1
|
Han Y, Xu S, Ye W, Wang Y, Zhang X, Deng J, Zhang Z, Liu L, Liu S. Targeting LSD1 suppresses stem cell-like properties and sensitizes head and neck squamous cell carcinoma to PD-1 blockade. Cell Death Dis 2021; 12:993. [PMID: 34689153 PMCID: PMC8542042 DOI: 10.1038/s41419-021-04297-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly aggressive tumor with poor clinical outcomes due to recurrence, metastasis, and treatment resistance. Cancer stem cells (CSCs), a small population among tumor cells, are proposed to be responsible for tumor initiation, progression, metastasis, drug resistance, and recurrence. Here we show that high LSD1 expression was a predictor of poor prognosis for HNSCC patients. We found that high expression of LSD1 is essential for the maintenance of the CSC properties by regulating Bmi-1 expression. Moreover, tumor LSD1 ablation suppresses CSC-like characteristics in vitro and inhibits tumorigenicity in vivo in immune-deficient xenografts. However, this deletion induces the upregulation of PDL1 levels, which compromises antitumor immunity and reduces antitumor efficacy in an immune-competent mouse model. Functionally, the combination of LSD1 inhibitor and anti-PD-1 monoclonal antibody can overcome tumor immune evasion and greatly inhibit tumor growth, which was associated with reduced Ki-67 level and augmented CD8+ T cell infiltration in immunocompetent tumor-bearing mouse models. In summary, these findings provide a novel and promising combined strategy for the treatment of HNSCC using a combination of LSD1 inhibition and PD-1 blockade.
Collapse
Affiliation(s)
- Yong Han
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shengming Xu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Weimin Ye
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yang Wang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xiangkai Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jiong Deng
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Liu Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,National Clinical Research Center for Oral Diseases, Shanghai, China. .,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China. .,Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China.
| | - Shuli Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,National Clinical Research Center for Oral Diseases, Shanghai, China. .,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China. .,Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
2
|
Yu J, Wu Y, Li H, Zhou H, Shen C, Gao T, Lin M, Dai X, Ou J, Liu M, Huang X, Zheng B, Sun F. BMI1 Drives Steroidogenesis Through Epigenetically Repressing the p38 MAPK Pathway. Front Cell Dev Biol 2021; 9:665089. [PMID: 33928089 PMCID: PMC8076678 DOI: 10.3389/fcell.2021.665089] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 03/22/2021] [Indexed: 11/18/2022] Open
Abstract
Testosterone biosynthesis progressively decreases in aging males primarily as a result of functional changes to Leydig cells. Despite this, the mechanisms underlying steroidogenesis remain largely unclear. Using gene knock-out approaches, we and others have recently identified Bmi1 as an anti-aging gene. Herein, we investigate the role of BMI1 in steroidogenesis using mouse MLTC-1 and primary Leydig cells. We show that BMI1 can positively regulate testosterone production. Mechanistically, in addition to its known role in antioxidant activity, we also report that p38 mitogen-activated protein kinase (MAPK) signaling is activated, and testosterone levels reduced, in BMI1-deficient cells; however, the silencing of the p38 MAPK pathway restores testosterone production. Furthermore, we reveal that BMI1 directly binds to the promoter region of Map3k3, an upstream activator of p38, thereby modulating its chromatin status and repressing its expression. Consequently, this results in the inhibition of the p38 MAPK pathway and the promotion of steroidogenesis. Our study uncovered a novel epigenetic mechanism in steroidogenesis involving BMI1-mediated gene silencing and provides potential therapeutic targets for the treatment of hypogonadism.
