1
|
Zhang H, Zheng J, Fu Y, Ling J, Liu Z, Lin X, Dong X, Sun Y, Tan T, Guo Z, Xie G. Overexpression of POU3F2 promotes radioresistance in triple-negative breast cancer via Akt pathway activation. Breast Cancer Res Treat 2023; 198:437-446. [PMID: 36797433 DOI: 10.1007/s10549-023-06876-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023]
Abstract
PURPOSE POU3F2 is associated with malignant behaviors and poor prognosis in cancer. However, the function and mechanism of POU3F2 in breast cancer remain to be elucidated. Our study aimed to explore the role of POU3F2 in triple-negative breast cancer and radiotherapy. METHODS POU3F2 expression was examined by RT-PCR and Western blot. The proliferation of cancer cells was measured by MTT assay. Migration of cancer cells was determined by Transwell assay and wound healing assay. To determine which protein interacts with POU3F2, Co-IP was performed. Survival analysis was performed based on the online database GEPIA. DNA damage after radiation was examined by Comet Assay. Radiosensitivity was evaluated with clonogenic survival assays. A tumor xenograft model was established with MDA-MB-231 breast cancer cells in BALB/c nude mice to explore the effect of POU3F2 in vivo. RESULTS We found that the expression of POU3F2 was significantly elevated in breast cancer cells, especially in TNBC, and higher POU3F2 expression was related to poor prognosis of patients with breast cancer. Functional assays revealed that POU3F2 promoted proliferation, migration, and invasion of triple-negative breast cancer (TNBC) cells in vitro and in vivo. In addition, the knockdown of POU3F2 decreased the radioresistance of TNBC cells in vitro. Furthermore, POU3F2 could enhance the activation of the Akt pathway by interacting with ARNT2, thereby promoting proliferation and radioresistance in TNBC cells. CONCLUSIONS Our results provide evidence that high expression of POU3F2 promotes radioresistance in triple-negative breast cancer via Akt pathway activation by interacting with ARNT2.
Collapse
Affiliation(s)
- Han Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jieling Zheng
- Department of Radiology, First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yiming Fu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jing Ling
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - ZiShen Liu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xiaotong Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xin Dong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yao Sun
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Tingting Tan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhaoze Guo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Guozhu Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Alternate Roles of Sox Transcription Factors beyond Transcription Initiation. Int J Mol Sci 2021; 22:ijms22115949. [PMID: 34073089 PMCID: PMC8198692 DOI: 10.3390/ijms22115949] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/19/2022] Open
Abstract
Sox proteins are known as crucial transcription factors for many developmental processes and for a wide range of common diseases. They were believed to specifically bind and bend DNA with other transcription factors and elicit transcriptional activation or repression activities in the early stage of transcription. However, their functions are not limited to transcription initiation. It has been showed that Sox proteins are involved in the regulation of alternative splicing regulatory networks and translational control. In this review, we discuss the current knowledge on how Sox transcription factors such as Sox2, Sry, Sox6, and Sox9 allow the coordination of co-transcriptional splicing and also the mechanism of SOX4-mediated translational control in the context of RNA polymerase III.
Collapse
|
3
|
Snijders Blok L, Kleefstra T, Venselaar H, Maas S, Kroes HY, Lachmeijer AMA, van Gassen KLI, Firth HV, Tomkins S, Bodek S, Õunap K, Wojcik MH, Cunniff C, Bergstrom K, Powis Z, Tang S, Shinde DN, Au C, Iglesias AD, Izumi K, Leonard J, Abou Tayoun A, Baker SW, Tartaglia M, Niceta M, Dentici ML, Okamoto N, Miyake N, Matsumoto N, Vitobello A, Faivre L, Philippe C, Gilissen C, Wiel L, Pfundt R, Deriziotis P, Brunner HG, Fisher SE. De Novo Variants Disturbing the Transactivation Capacity of POU3F3 Cause a Characteristic Neurodevelopmental Disorder. Am J Hum Genet 2019; 105:403-412. [PMID: 31303265 PMCID: PMC6698880 DOI: 10.1016/j.ajhg.2019.06.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 06/07/2019] [Indexed: 01/22/2023] Open
Abstract
POU3F3, also referred to as Brain-1, is a well-known transcription factor involved in the development of the central nervous system, but it has not previously been associated with a neurodevelopmental disorder. Here, we report the identification of 19 individuals with heterozygous POU3F3 disruptions, most of which are de novo variants. All individuals had developmental delays and/or intellectual disability and impairments in speech and language skills. Thirteen individuals had characteristic low-set, prominent, and/or cupped ears. Brain abnormalities were observed in seven of eleven MRI reports. POU3F3 is an intronless gene, insensitive to nonsense-mediated decay, and 13 individuals carried protein-truncating variants. All truncating variants that we tested in cellular models led to aberrant subcellular localization of the encoded protein. Luciferase assays demonstrated negative effects of these alleles on transcriptional activation of a reporter with a FOXP2-derived binding motif. In addition to the loss-of-function variants, five individuals had missense variants that clustered at specific positions within the functional domains, and one small in-frame deletion was identified. Two missense variants showed reduced transactivation capacity in our assays, whereas one variant displayed gain-of-function effects, suggesting a distinct pathophysiological mechanism. In bioluminescence resonance energy transfer (BRET) interaction assays, all the truncated POU3F3 versions that we tested had significantly impaired dimerization capacities, whereas all missense variants showed unaffected dimerization with wild-type POU3F3. Taken together, our identification and functional cell-based analyses of pathogenic variants in POU3F3, coupled with a clinical characterization, implicate disruptions of this gene in a characteristic neurodevelopmental disorder.
Collapse
Affiliation(s)
- Lot Snijders Blok
- Human Genetics Department, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, the Netherlands; Language and Genetics Department, Max Planck Institute for Psycholinguistics, PO Box 310, 6500AH Nijmegen, the Netherlands; Donders Institute for Brain, Cognition, and Behaviour, PO Box 9104, 6500HE Nijmegen, the Netherlands.
| | - Tjitske Kleefstra
- Human Genetics Department, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, the Netherlands; Donders Institute for Brain, Cognition, and Behaviour, PO Box 9104, 6500HE Nijmegen, the Netherlands
| | - Hanka Venselaar
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, the Netherlands
| | - Saskia Maas
- Amsterdam University Medical Center, University of Amsterdam, Department of Clinical Genetics, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Hester Y Kroes
- Department of Genetics, University Medical Center Utrecht, PO Box 85090, 3508AB Utrecht, the Netherlands
| | - Augusta M A Lachmeijer
- Department of Genetics, University Medical Center Utrecht, PO Box 85090, 3508AB Utrecht, the Netherlands
| | - Koen L I van Gassen
- Department of Genetics, University Medical Center Utrecht, PO Box 85090, 3508AB Utrecht, the Netherlands
| | - Helen V Firth
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Susan Tomkins
- Clinical Genetics Service, University Hospitals Bristol National Health Service Foundation Trust, Bristol BS2 8HW, UK
| | - Simon Bodek
- Clinical Genetics Service, University Hospitals Bristol National Health Service Foundation Trust, Bristol BS2 8HW, UK
| | - Katrin Õunap
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital and Institute of Clinical Medicine, University of Tartu, Tartu 51014, Estonia; Institute of Clinical Medicine, University of Tartu, Tartu 51014, Estonia
| | - Monica H Wojcik
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Newborn Medicine, Division of Genetics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Christopher Cunniff
- Division of Medical Genetics, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Katherine Bergstrom
- Division of Medical Genetics, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Zoë Powis
- Clinical Genomics, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Sha Tang
- Clinical Genomics, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | | | - Catherine Au
- Division of Clinical Genetics, Department of Pediatrics, New York Presbyterian Hospital, Columbia University, New York, NY 10032, USA
| | - Alejandro D Iglesias
- Division of Clinical Genetics, Department of Pediatrics, New York Presbyterian Hospital, Columbia University, New York, NY 10032, USA
| | - Kosuke Izumi
- Division of Human Genetics, the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jacqueline Leonard
- Division of Human Genetics, the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ahmad Abou Tayoun
- Division of Genomic Diagnostics, the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Samuel W Baker
- Division of Genomic Diagnostics, the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00146 Rome, Italy
| | - Marcello Niceta
- Genetics and Rare Diseases Research Division, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00146 Rome, Italy
| | - Maria Lisa Dentici
- Genetics and Rare Diseases Research Division, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00146 Rome, Italy
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Antonio Vitobello
- UF Innovation en Diagnostic Génomique des Maladies Rares, Centre Hospitalier Universitaire Dijon Bourgogne, 21000 Dijon, France; INSERM UMR1231 Génétique des Anomalies du Développement, F-21000 Dijon, France
| | - Laurence Faivre
- INSERM UMR1231 Génétique des Anomalies du Développement, F-21000 Dijon, France; Centre de Référence Maladies Rares « Anomalies du Développement et Syndrome Malformatifs » de l'Est, Centre de Génétique, Hôpital d'Enfants, Fédération Hospitalo-Universitaire Médecine TRANSLationnelle et Anomalies du Développement, Centre Hospitalier Universitaire Dijon Bourgogne, 21000 Dijon, France
| | - Christophe Philippe
- UF Innovation en Diagnostic Génomique des Maladies Rares, Centre Hospitalier Universitaire Dijon Bourgogne, 21000 Dijon, France; INSERM UMR1231 Génétique des Anomalies du Développement, F-21000 Dijon, France
| | - Christian Gilissen
- Human Genetics Department, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, the Netherlands
| | - Laurens Wiel
- Human Genetics Department, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, the Netherlands; Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, the Netherlands
| | - Rolph Pfundt
- Human Genetics Department, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, the Netherlands
| | - Pelagia Deriziotis
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, PO Box 310, 6500AH Nijmegen, the Netherlands
| | - Han G Brunner
- Human Genetics Department, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, the Netherlands; Donders Institute for Brain, Cognition, and Behaviour, PO Box 9104, 6500HE Nijmegen, the Netherlands; Department of Clinical Genetics and GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, 6202AZ Maastricht, the Netherlands
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, PO Box 310, 6500AH Nijmegen, the Netherlands; Donders Institute for Brain, Cognition, and Behaviour, PO Box 9104, 6500HE Nijmegen, the Netherlands.
