests
- Promoter Regions, Genetic
- Protein Isoforms
- RNA, Messenger/metabolism
- Ribonucleases/metabolism
- TCF Transcription Factors
- Trans-Activators/chemistry
- Trans-Activators/physiology
- Transcription Factor 4
- Transcription Factors/metabolism
- Transcription, Genetic
- Transfection
- Up-Regulation
Collapse
Affiliation(s)
- M Furumura
- Pigment Cell Biology Section, Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
43
|
Liu J, Beqaj S, Yang Y, Honoré B, Schuger L. Heterogeneous nuclear ribonucleoprotein-H plays a suppressive role in visceral myogenesis. Mech Dev 2001; 104:79-87. [PMID: 11404082 DOI: 10.1016/s0925-4773(01)00377-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mouse embryonic mesenchymal cells undergo spontaneous smooth muscle (SM) differentiation upon spreading/elongation in culture (Relan et al., J. Cell Biol. 147 (1999) 1341; Yang et al., Development 125 (1998) 2621; Yang et al., Development 126 (1999) 3027). Using these cells we generated a subtracted cDNA library to identify potential suppressors of SM myogenesis. One of the differentially expressed genes was heterogeneous nuclear ribonucleoprotein-H (hnRNP-H), which is involved in pre-mRNA alternative splicing. hnRNP-H was highly expressed in mesenchymal cells prior to the onset of SM differentiation, but its expression rapidly decreased in mesenchymal cells undergoing SM myogenesis. In vivo, the drop in hnRNP-H expression was restricted to visceral SM cells. Antisense oligodeoxynucleotide and antisense RNA were used to inhibit hnRNP-H synthesis in SM-differentiating mesenchymal cells and in embryonic lung explants. A decrease in hnRNP-H levels resulted in upregulation of SM-specific gene expression and increased bronchial SM development in lung explants. hnRNP-H overexpression in cell cultures had the opposite effect. These studies, therefore, indicate a novel role for hnRNP-H in the control of visceral myogenesis.
Collapse
Affiliation(s)
- J Liu
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
44
|
Ridgeway AG, Petropoulos H, Wilton S, Skerjanc IS. Wnt signaling regulates the function of MyoD and myogenin. J Biol Chem 2000; 275:32398-405. [PMID: 10915791 DOI: 10.1074/jbc.m004349200] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The myogenic regulatory factors (MRFs), MyoD and myogenin, can induce myogenesis in a variety of cell lines but not efficiently in monolayer cultures of P19 embryonal carcinoma stem cells. Aggregation of cells expressing MRFs, termed P19[MRF] cells, results in an approximately 30-fold enhancement of myogenesis. Here we examine molecular events occurring during P19 cell aggregation to identify potential mechanisms regulating MRF activity. Although myogenin protein was continually present in the nuclei of >90% of P19[myogenin] cells, only a fraction of these cells differentiated. Consequently, it appears that post-translational regulation controls myogenin activity in a cell lineage-specific manner. A correlation was obtained between the expression of factors involved in somite patterning, including Wnt3a, Wnt5b, BMP-2/4, and Pax3, and the induction of myogenesis. Co-culturing P19[Wnt3a] cells with P19[MRF] cells in monolayer resulted in a 5- to 8-fold increase in myogenesis. Neither BMP-4 nor Pax3 was efficient in enhancing MRF activity in unaggregated P19 cultures. Furthermore, BMP-4 abrogated the enhanced myogenesis induced by Wnt signaling. Consequently, signaling events resulting from Wnt3a expression but not BMP-4 signaling or Pax3 expression, regulate MRF function. Therefore, the P19 cell culture system can be used to study the link between somite patterning events and myogenesis.
Collapse
Affiliation(s)
- A G Ridgeway
- Department of Biochemistry, Medical Sciences Building, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | | | | | |
Collapse
|
45
|
Petropoulos H, Skerjanc IS. Analysis of the inhibition of MyoD activity by ITF-2B and full-length E12/E47. J Biol Chem 2000; 275:25095-101. [PMID: 10833525 DOI: 10.1074/jbc.m004251200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
MyoD heterodimerizes with E type factors (E12/E47 and ITF-2A/ITF-2B) and binds E box sequences within promoters of muscle-specific genes. In transient transfection assays, MyoD activates transcription in the presence of ITF-2A but not ITF-2B, which contains a 182-amino acid N-terminal extension. The first 83 amino acids of the inhibitory N terminus of ITF-2B show high sequence homology to the N terminus of full-length E12/E47. Previous studies that showed activation of MyoD by E12 used an artificially N-terminally truncated form. Here we show that the full-length form of E12 inhibits MyoD function. A conserved alpha-helix motif, capable of interacting with the transcriptional machinery, was not essential for inhibition. Furthermore, the fusion of N-terminal ITF-2B sequences or non-inhibiting ITF-2A sequences to truncated E12 was sufficient in converting the activator into an inhibitor. Overexpression of ITF-2B did not inhibit C2C12 myogenesis or affect levels of endogenous muscle gene expression, consistent with the finding that inhibitory E type proteins are present in muscle. Furthermore, we found that MyoD co-transfected with either ITF-2B or ITF-2A converted fibroblasts into myoblasts with the same frequency. Our findings suggest that the ability of E type proteins to inhibit MyoD activity is dependent on the context of the E box.