Collapse
Affiliation(s)
- Jun Yu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, China
| | - Yibo Wu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Hong Li
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China
| | - Hui Zhou
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China
| | - Tingting Gao
- Center of Clinical Reproductive Medicine, The Affiliated Changzhou Maternity and Child Health Care Hospital of Nanjing Medical University, Changzhou, China
| | - Meng Lin
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xiuliang Dai
- Center of Clinical Reproductive Medicine, The Affiliated Changzhou Maternity and Child Health Care Hospital of Nanjing Medical University, Changzhou, China
| | - Jian Ou
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China
| | - Meiling Liu
- National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China
| | - Xiaoyan Huang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Bo Zheng
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China.,National Health Commission Key Laboratory of Male Reproductive Health, National Research Institute for Family Planning, Beijing, China
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
3
|
Zhou M, Xu Q, Huang D, Luo L. Regulation of gene transcription of B lymphoma Mo-MLV insertion region 1 homolog (Review). Biomed Rep 2021; 14:52. [PMID: 33884195 PMCID: PMC8056379 DOI: 10.3892/br.2021.1428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 02/19/2021] [Indexed: 12/18/2022] Open
Abstract
B lymphoma Mo-MLV insertion region 1 homolog (Bmi-1) is a core protein component of the polycomb repressive complex 1 that inhibits cell senescence and maintains the self-renewal ability of stem cells via downregulation of p16Ink4a and p19Arf expression. Bmi-1 serves an important role in hematopoietic stem cell maintenance and neurodevelopment during embryonic development, and it has been shown to enhance tumorigenesis by promoting cancer stem cell self-renewal and epithelial to mesenchymal transition. Emerging evidence suggests that Bmi-1 overexpression is closely related to the development and progression of various types of cancer, and that downregulation of Bmi-1 expression can inhibit the proliferation, invasion and metastasis of cancer cells. It is therefore important to elucidate the mechanisms underlying the regulation of Bmi-1 expression both under normal growth conditions and in malignant tissues. In the present review, the current body of knowledge pertaining to the transcriptional and post-transcriptional regulation of the BMI-1 gene is discussed, and the potential mechanisms by which Bmi-1 is dysregulated in various types of cancer are highlighted. Bmi-1 expression is primarily controlled via transcriptional regulation, and is regulated by the transcription https://www.ushuaia.pl/hyphen/?ln=en factors of the Myc family, including Myb, Twist1, SALL4 and E2F-1. Post-transcriptionally, regulation of Bmi-1 expression is inhibited by several microRNAs and certain small-molecule drugs. Thus, regulatory transcriptional factors are potential therapeutic targets to reduce Bmi-1 expression in cancer cells. Thus, the present review provides an up-to-date review on the regulation of BMI-1 gene expression at the transcriptional and post-transcriptional level.
Collapse
Affiliation(s)
- Meizhen Zhou
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qichao Xu
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Deqiang Huang
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lingyu Luo
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
4
|
Wang H, Hu Z, Wu J, Mei Y, Zhang Q, Zhang H, Miao D, Sun W. Sirt1 Promotes Osteogenic Differentiation and Increases Alveolar Bone Mass via Bmi1 Activation in Mice. J Bone Miner Res 2019; 34:1169-1181. [PMID: 30690778 DOI: 10.1002/jbmr.3677] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 12/27/2018] [Accepted: 01/11/2019] [Indexed: 12/16/2022]
Abstract
Sirtuin 1 (Sirt1), a protein deacetylase, is a novel target for bone metabolism. To investigate whether overexpression of Sirt1 in mandibular mesenchymal stem cells (M-MSCs) increased alveolar bone mass in vivo, we generated Sirt1 transgenic mice (Sirt1TG ), with Sirt1 gene expression driven by the Prx1 gene, which represents the mesenchymal lineage. Our results demonstrated that overexpression of Sirt1 in M-MSCs increased the alveolar bone volume in 1-month-old, 9-month-old, and 18-month-old Sirt1TG mice compared with age-matched wild-type (WT) mice, and in ovariectomized Sirt1TG mice compared with ovariectomized WT mice by stimulating M-MSC differentiation into osteoblasts. Treatment with resveratrol, a Sirt1 activator, increased Sirt1 binding with Bmi1 and reduced Bmi1 acetylation in a dose-dependent manner demonstrated in M-MSC cultures. Both treatment with resveratrol in M-MSC cultures and overexpressed Sirt1 in M-MSCs ex vivo cultures increased nuclear translocation of Bmi1. Furthermore, we demonstrated that deletion of Bmi1 blocked the increased alveolar bone volume in Sirt1TG mice. The Sirt1 activator resveratrol inhibited human MSC senescence and promoted their differentiation into osteoblasts, which were associated with upregulating the expression levels of Sirt1 and nuclear translocation of Bmi1. The present results suggested that Sirt1 promotes MSC proliferation and osteogenic differentiation, inhibits MSC senescence to increase alveolar bone volume by promoting the deacetylation and nuclear translocation of Bmi1. Thus, our study elucidated the mechanism by which Sirt1 increases alveolar bone mass, and these findings are important for the clinical application of the Sirt1 activator resveratrol for the promotion of alveolar bone formation and prevention of alveolar bone loss. