1
|
Hinostroza F, Araya-Duran I, Piñeiro A, Lobos I, Pastenes L. Transcription factor roles in the local adaptation to temperature in the Andean Spiny Toad Rhinella spinulosa. Sci Rep 2024; 14:15158. [PMID: 38956427 PMCID: PMC11220030 DOI: 10.1038/s41598-024-66127-5] [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: 05/31/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
Environmental temperature strongly influences the adaptation dynamics of amphibians, whose limited regulation capabilities render them susceptible to thermal oscillations. A central element of the adaptive strategies is the transcription factors (TFs), which act as master regulators that orchestrate stress responses, enabling species to navigate the fluctuations of their environment skillfully. Our study delves into the intricate relationship between TF expression and thermal adaptation mechanisms in the Rhinella spinulosa populations. We sought to elucidate the dynamic modulations of TF expression in prometamorphic and metamorphic tadpoles that inhabit two thermally contrasting environments (Catarpe and El Tatio Geyser, Chile) and which were exposed to two thermal treatments (25 °C vs. 20 °C). Our findings unravel an intriguing dichotomy in response strategies between these populations. First, results evidence the expression of 1374 transcription factors. Regarding the temperature shift, the Catarpe tadpoles show a multifaceted approach by up-regulating crucial TFs, including fosB, atf7, and the androgen receptor. These dynamic regulatory responses likely underpin the population's ability to navigate thermal fluctuations effectively. In stark contrast, the El Tatio tadpoles exhibit a more targeted response, primarily up-regulating foxc1. This differential expression suggests a distinct focus on specific TFs to mitigate the effects of temperature variations. Our study contributes to understanding the molecular mechanisms governing thermal adaptation responses and highlights the resilience and adaptability of amphibians in the face of ever-changing environmental conditions.
Collapse
Affiliation(s)
- Fernando Hinostroza
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
- Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Facultad de Ciencias de la Salud, Universidad Católica del Maule, Talca, Chile
- Escuela de Química y Farmacia, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
- Centro Para la Investigación Traslacional en Neurofarmacología, Universidad de Valparaíso, Valparaíso, Chile
| | - Ingrid Araya-Duran
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alejandro Piñeiro
- Laboratorio de Genética y Microevolución, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile
| | - Isabel Lobos
- Laboratorio de Genética y Microevolución, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile
| | - Luis Pastenes
- Laboratorio de Genética y Microevolución, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile.
| |
Collapse
|
2
|
Wang H, Lin Z, Nian Z, Zhang W, Liu W, Yan F, Xiao Z, Wang X, Zhang Z, Ma Z, Liu Z. Hematopoietic transcription factor GFI1 promotes anchorage independence by sustaining ERK activity in cancer cells. J Clin Invest 2022; 132:149551. [PMID: 35819844 PMCID: PMC9433100 DOI: 10.1172/jci149551] [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: 03/15/2021] [Accepted: 07/07/2022] [Indexed: 11/26/2022] Open
Abstract
The switch from anchorage-dependent to anchorage-independent growth is essential for epithelial metastasis. The underlying mechanism, however, is not fully understood. In this study, we identified growth factor independent-1 (GFI1), a transcription factor that drives the transition from adherent endothelial cells to suspended hematopoietic cells during hematopoiesis, as a critical regulator of anchorage independence in lung cancer cells. GFI1 elevated the numbers of circulating and lung-infiltrating tumor cells in xenograft models and predicted poor prognosis of patients with lung cancer. Mechanistically, GFI1 inhibited the expression of multiple adhesion molecules and facilitated substrate detachment. Concomitantly, GFI1 reconfigured the chromatin structure of the RASGRP2 gene and increased its expression, causing Rap1 activation and subsequent sustained ERK activation upon detachment, and this led to ERK signaling dependency in tumor cells. Our studies unveiled a mechanism by which carcinoma cells hijacked a hematopoietic factor to gain anchorage independence and suggested that the intervention of ERK signaling may suppress metastasis and improve the therapeutic outcome of patients with GFI1-positive lung cancer.
Collapse
Affiliation(s)
- Hao Wang
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Zhenzhen Lin
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Zhe Nian
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Wei Zhang
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Wenxu Liu
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Fei Yan
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Zengtuan Xiao
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xia Wang
- Department of Gastrointestinal Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhenfa Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhenyi Ma
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Zhe Liu
- Department of Immunology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
3
|
Curcumin as an Epigenetic Therapeutic Agent in Myelodysplastic Syndromes (MDS). Int J Mol Sci 2021; 23:ijms23010411. [PMID: 35008835 PMCID: PMC8745143 DOI: 10.3390/ijms23010411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/19/2022] Open
Abstract
Growth Factor Independence 1 (GFI1) is a transcription factor with an important role in the regulation of development of myeloid and lymphoid cell lineages and was implicated in the development of myelodysplastic syndrome (MDS) and acute myeloid leukaemia (AML). Reduced expression of GFI1 or presence of the GFI1-36N (serine replaced with asparagine) variant leads to epigenetic changes in human and murine AML blasts and accelerated the development of leukaemia in a murine model of human MDS and AML. We and other groups previously showed that the GFI1-36N allele or reduced expression of GFI1 in human AML blasts is associated with an inferior prognosis. Using GFI1-36S, -36N -KD, NUP98-HOXD13-tg mice and curcumin (a natural histone acetyltransferase inhibitor (HATi)), we now demonstrate that expansion of GFI1-36N or –KD, NUP98-HODXD13 leukaemic cells can be delayed. Curcumin treatment significantly reduced AML progression in GFI1-36N or -KD mice and prolonged AML-free survival. Of note, curcumin treatment had no effect in GFI1-36S, NUP98-HODXD13 expressing mice. On a molecular level, curcumin treatment negatively affected open chromatin structure in the GFI1-36N or -KD haematopoietic cells but not GFI1-36S cells. Taken together, our study thus identified a therapeutic role for curcumin treatment in the treatment of AML patients (homo or heterozygous for GFI1-36N or reduced GFI1 expression) and possibly improved therapy outcome.
Collapse
|
4
|
Timmerman DM, Remmers TL, Hillenius S, Looijenga LHJ. Mechanisms of TP53 Pathway Inactivation in Embryonic and Somatic Cells-Relevance for Understanding (Germ Cell) Tumorigenesis. Int J Mol Sci 2021; 22:ijms22105377. [PMID: 34065345 PMCID: PMC8161298 DOI: 10.3390/ijms22105377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 01/10/2023] Open
Abstract
The P53 pathway is the most important cellular pathway to maintain genomic and cellular integrity, both in embryonic and non-embryonic cells. Stress signals induce its activation, initiating autophagy or cell cycle arrest to enable DNA repair. The persistence of these signals causes either senescence or apoptosis. Over 50% of all solid tumors harbor mutations in TP53 that inactivate the pathway. The remaining cancers are suggested to harbor mutations in genes that regulate the P53 pathway such as its inhibitors Mouse Double Minute 2 and 4 (MDM2 and MDM4, respectively). Many reviews have already been dedicated to P53, MDM2, and MDM4, while this review additionally focuses on the other factors that can deregulate P53 signaling. We discuss that P14ARF (ARF) functions as a negative regulator of MDM2, explaining the frequent loss of ARF detected in cancers. The long non-coding RNA Antisense Non-coding RNA in the INK4 Locus (ANRIL) is encoded on the same locus as ARF, inhibiting ARF expression, thus contributing to the process of tumorigenesis. Mutations in tripartite motif (TRIM) proteins deregulate P53 signaling through their ubiquitin ligase activity. Several microRNAs (miRNAs) inactivate the P53 pathway through inhibition of translation. CCCTC-binding factor (CTCF) maintains an open chromatin structure at the TP53 locus, explaining its inactivation of CTCF during tumorigenesis. P21, a downstream effector of P53, has been found to be deregulated in different tumor types. This review provides a comprehensive overview of these factors that are known to deregulate the P53 pathway in both somatic and embryonic cells, as well as their malignant counterparts (i.e., somatic and germ cell tumors). It provides insights into which aspects still need to be unraveled to grasp their contribution to tumorigenesis, putatively leading to novel targets for effective cancer therapies.
Collapse
|
5
|
Ferone G, Song JY, Krijgsman O, van der Vliet J, Cozijnsen M, Semenova EA, Adams DJ, Peeper D, Berns A. FGFR1 Oncogenic Activation Reveals an Alternative Cell of Origin of SCLC in Rb1/p53 Mice. Cell Rep 2021; 30:3837-3850.e3. [PMID: 32187553 PMCID: PMC7090386 DOI: 10.1016/j.celrep.2020.02.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/02/2019] [Accepted: 02/11/2020] [Indexed: 12/28/2022] Open
Abstract
Fibroblast growth factor receptor 1 (FGFR1) is frequently amplified in human small-cell lung cancer (SCLC), but its contribution to SCLC and other lung tumors has remained elusive. Here, we assess the tumorigenic capacity of constitutive-active FGFR1 (FGFR1K656E) with concomitant RB and P53 depletion in mouse lung. Our results reveal a context-dependent effect of FGFR1K656E: it impairs SCLC development from CGRPPOS neuroendocrine (NE) cells, which are considered the major cell of origin of SCLC, whereas it promotes SCLC and low-grade NE bronchial lesions from tracheobronchial-basal cells. Moreover, FGFR1K656E induces lung adenocarcinoma (LADC) from most lung cell compartments. However, its expression is not sustained in LADC originating from CGRPPOS cells. Therefore, cell context and tumor stage should be taken into account when considering FGFR1 inhibition as a therapeutic option. FGRF signaling impairs SCLC development initiated from CGRPPOS NE cells FGRF signaling promotes development of NE tumors initiated from K14POS cells Rb;p53;Fgfr1 mice develop ADCs that retain signatures of their cell of origin Low expression of Fgfr1 in progressed tumors suggests a role in tumor initiation
Collapse
Affiliation(s)
- Giustina Ferone
- Oncode Institute, Division of Molecular Genetics, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Ji-Ying Song
- Division of Molecular Oncology, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Oscar Krijgsman
- Oncode Institute, Department of Experimental Animal Pathology, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Jan van der Vliet
- Oncode Institute, Division of Molecular Genetics, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Miranda Cozijnsen
- Oncode Institute, Division of Molecular Genetics, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Ekaterina A Semenova
- Oncode Institute, Division of Molecular Genetics, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - David J Adams
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Daniel Peeper
- Oncode Institute, Department of Experimental Animal Pathology, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Anton Berns
- Oncode Institute, Division of Molecular Genetics, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Ashour N, Angulo JC, González-Corpas A, Orea MJ, Lobo MVT, Colomer R, Colás B, Esteller M, Ropero S. Epigenetic Regulation of Gfi1 in Endocrine-Related Cancers: a Role Regulating Tumor Growth. Int J Mol Sci 2020; 21:ijms21134687. [PMID: 32630147 PMCID: PMC7370116 DOI: 10.3390/ijms21134687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023] Open
Abstract
Prostate and breast cancer constitute the most common cancers among men and women worldwide. The aging population is one of the main risk factors for prostate and breast cancer development and accumulating studies link aging with epigenetic changes. Growth factor independence-1 (Gfi1) is a transcriptional repressor with an important role in human malignancies, including leukemia, colorectal carcinoma, and lung cancer, but its role in prostate and breast cancer is unknown. We have found that Gfi1 epigenetic silencing is a common event in prostate and breast cancer. Gfi1 re-expression in prostate and breast cancer cell lines displaying Gfi1 epigenetic silencing decreases cell proliferation, reduced colony formation density, and tumor growth in nude mice xenografts. In addition, we found that Gfi1 repress alpha 1-anti-trypsin (AAT) and alpha 1-anti-chymotrypsin (ACT) expression, two genes with important functions in cancer development, suggesting that Gfi1 silencing promotes tumor growth by increasing AAT and ACT expression in our system. Finally, Gfi1 epigenetic silencing could be a promising biomarker for prostate cancer progression because it is associated with shorter disease-free survival. In conclusion, our findings strongly indicate that Gfi1 epigenetic silencing in prostate and breast cancer could be a crucial step in the development of these two-well characterized endocrine related tumors.