| |
Collapse
|
4
|
Herbert K, Binet R, Lambert JP, Louphrasitthiphol P, Kalkavan H, Sesma-Sanz L, Robles-Espinoza CD, Sarkar S, Suer E, Andrews S, Chauhan J, Roberts ND, Middleton MR, Gingras AC, Masson JY, Larue L, Falletta P, Goding CR. BRN2 suppresses apoptosis, reprograms DNA damage repair, and is associated with a high somatic mutation burden in melanoma. Genes Dev 2019; 33:310-332. [PMID: 30804224 PMCID: PMC6411009 DOI: 10.1101/gad.314633.118] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 01/04/2019] [Indexed: 01/04/2023]
Abstract
Herbert et al. show that BRN2 is associated with DNA damage response proteins and suppresses an apoptosis-associated gene expression program to protect against UVB-, chemotherapy-, and vemurafenib-induced apoptosis. Whether cell types exposed to a high level of environmental insults possess cell type-specific prosurvival mechanisms or enhanced DNA damage repair capacity is not well understood. BRN2 is a tissue-restricted POU domain transcription factor implicated in neural development and several cancers. In melanoma, BRN2 plays a key role in promoting invasion and regulating proliferation. Here we found, surprisingly, that rather than interacting with transcription cofactors, BRN2 is instead associated with DNA damage response proteins and directly binds PARP1 and Ku70/Ku80. Rapid PARP1-dependent BRN2 association with sites of DNA damage facilitates recruitment of Ku80 and reprograms DNA damage repair by promoting Ku-dependent nonhomologous end-joining (NHEJ) at the expense of homologous recombination. BRN2 also suppresses an apoptosis-associated gene expression program to protect against UVB-, chemotherapy- and vemurafenib-induced apoptosis. Remarkably, BRN2 expression also correlates with a high single-nucleotide variation prevalence in human melanomas. By promoting error-prone DNA damage repair via NHEJ and suppressing apoptosis of damaged cells, our results suggest that BRN2 contributes to the generation of melanomas with a high mutation burden. Our findings highlight a novel role for a key transcription factor in reprogramming DNA damage repair and suggest that BRN2 may impact the response to DNA-damaging agents in BRN2-expressing cancers.
Collapse
Affiliation(s)
- Katharine Herbert
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Romuald Binet
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Jean-Philippe Lambert
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada.,Department of Molecular Medicine, Cancer Research Centre, Université Laval, Quebec G1V 0A6, Canada; CHU de Québec Research Center, CHUL, Quebec G1V 4G2, Canada
| | - Pakavarin Louphrasitthiphol
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Halime Kalkavan
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Laura Sesma-Sanz
- Genome Stability Laboratory, CHU de Oncology Division, Québec Research Center, Québec City, Quebec G1R 3S3, Canada.,Department of Molecular Biology, Medical Biochemistry, and Pathology, Laval University Cancer Research Center, Québec City, Quebec G1V 0A6, Canada
| | - Carla Daniela Robles-Espinoza
- Laboratorio Internacional de Investigación Sobre el Genoma Humano, Universidad Nacional Autónoma de México, Santiago de Querétaro 76230, Mexico.,Experimental Cancer Genetics, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom
| | - Sovan Sarkar
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Eda Suer
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Sarah Andrews
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Jagat Chauhan
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Nicola D Roberts
- The Cancer Genome Project, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, United Kingdom
| | - Mark R Middleton
- Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jean-Yves Masson
- Genome Stability Laboratory, CHU de Oncology Division, Québec Research Center, Québec City, Quebec G1R 3S3, Canada.,Department of Molecular Biology, Medical Biochemistry, and Pathology, Laval University Cancer Research Center, Québec City, Quebec G1V 0A6, Canada
| | - Lionel Larue
- Institut Curie, PSL Research University, Normal and Pathological Development of Melanocytes, U1021, Institut National de la Santé et de la Recherche Médicale (INSERM), 91405 Orsay, France.,University Paris-Sud, University Paris-Saclay, UMR 3347, Centre National de la Recherche Scientifique (CNRS), 91505 Orsay, France.,Equipe Labellisée Ligue Contre le Cancer, 91405 Orsay, France
| | - Paola Falletta
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom.,Università Vita-Salute San Raffaele, Milano, 20132 Milano MI, Italy
| | - Colin R Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
5
|
Chhabra Y, Yong HXL, Fane ME, Soogrim A, Lim W, Mahiuddin DN, Kim RSQ, Ashcroft M, Beatson SA, Ainger SA, Smit DJ, Jagirdar K, Walker GJ, Sturm RA, Smith AG. Genetic variation in IRF4 expression modulates growth characteristics, tyrosinase expression and interferon-gamma response in melanocytic cells. Pigment Cell Melanoma Res 2017; 31:51-63. [PMID: 28755520 DOI: 10.1111/pcmr.12620] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/24/2017] [Indexed: 12/29/2022]
Abstract
A SNP within intron4 of the interferon regulatory factor4 (IRF4) gene, rs12203592*C/T, has been independently associated with pigmentation and age-specific effects on naevus count in European-derived populations. We have characterized the cis-regulatory activity of this intronic region and using human foreskin-derived melanoblast strains, we have explored the correlation between IRF4 rs12203592 homozygous C/C and T/T genotypes with TYR enzyme activity, supporting its association with pigmentation traits. Further, higher IRF4 protein levels directed by the rs12203592*C allele were associated with increased basal proliferation but decreased cell viability following UVR, an etiological factor in melanoma development. Since UVR, and accompanying IFNγ-mediated inflammatory response, is associated with melanomagenesis, we evaluated its effects in the context of IRF4 status. Manipulation of IRF4 levels followed by IFNγ treatment revealed a subset of chemokines and immuno-evasive molecules that are sensitive to IRF4 expression level and genotype including CTLA4 and PD-L1.