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hua Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zixuan Hu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Jun Wu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Yukun Mei
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Qian Zhang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Hengwei Zhang
- Center for Musculoskeletal Research (CMSR), Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Dengshun Miao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Banerjee Mustafi S, Aznar N, Dwivedi SKD, Chakraborty PK, Basak R, Mukherjee P, Ghosh P, Bhattacharya R. Mitochondrial BMI1 maintains bioenergetic homeostasis in cells. FASEB J 2016; 30:4042-4055. [PMID: 27613804 PMCID: PMC5102112 DOI: 10.1096/fj.201600321r] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/15/2016] [Indexed: 12/14/2022]
Abstract
The polycomb complex proto-oncogene BMI1 [B lymphoma Mo-MLV insertion region 1 homolog (mouse)] is essential for self-renewal of normal and cancer stem cells. BMI1-null mice show severe defects in growth, development, and survival. Although BMI1 is known to exert its effect in the nucleus via repression of 2 potent cell-cycle regulators that are encoded by the Ink4a/Arf locus, deletion of this locus only partially rescues BMI1-null phenotypes, which is indicative of alternate mechanisms of action of BMI1. Here, we show that an extranuclear pool of BMI1 localizes to inner mitochondrial membrane and directly regulates mitochondrial RNA (mtRNA) homeostasis and bioenergetics. These mitochondrial functions of BMI1 are independent of its previously described nuclear functions because a nuclear localization-defective mutant BMI1 rescued several bioenergetic defects that we observed in BMI1-depleted cells, for example, mitochondrial respiration, cytochrome c oxidase activity, and ATP production. Mechanistically, BMI1 coprecipitated with polynucleotide phosphorylase, a ribonuclease that is responsible for decay of mtRNA transcripts. Loss of BMI1 enhanced ribonuclease activity of polynucleotide phosphorylase and reduced mtRNA stability. These findings not only establish a novel extranuclear role of BMI1 in the regulation of mitochondrial bioenergetics, but also provide new mechanistic insights into the role of this proto-oncogene in stem cell differentiation, neuronal aging, and cancer.-Banerjee Mustafi, S., Aznar, N., Dwivedi, S. K. D., Chakraborty, P. K., Basak, R., Mukherjee, P., Ghosh, P., Bhattacharya, R. Mitochondrial BMI1 maintains bioenergetic homeostasis in cells.
Collapse
Affiliation(s)
- Soumyajit Banerjee Mustafi
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA
| | - Nicolas Aznar
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Shailendra Kumar Dhar Dwivedi
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA
| | - Prabir Kumar Chakraborty
- Department of Pathology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and
| | - Rumki Basak
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA
| | - Priyabrata Mukherjee
- Department of Pathology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and
| | - Pradipta Ghosh
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA;
- Department of Cell Biology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma, USA
| |
Collapse
|
6
|
Gray F, Cho HJ, Shukla S, He S, Harris A, Boytsov B, Jaremko Ł, Jaremko M, Demeler B, Lawlor ER, Grembecka J, Cierpicki T. BMI1 regulates PRC1 architecture and activity through homo- and hetero-oligomerization. Nat Commun 2016; 7:13343. [PMID: 27827373 PMCID: PMC5105191 DOI: 10.1038/ncomms13343] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 09/26/2016] [Indexed: 12/21/2022] Open
Abstract
BMI1 is a core component of the polycomb repressive complex 1 (PRC1) and emerging data support a role of BMI1 in cancer. The central domain of BMI1 is involved in protein-protein interactions and is essential for its oncogenic activity. Here, we present the structure of BMI1 bound to the polyhomeotic protein PHC2 illustrating that the central domain of BMI1 adopts an ubiquitin-like (UBL) fold and binds PHC2 in a β-hairpin conformation. Unexpectedly, we find that the UBL domain is involved in homo-oligomerization of BMI1. We demonstrate that both the interaction of BMI1 with polyhomeotic proteins and homo-oligomerization via UBL domain are necessary for H2A ubiquitination activity of PRC1 and for clonogenic potential of U2OS cells. Here, we also emphasize need for joint application of NMR spectroscopy and X-ray crystallography to determine the overall structure of the BMI1-PHC2 complex.