Collapse
Affiliation(s)
- Nadia Ashour
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
| | - Javier C. Angulo
- Servicio de Urología, Hospital Universitario de Getafe, Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Universidad Europea de Madrid, Getafe, 28905 Madrid, Spain;
| | - Ana González-Corpas
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
| | - María J. Orea
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
| | - María V. T. Lobo
- Departamento de Biomedicina y Biotecnología, Universidad de Alcalá; Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), 28054 Madrid, Spain;
| | - Ramón Colomer
- Medical Oncology Department, Instituto De Investigación Sanitaria La Princesa, HU La Princesa, 28029 Madrid, Spain;
- Department of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Begoña Colás
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Catalonia, Spain;
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28040 Madrid, Spain
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Catalonia, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), 08028 Barcelona, Catalonia, Spain
| | - Santiago Ropero
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
- Correspondence:
| |
Collapse
|
7
|
Tang Q, Xu M, Xu J, Xie X, Yang H, Gan L. Gfi1-GCE inducible Cre line for hair cell-specific gene manipulation in mouse inner ear. Genesis 2019; 57:e23304. [PMID: 31077553 DOI: 10.1002/dvg.23304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 01/16/2023]
Abstract
Tissue-specific inducible Cre recombinase mouse lines allow precise genetic manipulations in spatiotemporal manners and are pivotal for functional studies of genes during development and in adults. Growth factor independence 1 (GFI1) is an essential transcription factor expressed in the hair cells of mouse inner ear and Gfi1 locus serves as an excellent anchor site to drive the expression of inducible Cre recombinase in mouse inner hair cells. In this study, we have generated Gfi1-P2A-GFP-CreERT2 (Gfi1-GCE) knock-in mouse line by in-frame fusion of a self-cleaving GCE to the C-terminus of GFI1. We have shown that as predicted, the expression of GCE and GFI1 was detected specifically in the cytosol and nuclei of hair cells, respectively, of uninduced Gfi1-GCE mice, suggesting the successful cleavage and simultaneous expression of GFI1 and GCE. In addition, the in-frame fusion of the self-cleaving GCE does not interrupt the function of Gfi1 in the inner ear. Administration of tamoxifen leads to nuclear translocation of GCE and results in an efficient activation of tdTomato reporter gene expression specifically in most hair cells throughout development and in adults. Thus, this inducible Gfi1-GCE mouse line is a highly efficient Cre deleter and is suitable for gene manipulation in developing and adult inner ear hair cells.
Collapse
Affiliation(s)
- Qi Tang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York
| | - Mei Xu
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York.,Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jiadong Xu
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York.,Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaoling Xie
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York
| | - Hua Yang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Gan
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York
| |
Collapse
|
8
|
García-Gutiérrez L, Delgado MD, León J. MYC Oncogene Contributions to Release of Cell Cycle Brakes. Genes (Basel) 2019; 10:E244. [PMID: 30909496 PMCID: PMC6470592 DOI: 10.3390/genes10030244] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Promotion of the cell cycle is a major oncogenic mechanism of the oncogene c-MYC (MYC). MYC promotes the cell cycle by not only activating or inducing cyclins and CDKs but also through the downregulation or the impairment of the activity of a set of proteins that act as cell-cycle brakes. This review is focused on the role of MYC as a cell-cycle brake releaser i.e., how MYC stimulates the cell cycle mainly through the functional inactivation of cell cycle inhibitors. MYC antagonizes the activities and/or the expression levels of p15, ARF, p21, and p27. The mechanism involved differs for each protein. p15 (encoded by CDKN2B) and p21 (CDKN1A) are repressed by MYC at the transcriptional level. In contrast, MYC activates ARF, which contributes to the apoptosis induced by high MYC levels. At least in some cells types, MYC inhibits the transcription of the p27 gene (CDKN1B) but also enhances p27's degradation through the upregulation of components of ubiquitin ligases complexes. The effect of MYC on cell-cycle brakes also opens the possibility of antitumoral therapies based on synthetic lethal interactions involving MYC and CDKs, for which a series of inhibitors are being developed and tested in clinical trials.
Collapse
Affiliation(s)
- Lucía García-Gutiérrez
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Department of Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain.
- Current address: Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland.
| | - María Dolores Delgado
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Department of Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain.
| | - Javier León
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Department of Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain.
| |
Collapse
|
9
|
Chen MS, Lo YH, Chen X, Williams CS, Donnelly JM, Criss ZK, Patel S, Butkus JM, Dubrulle J, Finegold MJ, Shroyer NF. Growth Factor-Independent 1 Is a Tumor Suppressor Gene in Colorectal Cancer. Mol Cancer Res 2019; 17:697-708. [PMID: 30606770 DOI: 10.1158/1541-7786.mcr-18-0666] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/20/2018] [Accepted: 12/19/2018] [Indexed: 12/27/2022]
Abstract
Colorectal cancer is the third most common cancer and the third leading cause of cancer death in the United States. Growth factor-independent 1 (GFI1) is a zinc finger transcriptional repressor responsible for controlling secretory cell differentiation in the small intestine and colon. GFI1 plays a significant role in the development of human malignancies, including leukemia, lung cancer, and prostate cancer. However, the role of GFI1 in colorectal cancer progression is largely unknown. Our results demonstrate that RNA and protein expression of GFI1 are reduced in advanced-stage nonmucinous colorectal cancer. Subcutaneous tumor xenograft models demonstrated that the reexpression of GFI1 in 4 different human colorectal cancer cell lines inhibits tumor growth. To further investigate the role of Gfi1 in de novo colorectal tumorigenesis, we developed transgenic mice harboring a deletion of Gfi1 in the colon driven by CDX2-cre (Gfi1F/F; CDX2-cre) and crossed them with ApcMin/+ mice (ApcMin/+; Gfi1F/F; CDX2-cre). Loss of Gfi1 significantly increased the total number of colorectal adenomas compared with littermate controls with an APC mutation alone. Furthermore, we found that compound (ApcMin/+; Gfi1F/F; CDX2-cre) mice develop larger adenomas, invasive carcinoma, as well as hyperplastic lesions expressing the neuroendocrine marker chromogranin A, a feature that has not been previously described in APC-mutant tumors in mice. Collectively, these results demonstrate that GFI1 acts as a tumor suppressor gene in colorectal cancer, where deficiency of Gfi1 promotes malignancy in the colon. IMPLICATIONS: These findings reveal that GFI1 functions as a tumor suppressor gene in colorectal tumorigenesis.
Collapse
Affiliation(s)
- Min-Shan Chen
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas
| | - Yuan-Hung Lo
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas
| | - Xi Chen
- Department of Public Health Sciences, University of Miami, Miami, Florida
| | - Christopher S Williams
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University, Nashville, Tennessee
| | - Jessica M Donnelly
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas
| | - Zachary K Criss
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas
| | - Shreena Patel
- Department of Pediatrics, Section of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas
| | - Joann M Butkus
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas.,Summer Undergraduate Research Training Program, Baylor College of Medicine, Houston Texas.,Susquehanna University, Selinsgrove, Pennsylvania
| | - Julien Dubrulle
- Integrated Microscopy Core, Baylor College of Medicine, Houston, Texas
| | - Milton J Finegold
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Noah F Shroyer
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas. .,Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas.,Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
10
|
Ningning S, Libo S, Chuanbin W, Haijiang S, Qing Z. RETRACTED: MiR-650 regulates the proliferation, migration and invasion of human oral cancer by targeting growth factor independent 1 (Gfi1). Biochimie 2019; 156:69-78. [PMID: 30296465 DOI: 10.1016/j.biochi.2018.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/04/2018] [Indexed: 01/11/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief. Following concerns raised by Dr. E. Bik and an anonymous reader, the journal conducted an investigation and found evidence that there had been improper manipulation and duplication of images in Figures 2A, 5C and 6C, as shown by partial overlap between images representing different experiments. An anonymous reader has also drawn attention to flow-cytometry data in Figure 1 similar to previously published data from a different laboratory (Med Sci Monit, 2018; 24: 5874-5880 DOI: 10.12659/MSM.909682, Figure 2B). After raising the complaint with the authors, the corresponding author agreed that the paper needed to be retracted. The authors apologise for any confusion that may have arisen from their article.
Collapse
Affiliation(s)
- Sun Ningning
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, Liaoning, China
| | - Sun Libo
- Department of Oral and Maxillofacial Surgery, Southwest Medical University Affiliated Stomatological Hospital, Luzhou, Sichuan, 646000, China
| | - Wu Chuanbin
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, Liaoning, China
| | - Sun Haijiang
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, Liaoning, China
| | - Zhou Qing
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, Liaoning, China.
| |
Collapse
|
11
|
Petrusca DN, Toscani D, Wang FM, Park C, Crean CD, Anderson JL, Marino S, Mohammad KS, Zhou D, Silbermann R, Sun Q, Kurihara N, Galson DL, Giuliani N, Roodman GD. Growth factor independence 1 expression in myeloma cells enhances their growth, survival, and osteoclastogenesis. J Hematol Oncol 2018; 11:123. [PMID: 30286780 PMCID: PMC6172782 DOI: 10.1186/s13045-018-0666-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/19/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In spite of major advances in treatment, multiple myeloma (MM) is currently an incurable malignancy due to the emergence of drug-resistant clones. We previously showed that MM cells upregulate the transcriptional repressor, growth factor independence 1 (Gfi1), in bone marrow stromal cells (BMSCs) that induces prolonged inhibition of osteoblast differentiation. However, the role of Gfi1 in MM cells is unknown. METHODS Human primary CD138+ and BMSC were purified from normal donors and MM patients' bone marrow aspirates. Gfi1 knockdown and overexpressing cells were generated by lentiviral-mediated shRNA. Proliferation/apoptosis studies were done by flow cytometry, and protein levels were determined by Western blot and/or immunohistochemistry. An experimental MM mouse model was generated to investigate the effects of MM cells overexpressing Gfi1 on tumor burden and osteolysis in vivo. RESULTS We found that Gfi1 expression is increased in patient's MM cells and MM cell lines and was further increased by co-culture with BMSC, IL-6, and sphingosine-1-phosphate. Modulation of Gfi1 in MM cells had major effects on their survival and growth. Knockdown of Gfi1 induced apoptosis in p53-wt, p53-mutant, and p53-deficient MM cells, while Gfi1 overexpression enhanced MM cell growth and protected MM cells from bortezomib-induced cell death. Gfi1 enhanced cell survival of p53-wt MM cells by binding to p53, thereby blocking binding to the promoters of the pro-apoptotic BAX and NOXA genes. Further, Gfi1-p53 binding could be blocked by HDAC inhibitors. Importantly, inoculation of MM cells overexpressing Gfi1 in mice induced increased bone destruction, increased osteoclast number and size, and enhanced tumor growth. CONCLUSIONS These results support that Gfi1 plays a key role in MM tumor growth, survival, and bone destruction and contributes to bortezomib resistance, suggesting that Gfi1 may be a novel therapeutic target for MM.