Collapse
Affiliation(s)
- Yash Chhabra
- Dermatology Research Centre, UQ Diamantina Institute, The University of Queensland, TRI, Brisbane, QLD, Australia.,School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, at the Translational Research Institute, Brisbane, QLD, Australia
| | - Hilary X L Yong
- Dermatology Research Centre, UQ Diamantina Institute, The University of Queensland, TRI, Brisbane, QLD, Australia
| | - Mitchell E Fane
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, at the Translational Research Institute, Brisbane, QLD, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Arish Soogrim
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Wen Lim
- Dermatology Research Centre, UQ Diamantina Institute, The University of Queensland, TRI, Brisbane, QLD, Australia
| | - Dayana Nur Mahiuddin
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Reuben S Q Kim
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Melinda Ashcroft
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Scott A Beatson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen A Ainger
- Dermatology Research Centre, UQ Diamantina Institute, The University of Queensland, TRI, Brisbane, QLD, Australia
| | - Darren J Smit
- Dermatology Research Centre, UQ Diamantina Institute, The University of Queensland, TRI, Brisbane, QLD, Australia
| | - Kasturee Jagirdar
- Dermatology Research Centre, UQ Diamantina Institute, The University of Queensland, TRI, Brisbane, QLD, Australia
| | - Graeme J Walker
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Richard A Sturm
- Dermatology Research Centre, UQ Diamantina Institute, The University of Queensland, TRI, Brisbane, QLD, Australia
| | - Aaron G Smith
- Dermatology Research Centre, UQ Diamantina Institute, The University of Queensland, TRI, Brisbane, QLD, Australia.,School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, at the Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
6
|
Benítez-Burraco A, Lattanzi W, Murphy E. Language Impairments in ASD Resulting from a Failed Domestication of the Human Brain. Front Neurosci 2016; 10:373. [PMID: 27621700 PMCID: PMC5002430 DOI: 10.3389/fnins.2016.00373] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/02/2016] [Indexed: 11/16/2022] Open
Abstract
Autism spectrum disorders (ASD) are pervasive neurodevelopmental disorders entailing social and cognitive deficits, including marked problems with language. Numerous genes have been associated with ASD, but it is unclear how language deficits arise from gene mutation or dysregulation. It is also unclear why ASD shows such high prevalence within human populations. Interestingly, the emergence of a modern faculty of language has been hypothesized to be linked to changes in the human brain/skull, but also to the process of self-domestication of the human species. It is our intention to show that people with ASD exhibit less marked domesticated traits at the morphological, physiological, and behavioral levels. We also discuss many ASD candidates represented among the genes known to be involved in the “domestication syndrome” (the constellation of traits exhibited by domesticated mammals, which seemingly results from the hypofunction of the neural crest) and among the set of genes involved in language function closely connected to them. Moreover, many of these genes show altered expression profiles in the brain of autists. In addition, some candidates for domestication and language-readiness show the same expression profile in people with ASD and chimps in different brain areas involved in language processing. Similarities regarding the brain oscillatory behavior of these areas can be expected too. We conclude that ASD may represent an abnormal ontogenetic itinerary for the human faculty of language resulting in part from changes in genes important for the “domestication syndrome” and, ultimately, from the normal functioning of the neural crest.
Collapse
Affiliation(s)
| | - Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Elliot Murphy
- Division of Psychology and Language Sciences, University College London London, UK
| |
Collapse
|
7
|
Chen HY, Lee YH, Chen HY, Yeh CA, Chueh PJ, Lin YMJ. Capsaicin Inhibited Aggressive Phenotypes through Downregulation of Tumor-Associated NADH Oxidase (tNOX) by POU Domain Transcription Factor POU3F2. Molecules 2016; 21:molecules21060733. [PMID: 27271588 PMCID: PMC6273514 DOI: 10.3390/molecules21060733] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/26/2016] [Accepted: 05/31/2016] [Indexed: 11/22/2022] Open
Abstract
Capsaicin has been reported to preferentially inhibit the activity of tumor-associated NADH oxidase (tNOX), which belongs to a family of growth-related plasma membrane hydroquinone oxidases in cancer/transformed cells. The inhibitory effect of capsaicin on tNOX is associated with cell growth attenuation and apoptosis. However, no previous study has examined the transcriptional regulation of tNOX protein expression. Bioinformatic analysis has indicated that the tNOX promoter sequence harbors a binding motif for POU3F2, which is thought to play important roles in neuronal differentiation, melanocytes growth/differentiation and tumorigenesis. In this study, we found that capsaicin-mediated tNOX downregulation and cell migration inhibition were through POU3F2. The protein expression levels of POU3F2 and tNOX are positively correlated, and that overexpression of POU3F2 (and the corresponding upregulation of tNOX) enhanced the proliferation, migration and invasion in AGS (human gastric carcinoma) cells. In contrast, knockdown of POU3F2 downregulates tNOX, and the cancer phenotypes are affected. These findings not only shed light on the molecular mechanism of the anticancer properties of capsaicin, but also the transcription regulation of tNOX expression that may potentially explain how POU3F2 is associated with tumorigenesis.
Collapse
Affiliation(s)
- Hung Yen Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Yi Hui Lee
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Huei Yu Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Chia An Yeh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Pin Ju Chueh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Yi-Mei J Lin
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
8
|
Kubic JD, Little EC, Kaiser RS, Young KP, Lang D. FOXD3 Promotes PAX3 Expression in Melanoma Cells. J Cell Biochem 2015; 117:533-41. [PMID: 26252164 DOI: 10.1002/jcb.25306] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/04/2015] [Indexed: 01/01/2023]
Abstract
Several key transcription factors regulate cell growth, survival, and differentiation during neural crest and melanoblast development in the embryo, and these same pathways may be reactivated in tumors arising from the progenitors of these cells. The transcription factors PAX3 and FOXD3 have essential roles in melanoblasts and melanoma. In this study, we define a regulatory pathway where FOXD3 promotes the expression of PAX3. Both factors are expressed in melanoma cells and there is a positive correlation between the transcript levels of PAX3 and FOXD3. The PAX3 gene contains two FOX binding motifs within highly conserved enhancer regulatory elements that are essential for neural crest development. FOXD3 binds to both of these motifs in vitro but only one of these sites is preferentially utilized in melanoma cells. Overexpression of FOXD3 upregulates PAX3 levels while inhibition of FOXD3 function does not alter PAX3 protein levels, supporting that FOXD3 is sufficient but not necessary to drive PAX3 expression in melanoma cells. Here, we identify a molecular pathway where FOXD3 upregulates PAX3 expression and therefore contributes to melanoma progression.
Collapse
Affiliation(s)
- Jennifer D Kubic
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, 60637
| | - Elizabeth C Little
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, 60637
| | - Rebecca S Kaiser
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, 60637
| | - Kacey P Young
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, 60637
| | - Deborah Lang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, 60637
| |
Collapse
|
9
|
Sokolik C, Liu Y, Bauer D, McPherson J, Broeker M, Heimberg G, Qi LS, Sivak DA, Thomson M. Transcription factor competition allows embryonic stem cells to distinguish authentic signals from noise. Cell Syst 2015; 1:117-129. [PMID: 26405695 DOI: 10.1016/j.cels.2015.08.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stem cells occupy variable environments where they must distinguish stochastic fluctuations from developmental cues. Here, we use optogenetics to investigate how the pluripotency network in embryonic stem (ES) cells achieves a robust response to differentiation cues but not to gene expression fluctuations. We engineered ES cells in which we could quantitatively ontrol the endogenous mechanism of neural differentiation through a light-inducible Brn2 transgene and monitor differentiation status through a genome-integrated Nanog-GFP reporter. By exposing cells to pulses of Brn2, we find that the pluripotency network rejects Brn2 inputs that are below specific magnitude or duration thresholds, but allows rapid differentiation when both thresholds are satisfied. The filtering properties of the network arise through its positive feedback architecture and the intrinsic half-life of Nanog, which determines the duration threshold in the network. Together our results suggest that the dynamic properties of positive-feedback networks might determine how inputs are classified as signal or noise by stem cells.