Collapse
Affiliation(s)
- Felicia Gray
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Hyo Je Cho
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Shirish Shukla
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Shihan He
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ashley Harris
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Bohdan Boytsov
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Łukasz Jaremko
- Deutsches Zentrum fur Neurodegenerative Erkrankungen (DZNE), Am Fassberg 11, 37077 Goettingen, Germany
- Max-Planck Institute of Biophysical Chemistry, NMR-based Department for Structural Biology, Am Fassberg 11, 37077 Goettingen, Germany
| | - Mariusz Jaremko
- Max-Planck Institute of Biophysical Chemistry, NMR-based Department for Structural Biology, Am Fassberg 11, 37077 Goettingen, Germany
| | - Borries Demeler
- Department of Biochemistry, The University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | - Elizabeth R. Lawlor
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
7
|
Zhou X, Dai X, Wu X, Ji J, Karaplis A, Goltzman D, Yang X, Miao D. Overexpression of Bmi1 in Lymphocytes Stimulates Skeletogenesis by Improving the Osteogenic Microenvironment. Sci Rep 2016; 6:29171. [PMID: 27373231 PMCID: PMC4931581 DOI: 10.1038/srep29171] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/15/2016] [Indexed: 12/13/2022] Open
Abstract
To investigate whether overexpression of Bmi1 in lymphocytes can stimulate skeletogenesis by improving the osteogenic microenvironment, we examined the skeletal phenotype of EμBmi1 transgenic mice with overexpression of Bmi1 in lymphocytes. The size of the skeleton, trabecular bone volume and osteoblast number, indices of proliferation and differentiation of bone marrow mesenchymal stem cells (BM-MSCs) were increased significantly, ROS levels were reduced and antioxidative capacity was enhanced in EμBmi1 mice compared to WT mice. In PTHrP1-84 knockin (Pthrp(KI/KI)) mice, the expression levels of Bmi1 are reduced and potentially can mediate the premature osteoporosis observed. We therefore generated a Pthrp(KI/KI) mice overexpressing Bmi1 in lymphocytes and compared them with Pthrp(KI/KI) and WT littermates. Overexpression of Bmi1 in Pthrp(KI/KI) mice resulted in a longer lifespan, increased body weight and improvement in skeletal growth and parameters of osteoblastic bone formation with reduced ROS levels and DNA damage response parameters. Our results demonstrate that overexpression of Bmi1 in lymphocytes can stimulate osteogenesis in vivo and partially rescue defects in skeletal growth and osteogenesis in Pthrp(KI/KI) mice. These studies therefore indicate that overexpression of Bmi1 in lymphocytes can stimulate skeletogenesis by inhibiting oxidative stress and improving the osteogenic microenvironment.
Collapse
Affiliation(s)
- Xichao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xiuliang Dai
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xuan Wu
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Ji Ji
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
- Department of Fundamentals of Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Andrew Karaplis
- The Department of Medicine, McGill University, Montreal, Canada
| | - David Goltzman
- The Department of Medicine, McGill University, Montreal, Canada
| | - Xiangjiao Yang
- The Department of Medicine, McGill University, Montreal, Canada
- Rosalind & Morris Goodman Cancer Research Center, Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Dengshun Miao
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Balasubramanian S, Scharadin TM, Han B, Xu W, Eckert RL. The Bmi-1 helix-turn and ring finger domains are required for Bmi-1 antagonism of (-) epigallocatechin-3-gallate suppression of skin cancer cell survival. Cell Signal 2015; 27:1336-44. [PMID: 25843776 PMCID: PMC4756650 DOI: 10.1016/j.cellsig.2015.03.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/18/2015] [Accepted: 03/21/2015] [Indexed: 01/01/2023]
Abstract
The Bmi-1 Polycomb group (PcG) protein is an important epigenetic regulator of chromatin status. Elevated Bmi-1 expression is observed in skin cancer and contributes to cancer cell survival. (-) Epigallocatechin-3-gallate (EGCG), an important green tea-derived cancer prevention agent, reduces Bmi-1 level resulting in reduced skin cancer cell survival. This is associated with increased p21(Cip1) and p27(Kip1) expression, reduced cyclin, and cyclin dependent kinase expression, and increased cleavage of apoptotic markers. These EGCG-dependent changes are attenuated by vector-mediated maintenance of Bmi-1 expression. In the present study, we identify Bmi-1 functional domains that are required for this response. Bmi-1 expression reverses the EGCG-dependent reduction in SCC-13 cell survival, but Bmi-1 mutants lacking the helix-turn-helix-turn-helix-turn (Bmi-1ΔHT) or ring finger (Bmi-1ΔRF) domains do not reverse the EGCG impact. The reduction in Ring1B ubiquitin ligase activity, observed in the presence of mutant Bmi-1, is associated with reduced ability of these mutants to interact with and activate Ring1B ubiquitin ligase, the major ligase responsible for the ubiquitination of histone H2A during chromatin condensation. This results in less chromatin condensation leading to increased tumor suppressor gene expression and reduced cell survival; thereby making the cells more susceptible to the anti-survival action of EGCG. We further show that these mutants act in a dominant-negative manner to inhibit the action of endogenous Bmi-1. Our results suggest that the HT and RF domains are required for Bmi-1 ability to maintain skin cancer cell survival in response to cancer preventive agents.