Collapse
Affiliation(s)
- Daniela N Petrusca
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA.
| | - Denise Toscani
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA.,Myeloma Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Feng-Ming Wang
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA.,Endodontics, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - Cheolkyu Park
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA
| | - Colin D Crean
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA
| | - Judith L Anderson
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA
| | - Silvia Marino
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA
| | - Khalid S Mohammad
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dan Zhou
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA
| | - Rebecca Silbermann
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA
| | - Quanhong Sun
- Department of Medicine, Division of Hematology-Oncology, UPMC Hillman Cancer Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Noriyoshi Kurihara
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA
| | - Deborah L Galson
- Department of Medicine, Division of Hematology-Oncology, UPMC Hillman Cancer Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicola Giuliani
- Myeloma Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - G David Roodman
- Department of Medicine, Division of Hematology-Oncology, Indiana University School of Medicine, 980 Walnut Street, Walther Hall, Room C346, Indianapolis, IN, 46202, USA.,Rodebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
12
|
Vadnais C, Chen R, Fraszczak J, Yu Z, Boulais J, Pinder J, Frank D, Khandanpour C, Hébert J, Dellaire G, Côté JF, Richard S, Orthwein A, Drobetsky E, Möröy T. GFI1 facilitates efficient DNA repair by regulating PRMT1 dependent methylation of MRE11 and 53BP1. Nat Commun 2018; 9:1418. [PMID: 29651020 PMCID: PMC5897347 DOI: 10.1038/s41467-018-03817-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 03/08/2018] [Indexed: 01/01/2023] Open
Abstract
GFI1 is a transcriptional regulator expressed in lymphoid cells, and an “oncorequisite” factor required for development and maintenance of T-lymphoid leukemia. GFI1 deletion causes hypersensitivity to ionizing radiation, for which the molecular mechanism remains unknown. Here, we demonstrate that GFI1 is required in T cells for the regulation of key DNA damage signaling and repair proteins. Specifically, GFI1 interacts with the arginine methyltransferase PRMT1 and its substrates MRE11 and 53BP1. We demonstrate that GFI1 enables PRMT1 to bind and methylate MRE11 and 53BP1, which is necessary for their function in the DNA damage response. Thus, our results provide evidence that GFI1 can adopt non-transcriptional roles, mediating the post-translational modification of proteins involved in DNA repair. These findings have direct implications for treatment responses in tumors overexpressing GFI1 and suggest that GFI1’s activity may be a therapeutic target in these malignancies. The transcription factor GFI1 mediates the DNA damage response (DDR) of T cells through a yet unknown mechanism. Here the authors show that GFI1 can adopt non-transcriptional roles during DDR, enabling PRMT1 to bind and methylate the DNA repair proteins MRE11 and 53BP1.
Collapse
Affiliation(s)
- Charles Vadnais
- Institut de Recherches Cliniques de Montréal, IRCM, Montréal, QC, H2W 1R7, Canada
| | - Riyan Chen
- Institut de Recherches Cliniques de Montréal, IRCM, Montréal, QC, H2W 1R7, Canada
| | - Jennifer Fraszczak
- Institut de Recherches Cliniques de Montréal, IRCM, Montréal, QC, H2W 1R7, Canada
| | - Zhenbao Yu
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, H3T 1E2, Canada
| | - Jonathan Boulais
- Institut de Recherches Cliniques de Montréal, IRCM, Montréal, QC, H2W 1R7, Canada
| | - Jordan Pinder
- Departments of Pathology and Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Daria Frank
- Department of Hematology, University Hospital, Essen, 45147, Germany
| | - Cyrus Khandanpour
- Department of Hematology, University Hospital, Essen, 45147, Germany
| | - Josée Hébert
- Department of Medicine, Université de Montréal, Montreal, H3T 1J4, QC, Canada.,Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, H1T 2M4, QC, Canada.,Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montreal, H1T 2M4, QC, Canada
| | - Graham Dellaire
- Departments of Pathology and Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Jean-François Côté
- Institut de Recherches Cliniques de Montréal, IRCM, Montréal, QC, H2W 1R7, Canada.,Département de Médecine, Université de Montréal and Centre de Recherche, Hôpital Maisonneuve Rosemont, Montréal, QC, H1T 2M4, Canada
| | - Stéphane Richard
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, H3T 1E2, Canada.,Deparment of Medicine, McGill University, Montreal, H4A 3J1, QC, Canada.,Department of Oncology, McGill University, Montreal, QC, H4A 3T2, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Alexandre Orthwein
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, H3T 1E2, Canada.,Department of Oncology, McGill University, Montreal, QC, H4A 3T2, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, H3A 1A3, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Elliot Drobetsky
- Département de Médecine, Université de Montréal and Centre de Recherche, Hôpital Maisonneuve Rosemont, Montréal, QC, H1T 2M4, Canada
| | - Tarik Möröy
- Institut de Recherches Cliniques de Montréal, IRCM, Montréal, QC, H2W 1R7, Canada. .,Division of Experimental Medicine, McGill University, Montreal, QC, H3A 1A3, Canada. .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H3C 3J7, Canada.
| |
Collapse
|
13
|
Kim DW, Kim KC, Kim KB, Dunn CT, Park KS. Transcriptional deregulation underlying the pathogenesis of small cell lung cancer. Transl Lung Cancer Res 2018. [PMID: 29535909 DOI: 10.21037/tlcr.2017.10.07] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The discovery of recurrent alterations in genes encoding transcription regulators and chromatin modifiers is one of the most important recent developments in the study of the small cell lung cancer (SCLC) genome. With advances in models and analytical methods, the field of SCLC biology has seen remarkable progress in understanding the deregulated transcription networks linked to the tumor development and malignant progression. This review will discuss recent discoveries on the roles of RB and P53 family of tumor suppressors and MYC family of oncogenes in tumor initiation and development. It will also describe the roles of lineage-specific factors in neuroendocrine (NE) cell differentiation and homeostasis and the roles of epigenetic alterations driven by changes in NFIB and chromatin modifiers in malignant progression and chemoresistance. These recent findings have led to a model of transcriptional network in which multiple pathways converge on regulatory regions of crucial genes linked to tumor development. Validation of this model and characterization of target genes will provide critical insights into the biology of SCLC and novel strategies for tumor intervention.
Collapse
Affiliation(s)
- Dong-Wook Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Keun-Cheol Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA.,Department of Biological Sciences, Kangwon National University, Chuncheon, Korea
| | - Kee-Beom Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Colin T Dunn
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Kwon-Sik Park
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
14
|
Westerman BA, Breuer RHJ, Poutsma A, Chhatta A, Noorduyn LA, Koolen MGJ, Postmus PE, Blankenstein MA, Oudejans CBM. Basic Helix-Loop-Helix Transcription Factor Profiling of Lung Tumors Shows Aberrant Expression of the Proneural Gene Atonal Homolog 1 (ATOH1, HATH1, MATH1) in Neuroendocrine Tumors. Int J Biol Markers 2018; 22:114-23. [PMID: 17549667 DOI: 10.1177/172460080702200205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Microarray-based expression profiling studies of lung adenocarcinomas have defined neuroendocrine subclasses with poor prognosis. As neuroendocrine development is regulated by members of the achaetescute and atonal classes of basic helix-loop-helix (bHLH) transcription factors, we analyzed lung tumors for expression of these factors. Out of 13 bHLH genes tested, 4 genes, i.e., achaetescute complex-like 1 (ASCL1, HASH1, Mash1), atonal homolog 1 (ATOH1, HATH1, MATH1), NEUROD4 (ATH-3, Atoh3, MATH-3) and neurogenic differentiation factor 1 (NEUROD1, NEUROD, BE-TA2), showed differential expression among lung tumors and absent or low expression in normal lung. As expected, tumors that have high levels of ASCL1 also express neuroendocrine markers, and we found that this is accompanied by increased levels of NEUROD1. In addition, we found ATOH1 expression in 9 (16%) out of 56 analyzed adenocarcinomas and these tumors showed neuroendocrine features as shown by dopa decarboxylase mRNA expression and immunostaining for neuroendocrine markers. ATOH1 expression as well as NEUROD4 was observed in small cell lung carcinoma (SCLC), a known neuroendocrine tumor. Since ATOH1 is not known to be involved in normal lung development, our results suggest that aberrant activation of ATOH1 leads to a neuroendocrine phenotype similar to what is observed for ASCL1 activation during normal neuroendocrine development and in lung malignancies. Our preliminary data indicate that patients with ATOH1-expressing adenocarcinomas might have a worse prognosis.
Collapse
Affiliation(s)
- B A Westerman
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Costa A, Powell LM, Lowell S, Jarman AP. Atoh1 in sensory hair cell development: constraints and cofactors. Semin Cell Dev Biol 2017; 65:60-68. [DOI: 10.1016/j.semcdb.2016.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 09/26/2016] [Accepted: 10/13/2016] [Indexed: 11/28/2022]
|
16
|
Škalamera D, Dahmer-Heath M, Stevenson AJ, Pinto C, Shah ET, Daignault SM, Said NAB, Davis M, Haass NK, Williams ED, Hollier BG, Thompson EW, Gabrielli B, Gonda TJ. Genome-wide gain-of-function screen for genes that induce epithelial-to-mesenchymal transition in breast cancer. Oncotarget 2016; 7:61000-61020. [PMID: 27876705 PMCID: PMC5308632 DOI: 10.18632/oncotarget.11314] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/27/2016] [Indexed: 01/08/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a developmental program that has been implicated in progression, metastasis and therapeutic resistance of some carcinomas. To identify genes whose overexpression drives EMT, we screened a lentiviral expression library of 17000 human open reading frames (ORFs) using high-content imaging to quantitate cytoplasmic vimentin. Hits capable of increasing vimentin in the mammary carcinoma-derived cell line MDA-MB-468 were confirmed in the non-tumorigenic breast-epithelial cell line MCF10A. When overexpressed in this model, they increased the rate of cell invasion through Matrigel™, induced mesenchymal marker expression and reduced expression of the epithelial marker E-cadherin. In gene-expression datasets derived from breast cancer patients, the expression of several novel genes correlated with expression of known EMT marker genes, indicating their in vivo relevance. As EMT-associated properties are thought to contribute in several ways to cancer progression, genes identified in this study may represent novel targets for anti-cancer therapy.
Collapse
Affiliation(s)
- Dubravka Škalamera
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Mareike Dahmer-Heath
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Alexander J. Stevenson
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Cletus Pinto
- St Vincent's Institute of Medical Research and University of Melbourne Department of Surgery, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Esha T. Shah
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, QLD, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Sheena M. Daignault
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Nur Akmarina B.M. Said
- Monash Institute of Medical Research (now Hudson Institute of Medical Research), Monash University, Melbourne, VIC, Australia
- University of Malaya, Kuala Lumpur, Malaysia
| | - Melissa Davis
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Nikolas K. Haass
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Elizabeth D. Williams
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, QLD, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
- Monash Institute of Medical Research (now Hudson Institute of Medical Research), Monash University, Melbourne, VIC, Australia
| | - Brett G. Hollier
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, QLD, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Erik W. Thompson
- St Vincent's Institute of Medical Research and University of Melbourne Department of Surgery, St. Vincent's Hospital, Melbourne, VIC, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Brian Gabrielli
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Thomas J. Gonda
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- School of Pharmacy, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
17
|
Thambyrajah R, Patel R, Mazan M, Lie-a-Ling M, Lilly A, Eliades A, Menegatti S, Garcia-Alegria E, Florkowska M, Batta K, Kouskoff V, Lacaud G. New insights into the regulation by RUNX1 and GFI1(s) proteins of the endothelial to hematopoietic transition generating primordial hematopoietic cells. Cell Cycle 2016; 15:2108-2114. [PMID: 27399214 PMCID: PMC4993433 DOI: 10.1080/15384101.2016.1203491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 10/26/2022] Open
Abstract
The first hematopoietic cells are generated very early in ontogeny to support the growth of the embryo and to provide the foundation to the adult hematopoietic system. There is a considerable therapeutic interest in understanding how these first blood cells are generated in order to try to reproduce this process in vitro. This would allow generating blood products, or hematopoietic cell populations from embryonic stem (ES) cells, induced pluripotent stem cells or through directed reprogramming. Recent studies have clearly established that the first hematopoietic cells originate from a hemogenic endothelium (HE) through an endothelial to hematopoietic transition (EHT). The molecular mechanisms underlining this transition remain largely unknown with the exception that the transcription factor RUNX1 is critical for this process. In this Extra Views report, we discuss our recent studies demonstrating that the transcriptional repressors GFI1 and GFI1B have a critical role in the EHT. We established that these RUNX1 transcriptional targets are actively implicated in the downregulation of the endothelial program and the loss of endothelial identity during the formation of the first blood cells. In addition, our results suggest that GFI1 expression provides an ideal novel marker to identify, isolate and study the HE cell population.