Collapse
Affiliation(s)
- Cameron Sokolik
- Center for Systems and Synthetic Biology, University of California, San Francisco; San Francisco, California, 94158. USA ; Department of Cellular and Molecular Pharmacology, University of California, San Francisco; San Francisco, California, 94158. USA
| | - Yanxia Liu
- Center for Systems and Synthetic Biology, University of California, San Francisco; San Francisco, California, 94158. USA
| | - David Bauer
- Center for Systems and Synthetic Biology, University of California, San Francisco; San Francisco, California, 94158. USA ; Department of Cellular and Molecular Pharmacology, University of California, San Francisco; San Francisco, California, 94158. USA
| | - Jade McPherson
- Center for Systems and Synthetic Biology, University of California, San Francisco; San Francisco, California, 94158. USA ; Department of Cellular and Molecular Pharmacology, University of California, San Francisco; San Francisco, California, 94158. USA
| | - Michael Broeker
- Center for Systems and Synthetic Biology, University of California, San Francisco; San Francisco, California, 94158. USA ; Department of Cellular and Molecular Pharmacology, University of California, San Francisco; San Francisco, California, 94158. USA
| | - Graham Heimberg
- Center for Systems and Synthetic Biology, University of California, San Francisco; San Francisco, California, 94158. USA
| | - Lei S Qi
- Center for Systems and Synthetic Biology, University of California, San Francisco; San Francisco, California, 94158. USA
| | - David A Sivak
- Center for Systems and Synthetic Biology, University of California, San Francisco; San Francisco, California, 94158. USA
| | - Matt Thomson
- Center for Systems and Synthetic Biology, University of California, San Francisco; San Francisco, California, 94158. USA ; Department of Cellular and Molecular Pharmacology, University of California, San Francisco; San Francisco, California, 94158. USA
| |
Collapse
|
10
|
Li R, Wu F, Ruonala R, Sapkota D, Hu Z, Mu X. Isl1 and Pou4f2 form a complex to regulate target genes in developing retinal ganglion cells. PLoS One 2014; 9:e92105. [PMID: 24643061 PMCID: PMC3958441 DOI: 10.1371/journal.pone.0092105] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/17/2014] [Indexed: 02/01/2023] Open
Abstract
Precise regulation of gene expression during biological processes, including development, is often achieved by combinatorial action of multiple transcription factors. The mechanisms by which these factors collaborate are largely not known. We have shown previously that Isl1, a Lim-Homeodomain transcription factor, and Pou4f2, a class IV POU domain transcription factor, co-regulate a set of genes required for retinal ganglion cell (RGC) differentiation. Here we further explore how these two factors interact to precisely regulate gene expression during RGC development. By GST pulldown assays, co-immunoprecipitation, and electrophoretic mobility shift assays, we show that Isl1 and Pou4f2 form a complex in vitro and in vivo, and identify the domains within these two proteins that are responsible for this interaction. By luciferase assay, in situ hybridization, and RNA-seq, we further demonstrate that the two factors contribute quantitatively to gene expression in the developing RGCs. Although each factor alone can activate gene expression, both factors are required to achieve optimal expression levels. Finally, we discover that Isl1 and Pou4f2 can interact with other POU and Lim-Homeodomain factors respectively, indicating the interactions between these two classes of transcription factors are prevalent in development and other biological processes.
Collapse
Affiliation(s)
- Renzhong Li
- Department of Ophthalmology/Ross Eye Institute, University of Buffalo, Buffalo, New York, United States of America
- Developmental Genomics Group, New York State Center of Excellence in Bioinformatics and Life Sciences, University of Buffalo, Buffalo, New York, United States of America
- SUNY Eye Institute, University of Buffalo, Buffalo, New York, United States of America
| | - Fuguo Wu
- Department of Ophthalmology/Ross Eye Institute, University of Buffalo, Buffalo, New York, United States of America
- Developmental Genomics Group, New York State Center of Excellence in Bioinformatics and Life Sciences, University of Buffalo, Buffalo, New York, United States of America
- SUNY Eye Institute, University of Buffalo, Buffalo, New York, United States of America
| | - Raili Ruonala
- Department of Ophthalmology/Ross Eye Institute, University of Buffalo, Buffalo, New York, United States of America
- Developmental Genomics Group, New York State Center of Excellence in Bioinformatics and Life Sciences, University of Buffalo, Buffalo, New York, United States of America
- SUNY Eye Institute, University of Buffalo, Buffalo, New York, United States of America
| | - Darshan Sapkota
- Department of Ophthalmology/Ross Eye Institute, University of Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, University of Buffalo, Buffalo, New York, United States of America
- Developmental Genomics Group, New York State Center of Excellence in Bioinformatics and Life Sciences, University of Buffalo, Buffalo, New York, United States of America
- SUNY Eye Institute, University of Buffalo, Buffalo, New York, United States of America
| | - Zihua Hu
- Department of Ophthalmology/Ross Eye Institute, University of Buffalo, Buffalo, New York, United States of America
- Department of Biostatistics, University of Buffalo, Buffalo, New York, United States of America
- Department of Medicine, University of Buffalo, Buffalo, New York, United States of America
- Center of Computational Research, New York State Center of Excellence in Bioinformatics and Life Sciences, University of Buffalo, Buffalo, New York, United States of America
- SUNY Eye Institute, University of Buffalo, Buffalo, New York, United States of America
| | - Xiuqian Mu
- Department of Ophthalmology/Ross Eye Institute, University of Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, University of Buffalo, Buffalo, New York, United States of America
- Developmental Genomics Group, New York State Center of Excellence in Bioinformatics and Life Sciences, University of Buffalo, Buffalo, New York, United States of America
- SUNY Eye Institute, University of Buffalo, Buffalo, New York, United States of America
- CCSG Cancer Genetics Program, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
11
|
Su A, Dry SM, Binder SW, Said J, Shintaku P, Sarantopoulos GP. Malignant melanoma with neural differentiation: an exceptional case report and brief review of the pertinent literature. Am J Dermatopathol 2014; 36:e5-9. [PMID: 23782676 PMCID: PMC4079032 DOI: 10.1097/dad.0b013e31828cf90a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
: The term neurotropic melanoma has been used to refer to malignant melanoma with associated infiltration of nerve or "neural differentiation"--that is, melanoma cells exhibiting cytological characteristics of nerve cells. Historically, neurotropic melanoma has generally been discussed within the context of desmoplastic melanoma. We report an exceptional case of melanoma notable for a very well-differentiated neural component that was contiguous with obvious overlying melanoma. After careful consideration of all pertinent histological features, the overall diagnostic impression was that of melanoma with associated "malignant neurotization." We have not encountered a previously reported case with such a well-differentiated neural component. The following article details our exceptional case of melanoma with "malignant neurotization" and presents a discussion of the differential diagnosis and brief review of the pertinent literature.
Collapse
Affiliation(s)
- Albert Su
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | | | | | | | | | | |
Collapse
|
12
|
Bery A, Martynoga B, Guillemot F, Joly JS, Rétaux S. Characterization of enhancers active in the mouse embryonic cerebral cortex suggests Sox/Pou cis-regulatory logics and heterogeneity of cortical progenitors. Cereb Cortex 2013; 24:2822-34. [PMID: 23720416 DOI: 10.1093/cercor/bht126] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We aimed to identify cis-regulatory elements that control gene expression in progenitors of the cerebral cortex. A list of 975 putative enhancers were retrieved from a ChIP-Seq experiment performed in NS5 mouse stem cells with antibodies to Sox2, Brn2/Pou3f2, or Brn1/Pou3f3. Through a selection pipeline including gene ontology and expression pattern, we reduced the number of candidate enhancer sequences to 20. Ex vivo electroporation of green fluorescent pProtein (GFP) reporter constructs in the telencephalon of mouse embryos showed that 35% of the 20 selected candidate sequences displayed enhancer activity in the developing cortex at E13.5. In silico transcription factor binding site (TFBS) searches and mutagenesis experiments showed that enhancer activity is related to the presence of Sox/Pou TFBS pairs in the sequence. Comparative genomic analyses showed that enhancer activity is not related to the evolutionary conservation of the sequence. Finally, the combination of in utero electroporation of GFP reporter constructs with immunostaining for Tbr2 (basal progenitor marker) and phospho-histoneH3 (mitotic activity marker) demonstrated that each enhancer is specifically active in precise subpopulations of progenitors in the cortical germinal zone, highlighting the heterogeneity of these progenitors in terms of cis-regulation.
Collapse
Affiliation(s)
| | | | | | - Jean-Stéphane Joly
- Equipe Morphogenesis of the Chordate Nervous System, UPR3294 N&D, Institut de Neurobiologie Alfred Fessard, CNRS, Gif-sur-Yvette, France and
| | | |
Collapse
|
13
|
Zhao J, Lambert G, Meijer AH, Rosa FM. The transcription factor Vox represses endoderm development by interacting with Casanova and Pou2. Development 2013; 140:1090-9. [DOI: 10.1242/dev.082008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Endoderm and mesoderm are both formed upon activation of Nodal signaling but how endoderm differentiates from mesoderm is still poorly explored. The sox-related gene casanova (sox32) acts downstream of the Nodal signal, is essential for endoderm development and requires the co-factor Pou2 (Pou5f1, Oct3, Oct4) in this process. Conversely, BMP signals have been shown to inhibit endoderm development by an as yet unexplained mechanism. In a search for Casanova regulators in zebrafish, we identified two of its binding partners as the transcription factors Pou2 and Vox, a member of the Vent group of proteins also involved in the patterning of the gastrula. In overexpression studies we show that vox and/or Vent group genes inhibit the capacity of Casanova to induce endoderm, even in the presence of its co-factor Pou2, and that Vox acts as a repressor in this process. We further show that vox, but not other members of the Vent group, is essential for defining the proper endodermal domain size at gastrulation. In this process, vox acts downstream of BMPs. Cell fate analysis further shows that Vox plays a key role downstream of BMP signals in regulating the capacity of Nodal to induce endoderm versus mesoderm by modulating the activity of the Casanova/Pou2 regulatory system.