Collapse
Affiliation(s)
| | - Tiffany M Scharadin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bingshe Han
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wen Xu
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard L Eckert
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Dermatology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Obstetrics and Gynecology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Bhattacharya R, Mustafi SB, Street M, Dey A, Dwivedi SKD. Bmi-1: At the crossroads of physiological and pathological biology. Genes Dis 2015; 2:225-239. [PMID: 26448339 PMCID: PMC4593320 DOI: 10.1016/j.gendis.2015.04.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bmi-1 is a member of the Polycomb repressor complex 1 that mediates gene silencing by regulating chromatin structure and is indispensable for self-renewal of both normal and cancer stem cells. Despite three decades of research that have elucidated the transcriptional regulation, post-translational modifications and functions of Bmi-1 in regulating the DNA damage response, cellular bioenergetics, and pathologies, the entire potential of a protein with such varied functions remains to be realized. This review attempts to synthesize the current knowledge on Bmi-1 with an emphasis on its role in both normal physiology and cancer. Additionally, since cancer stem cells are emerging as a new paradigm for therapy resistance, the role of Bmi-1 in this perspective is also highlighted. The wide spectrum of malignancies that implicate Bmi-1 as a signature for stemness and oncogenesis also make it a suitable candidate for therapy. Nonetheless, new approaches are vitally needed to further characterize physiological roles of Bmi-1 with the long-term goal of using Bmi-1 as a prognostic marker and a therapeutic target.
Collapse
Affiliation(s)
- Resham Bhattacharya
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, United States of America
| | - Soumyajit Banerjee Mustafi
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, United States of America
| | - Mark Street
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, United States of America
| | - Anindya Dey
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, United States of America
| | - Shailendra Kumar Dhar Dwivedi
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, United States of America
| |
Collapse
|
10
|
Wei F, Ojo D, Lin X, Wong N, He L, Yan J, Xu S, Major P, Tang D. BMI1 attenuates etoposide-induced G2/M checkpoints via reducing ATM activation. Oncogene 2014; 34:3063-75. [PMID: 25088203 DOI: 10.1038/onc.2014.235] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/11/2014] [Accepted: 06/20/2014] [Indexed: 12/17/2022]
Abstract
The BMI1 protein contributes to stem cell pluripotency and oncogenesis via multiple functions, including its newly identified role in DNA damage response (DDR). Although evidence clearly demonstrates that BMI1 facilitates the repair of double-stranded breaks via homologous recombination (HR), it remains unclear how BMI1 regulates checkpoint activation during DDR. We report here that BMI1 has a role in G2/M checkpoint activation in response to etoposide (ETOP) treatment. Ectopic expression of BMI1 in MCF7 breast cancer and DU145 prostate cancer cells significantly reduced ETOP-induced G2/M arrest. Conversely, knockdown of BMI1 in both lines enhanced the arrest. Consistent with ETOP-induced activation of the G2/M checkpoints via the ATM pathway, overexpression and knockdown of BMI1, respectively, reduced and enhanced ETOP-induced phosphorylation of ATM at serine 1981 (ATM pS1981). Furthermore, the phosphorylation of ATM targets, including γH2AX, threonine 68 (T68) on CHK2 (CHK2 pT68) and serine 15 (S15) on p53 were decreased in overexpression and increased in knockdown BMI1 cells in response to ETOP. In line with the requirement of NBS1 in ATM activation, we were able to show that BMI1 associates with NBS1 and that this interaction altered the binding of NBS1 with ATM. BMI1 consists of a ring finger (RF), helix-turn-helix-turn-helix-turn (HT), proline/serine (PS) domain and two nuclear localization signals (NLS). Although deletion of either RF or HT did not affect the association of BMI1 with NBS1, the individual deletions of PS and one NLS (KRMK) robustly reduced the interaction. Stable expression of these BMI1 mutants decreased ETOP-induced ATM pS1981 and CHK2 pT68, but not ETOP-elicited γH2AX in MCF7 cells. Furthermore, ectopic expression of BMI1 in non-transformed breast epithelial MCF10A cells also compromised ETOP-initiated ATM pS1981 and γH2AX. Taken together, we provide compelling evidence that BMI1 decreases ETOP-induced G2/M checkpoint activation via reducing NBS1-mediated ATM activation.