Collapse
Affiliation(s)
- Roshana Thambyrajah
- CRUK Stem Cell Biology, Cancer Research UK Manchester Institute, Manchester, UK
| | - Rahima Patel
- CRUK Stem Cell Biology, Cancer Research UK Manchester Institute, Manchester, UK
| | - Milena Mazan
- CRUK Stem Cell Biology, Cancer Research UK Manchester Institute, Manchester, UK
| | - Michael Lie-a-Ling
- CRUK Stem Cell Biology, Cancer Research UK Manchester Institute, Manchester, UK
| | - Andrew Lilly
- CRUK Stem Cell Haematopoiesis, Cancer Research UK Manchester Institute, Manchester, UK
| | - Alexia Eliades
- CRUK Stem Cell Haematopoiesis, Cancer Research UK Manchester Institute, Manchester, UK
| | - Sara Menegatti
- CRUK Stem Cell Haematopoiesis, Cancer Research UK Manchester Institute, Manchester, UK
| | - Eva Garcia-Alegria
- CRUK Stem Cell Haematopoiesis, Cancer Research UK Manchester Institute, Manchester, UK
| | | | - Kiran Batta
- CRUK Stem Cell Biology, Cancer Research UK Manchester Institute, Manchester, UK
| | - Valerie Kouskoff
- CRUK Stem Cell Haematopoiesis, Cancer Research UK Manchester Institute, Manchester, UK
| | - Georges Lacaud
- CRUK Stem Cell Biology, Cancer Research UK Manchester Institute, Manchester, UK
| |
Collapse
|
18
|
Botezatu L, Michel LC, Helness A, Vadnais C, Makishima H, Hönes JM, Robert F, Vassen L, Thivakaran A, Al-Matary Y, Lams RF, Schütte J, Giebel B, Görgens A, Heuser M, Medyouf H, Maciejewski J, Dührsen U, Möröy T, Khandanpour C. Epigenetic therapy as a novel approach for GFI136N-associated murine/human AML. Exp Hematol 2016; 44:713-726.e14. [PMID: 27216773 DOI: 10.1016/j.exphem.2016.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 02/02/2023]
Abstract
Epigenetic changes can contribute to development of acute myeloid leukemia (AML), a malignant disease of the bone marrow. A single-nucleotide polymorphism of transcription factor growth factor independence 1 (GFI1) generates a protein with an asparagine at position 36 (GFI1(36N)) instead of a serine at position 36 (GFI1(36S)), which is associated with de novo AML in humans. However, how GFI1(36N) predisposes to AML is poorly understood. To explore the mechanism, we used knock-in mouse strains expressing GFI1(36N) or GFI1(36S). Presence of GFI1(36N) shortened the latency and increased the incidence of AML in different murine models of myelodysplastic syndrome/AML. On a molecular level, GFI1(36N) induced genomewide epigenetic changes, leading to expression of AML-associated genes. On a therapeutic level, use of histone acetyltransferase inhibitors specifically impeded growth of GFI1(36N)-expressing human and murine AML cells in vitro and in vivo. These results establish, as a proof of principle, how epigenetic changes in GFI1(36N)-induced AML can be targeted.
Collapse
Affiliation(s)
- Lacramioara Botezatu
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lars C Michel
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Anne Helness
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Charles Vadnais
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Hideki Makishima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH
| | - Judith M Hönes
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - François Robert
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada; Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Lothar Vassen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Aniththa Thivakaran
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Yahya Al-Matary
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Robert F Lams
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Judith Schütte
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - André Görgens
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Hind Medyouf
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Jaroslaw Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Tarik Möröy
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada; Department of Hematology and Oncology, University Hospital Düsseldorf, Düsseldorf, Germany; Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada.
| | - Cyrus Khandanpour
- Department of Hematology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
19
|
SUMOylation Regulates Growth Factor Independence 1 in Transcriptional Control and Hematopoiesis. Mol Cell Biol 2016; 36:1438-50. [PMID: 26951200 DOI: 10.1128/mcb.01001-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/20/2016] [Indexed: 01/08/2023] Open
Abstract
Cell fate specification requires precise coordination of transcription factors and their regulators to achieve fidelity and flexibility in lineage allocation. The transcriptional repressor growth factor independence 1 (GFI1) is comprised of conserved Snail/Slug/Gfi1 (SNAG) and zinc finger motifs separated by a linker region poorly conserved with GFI1B, its closest homolog. Moreover, GFI1 and GFI1B coordinate distinct developmental fates in hematopoiesis, suggesting that their functional differences may derive from structures within their linkers. We show a binding interface between the GFI1 linker and the SP-RING domain of PIAS3, an E3-SUMO (small ubiquitin-related modifier) ligase. The PIAS3 binding region in GFI1 contains a conserved type I SUMOylation consensus element, centered on lysine-239 (K239). In silico prediction algorithms identify K239 as the only high-probability site for SUMO modification. We show that GFI1 is modified by SUMO at K239. SUMOylation-resistant derivatives of GFI1 fail to complement Gfi1 depletion phenotypes in zebrafish primitive erythropoiesis and granulocytic differentiation in cultured human cells. LSD1/CoREST recruitment and MYC repression by GFI1 are profoundly impaired for SUMOylation-resistant GFI1 derivatives, while enforced expression of MYC blocks granulocytic differentiation. These findings suggest that SUMOylation within the GFI1 linker favors LSD1/CoREST recruitment and MYC repression to govern hematopoietic differentiation.
Collapse
|
20
|
Wang L, Liu JL, Yu L, Liu XX, Wu HM, Lei FY, Wu S, Wang X. Downregulated miR-495 [Corrected] Inhibits the G1-S Phase Transition by Targeting Bmi-1 in Breast Cancer. Medicine (Baltimore) 2015; 94:e718. [PMID: 26020378 PMCID: PMC4616407 DOI: 10.1097/md.0000000000000718] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bmi-1 (B cell-specific Moloney murine leukemia virus integration site 1) is upregulated in breast cancer and was involved in many malignant progressions of breast cells, including cell proliferation, stem cell pluripotency, and cancer initiation. However, the epigenetic regulatory mechanism of Bmi-1 in breast cancer remains unclear. After analysis of the ArrayExpress dataset GSE45666, we comparatively detected the expression levels of miR-495 in 9 examined breast cancer cell lines, normal breast epithelial cells and 8 pairs of fresh clinical tumor samples. Furthermore, to evaluate the effect of miR-495 on the progression of breast cancer, MCF-7 and MDA-MB-231 were transduced to stably overexpress miR-495. The 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay, colony formation assays, 5-Bromo-2-deoxyUridine labeling and immunofluorescence, anchorage-independent growth ability assay, flow cytometry analysis, and luciferase assays were used to test the effect of miR-495 in MCF-7 and MDA-MB-231 cells in vitro. Xenografted tumor model was also used to evaluate the effect of miR-495 in breast cancer. Herein, we found that miR-495, a predicted regulator of Bmi-1, was frequently downregulated in malignant cells and tissues of breast. Upregulation of miR-495 significantly suppressed breast cancer cell proliferation and tumorigenicity via G1-S arrest. Further analysis revealed that miR-495 targeted Bmi-1 through its 3' untranslated region. Moreover, Bmi-1 could neutralize the suppressive effect of miR-495 on cell proliferation and tumorigenicity of breast cancer in vivo. These data suggested that miR-495 could inhibit the G1-S phase transition that leads to proliferation and tumorigenicity inhibition by targeting and suppressing Bmi-1 in breast cancer.
Collapse
Affiliation(s)
- Lan Wang
- From the Department of Pathogen Biology and Immunology, School of Basic Courses, Guangdong Pharmaceutical University (LW, H-MW); Department of Medical Oncology, Sun Yat-sen University Cancer Center (JLL); Department of Vascular and Breast Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou (LY); Department of Plastic Surgery, the First Affiliated Hospital of Sun Yat-sen University (X-XL); State Key Laboratory of Oncology in South China (F-YL, SW); State Key Laboratory of Oncology in Southern China, Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China (XW)
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Velu CS, Chaubey A, Phelan JD, Horman SR, Wunderlich M, Guzman ML, Jegga AG, Zeleznik-Le NJ, Chen J, Mulloy JC, Cancelas JA, Jordan CT, Aronow BJ, Marcucci G, Bhat B, Gebelein B, Grimes HL. Therapeutic antagonists of microRNAs deplete leukemia-initiating cell activity. J Clin Invest 2014; 124:222-36. [PMID: 24334453 PMCID: PMC3871218 DOI: 10.1172/jci66005] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/10/2013] [Indexed: 12/14/2022] Open
Abstract
Acute myelogenous leukemia (AML) subtypes that result from oncogenic activation of homeobox (HOX) transcription factors are associated with poor prognosis. The HOXA9 transcription activator and growth factor independent 1 (GFI1) transcriptional repressor compete for occupancy at DNA-binding sites for the regulation of common target genes. We exploited this HOXA9 versus GFI1 antagonism to identify the genes encoding microRNA-21 and microRNA-196b as transcriptional targets of HOX-based leukemia oncoproteins. Therapeutic inhibition of microRNA-21 and microRNA-196b inhibited in vitro leukemic colony forming activity and depleted in vivo leukemia-initiating cell activity of HOX-based leukemias, which led to leukemia-free survival in a murine AML model and delayed disease onset in xenograft models. These data establish microRNA as functional effectors of endogenous HOXA9 and HOX-based leukemia oncoproteins, provide a concise in vivo platform to test RNA therapeutics, and suggest therapeutic value for microRNA antagonists in AML.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Base Sequence
- Binding Sites
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Combined Modality Therapy
- Cytarabine/administration & dosage
- DNA-Binding Proteins/metabolism
- Doxorubicin/administration & dosage
- Gene Expression Regulation, Leukemic
- Homeodomain Proteins/metabolism
- Humans
- Induction Chemotherapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Myeloid Ecotropic Viral Integration Site 1 Protein
- Neoplasm Proteins/metabolism
- Neoplastic Stem Cells/physiology
- Phosphorothioate Oligonucleotides/genetics
- Pre-B-Cell Leukemia Transcription Factor 1
- Protein Binding
- Proto-Oncogene Proteins/metabolism
- Regulatory Sequences, Nucleic Acid
- Transcription Factors/metabolism
- Transcriptome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chinavenmeni S. Velu
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Aditya Chaubey
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - James D. Phelan
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Shane R. Horman
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mark Wunderlich
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Monica L. Guzman
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Anil G. Jegga
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Nancy J. Zeleznik-Le
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jianjun Chen
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - James C. Mulloy
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jose A. Cancelas
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Craig T. Jordan
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Bruce J. Aronow
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Guido Marcucci
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Balkrishen Bhat
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Brian Gebelein
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - H. Leighton Grimes
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
22
|
Du P, Tang F, Qiu Y, Dong F. GFI1 is repressed by p53 and inhibits DNA damage-induced apoptosis. PLoS One 2013; 8:e73542. [PMID: 24023884 PMCID: PMC3762790 DOI: 10.1371/journal.pone.0073542] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 07/20/2013] [Indexed: 12/12/2022] Open
Abstract
GFI1 is a transcriptional repressor that plays a critical role in hematopoiesis and has also been implicated in lymphomagenesis. It is still poorly understood how GFI1 expression is regulated in the hematopoietic system. We show here that GFI1 transcription was repressed by the tumor suppressor p53 in hematopoietic cells. Knockdown of p53 resulted in increased GFI1 expression and abolished DNA damage-induced GFI1 downregulation. In contrast, GFI1 expression was reduced and its downregulation in response to DNA damage was rescued upon restoration of p53 function in p53-deficient cells. In luciferase reporter assays, wild type p53, but not a DNA binding-defective p53 mutant, repressed the GFI1 promoter. Chromatin immunoprecipitation (ChIP) assays demonstrated that p53 bound to the proximal region of the GFI1 promoter. Detailed mapping of the GFI1 promoter indicated that GFI1 core promoter region spanning from -33 to +6 bp is sufficient for p53-mediated repression. This core promoter region contains a putative p53 repressive response element, mutation of which abolished p53 binding to and repression of GFI1 promoter. Significantly, apoptosis induced by DNA damage was inhibited upon Gfi1 overexpression, but augmented following GFI1 knockdown. Our data establish for the first time that GFI1 is repressed by p53 and add to our understanding of the roles of GFI1 in normal hematopoiesis and lymphomagenesis.