Collapse
Affiliation(s)
- Jue Zhao
- INSERM U1024, F-75005 Paris, France
- CNRS UMR 8197, F-75005 Paris, France
- IBENS, Institut de Biologie de l’Ecole Normale Supérieure, F-75230 Paris, France
- College of Life Sciences, Peking University, Beijing 100871, P. R. China
| | - Guillaume Lambert
- INSERM U1024, F-75005 Paris, France
- CNRS UMR 8197, F-75005 Paris, France
- IBENS, Institut de Biologie de l’Ecole Normale Supérieure, F-75230 Paris, France
| | | | - Frederic M. Rosa
- INSERM U1024, F-75005 Paris, France
- CNRS UMR 8197, F-75005 Paris, France
- IBENS, Institut de Biologie de l’Ecole Normale Supérieure, F-75230 Paris, France
| |
Collapse
|
14
|
Liu F, Cao J, Lv J, Dong L, Pier E, Xu GX, Wang RA, Xu Z, Goding C, Cui R. TBX2 expression is regulated by PAX3 in the melanocyte lineage. Pigment Cell Melanoma Res 2013; 26:67-77. [PMID: 23020925 PMCID: PMC3527652 DOI: 10.1111/pcmr.12029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 09/25/2012] [Indexed: 11/28/2022]
Abstract
The paired box homeotic gene 3 (PAX3) is a crucial regulator for the maintenance of melanocytic progenitor cells and has a poorly defined role in melanoma. To understand how PAX3 affects melanocyte and melanoma proliferation, we identified potential PAX3 downstream targets through gene expression profiling. Here, we identify T-box 2 (TBX2), a key developmental regulator of cell identity and an antisenescence factor in melanoma, as a directly regulated PAX3 target. We also found that TBX2 is involved in the survival of melanoma cells and is overexpressed in some melanoma specimens. The identification of TBX2 as a target for PAX3 provides a key insight into how PAX3 may contribute to melanoma evolution and may provide opportunities for prosenescence therapeutic intervention aimed at disrupting the ability of PAX3 to regulate TBX2.
Collapse
Affiliation(s)
- Fang Liu
- Department of Dermatology, Boston University School of Medicine 609 Albany St, Boston, MA 02118
- Department of Dermatology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China,100020
| | - Juxiang Cao
- Department of Dermatology, Boston University School of Medicine 609 Albany St, Boston, MA 02118
| | - Jinghu Lv
- Rizhao General Hospital, Rizhao, Shandong Province, China
| | - Liang Dong
- Department of Dermatology, Boston University School of Medicine 609 Albany St, Boston, MA 02118
| | - Eric Pier
- Department of Dermatology, Boston University School of Medicine 609 Albany St, Boston, MA 02118
| | - George X. Xu
- Department of Pathology and Lab Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Rui-an Wang
- Department of Pathology, Fourth Military Medical University, Xian, Shanxi, China
| | - Zhixiang Xu
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham School of Medicine, 17 Ave S, Birmingham, Al 35233
| | - Colin Goding
- Ludwig Institute for Cancer Research, University of Oxford, Headington, Oxford, OX3 7DQ, UK
| | - Rutao Cui
- Department of Dermatology, Boston University School of Medicine 609 Albany St, Boston, MA 02118
| |
Collapse
|
15
|
Ellmann L, Joshi MB, Resink TJ, Bosserhoff AK, Kuphal S. BRN2 is a transcriptional repressor of CDH13 (T-cadherin) in melanoma cells. J Transl Med 2012; 92:1788-800. [PMID: 23069940 DOI: 10.1038/labinvest.2012.140] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
T-cadherin (cadherin 13, H-cadherin, gene name CDH13) has been proposed to act as a tumor-suppressor gene as its expression is significantly diminished in several types of carcinomas, including melanomas. Allelic loss and promoter hypermethylation have been proposed as mechanisms for silencing of CDH13. However, they do not account for loss of T-cadherin expression in all carcinomas, and other genetic or epigenetic alterations can be presumed. The present study investigated transcriptional regulation of CDH13 in melanoma. Bioinformatical analysis pointed to the presence of known BRN2 (also known as POU3F2 and N-Oct-3)-binding motifs in the CDH13 promoter sequence. We found an inverse correlation between BRN2 and T-cadherin protein and transcript expression. Reporter gene analysis and electrophoretic mobility shift assays in melanoma cells demonstrated that CDH13 is a direct target of BRN2 and that BRN2 is a functional transcriptional repressor of CDH13 promoter activity. The regulatory binding element of BRN2 was located -219 bp of the CDH13 promoter proximal to the start codon and was identified as 5'-CATGCAAAA-3'. Ectopic expression of BRN2 in BRN2-negative/T-cadherin-positive melanoma cells resulted in suppression of CDH13 promoter activity, whereas BRN2 knockdown in BRN2-positive/T-cadherin-negative melanoma cells resulted in re-expression of T-cadherin transcripts and protein. Transcriptional repression of CDH13 by BRN2 may participate in malignant transformation of melanoma by increasing invasion and migration potentials of melanoma cells. The study has identified CDH13 as a novel direct BRN2 transcriptional target gene and has advanced knowledge of mechanisms underlying loss of T-cadherin expression in melanoma.
Collapse
Affiliation(s)
- Lisa Ellmann
- Institute of Pathology, Molecular Pathology, University of Regensburg, Regensburg, Germany
| | | | | | | | | |
Collapse
|
16
|
Epigenetic obstacles encountered by transcription factors: reprogramming against all odds. Curr Opin Genet Dev 2012; 22:409-15. [PMID: 22922161 DOI: 10.1016/j.gde.2012.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 07/24/2012] [Accepted: 08/02/2012] [Indexed: 12/24/2022]
Abstract
Reprogramming of a somatic nucleus to an induced pluripotent state can be achieved in vitro through ectopic expression of Oct4 (Pou5f1), Sox2, Klf4 and c-Myc. While the ability of these factors to regulate transcription in a pluripotent context has been studied extensively, their ability to interact with and remodel a somatic genome remains underexplored. Several recent studies have begun to provide mechanistic insights that will eventually lead to a more rational design and improved understanding of nuclear reprogramming.
Collapse
|
17
|
FGF2 regulates melanocytes viability through the STAT3-transactivated PAX3 transcription. Cell Death Differ 2011; 19:616-22. [PMID: 21997191 PMCID: PMC3307977 DOI: 10.1038/cdd.2011.132] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PAX3 (paired box 3) is known to have an important role in melanocyte development through modulation of microphthalmia-associated transcription factor transcription. Here we found that PAX3 transcriptional activity could be regulated through FGF2 (basic fibroblast growth factor)-STAT3 (signal transducer and activator of transcription 3) signaling in the pigment cells. To study its function in vivo, we have generated a transgenic mouse model expressing PAX3 driven by tyrosinase promoter in a tissue-specific fashion. These animals exhibit hyperpigmentation in the epidermis, evident in the skin color of their ears and tails. We showed that the darker skin color results from both increased melanocyte numbers and melanin synthesis. Together, our study delineated a novel pathway in the melanocyte lineage, linking FGF2-STAT3 signaling to increased PAX3 transcription. Moreover, our results suggest that this pathway might contribute to the regulation of melanocyte numbers and melanin levels, and thereby provide an alternative strategy to induce pigmentation.
Collapse
|
18
|
DNA demethylation in PD-1 gene promoter induced by 5-azacytidine activates PD-1 expression on Molt-4 cells. Cell Immunol 2011; 271:450-4. [PMID: 21907333 DOI: 10.1016/j.cellimm.2011.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 08/15/2011] [Accepted: 08/17/2011] [Indexed: 01/20/2023]
Abstract
The expression of the programmed death 1 (PD-1) gene is an indicator of exhausted T-cells with decreased activation and function. It remains unknown, however, whether the methylation status of the PD-1 gene promoter is associated with PD-1 expression level. This study shows the changes of PD-1 expression levels and the demethylation status of the PD-1 promoter region in Molt-4 cells under different concentrations of 5-azacytidine (5-Zac). The result demonstrated that DNA demethylation at PD-1 promoter may contribute to PD-1 overexpression.