Collapse
Affiliation(s)
- F Wei
- 1] Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada [2] Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada [3] The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada [4] The Genetics Laboratory, Institute of Women and Children's Health, Longgang District, Shenzhen, Guangdong, P.R. China
| | - D Ojo
- 1] Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada [2] Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada [3] The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - X Lin
- 1] Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada [2] Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada [3] The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - N Wong
- 1] Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada [2] Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada [3] The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - L He
- 1] Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada [2] Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada [3] The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada [4] Massachusetts General Hospital (MGH), Harvard Medical School, Boston, MA, USA
| | - J Yan
- 1] Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada [2] Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada [3] The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - S Xu
- 1] Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada [2] Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada [3] The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - P Major
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | - D Tang
- 1] Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada [2] Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada [3] The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| |
Collapse
|
11
|
Yu F, Hsieh WS, Petersson F, Yang H, Li Y, Li C, Low SW, Liu J, Yan Y, Wang DY, Loh KS. Malignant cells derived from 3T3 fibroblast feeder layer in cell culture for nasopharyngeal carcinoma. Exp Cell Res 2013; 322:193-201. [PMID: 24368151 DOI: 10.1016/j.yexcr.2013.12.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/06/2013] [Accepted: 12/11/2013] [Indexed: 11/29/2022]
Abstract
In this study, we discovered a subpopulation of 3T3 feeder cells were malignantly transformed by nasopharyngeal carcinoma (NPC) tumor cells during co-culture. The transformed 3T3 cells acquired an accelerated growth rate, displayed loosely attached multilayer growth in vitro and highly tumorigenic in vivo. Most strikingly, instead of forming sarcomas, they developed into carcinoma-like tumors somewhat resembling the original NPC. We further demonstrated the transformation is not a single isolated event, rather a common reproducible, cell contact dispensable phenomena among NPC tumor cells. However, NPC tumor cells alone were not sufficient to confer the transformed characteristics onto normal human cells.
Collapse
Affiliation(s)
- Fenggang Yu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Wen-son Hsieh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Fredrik Petersson
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Yingying Li
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chunwei Li
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Soon Wah Low
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jing Liu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yan Yan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kwok Seng Loh
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Head & Neck Tumor group, National Cancer Institute of Singapore, Head & Neck Surgery, NUHS, Singapore
| |
Collapse
|
12
|
Choy B, Bandla S, Xia Y, Tan D, Pennathur A, Luketich JD, Godfrey TE, Peters JH, Sun J, Zhou Z. Clinicopathologic characteristics of high expression of Bmi-1 in esophageal adenocarcinoma and squamous cell carcinoma. BMC Gastroenterol 2012; 12:146. [PMID: 23078618 PMCID: PMC3544684 DOI: 10.1186/1471-230x-12-146] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 10/08/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND High expression of Bmi-1, a key regulatory component of the polycomb repressive complex-1, has been associated with many solid and hematologic malignancies including esophageal squamous cell carcinoma. However, little is known about the role of Bmi-1 in esophageal adenocarcinoma. The aim of this study is to investigate the amplification and high expression of Bmi-1 and the associated clinicopathologic characteristics in esophageal adenocarcinoma and squamous cell carcinoma. METHODS The protein expression level of Bmi-1 was detected by immunohistochemistry (IHC) from tissue microarrays (TMA) constructed at the University of Rochester from using tissues accrued between 1997 and 2005. Types of tissues included adenocarcinoma, squamous cell carcinoma and precancerous lesions. Patients' survival data, demographics, histologic diagnoses and