Collapse
Affiliation(s)
- Pei Du
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Fangqiang Tang
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Yaling Qiu
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Fan Dong
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
- * E-mail:
| |
Collapse
|
23
|
Potential roles for Gfi1 in the pathogenesis and proliferation of glioma. Med Hypotheses 2013; 80:629-32. [PMID: 23466061 DOI: 10.1016/j.mehy.2013.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 01/27/2013] [Accepted: 02/07/2013] [Indexed: 11/21/2022]
Abstract
Glioblastoma multiforme (GBM) is a major form of adult brain tumour with relatively poor prognosis and high mortality. Temozolomide (TMZ)-based chemotherapy following neurosurgery and radiotherapy has been suggested as the first line of treatment and is proven to effectively prolong overall survival and enhance patient quality of life. However, not all patients benefit from this treatment because of drug resistance. Even patients with TMZ-sensitive GBM may become resistant, which is partly due to the restoration of activity of the DNA repair enzyme O(6)-methylguanine-DNA-methyltransferase (MGMT); thus, patients cannot evade eventual tumour recurrence. The cellular activity of MGMT is the most important determinant of TMZ-resistance. However, some patients with a low level of activated MGMT are also TMZ-resistant. The aberrant expression of HOXA9, one of the 39 class I homeobox genes, is a marker of poor prognosis, and its level gradually increases with histologic malignant progression, shorter time to overall survival (OS) and free progression survival (FPS) in glioma patients, which further supports an oncogenic role for HOXA9 in gliomas. The HOXA9-PI3K signalling pathway is an important mechanism in GBM that is independent of MGMT promoter methylation status. The DNA binding sites of growth factor independent-1 (Gfi1) can overlap with the HOXA9 promoter through the "AATC" versus "GATT" core sequence. The competition for this binding site inhibits the expression of HOXA9 and induces different transcriptional outcomes, which suggests a new direction for investigation of the mechanism underlying targeted therapy of malignant gliomas.
Collapse
|
24
|
Chowdhury AH, Ramroop JR, Upadhyay G, Sengupta A, Andrzejczyk A, Saleque S. Differential transcriptional regulation of meis1 by Gfi1b and its co-factors LSD1 and CoREST. PLoS One 2013; 8:e53666. [PMID: 23308270 PMCID: PMC3538684 DOI: 10.1371/journal.pone.0053666] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 12/03/2012] [Indexed: 02/01/2023] Open
Abstract
Gfi1b (growth factor independence 1b) is a zinc finger transcription factor essential for development of the erythroid and megakaryocytic lineages. To elucidate the mechanism underlying Gfi1b function, potential downstream transcriptional targets were identified by chromatin immunoprecipitation and expression profiling approaches. The combination of these approaches revealed the oncogene meis1, which encodes a homeobox protein, as a direct and prominent target of Gfi1b. Examination of the meis1 promoter sequence revealed multiple Gfi1/1b consensus binding motifs. Distinct regions of the promoter were occupied by Gfi1b and its cofactors LSD1 and CoREST/Rcor1, in erythroid cells but not in the closely related megakaryocyte lineage. Accordingly, Meis1 was significantly upregulated in LSD1 inhibited erythroid cells, but not in megakaryocytes. This lineage specific upregulation in Meis1 expression was accompanied by a parallel increase in di-methyl histone3 lysine4 levels in the Meis1 promoter in LSD1 inhibited, erythroid cells. Meis1 was also substantially upregulated in gfi1b−/− fetal liver cells along with its transcriptional partners Pbx1 and several Hox messages. Elevated Meis1 message levels persisted in gfi1b mutant fetal liver cells differentiated along the erythroid lineage, relative to wild type. However, cells differentiated along the megakaryocytic lineage, exhibited no difference in Meis1 levels between controls and mutants. Transfection experiments further demonstrated specific repression of meis1 promoter driven reporters by wild type Gfi1b but neither by a SNAG domain mutant nor by a DNA binding deficient one, thus confirming direct functional regulation of this promoter by the Gfi1b transcriptional complex. Overall, our results demonstrate direct yet differential regulation of meis1 transcription by Gfi1b in distinct hematopoietic lineages thus revealing it to be a common, albeit lineage specific, target of both Gfi1b and its paralog Gfi1.
Collapse
Affiliation(s)
- Asif H. Chowdhury
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Johnny R. Ramroop
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Ghanshyam Upadhyay
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Ananya Sengupta
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Anna Andrzejczyk
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Shireen Saleque
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
25
|
The SUMO pathway promotes basic helix-loop-helix proneural factor activity via a direct effect on the Zn finger protein senseless. Mol Cell Biol 2012; 32:2849-60. [PMID: 22586269 DOI: 10.1128/mcb.06595-11] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During development, proneural transcription factors of the basic helix-loop-helix (bHLH) family are required to commit cells to a neural fate. In Drosophila neurogenesis, a key mechanism promoting sense organ precursor (SOP) fate is the synergy between proneural factors and their coactivator Senseless in transcriptional activation of target genes. Here we present evidence that posttranslational modification by SUMO enhances this synergy via an effect on Senseless protein. We show that Senseless is a direct target for SUMO modification and that mutagenesis of a predicted SUMOylation motif in Senseless reduces Senseless/proneural synergy both in vivo and in cell culture. We propose that SUMOylation of Senseless via lysine 509 promotes its synergy with proneural proteins during transcriptional activation and hence regulates an important step in neurogenesis leading to the formation and maturation of the SOPs.
Collapse
|
26
|
Characterization of the colorectal cancer-associated enhancer MYC-335 at 8q24: the role of rs67491583. Cancer Genet 2012; 205:25-33. [PMID: 22429595 DOI: 10.1016/j.cancergen.2012.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 12/23/2011] [Accepted: 01/09/2012] [Indexed: 12/12/2022]
Abstract
Recent genome-wide association studies have identified multiple regions at 8q24 that confer susceptibility to many cancers. In our previous work, we showed that the colorectal cancer (CRC) risk variant rs6983267 at 8q24 resides within a TCF4 binding site at the MYC-335 enhancer, with the risk allele G having a stronger binding capacity and Wnt responsiveness. Here, we searched for other potential functional variants within MYC-335. Genetic variation within MYC-335 was determined in samples from individuals of European, African, and Asian descent, with emphasis on variants in putative transcription factor binding sites. A 2-bp GA deletion rs67491583 was found to affect a growth factor independent (GFI) binding site and was present only in individuals with African ancestry. Chromatin immunoprecipitation performed in heterozygous cells showed that the GA deletion had an ability to reduce binding of the transcriptional repressors GFI1 and GFI1b. Screening of 1,027 African American colorectal cancer cases and 1,773 healthy controls did not reveal evidence for association (odds ratio: 1.17, 95% confidence interval: 0.97-1.41, P = 0.095). In this study, rs67491583 was identified as another functional variant in the CRC-associated enhancer MYC-335, but further studies are needed to establish the role of rs67491583 in the colorectal cancer predisposition of African Americans.
Collapse
|
27
|
Anderson AE, Karandikar UC, Pepple KL, Chen Z, Bergmann A, Mardon G. The enhancer of trithorax and polycomb gene Caf1/p55 is essential for cell survival and patterning in Drosophila development. Development 2011; 138:1957-66. [PMID: 21490066 PMCID: PMC3082301 DOI: 10.1242/dev.058461] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2011] [Indexed: 12/18/2022]
Abstract
In vitro data suggest that the human RbAp46 and RbAp48 genes encode proteins involved in multiple chromatin remodeling complexes and are likely to play important roles in development and tumor suppression. However, to date, our understanding of the role of RbAp46/RbAp48 and its homologs in metazoan development and disease has been hampered by a lack of insect and mammalian mutant models, as well as redundancy due to multiple orthologs in most organisms studied. Here, we report the first mutations in the single Drosophila RbAp46/RbAp48 homolog Caf1, identified as strong suppressors of a senseless overexpression phenotype. Reduced levels of Caf1 expression result in flies with phenotypes reminiscent of Hox gene misregulation. Additionally, analysis of Caf1 mutant tissue suggests that Caf1 plays important roles in cell survival and segment identity, and loss of Caf1 is associated with a reduction in the Polycomb Repressive Complex 2 (PRC2)-specific histone methylation mark H3K27me3. Taken together, our results suggest suppression of senseless overexpression by mutations in Caf1 is mediated by participation of Caf1 in PRC2-mediated silencing. More importantly, our mutant phenotypes confirm that Caf1-mediated silencing is vital to Drosophila development. These studies underscore the importance of Caf1 and its mammalian homologs in development and disease.
Collapse
Affiliation(s)
- Aimée E. Anderson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Kathryn L. Pepple
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhihong Chen
- Department of Biochemistry and Molecular Biology, M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Andreas Bergmann
- Department of Biochemistry and Molecular Biology, M. D. Anderson Cancer Center, Houston, TX 77030, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Graeme Mardon
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology, Baylor College of Medicine, Houston, TX 77030, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
28
|
Kenth G, Puzhko S, Goodyer CG. Human growth hormone receptor gene expression is regulated by Gfi-1/1b and GAGA cis-elements. Mol Cell Endocrinol 2011; 335:135-47. [PMID: 21238539 DOI: 10.1016/j.mce.2011.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Revised: 12/20/2010] [Accepted: 01/07/2011] [Indexed: 12/15/2022]
Abstract
Human growth hormone receptor (hGHR) gene regulation is complex: mRNAs are transcribed from multiple variant (V) 5'UTR exons, several ubiquitously while others only in the postnatal hepatocyte. The liver-specific V1 exon promoter contains Gfi-1/1b repressor sites adjacent to a GAGA box, a GH response element (GHRE) in several mammalian genes. GAGA boxes are also present in the ubiquitously expressing V3 exon promoter. Heterologous sites in bovine, ovine and murine GHR genes suggest conserved roles. GAGA factor stimulated V1 and V3 promoters while Gfi-1/1b repressed basal and GAF-stimulated V1 transcription. HGH treatment of HepG2 cells resulted in a new complex forming with V3 GAGA elements, suggesting a functional GHRE. Data suggest liver-specific V1 transcription is regulated by inhibitory Gfi-1/1b and stimulatory GAGA cis-elements and Gfi-1/1b may control the lack of V1 expression in fetal liver, hepatic tumours and non-hepatic tissues. In addition, hGH may regulate hGHR expression through V3 GAGA boxes.