Collapse
|
19
|
Smith AG, Lim W, Pearen M, Muscat GEO, Sturm RA. Regulation of NR4A nuclear receptor expression by oncogenic BRAF in melanoma cells. Pigment Cell Melanoma Res 2011; 24:551-63. [PMID: 21362156 DOI: 10.1111/j.1755-148x.2011.00843.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Activating mutations in the MAPK pathway effectors, NRAS or BRAF, are detected in over 70% of melanomas. Accordingly, the identification of downstream targets of constitutive MAPK signalling in melanoma represents a major goal in understanding the genesis of this disease. We report here the regulation of members of the NR4A family of nuclear receptors by the BRAF-MEK-ERK cascade in melanoma cells. Expression profiling of melanoma cells in which both the NR4A1 and NR4A2 family members have been down-regulated by siRNA revealed alterations in genes associated with proliferation, survival and invasiveness of tumour cells. Notably, the up-regulation of Wnt/β-catenin pathway antagonists, DACT1 and CITED1, following NR4A1/2 ablation suggests a possible link between NR4A and β-catenin activity in melanoma cells. Taken together, these data suggest that dysregulation of NR4A nuclear receptors expression and function by the MAPK pathway may contribute to melanoma tumourigenicity.
Collapse
Affiliation(s)
- Aaron G Smith
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| | | | | | | | | |
Collapse
|
20
|
Liu Y, Zhu H, Liu M, Du J, Qian Y, Wang Y, Ding F, Gu X. Downregulation of Pax3 expression correlates with acquired GFAP expression during NSC differentiation towards astrocytes. FEBS Lett 2011; 585:1014-20. [PMID: 21371476 DOI: 10.1016/j.febslet.2011.02.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 02/21/2011] [Accepted: 02/24/2011] [Indexed: 10/18/2022]
Abstract
Glial fibrillary acidic protein (GFAP) is a principal intermediate filament in mature astrocytes of the central nervous system (CNS), and the regulation of GFAP transcription has not been well understood yet. In the present study, we reported paired box 3 protein (Pax3) as a novel regulator of GFAP transcription, which could bind the promoter region of GFAP and down regulate the GFAP level during the serum-induced astrocyte differentiation of neural stem cells (NSCs). Moreover, the overexpression and suppression of Pax3 could inhibit and promote NSC differentiation, respectively. These data suggest that Pax3 negatively regulates GFAP expression during astrocyte differentiation in vitro.
Collapse
Affiliation(s)
- Yan Liu
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Harris ML, Baxter LL, Loftus SK, Pavan WJ. Sox proteins in melanocyte development and melanoma. Pigment Cell Melanoma Res 2010; 23:496-513. [PMID: 20444197 DOI: 10.1111/j.1755-148x.2010.00711.x] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Over 10 years have passed since the first Sox gene was implicated in melanocyte development. Since then, we have discovered that SOX5, SOX9, SOX10 and SOX18 all participate as transcription factors that affect key melanocytic genes in both regulatory and modulatory fashions. Both SOX9 and SOX10 play major roles in the establishment and normal function of the melanocyte; SOX10 has been shown to heavily influence melanocyte development and SOX9 has been implicated in melanogenesis in the adult. Despite these advances, the precise cellular and molecular details of how these SOX proteins are regulated and interact during all stages of the melanocyte life cycle remain unknown. Improper regulation of SOX9 or SOX10 is also associated with cancerous transformation, and thus understanding the normal function of SOX proteins in the melanocyte will be key to revealing how these proteins contribute to melanoma.
Collapse
Affiliation(s)
- Melissa L Harris
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
22
|
Kobi D, Steunou AL, Dembélé D, Legras S, Larue L, Nieto L, Davidson I. Genome-wide analysis of POU3F2/BRN2 promoter occupancy in human melanoma cells reveals Kitl as a novel regulated target gene. Pigment Cell Melanoma Res 2010; 23:404-18. [PMID: 20337985 DOI: 10.1111/j.1755-148x.2010.00697.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
POU3F2 is a POU-Homeodomain transcription factor expressed in neurons and melanoma cells. In melanoma lesions, cells expressing high levels of POU3F2 show enhanced invasive and metastatic capacity that can in part be explained by repression of Micropthalmia-associated Transcription Factor (MITF) expression via POU3F2 binding to its promoter. To identify other POU3F2 target genes that may be involved in modulating the properties of melanoma cells, we performed ChIP-chip experiments in 501Mel melanoma cells. 2108 binding loci located in the regulatory regions of 1700 potential target genes were identified. Bioinformatic and experimental assays showed the presence of known POU3F2-binding motifs, but also many AT-rich sequences with only partial similarity to the known motifs at the occupied loci. Functional analysis indicates that POU3F2 regulates the stem cell factor (Kit ligand, Kitl) promoter via a cluster of four closely spaced binding sites located in the proximal promoter. Our results suggest that POU3F2 may regulate the properties of melanoma cells via autocrine KIT ligand signalling.
Collapse
Affiliation(s)
- Dominique Kobi
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, Illkirch Cédex, France
| | | | | | | | | | | | | |
Collapse
|
23
|
Smith AG, Beaumont KA, Smit DJ, Thurber AE, Cook AL, Boyle GM, Parsons PG, Sturm RA, Muscat GE. PPARγ agonists attenuate proliferation and modulate Wnt/β-catenin signalling in melanoma cells. Int J Biochem Cell Biol 2009; 41:844-52. [DOI: 10.1016/j.biocel.2008.08.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 08/18/2008] [Accepted: 08/21/2008] [Indexed: 11/26/2022]
|
24
|
Cabos-Siguier B, Steunou AL, Joseph G, Alazard R, Ducoux-Petit M, Nieto L, Monsarrat B, Erard M, Clottes E. Expression and purification of human full-length N Oct-3, a transcription factor involved in melanoma growth. Protein Expr Purif 2009; 64:39-46. [DOI: 10.1016/j.pep.2008.10.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 10/09/2008] [Accepted: 10/20/2008] [Indexed: 11/28/2022]
|
25
|
Yang G, Li Y, Nishimura EK, Xin H, Zhou A, Guo Y, Dong L, Denning MF, Nickoloff BJ, Cui R. Inhibition of PAX3 by TGF-beta modulates melanocyte viability. Mol Cell 2009; 32:554-63. [PMID: 19026785 DOI: 10.1016/j.molcel.2008.11.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 07/25/2008] [Accepted: 11/04/2008] [Indexed: 11/18/2022]
Abstract
The protein encoded by paired-box homeotic gene 3 (PAX3) is a key regulator of the microphthalmia-associated transcription factor (Mitf) in the melanocyte lineage. Here, we show that PAX3 expression in skin is directly inhibited by TGF-beta/Smads. UV irradiation represses TGF-beta in keratinocytes, and the repression of TGF-beta/Smads upregulates PAX3 in melanocytes, which is associated with a UV-induced melanogenic response and consequent pigmentation. Furthermore, the TGF-beta-PAX3 signaling pathway interacts with the p53-POMC/MSH-MC1R signaling pathway, and both are crucial in melanogenesis. The activation of p53-POMC/MSH-MC1R signaling is required for the UV-induced melanogenic response because PAX3 functions in synergy with SOX10 in a cAMP-response element (CRE)-dependent manner to regulate the transcription of Mitf. This study will provide a rich foundation for further research on skin cancer prevention by enabling us to identify targeted small molecules in the signaling pathways of the UV-induced melanogenic response that are highly likely to induce naturally protective pigmentation.
Collapse
Affiliation(s)
- Guang Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Children's Hospital Boston, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Kubic JD, Young KP, Plummer RS, Ludvik AE, Lang D. Pigmentation PAX-ways: the role of Pax3 in melanogenesis, melanocyte stem cell maintenance, and disease. Pigment Cell Melanoma Res 2009; 21:627-45. [PMID: 18983540 DOI: 10.1111/j.1755-148x.2008.00514.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Transcription factors initiate programs of gene expression and are catalysts in downstream molecular cascades that modulate a variety of cellular processes. Pax3 is a transcription factor that is important in the melanocyte and influences melanocytic proliferation, resistance to apoptosis, migration, lineage specificity and differentiation. In this review, we focus on Pax3 and the molecular pathways that Pax3 is a part of during melanogenesis and in the melanocyte stem cell. These roles of Pax3 are emphasized during the development of diseases and syndromes resulting from either too much or too little Pax3 function. Due to its key task in melanocyte stem cells and tumors, the Pax3 pathway may provide an ideal target for either stem cell or cancer therapies.