Collapse
Affiliation(s)
- H Petropoulos
- Department of Biochemistry, University of Western Ontario, London, Canada
| | | |
Collapse
|
46
|
Persson P, Jögi A, Grynfeld A, Påhlman S, Axelson H. HASH-1 and E2-2 are expressed in human neuroblastoma cells and form a functional complex. Biochem Biophys Res Commun 2000; 274:22-31. [PMID: 10903890 DOI: 10.1006/bbrc.2000.3090] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The basic helix-loop-helix (bHLH) transcription factor mammalian achaete-scute homolog-1 (MASH-1 in mouse and HASH-1 in human) is essential for proper development of olfactory and most peripheral autonomic neurons, and for the formation of distinct neuronal circuits within the central nervous system. We have previously shown that HASH-1 is expressed in neuroblastoma tumors and cell lines, and in this study we have used the yeast two-hybrid system to isolate HASH-1 interacting proteins from a human neuroblastoma cDNA library. Two of the isolated clones contained cDNA from the E2-2 gene (also known as ITF2/SEF2-1). We show that E2-2 interacts with HASH-1 in both yeast and mammalian cells. The HASH-1/E2-2 complex binds an E-box (CACCTG) in vitro, and transactivates an E-box containing reporter construct in vivo. Furthermore, E2-2 seems to be one of the major HASH-1 interacting proteins in extracts from neuroblastoma cells. In conclusion, E2-2 forms a functional complex with HASH-1, and might therefore be involved in the development of specific parts of the central and peripheral nervous systems.
Collapse
Affiliation(s)
- P Persson
- Department of Laboratory Medicine, Lund University, Malmö, University Hospital MAS, S-205 02, Sweden
| | | | | | | | | |
Collapse
|
47
|
Anderson MK, Rothenberg EV. Transcription factor expression in lymphocyte development: clues to the evolutionary origins of lymphoid cell lineages? Curr Top Microbiol Immunol 2000; 248:137-55. [PMID: 10793477 DOI: 10.1007/978-3-642-59674-2_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- M K Anderson
- Department of Biology, California Institute of Technology, Pasadena 91125, USA
| | | |
Collapse
|
48
|
Lin F, Blake DL, Callebaut I, Skerjanc IS, Holmer L, McBurney MW, Paulin-Levasseur M, Worman HJ. MAN1, an inner nuclear membrane protein that shares the LEM domain with lamina-associated polypeptide 2 and emerin. J Biol Chem 2000; 275:4840-7. [PMID: 10671519 DOI: 10.1074/jbc.275.7.4840] [Citation(s) in RCA: 268] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The "MAN antigens" are polypeptides recognized by autoantibodies from a patient with a collagen vascular disease and localized to the nuclear envelope. We now show that one of the human MAN antigens termed MAN1 is a 82.3-kDa protein with an amino-terminal domain followed by two hydrophobic segments and a carboxyl-terminal tail. The MAN1 gene contains seven protein-coding exons and is assigned to human chromosome 12q14. Its mRNA is approximately 5.5 kilobases and is detected in several different cell types that were examined. Cell extraction experiments show that MAN1 is an integral membrane protein. When expressed in transfected cells, MAN1 is exclusively targeted to the nuclear envelope, consistent with an inner nuclear membrane localization. Protein sequence analysis reveals that MAN1 shares a conserved globular domain of approximately 40 amino acids, which we term the LEM module, with inner nuclear membrane proteins lamina-associated polypeptide 2 and emerin. The LEM module is also present in two proteins of Caenorhabditis elegans. These results show that MAN1 is an integral protein of the inner nuclear membrane that shares the LEM module with other proteins of this subcellular localization.
Collapse
Affiliation(s)
- F Lin
- Departments of Medicine and of Anatomy and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The development of T cells and B cells from pluripotent hematopoietic precursors occurs through a stepwise narrowing of developmental potential that ends in lineage commitment. During this process, lineage-specific genes are activated asynchronously, and lineage-inappropriate genes, although initially expressed, are asynchronously turned off. These complex gene expression events are the outcome of the changes in expression of multiple transcription factors with partially overlapping roles in early lymphocyte and myeloid cell development. Key transcription factors promoting B-cell development and candidates for this role in T-cell development are discussed in terms of their possible modes of action in fate determination. We discuss how a robust, stable, cell-type-specific gene expression pattern may be established in part by the interplay between endogenous transcription factors and signals transduced by cytokine receptors, and in part by the network of effects of particular transcription factors on each other.
Collapse
Affiliation(s)
- E V Rothenberg
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125, USA.
| | | | | |
Collapse
|
50
|
Ueba T, Kaspar B, Zhao X, Gage FH. Repression of human fibroblast growth factor 2 by a novel transcription factor. J Biol Chem 1999; 274:10382-7. [PMID: 10187827 DOI: 10.1074/jbc.274.15.10382] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Here we describe the cloning of the regulator of fibroblast growth factor 2 (FGF-2) transcription (RFT) using a yeast one-hybrid screening with a defined motif in FGF-2 promoter as a target sequence. Overexpression of human RFT (RFT-A) reduces FGF-2 RNA and protein levels in both normal and tumor cell lines. Its splice variants, RFT-A' and RFT-B, have deletions in the putative DNA binding domain and fail to bind FGF-2 promoter and repress FGF-2 gene expression. The ratios of RFT isoforms differ between normal and tumor cells, with the splice variants dominating in tumor cells. Overexpression of RFT-A induces glioma cell death. Our data suggest that regulation of FGF-2 by RFT is important for cellular functions and may be impaired in certain tumors.
Collapse
Affiliation(s)
- T Ueba
- Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|