Collapse
Affiliation(s)
- Gurvinder Kenth
- Department of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
29
|
Dai Y, Han B. [Research advance on non-small cell lung carcinoma with neuroendocrine differentiation]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2011; 14:165-9. [PMID: 21342649 PMCID: PMC5999768 DOI: 10.3779/j.issn.1009-3419.2011.02.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Yun Dai
- Department of Respiratory Medicine, Shanghai Wujiaochang Hospital, Shanghai 200438, China
| | | |
Collapse
|
30
|
Wilson NK, Calero-Nieto FJ, Ferreira R, Göttgens B. Transcriptional regulation of haematopoietic transcription factors. Stem Cell Res Ther 2011; 2:6. [PMID: 21345252 PMCID: PMC3092146 DOI: 10.1186/scrt47] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The control of differential gene expression is central to all metazoan biology. Haematopoiesis represents one of the best understood developmental systems where multipotent blood stem cells give rise to a range of phenotypically distinct mature cell types, all characterised by their own distinctive gene expression profiles. Small combinations of lineage-determining transcription factors drive the development of specific mature lineages from multipotent precursors. Given their powerful regulatory nature, it is imperative that the expression of these lineage-determining transcription factors is under tight control, a fact underlined by the observation that their misexpression commonly leads to the development of leukaemia. Here we review recent studies on the transcriptional control of key haematopoietic transcription factors, which demonstrate that gene loci contain multiple modular regulatory regions within which specific regulatory codes can be identified, that some modular elements cooperate to mediate appropriate tissue-specific expression, and that long-range approaches will be necessary to capture all relevant regulatory elements. We also explore how changes in technology will impact on this area of research in the future.
Collapse
Affiliation(s)
- Nicola K Wilson
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge, CB2 0XY, UK
| | - Fernando J Calero-Nieto
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge, CB2 0XY, UK
| | - Rita Ferreira
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge, CB2 0XY, UK
| | - Berthold Göttgens
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge, CB2 0XY, UK
| |
Collapse
|
31
|
Cakir M, Grossman A. The molecular pathogenesis and management of bronchial carcinoids. Expert Opin Ther Targets 2011; 15:457-91. [DOI: 10.1517/14728222.2011.555403] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
32
|
van der Meer LT, Jansen JH, van der Reijden BA. Gfi1 and Gfi1b: key regulators of hematopoiesis. Leukemia 2010; 24:1834-43. [DOI: 10.1038/leu.2010.195] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
33
|
Wilson NK, Timms RT, Kinston SJ, Cheng YH, Oram SH, Landry JR, Mullender J, Ottersbach K, Gottgens B. Gfi1 expression is controlled by five distinct regulatory regions spread over 100 kilobases, with Scl/Tal1, Gata2, PU.1, Erg, Meis1, and Runx1 acting as upstream regulators in early hematopoietic cells. Mol Cell Biol 2010; 30:3853-63. [PMID: 20516218 PMCID: PMC2916401 DOI: 10.1128/mcb.00032-10] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 03/06/2010] [Accepted: 05/06/2010] [Indexed: 01/30/2023] Open
Abstract
The growth factor independence 1 (Gfi1) gene was originally discovered in the hematopoietic system, where it functions as a key regulator of stem cell homeostasis, as well as neutrophil and T-cell development. Outside the blood system, Gfi1 is essential for inner-ear hair and intestinal secretory cell differentiation. To understand the regulatory hierarchies within which Gfi1 operates to control these diverse biological functions, we used a combination of comparative genomics, locus-wide chromatin immunoprecipitation assays, functional validation in cell lines, and extensive transgenic mouse assays to identify and characterize the complete ensemble of Gfi1 regulatory elements. This concerted effort identified five distinct regulatory elements spread over 100kb each driving expression in transgenic mice to a subdomain of endogenous Gfi1. Detailed characterization of an enhancer 35 kb upstream of Gfi1 demonstrated activity in the dorsal aorta region and fetal liver in transgenic mice, which was bound by key stem cell transcription factors Scl/Tal1, PU.1/Sfpi1, Runx1, Erg, Meis1, and Gata2. Taken together, our results reveal the regulatory regions responsible for Gfi1 expression and importantly establish that Gfi1 expression at the sites of hematopoietic stem cell (HSC) emergence is controlled by key HSC regulators, thus integrating Gfi1 into the wider HSC regulatory networks.
Collapse
Affiliation(s)
- Nicola K. Wilson
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, United Kingdom
| | - Richard T. Timms
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, United Kingdom
| | - Sarah J. Kinston
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, United Kingdom
| | - Yi-Han Cheng
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, United Kingdom
| | - S. Helen Oram
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, United Kingdom
| | - Josette-Renee Landry
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, United Kingdom
| | - Joanne Mullender
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, United Kingdom
| | - Katrin Ottersbach
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, United Kingdom
| | - Berthold Gottgens
- University of Cambridge Department of Haematology, Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, United Kingdom
| |
Collapse
|
34
|
Phelan JD, Shroyer NF, Cook T, Gebelein B, Grimes HL. Gfi1-cells and circuits: unraveling transcriptional networks of development and disease. Curr Opin Hematol 2010; 17:300-7. [PMID: 20571393 PMCID: PMC2910316 DOI: 10.1097/moh.0b013e32833a06f8] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The review will integrate current knowledge of transcriptional circuits whose dysregulation leads to autoimmunity, neutropenia and leukemia. RECENT FINDINGS Growth factor independent-1 (Gfi1) is a transcriptional repressor with essential roles in controlling hematopoietic stem cell biology, myeloid and lymphoid differentiation and lymphocyte effector functions. Recent work has suggested that Gfi1 competes or collaborates with other transcription factors to modulate transcription programs and lineage decisions. SUMMARY Gfi1 is central to several transcriptional circuits whose dysregulation leads to abnormal or malignant hematopoiesis. These functional relationships are conserved from Drosophila development. Such conserved pathways represent central oncogenic or 'gatekeeper' pathways that are pivotal to understanding the process of cellular transformation, and illustrate key targets for clinical intervention.
Collapse
Affiliation(s)
- James D. Phelan
- Immunobiology Graduate Program Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Noah F. Shroyer
- Division of Gastroenterology, Hepatology and Nutrition Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Tiffany Cook
- Division of Developmental Biology Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Brian Gebelein
- Division of Developmental Biology Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - H. Leighton Grimes
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
- Division of Experimental Hematology and Cancer Biology Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| |
Collapse
|
35
|
Yang H, Gan J, Xie X, Deng M, Feng L, Chen X, Gao Z, Gan L. Gfi1-Cre knock-in mouse line: A tool for inner ear hair cell-specific gene deletion. Genesis 2010; 48:400-6. [PMID: 20533399 PMCID: PMC2885521 DOI: 10.1002/dvg.20632] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Gfi1 encodes a zinc-finger transcription factor essential for the development and maintenance of haematopoiesis and the inner ear. In mouse inner ear, Gfi1 expression is confined to hair cells during development and in adulthood. To construct a genetic tool for inner ear hair cell-specific gene deletion, we generated a Gfi1-Cre mouse line by knocking-in Cre coding sequences into the Gfi1 locus and inactivating the endogenous Gfi1. The specificity and efficiency of Gfi1-Cre recombinase-mediated recombination in the developing inner ear was revealed through the expression of the conditional R26R-lacZ reporter gene. The onset of lacZ expression in the Gfi1(Cre/+) inner ear was first detected at E13.5 in the vestibule and at E15.5 in the cochlea, coinciding with the generation of hair cells. Throughout inner ear development, lacZ expression was detected only in hair cells. Thus, Gfi1-Cre knock-in mouse line provides a useful tool for gene manipulations specifically in inner ear hair cells.
Collapse
Affiliation(s)
- Hua Yang
- Flaum Eye Institute, University of Rochester, Rochester, NY 14642
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China 100730
| | - Jean Gan
- Flaum Eye Institute, University of Rochester, Rochester, NY 14642
| | - Xiaoling Xie
- Flaum Eye Institute, University of Rochester, Rochester, NY 14642
| | - Min Deng
- Flaum Eye Institute, University of Rochester, Rochester, NY 14642
| | - Liang Feng
- Flaum Eye Institute, University of Rochester, Rochester, NY 14642
| | - Xiaowei Chen
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China 100730
| | - Zhiqiang Gao
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China 100730
| | - Lin Gan
- Flaum Eye Institute, University of Rochester, Rochester, NY 14642
- Center for Visual Science, University of Rochester, Rochester, NY 14642
- Department of Neurobiology and Anatomy, University of Rochester, Rochester, NY 14642
| |
Collapse
|
36
|
Liu Q, Basu S, Qiu Y, Tang F, Dong F. A role of Miz-1 in Gfi-1-mediated transcriptional repression of CDKN1A. Oncogene 2010; 29:2843-52. [PMID: 20190815 PMCID: PMC2869400 DOI: 10.1038/onc.2010.48] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 01/13/2010] [Accepted: 01/29/2010] [Indexed: 12/11/2022]
Abstract
Zinc-finger (ZF) transcriptional repressor growth factor independence 1 (Gfi-1) has an important role in hematopoiesis and inner ear development, and also functions as an oncoprotein that cooperates with c-Myc in lymphomagenesis. Gfi-1 represses transcription by directly binding to conserved sequences in the promoters of its target genes. CDKN1A encoding p21(Cip1) has been identified as a Gfi-1 target gene and has been shown to contain Gfi-1 binding sites in the upstream promoter region. We show here that Gfi-1 represses CDKN1A in a manner that is independent of its DNA binding activity. Gfi-1 interacts with POZ-ZF transcription factor Miz-1, originally shown to be a c-Myc-interacting partner, and through Miz-1 binds to the CDKN1A core promoter. Interestingly, Gfi-1 and c-Myc, through Miz-1, form a ternary complex on the CDKN1A promoter, and function in collaboration to repress CDKN1A. Gfi-1 knockdown results in enhanced levels of p21(Cip1) and attenuated cell proliferation. Notably, similar to c-Myc, the expression of Gfi-1 is downregulated by transforming growth factor-beta (TGFbeta) and the level of Gfi-1 influences the response of cells to the cytostatic effect of TGFbeta. Our data reveal an important mechanism by which Gfi-1 regulates cell proliferation and may also have implications for understanding the role of Gfi-1 in lymphomagenesis.