Collapse
Affiliation(s)
- Jennifer D Kubic
- Section of Dermatology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | | | | | | |
Collapse
|
27
|
Cook AL, Sturm RA. POU domain transcription factors: BRN2 as a regulator of melanocytic growth and tumourigenesis. Pigment Cell Melanoma Res 2008; 21:611-26. [DOI: 10.1111/j.1755-148x.2008.00510.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
28
|
Goodall J, Carreira S, Denat L, Kobi D, Davidson I, Nuciforo P, Sturm RA, Larue L, Goding CR. Brn-2 represses microphthalmia-associated transcription factor expression and marks a distinct subpopulation of microphthalmia-associated transcription factor-negative melanoma cells. Cancer Res 2008; 68:7788-94. [PMID: 18829533 DOI: 10.1158/0008-5472.can-08-1053] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The origin of tumor heterogeneity is poorly understood, yet it represents a major barrier to effective therapy. In melanoma and in melanocyte development, the microphthalmia-associated transcription factor (Mitf) controls survival, differentiation, proliferation, and migration/metastasis. The Brn-2 (N-Oct-3, POU3F2) transcription factor also regulates melanoma proliferation and is up-regulated by BRAF and beta-catenin, two key melanoma-associated signaling molecules. Here, we show that Brn-2 also regulates invasiveness and directly represses Mitf expression. Remarkably, in melanoma biopsies, Mitf and Brn-2 each mark a distinct subpopulation of melanoma cells, providing a striking illustration of melanoma tumor heterogeneity with implications for melanoma therapy.
Collapse
Affiliation(s)
- Jane Goodall
- Signaling and Development Laboratory, Marie Curie Research Institute, The Chart, Oxted, Surrey, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
SOX9 and SOX10 but not BRN2 are required for nestin expression in human melanoma cells. J Invest Dermatol 2008; 129:945-53. [PMID: 18923447 DOI: 10.1038/jid.2008.316] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nestin is an intermediate filament protein and a marker of neuroectodermal stem cells indicating multipotentiality and regenerative capability. In melanoma tissues, nestin re-expression was correlated with tumor progression. Activation of the nestin neural enhancer was shown to be dependent on the binding of class III POU transcription factors, with brain-2 (BRN2) suggested to play a key role. We found both nestin and BRN2 mRNA in almost all of 13 analyzed melanoma cell lines of different progression stages, but expression levels did not correlate. Nestin protein was detected in 11 of 13 and BRN2 protein in 7 of 13 melanoma cell lines independent of progression stage. Downregulation of BRN2 by small-interfering RNA did not alter nestin expression in melanoma cells. However, POU proteins, such as BRN2, commonly cooperate with transcription factors of the Sry-box (SOX) family by binding to a nearby DNA site necessary for their action. SOX9 and SOX10 have been shown to be expressed in melanocyte precursors, with SOX10 downregulated upon differentiation. We now demonstrate SOX9 and SOX10 protein expression in melanoma tissues and cell lines. Downregulation of SOX9 and of SOX10 markedly decreased nestin levels in melanoma cells in a cooperative manner. Thus, SOX9 and SOX10 but not BRN2 seem to be required for nestin expression in human melanoma.
Collapse
|
30
|
Blake JA, Thomas M, Thompson JA, White R, Ziman M. Perplexing Pax: From puzzle to paradigm. Dev Dyn 2008; 237:2791-803. [DOI: 10.1002/dvdy.21711] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
31
|
Analysis of cultured human melanocytes based on polymorphisms within the SLC45A2/MATP, SLC24A5/NCKX5, and OCA2/P loci. J Invest Dermatol 2008; 129:392-405. [PMID: 18650849 DOI: 10.1038/jid.2008.211] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Single nucleotide polymorphisms (SNPs) within the SLC45A2/MATP, SLC24A5/NCKX5, and OCA2/P genes have been associated with natural variation of pigmentation traits in human populations. Here, we describe the characterization of human primary melanocytic cells genotyped for polymorphisms within the MATP, NCKX5, or OCA2 loci. On the basis of genotype, these cultured cells reflect the phenotypes observed by others in terms of both melanin content and tyrosinase (TYR) activity when comparing skin designated as either "White" or "Black". We found a statistically significant association of MATP-374L (darker skin) with higher TYR protein abundance that was not observed for any NCKX5-111 or OCA2 rs12913832 allele. MATP-374L/L homozygous strains displayed significantly lower MATP transcript levels compared to MATP-374F/F homozygous cells, but this did not reach statistical significance based on NCKX5 or OCA2 genotype. Similarly, we observed significantly increased levels of OCA2 mRNA in rs12913832-T (brown eye) homozygotes compared to rs12913832-C (blue eye) homozygous strains, which was not observed for MATP or NCKX5 gene transcripts. In genotype-phenotype associations performed on a collection of 226 southern European individuals using these same SNPs, we were able to show strong correlations in MATP-L374F, OCA2, and melanocortin-1 receptor with skin, eye, and hair color variation, respectively.
Collapse
|
32
|
Differential effects of phosphorylation on DNA binding properties of N Oct-3 are dictated by protein/DNA complex structures. J Mol Biol 2007; 370:687-700. [PMID: 17543985 DOI: 10.1016/j.jmb.2007.04.072] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 03/22/2007] [Accepted: 04/29/2007] [Indexed: 10/23/2022]
Abstract
N Oct-3, a transcription factor member of the POU protein family, is implicated in normal central nervous system development but also in melanoma growth. Its DNA-binding domain (DBD) comprises two subdomains, POUs and POUh, joined by a linker peptide. We have previously shown that N Oct-3 can interact with the already described PORE and MORE DNA motifs, but also with a new structural element we have termed NORE. Having observed that both the PORE and NORE DNA-association modes depend on a strong anchoring of the POUh subdomain rigid arm into the DNA-target minor groove, in contrast to the MORE mode, we have formulated the hypothesis that phosphorylation of the conserved Ser101 residue located in the N Oct-3 POUh arm could lead to differential results in DNA binding according to the type of target. Here we demonstrate that, in vitro, Ser101 is phosphorylated by protein kinase A (PKA), either purified or contained in melanoma (624 mel) nuclear extract, and that this phosphorylation indeed significantly reduced N Oct-3 DBD binding to PORE and NORE motifs, most likely by hampering the POUh rigid arm insertion in the DNA minor groove. Conversely, no effect was observed on the binding of N Oct-3 DBD to MORE sequences. Finally, once bound to its DNA targets, N Oct-3 DBD is less susceptible to PKA activity. We conclude that transcription of genes exhibiting a MORE motif in their promoter should be less affected by N Oct-3 phosphorylation than that of genes switched on by PORE or NORE sequences.
Collapse
|
33
|
Kasper LH, Fukuyama T, Biesen MA, Boussouar F, Tong C, de Pauw A, Murray PJ, van Deursen JMA, Brindle PK. Conditional knockout mice reveal distinct functions for the global transcriptional coactivators CBP and p300 in T-cell development. Mol Cell Biol 2006; 26:789-809. [PMID: 16428436 PMCID: PMC1347027 DOI: 10.1128/mcb.26.3.789-809.2006] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The global transcriptional coactivators CREB-binding protein (CBP) and the closely related p300 interact with over 312 proteins, making them among the most heavily connected hubs in the known mammalian protein-protein interactome. It is largely uncertain, however, if these interactions are important in specific cell lineages of adult animals, as homozygous null mutations in either CBP or p300 result in early embryonic lethality in mice. Here we describe a Cre/LoxP conditional p300 null allele (p300flox) that allows for the temporal and tissue-specific inactivation of p300. We used mice carrying p300flox and a CBP conditional knockout allele (CBPflox) in conjunction with an Lck-Cre transgene to delete CBP and p300 starting at the CD4- CD8- double-negative thymocyte stage of T-cell development. Loss of either p300 or CBP led to a decrease in CD4+ CD8+ double-positive thymocytes, but an increase in the percentage of CD8+ single-positive thymocytes seen in CBP mutant mice was not observed in p300 mutants. T cells completely lacking both CBP and p300 did not develop normally and were nonexistent or very rare in the periphery, however. T cells lacking CBP or p300 had reduced tumor necrosis factor alpha gene expression in response to phorbol ester and ionophore, while signal-responsive gene expression in CBP- or p300-deficient macrophages was largely intact. Thus, CBP and p300 each supply a surprising degree of redundant coactivation capacity in T cells and macrophages, although each gene has also unique properties in thymocyte development.