Collapse
Affiliation(s)
- Qingquan Liu
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Suchitra Basu
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Yaling Qiu
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Fangqiang Tang
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Fan Dong
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| |
Collapse
|
37
|
Lee S, Doddapaneni K, Hogue A, McGhee L, Meyers S, Wu Z. Solution Structure of Gfi-1 Zinc Domain Bound to Consensus DNA. J Mol Biol 2010; 397:1055-66. [DOI: 10.1016/j.jmb.2010.02.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Revised: 02/02/2010] [Accepted: 02/04/2010] [Indexed: 01/29/2023]
|
38
|
Chen G, Korfhagen TR, Xu Y, Kitzmiller J, Wert SE, Maeda Y, Gregorieff A, Clevers H, Whitsett JA. SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production. J Clin Invest 2009; 119:2914-24. [PMID: 19759516 DOI: 10.1172/jci39731] [Citation(s) in RCA: 228] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 07/15/2009] [Indexed: 12/13/2022] Open
Abstract
Various acute and chronic inflammatory stimuli increase the number and activity of pulmonary mucus-producing goblet cells, and goblet cell hyperplasia and excess mucus production are central to the pathogenesis of chronic pulmonary diseases. However, little is known about the transcriptional programs that regulate goblet cell differentiation. Here, we show that SAM-pointed domain-containing Ets-like factor (SPDEF) controls a transcriptional program critical for pulmonary goblet cell differentiation in mice. Initial cell-lineage-tracing analysis identified nonciliated secretory epithelial cells, known as Clara cells, as the progenitors of goblet cells induced by pulmonary allergen exposure in vivo. Furthermore, in vivo expression of SPDEF in Clara cells caused rapid and reversible goblet cell differentiation in the absence of cell proliferation. This was associated with enhanced expression of genes regulating goblet cell differentiation and protein glycosylation, including forkhead box A3 (Foxa3), anterior gradient 2 (Agr2), and glucosaminyl (N-acetyl) transferase 3, mucin type (Gcnt3). Consistent with these findings, levels of SPDEF and FOXA3 were increased in mouse goblet cells after sensitization with pulmonary allergen, and the proteins were colocalized in goblet cells lining the airways of patients with chronic lung diseases. Deletion of the mouse Spdef gene resulted in the absence of goblet cells in tracheal/laryngeal submucosal glands and in the conducting airway epithelium after pulmonary allergen exposure in vivo. These data show that SPDEF plays a critical role in regulating a transcriptional network mediating the goblet cell differentiation and mucus hyperproduction associated with chronic pulmonary disorders.
Collapse
Affiliation(s)
- Gang Chen
- The Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati School of Medicine, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Loss of MicroRNA targets in the 3' untranslated region as a mechanism of retroviral insertional activation of growth factor independence 1. J Virol 2009; 83:8051-61. [PMID: 19474094 DOI: 10.1128/jvi.00427-09] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The non-oncogene-bearing retrovirus SL3-3 murine leukemia virus induces strictly T-cell lymphomas with a mean latency of 2 to 4 months in mice of the NMRI-inbred (NMRI-i) strain. By high-throughput sequencing of retroviral tags, we have identified the genomic region carrying the transcriptional repressor and oncogene growth factor independence 1 (Gfi1) as a frequent target for SL3-3 in the NMRI-i mouse genome. Twenty-four SL3-3 insertions were identified within a 1-kb window of the 3' untranslated region (3'UTR) of the Gfi1 gene, a clustering pattern unique for this lymphoma model. Expression analysis determined that the Gfi1 gene was transcriptionally activated by SL3-3 insertions, and an upregulation of Gfi1 protein expression was detected for tumors harboring insertions in the Gfi1 3'UTR. Here we provide data in support of a mechanism by which retroviral insertions in the Gfi1 3'UTR decouple microRNA-mediated posttranscriptional regulation.
Collapse
|
40
|
Loss of the transcriptional repressor PAG-3/Gfi-1 results in enhanced neurosecretion that is dependent on the dense-core vesicle membrane protein IDA-1/IA-2. PLoS Genet 2009; 5:e1000447. [PMID: 19343207 PMCID: PMC2657203 DOI: 10.1371/journal.pgen.1000447] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Accepted: 03/06/2009] [Indexed: 01/09/2023] Open
Abstract
It is generally accepted that neuroendocrine cells regulate dense core vesicle (DCV) biogenesis and cargo packaging in response to secretory demands, although the molecular mechanisms of this process are poorly understood. One factor that has previously been implicated in DCV regulation is IA-2, a catalytically inactive protein phosphatase present in DCV membranes. Our ability to directly visualize a functional, GFP-tagged version of an IA-2 homolog in live Caenorhabditis elegans animals has allowed us to capitalize on the genetics of the system to screen for mutations that disrupt DCV regulation. We found that loss of activity in the transcription factor PAG-3/Gfi-1, which functions as a repressor in many systems, results in a dramatic up-regulation of IDA-1/IA-2 and other DCV proteins. The up-regulation of DCV components was accompanied by an increase in presynaptic DCV numbers and resulted in phenotypes consistent with increased neuroendocrine secretion. Double mutant combinations revealed that these PAG-3 mutant phenotypes were dependent on wild type IDA-1 function. Our results support a model in which IDA-1/IA-2 is a critical element in DCV regulation and reveal a novel genetic link to PAG-3-mediated transcriptional regulation. To our knowledge, this is the first mutation identified that results in increased neurosecretion, a phenotype that has clinical implications for DCV-mediated secretory disorders. Within secretory cells, hormones are packaged into vesicles (called DCVs) that are released upon stimulation. The number of DCVs is regulated to meet the secretory demands of the cell by a mechanism that is poorly understood, although a protein in the membrane of DCVs, called IA-2, is thought to play a role. A genetic screen in the nematode C. elegans is used, here, to find mutations that mis-regulate the corresponding worm protein called IDA-1. Capitalizing on the simple neuroanatomy of the nematode and its transparency, we visualize IDA-1 protein levels directly in the animal using a fluorescent tag. We find that mutations in the transcription factor PAG-3/Gfi-1 result in elevated levels of IDA-1 protein, increased numbers of presynaptic DCVs, and behaviors consistent with increased neurosecretion. Our results demonstrate that IDA-1/IA-2 protein levels correlate with the biogenesis, utilization, or stability of DCVs. We propose that PAG-3 normally down regulates the production of IDA-1, thus serving as part of the mechanism underlying DCV regulation. This is the first reported mutation that increases DCV numbers and secretion, offering insight into DCV homeostasis and a potential therapeutic target for diseases that would benefit from a boost in neuroendocrine secretion.
Collapse
|
41
|
Abstract
The zinc finger protein growth factor independent-1 (Gfi1) is a transcriptional repressor that is critically required for normal granulocytic differentiation. GFI1 loss-of-function mutations are found in some patients with severe congenital neutropenia (SCN). The SCN-associated GFI1-mutant proteins act as dominant negatives to block granulopoiesis through selective deregulation of a subset of GFI1 target genes. Here we show that Gfi1 is a master regulator of microRNAs, and that deregulated expression of these microRNAs recapitulates a Gfi1 loss-of-function block to granulocyte colony-stimulating factor (G-CSF)-stimulated granulopoiesis. Specifically, bone marrow cells from a GFI1-mutant SCN patient and Gfi1(-/-) mice display deregulated expression of miR-21 and miR-196B expression. Flow cytometric analysis and colony assays reveal that the overexpression or depletion of either miR induces changes in myeloid development. However, coexpression of miR-21 and miR-196b (as seen in Gfi1(-/-) mice and a GFI1N382S SCN patient) completely blocks G-CSF-induced granulopoiesis. Thus, our results not only identify microRNAs whose regulation is required during myelopoiesis, but also provide an example of synergy in microRNA biologic activity and illustrate potential mechanisms underlying SCN disease pathogenesis.
Collapse
|
42
|
Gfi-1 represses CDKN2B encoding p15INK4B through interaction with Miz-1. Proc Natl Acad Sci U S A 2009; 106:1433-8. [PMID: 19164764 DOI: 10.1073/pnas.0804863106] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Gfi-1 is a nuclear zinc finger (ZF) transcriptional repressor that plays an important role in hematopoiesis and inner ear development, and has been implicated in lymphomagenesis. Gfi-1 represses transcription by directly binding to the consensus DNA sequence in the promoters of its target genes. We report here an alternative mechanism by which Gfi-1 represses CDKN2B encoding p15(INK4B). Gfi-1 does not directly bind to CDKN2B, but interacts with Miz-1 and, via Miz-1, is recruited to the core promoter of CDKN2B. Miz-1 is a POZ-ZF transcription factor that has been shown to mediate transcriptional repression by c-Myc. Like c-Myc, upon recruitment to the CDKN2B promoter, Gfi-1 represses transcriptional activation of CDKN2B by Miz-1 and in response to TGFbeta. Consistent with its role in repressing CDKN2B transcription, knockdown of Gfi-1 in human leukemic cells or deficiency of Gfi-1 in mouse bone marrow cells results in augmented expression of p15(INK4B). Notably, Gfi-1 and c-Myc are both recruited to the CDKN2B core promoter and act in collaboration to repress CDKN2B. Our data reveal a mechanism of transcriptional repression by Gfi-1 and may have important implications for understanding the roles of Gfi-1 in normal development and tumorigenesis.
Collapse
|
43
|
Igwe E, Kosan C, Khandanpour C, Sharif-Askari E, Brüne B, Möröy T. The zinc finger protein Gfi1 is implicated in the regulation of IgG2b production and the expression of Igamma2b germline transcripts. Eur J Immunol 2009; 38:3004-14. [PMID: 18991277 DOI: 10.1002/eji.200838251] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gfi1 is a zinc finger transcription factor that is undetectable in B lymphocytes but its expression rises rapidly upon antigenic stimulation or treatment with lipopolysaccharide (LPS). Here we show that Gfi1(-/-) mice have higher serum levels of gamma isotype immunoglobulin than WT animals. When challenged with antigen, Gfi1(-/-) mice react with accelerated formation of PNA+/CD19+ germinal center B cells and an increased production of antigen-specific IgG2a and IgG2b. Moreover, Gfi1(-/-) B cells secrete more IgG2a and IgG2b than WT cells and produce higher levels of Igamma2b sterile germline transcripts when cultured with LPS. While the proliferative response to stimulation with anti-IgM antibodies and plasma cell differentiation was normal in Gfi1(-/-) B cells, we found that mRNA and protein levels of TGFbeta1 were significantly increased in the absence of Gfi1. TGFbeta1 has been shown to be essential for the regulation of IgG subclass production and was previously found to selectively stimulate IgG2b secretion. Our findings reveal a new function of Gfi1 in the control of IgG isotype production.
Collapse
Affiliation(s)
- Emeka Igwe
- Institut für Zellbiologie Tumorforschung, IFZ, Universitätsklinikum Essen, Essen, Germany.
| | | | | | | | | | | |
Collapse
|
44
|
Montoya-Durango DE, Velu CS, Kazanjian A, Rojas MEB, Jay CM, Longmore GD, Grimes HL. Ajuba functions as a histone deacetylase-dependent co-repressor for autoregulation of the growth factor-independent-1 transcription factor. J Biol Chem 2008; 283:32056-65. [PMID: 18805794 DOI: 10.1074/jbc.m802320200] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Growth factor independent-1 (Gfi1) is a zinc finger protein with a SNAG-transcriptional repressor domain. Ajuba is a LIM domain protein that shuttles between the cytoplasm and the nucleus. Ajuba functions as a co-repressor for synthetic Gfi1 SNAG-repressor domain-containing constructs, but a role for Ajuba co-repression of the cognate DNA bound Gfi1 protein has not been defined. Co-immunoprecipitation of synthetic and endogenous proteins and co-elution with gel filtration suggest that an endogenous Ajuba.Gfi1.HDAC multiprotein complex is possible. Active histone deacetylase activity co-immunoprecipitates with Ajuba or Gfi1, and both proteins depend upon histone deacetylases for full transcriptional repression activity. Ajuba LIM domains directly bind to Gfi1, but the association is not SNAG domain-dependent. ChIP analysis and reciprocal knockdown experiments suggest that Ajuba selectively functions as a co-repressor for Gfi1 autoregulation. The data suggest that Ajuba is utilized as a corepressor selectively on Gfi1 target genes.