Collapse
Affiliation(s)
- Lawryn H Kasper
- Department of Biochemistry, St. Jude Children's Research Hospital, 332 N. Lauderdale, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Zhu BK, Pruitt SC. Determination of transcription factors and their possible roles in the regulation of Pax3 gene expression in the mouse B16 F1 melanoma cell line. Melanoma Res 2005; 15:363-73. [PMID: 16179863 DOI: 10.1097/00008390-200510000-00004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The objective of this study was to determine which transcription factors regulate the expression of the Pax3 gene in the mouse B16 F1 melanoma cell line. The results showed that the -14 kilobase pair (kbp) Pax3 promoter, but not the -1.6 kbp Pax3 promoter, promoted Pax3 gene expression in B16 cells. Comparison of the sequence of the -14 kbp human Pax3 promoter with mouse Pax3 promoters indicated that homology sequences were located between -6.9 and -5.8 kbp, and also that the 1.1 kbp fragment (between -6.9 and -5.8 kbp), linked -1.6 kbp proximal to the Pax3 promoter [plasmid PGPax3PIV (N6.9/5.8) delta SST Lacz], could mimic the functions of plasmid PGPax3 -14(N-1.6) Lacz. Mutations of the core binding elements of either Pax3 site I or II or both sites I and II reduced significantly the beta-galactosidase (beta-gal) activity in the cells. However, mutations of the core binding sequences of site A or B increased significantly the beta-gal activity in the cells. Biochemistry analysis demonstrated that POU transcription factors (Oct-1 and Brn-2) bind to the specific binding elements of both sites I and II to stimulate Pax3 gene expression, whereas the TALE homeodomain-containing proteins (Pbx and Prep1) bind with the core binding sequences of sites A and B to repress the expression of the Pax3 gene in B16 cells.
Collapse
Affiliation(s)
- Bi-ke Zhu
- Faculty of Veterinary Science, University of Sydney, Camden, New South Wales, Australia.
| | | |
Collapse
|
35
|
Goodall J, Martinozzi S, Dexter TJ, Champeval D, Carreira S, Larue L, Goding CR. Brn-2 expression controls melanoma proliferation and is directly regulated by beta-catenin. Mol Cell Biol 2004; 24:2915-22. [PMID: 15024079 PMCID: PMC371132 DOI: 10.1128/mcb.24.7.2915-2922.2004] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Constitutive activation of the Wnt/beta-catenin signaling pathway is a notable feature of a large minority of cases of malignant melanoma, an aggressive and increasingly common cancer. The identification of target genes downstream from this pathway is therefore crucial to our understanding of the disease. The POU domain transcription factor Brn-2 has been implicated in control of proliferation and melanoma survival, and its expression is strongly upregulated in melanoma. We show here that in vivo Brn-2 is expressed in melanocytes but not in embryonic day 11.5 melanoblasts and that its expression is directly controlled by the Wnt/beta-catenin signaling pathway in melanoma cell lines and in transgenic mice. Moreover, silent interfering RNA-mediated inhibition of Brn-2 expression in melanoma cells overexpressing beta-catenin results in significantly decreased proliferation. These results, together with the observation that BRAF signaling also induces Brn-2 expression, reveal that Brn-2 is a focus for the convergence of two key melanoma-associated signaling pathways that are linked to cell proliferation.
Collapse
Affiliation(s)
- Jane Goodall
- Signaling and Development Laboratory, Marie Curie Research Institute, The Chart, Oxted, Surrey RH8 0TL, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
36
|
Cook AL, Donatien PD, Smith AG, Murphy M, Jones MK, Herlyn M, Bennett DC, Leonard JH, Sturm RA. Human melanoblasts in culture: expression of BRN2 and synergistic regulation by fibroblast growth factor-2, stem cell factor, and endothelin-3. J Invest Dermatol 2004; 121:1150-9. [PMID: 14708619 DOI: 10.1046/j.1523-1747.2003.12562.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The BRN2 transcription factor (POU3F2, N-Oct-3) has been implicated in development of the melanocytic lineage and in melanoma. Using a low calcium medium supplemented with stem cell factor, fibroblast growth factor-2, endothelin-3 and cholera toxin, we have established and partially characterised human melanocyte precursor cells, which are unpigmented, contain immature melanosomes and lack L-dihydroxyphenylalanine reactivity. Melanoblast cultures expressed high levels of BRN2 compared to melanocytes, which decreased to a level similar to that of melanocytes when cultured in medium that contained phorbol ester but lacked endothelin-3, stem cell factor and fibroblast growth factor-2. This decrease in BRN2 accompanied a positive L-dihydroxyphenylalanine reaction and induction of melanosome maturation consistent with melanoblast differentiation seen during development. Culture of primary melanocytes in low calcium medium supplemented with stem cell factor, fibroblast growth factor-2 and endothelin-3 caused an increase in BRN2 protein levels with a concomitant change to a melanoblast-like morphology. Synergism between any two of these growth factors was required for BRN2 protein induction, whereas all three factors were required to alter melanocyte morphology and for maximal BRN2 protein expression. These finding implicate BRN2 as an early marker of melanoblasts that may contribute to the hierarchy of melanocytic gene control.
Collapse
Affiliation(s)
- Anthony L Cook
- The Institute for Molecular Bioscience, Center for Functional and Applied Genomics, The University of Queensland, Brisbane, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kamachi Y, Uchikawa M, Tanouchi A, Sekido R, Kondoh H. Pax6 and SOX2 form a co-DNA-binding partner complex that regulates initiation of lens development. Genes Dev 2001; 15:1272-86. [PMID: 11358870 PMCID: PMC313803 DOI: 10.1101/gad.887101] [Citation(s) in RCA: 284] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pax6 is a key transcription factor in eye development, particularly in lens development, but its molecular action has not been clarified. We demonstrate that Pax6 initiates lens development by forming a molecular complex with SOX2 on the lens-specific enhancer elements, e.g., the delta-crystallin minimal enhancer DC5. DC5 shows a limited similarity to the binding consensus sequence of Pax6 and is bound poorly by Pax6 alone. However, Pax6 binds cooperatively with SOX2 to the DC5 sequence, resulting in formation of a high-mobility form of ternary complex in vitro, which correlates with the enhancer activation in vivo. We observed Pax6 and SOX2-interdependent factor occupancy of DC5 in a chromatin environment in vivo, providing the molecular basis of synergistic activation by Pax6 and SOX2. Subtle alterations of the Pax6-binding-site sequence of DC5 or of the inter-binding-sites distance diminished the cooperative binding and caused formation of a non-functional low-mobility form complex, suggesting DNA sequence-guided and protein interaction-induced conformation change of the Pax6 protein. When ectopically expressed in embryo ectoderm, Pax6 and SOX2 in combination activate delta-crystallin gene and elicit lens placode development, indicating that the complex of Pax6 and SOX2 formed on specific DNA sequences is the genetic switch for initiation of lens differentiation.
Collapse
Affiliation(s)
- Y Kamachi
- Institute for Molecular and Cellular Biology, Osaka University, Suita, Osaka 565-0871, Japan.
| | | | | | | | | |
Collapse
|
38
|
Dugast C, Weber MJ. NF-Y binding is required for transactivation of neuronal aromatic L-amino acid decarboxylase gene promoter by the POU-domain protein Brn-2. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 89:58-70. [PMID: 11311976 DOI: 10.1016/s0169-328x(01)00063-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have previously characterized binding sites for the NF-Y transcription factor (-71/-52) and Brn-2 POU-domain protein (-92/-71) in the neuronal promoter of the human aromatic L-amino acid decarboxylase gene [Mol. Brain Res. 56 (1998) 227]. We have now explored the functional role of these binding sites in transfected SK-N-BE neuroblastoma cells. Mutations of the NF-Y site that abolish binding depressed expression of a luciferase reporter gene up to 25-fold. The overexpression of a dominant negative mutant of NF-YA subunit depressed expression by 60%. Promoter activity was increased by the overexpression of Brn-2. Mutations or deletion of the binding site of Brn-2 did not suppress transcriptional activation by overexpressed Brn-2, while promoters defective in NF-Y binding were not transactivated by Brn-2. A GST-pulldown experiment showed that recombinant human Brn-2 protein weakly interacts with recombinant NF-Y outside of DNA. Cooperative binding of recombinant NF-Y and GST--Brn-2 proteins on the neuronal promoter was evidenced by an electrophoretic mobility shift assay. The POU-domain of Brn-2 was sufficient for such interaction. The results thus suggest that the activation of the neuronal promoter of the aromatic L-amino acid decarboxylase gene requires a direct interaction between the ubiquitous NF-Y factor and a cell-specific POU-domain protein. The NF-Y, but not the Brn-2 binding site, is essential for the recruitment of the NF-Y/Brn-2 complex on the promoter.
Collapse
Affiliation(s)
- C Dugast
- Laboratoire de Biologie Moléculaire Eucaryote, CNRS UMR 5099, 118 route de Narbonne, 31062 Toulouse, France
| | | |
Collapse
|