Collapse
Affiliation(s)
- Diego E Montoya-Durango
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Neptune ER, Podowski M, Calvi C, Cho JH, Garcia JGN, Tuder R, Linnoila RI, Tsai MJ, Dietz HC. Targeted disruption of NeuroD, a proneural basic helix-loop-helix factor, impairs distal lung formation and neuroendocrine morphology in the neonatal lung. J Biol Chem 2008; 283:21160-9. [PMID: 18339630 PMCID: PMC2475704 DOI: 10.1074/jbc.m708692200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Despite the importance of airspace integrity in vertebrate gas exchange,
the molecular pathways that instruct distal lung formation are poorly
understood. Recently, we found that fibrillin-1 deficiency in mice impairs
alveolar formation and recapitulates the pulmonary features of human Marfan
syndrome. To further elucidate effectors involved in distal lung formation, we
performed expression profiling analysis comparing the fibrillin-1-deficient
and wild-type developing lung. NeuroD, a basic helix-loop-helix transcription
factor, fulfilled the expression criteria for a candidate mediator of distal
lung development. We investigated its role in murine lung development using
genetically targeted NeuroD-deficient mice. We found that NeuroD deficiency
results in both impaired alveolar septation and altered morphology of the
pulmonary neuroendocrine cells. NeuroD-deficient mice had enlarged alveoli
associated with reduced epithelial proliferation in the airway and airspace
compartments during development. Additionally, the neuroendocrine compartment
in these mice manifested an increased number of neuroepithelial bodies but a
reduced number of solitary pulmonary neuroendocrine cells in the neonatal
lung. Overexpression of NeuroD in a murine lung epithelial cell line conferred
a neuroendocrine phenotype characterized by the induction of neuroendocrine
markers as well as increased proliferation. These results support an
unanticipated role for NeuroD in the regulation of pulmonary neuroendocrine
and alveolar morphogenesis and suggest an intimate connection between the
neuroendocrine compartment and distal lung development.
Collapse
Affiliation(s)
- Enid R Neptune
- Division of Pulmonary and Critical Care Medicine, Institute of Genetic Medicine, Howard Hughes Medical Institute, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang Z, Hao Y, Lowe AW. The adenocarcinoma-associated antigen, AGR2, promotes tumor growth, cell migration, and cellular transformation. Cancer Res 2008; 68:492-7. [PMID: 18199544 DOI: 10.1158/0008-5472.can-07-2930] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The AGR2 gene encodes a secretory protein that is highly expressed in adenocarcinomas of the esophagus, pancreas, breast, and prostate. This study explores the effect of AGR2 expression with well-established in vitro and in vivo assays that screen for cellular transformation and tumor growth. AGR2 expression in SEG-1 esophageal adenocarcinoma cells was reduced with RNA interference. Cellular transformation was examined using NIH3T3 cells that express AGR2 after stable transfection. The cell lines were studied in vitro with assays for density-dependent and anchorage-independent growth, and in vivo as tumor xenografts in nude mice. SEG-1 cells with reduced AGR2 expression showed an 82% decrease in anchorage-independent colony growth and a 60% reduction in tumor xenograft size. In vitro assays of AGR2-expressing NIH3T3 cells displayed enhanced foci formation and anchorage-independent growth. In vivo, AGR2-expressing NIH3T3 cells established tumors in nude mice. Thus, AGR2 expression promotes tumor growth in esophageal adenocarcinoma cells and is able to transform NIH3T3 cells. Immunohistochemistry of the normal mouse intestine detected AGR2 expression in proliferating and differentiated intestinal cells of secretory lineage. AGR2 may be important for the growth and development of the intestine as well as esophageal adenocarcinomas.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Medicine, Stanford University, Stanford, California 94305-5187, USA
| | | | | |
Collapse
|
47
|
Alkushi A, Clarke BA, Akbari M, Makretsov N, Lim P, Miller D, Magliocco A, Coldman A, van de Rijn M, Huntsman D, Parker R, Gilks CB. Identification of prognostically relevant and reproducible subsets of endometrial adenocarcinoma based on clustering analysis of immunostaining data. Mod Pathol 2007; 20:1156-65. [PMID: 17717550 DOI: 10.1038/modpathol.3800950] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Panels of immunomarkers can provide greater information than single markers, but the problem of how to optimally interpret data from multiple immunomarkers is unresolved. We examined the expression profile of 12 immunomarkers in 200 endometrial carcinomas using a tissue microarray. The outcomes of groups of patients were analyzed by using the Kaplan-Meier method, using the log-rank statistic for comparison of survival curves. Correlation between clustering results and traditional prognosticators of endometrial carcinoma was examined by either Fisher's exact test or chi2-test. Multivariate analysis was performed using a proportional hazards method (Cox regression modeling). Seven of the 12 immunomarkers studied showed prognostic significance in univariate analysis (P<0.05) and 1 marker showed a trend toward significance (P=0.06). These eight markers were used in unsupervised hierarchical clustering of the cases, and resulted in identification of three cluster groups. There was a statistically significant difference in patient survival between these cluster groups (P=0.0001). The prognostic significance of the cluster groups was independent of tumor stage and patient age on multivariate analysis (P=0.014), but was not of independent significance when either tumor grade or cell type was added to the model. The cluster group designation was strongly correlated with tumor grade, stage, and cell type (P<0.0001 for each). Interlaboratory reproducibility of subclassification of endometrial adenocarcinoma by hierarchical clustering analysis was verified by showing highly reproducible assignment of individual cases to specific cluster groups when the immunostaining was performed, interpreted, and clustered in a second laboratory (kappa=0.79, concordance rate=89.6%). Unsupervised hierarchical clustering of immunostaining data identifies prognostically relevant subsets of endometrial adenocarcinoma. Such analysis is reproducible, showing less interobserver variability than histopathological assessment of tumor cell type or grade.
Collapse
Affiliation(s)
- Abdulmohsen Alkushi
- Department of Pathology, King Fahad National Guard Hospital, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Saleque S, Kim J, Rooke HM, Orkin SH. Epigenetic regulation of hematopoietic differentiation by Gfi-1 and Gfi-1b is mediated by the cofactors CoREST and LSD1. Mol Cell 2007; 27:562-72. [PMID: 17707228 DOI: 10.1016/j.molcel.2007.06.039] [Citation(s) in RCA: 330] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Revised: 04/27/2007] [Accepted: 06/25/2007] [Indexed: 02/06/2023]
Abstract
Gfi-1 and Gfi-1b are homologous transcriptional repressors involved in diverse developmental contexts, including hematopoiesis and oncogenesis. Transcriptional repression by Gfi proteins requires the conserved SNAG domain. To elucidate the function of Gfi proteins, we purified Gfi-1b complexes and identified interacting proteins. Prominent among these is the corepressor CoREST, the histone demethylase LSD1, and HDACs 1 and 2. CoREST and LSD1 associate with Gfi-1/1b via the SNAG repression domain. Gfi-1b further recruits these cofactors to the majority of target gene promoters in vivo. Inhibition of CoREST and LSD1 perturbs differentiation of erythroid, megakaryocytic, and granulocytic cells as well as primary erythroid progenitors. LSD1 depletion derepresses Gfi targets in lineage-specific patterns, accompanied by enhanced histone 3 lysine 4 methylation at the respective promoters. Overall, we show that chromatin regulatory proteins CoREST and LSD1 mediate transcriptional repression by Gfi proteins. Lineage-restricted deployment of these cofactors through interaction with Gfi proteins controls hematopoietic differentiation.
Collapse
Affiliation(s)
- Shireen Saleque
- Division of Hematology-Oncology, Children's Hospital Boston, Boston, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
49
|
Marteijn JAF, van der Meer LT, van Emst L, van Reijmersdal S, Wissink W, de Witte T, Jansen JH, Van der Reijden BA. Gfi1 ubiquitination and proteasomal degradation is inhibited by the ubiquitin ligase Triad1. Blood 2007; 110:3128-35. [PMID: 17646546 DOI: 10.1182/blood-2006-11-058602] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Growth factor independence 1 (Gfi1) is a transcriptional repressor essential for the function and development of many different hematopoietic lineages. The Gfi1 protein expression is regulated by the ubiquitin-proteasome system. In granulocytes, Gfi1 is rapidly degraded by the proteasome, while it is more stable in monocytes. How the ubiquitination and degradation of Gfi1 is regulated is unclear. Here, we show that the ubiquitin ligase Triad1 interacts with the DNA-binding domain of Gfi1. Unexpectedly, we found that Triad1 inhibited Gfi1 ubiquitination, resulting in a prolonged half-life. Down-regulation of endogenous Triad1 by siRNAs resulted in increased Gfi1 ubiquitination. In U937 cells, Triad1 caused an increase in endogenous Gfi1 protein levels and slowed cell proliferation in a similar manner when Gfi1 itself was expressed. A Triad1 mutant that lacks the Gfi1-binding domain did not affect Gfi1 levels and proliferation. Because neither proteasome-ubiquitin nor Triad1 ubiquitin ligase activity was required for the inhibition of Gfi1 ubiquitination, these data suggest that Triad1 competes for Gfi1 binding with as yet to be identified E3 ubiquitin ligases that do mark Gfi1 for proteasomal degradation. The fine-tuning of Gfi1 protein levels regulated by Triad1 defines an unexpected role for this protein in hematopoiesis.
Collapse
Affiliation(s)
- Jurgen A F Marteijn
- Central Hematology Laboratory, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, 6500 HB Nijmegen, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Linnoila RI, Jensen-Taubman S, Kazanjian A, Grimes HL. Loss of GFI1 impairs pulmonary neuroendorine cell proliferation, but the neuroendocrine phenotype has limited impact on post-naphthalene airway repair. J Transl Med 2007; 87:336-44. [PMID: 17377622 PMCID: PMC2839158 DOI: 10.1038/labinvest.3700527] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Naphthalene exposure kills lung airway epithelial (Clara) cells, but is rapidly followed by Clara cell reconstitution coincident with proliferation of pulmonary neuroendocrine cells (PNEC). Although a role for mature PNEC in the reconstitution process has been excluded, the reconstituting progenitor cells have been suggested to enter a transient neuroendocrine (NE) differentiation phase before differentiating to Clara cells. Furthermore, these progenitors were suggested to be the target population for transformation to a NE tumor; small cell lung cancer (SCLC). Although the NE phenotype is central to SCLC oncogenesis, the relevance of NE differentiation to post naphthalene reconstitution remains to be determined. The Growth factor independent-1 (Gfi1) transcription factor is expressed in SCLC and is required for the NE differentiation of PNEC. Gfi1(-/-) mice display a 70% reduction in airway cells that express NE markers, and cells that stain for NE markers show weak expression of some markers. Therefore, to determine the relevance of the NE phenotype to post-naphthalene reconstitution, we examined post-naphthalene reconstitution in Gfi1(-/-) mice. Our analyses indicate that the post-naphthalene regeneration process includes both airway epithelial proliferation and apoptosis. Gfi1 deletion lowered both airway epithelial proliferation and apoptosis; however, the post-naphthalene rate of increase in growth and apoptosis was not significantly different between Gfi1(-/-) mice and wild-type littermates. Moreover, the timing and extent of CC10+ cell regeneration was unaffected by Gfi1 deletion. These data suggest that neither Gfi1 nor the NE phenotype play a dominant role in the regeneration process. However, the few Gfi1(-/-) cells capable of NE differentiation show a significant reduction in post-naphthalene proliferation. The modest proliferation seen in Gfi1(-/-) NE cells is consistent with the previously proposed role for Gfi1 in controlling neuroendocrine cancer growth.
Collapse
Affiliation(s)
- R Ilona Linnoila
- Experimental Pathology Section, Cell and Cancer Biology Branch, CCR, NCI, NIH, Bethesda, MD, USA
| | | | | | | |
Collapse
|