tumor staging data were collected. The intensity (0-3) and percentage of Bmi-1 expression on TMA slides were scored by two pathologists. Genomic DNA from 116 esophageal adenocarcinoma was analyzed for copy number aberrations using Affymetrix SNP 6.0 arrays. Fisher exact tests and Kaplan-Meier methods were used to analyze data. RESULTS By IHC, Bmi-1 was focally expressed in the basal layers of almost all esophageal squamous mucosa, which was similar to previous reports in other organs related to stem cells. High Bmi-1 expression significantly increased from squamous epithelium (7%), columnar cell metaplasia (22%), Barrett's esophagus (22%), to low- (45%) and high-grade dysplasia (43%) and adenocarcinoma (37%). The expression level of Bmi-1 was significantly associated with esophageal adenocarcinoma differentiation. In esophageal adenocarcinoma, Bmi-1 amplification was detected by DNA microarray in a low percentage (3%). However, high Bmi-1 expression did not show an association with overall survival in both esophageal adenocarcinoma and squamous cell carcinoma. CONCLUSIONS This study demonstrates that high expression Bmi-1 is associated with esophageal adenocarcinoma and precancerous lesions, which implies that Bmi-1 plays an important role in early carcinogenesis in esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Bonnie Choy
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Santhoshi Bandla
- Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Yinglin Xia
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Dongfeng Tan
- Department of Pathology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Arjun Pennathur
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center C-800, Presbyterian University Hospital, Pittsburgh, PA, 15213, USA
| | - James D Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center C-800, Presbyterian University Hospital, Pittsburgh, PA, 15213, USA
| | - Tony E Godfrey
- Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Jeffrey H Peters
- Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Jun Sun
- Department of Gastroenterology, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Biochemistry, Rush University Medical Center, Cohn Research Building, 1735 W. Harrison St., Room 506, Chicago, IL, 60612, USA
| | - Zhongren Zhou
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| |
Collapse
|
13
|
Kouzmina M, Häyry V, Leikola J, Haglund C, Böhling T, Koljonen V, Hagström J. BMI1 expression identifies subtypes of Merkel cell carcinoma. Virchows Arch 2012; 461:647-53. [PMID: 23064620 DOI: 10.1007/s00428-012-1327-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Revised: 07/31/2012] [Accepted: 09/28/2012] [Indexed: 12/24/2022]
Abstract
Merkel cell carcinoma (MCC) is a rare cutaneous neuroendocrine carcinoma. The aims of this study were to investigate the expression of the transcription factors B-lymphoma Moloney murine leukaemia virus insertion (BMI1), myelocytomatosis viral oncogene homologue (c-Myc) and Snail in MCC tumour specimens and to examine the relationship of these markers to Merkel cell polyoma virus (MCV). The study comprised of 133 patients with primary MCC. The expression of BMI1, Snail and c-Myc protein was assessed by immunohistochemistry and compared with clinical parameters, MCV status and patient survival. The presence of MCV was inversely correlated with the expression of BMI1 protein. Tumours expressing BMI1 protein more often presented with lymph node metastases. Snail protein expression was decreased in cases with metastatic dissemination. This study identified two subgroups of MCC: tumours expressing BMI1 but negative for MCV DNA and tumours negative for BMI1 expression but positive for MCV. Importantly, BMI1-positive cases often presented with lymph node metastases. Combined, these results suggest that subtypes of this malignancy exist.
Collapse
Affiliation(s)
- Maria Kouzmina
- Department of Oral and Maxillofacial Surgery, Helsinki University Central Hospital, Kasarminkatu 11-13, 00029 HUS, Helsinki, Finland.
| | | | | | | | | | | | | |
Collapse
|
14
|
Prezioso C, Orlando V. Polycomb proteins in mammalian cell differentiation and plasticity. FEBS Lett 2011; 585:2067-77. [PMID: 21575638 DOI: 10.1016/j.febslet.2011.04.062] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 04/22/2011] [Accepted: 04/27/2011] [Indexed: 12/31/2022]
Abstract
During development cell differentiation is accompanied by progressive restriction of the developmental potential and increased structural and functional specialization of cells. In this context, mechanisms of cell memory guarantee that cells maintain different identities previously determined by the integrated action of signalling and specific sets of transcription factors. Unraveling the molecular basis by which cells build and maintain their memory represents one of the most fascinating problems in biology. PcG proteins were originally identified as part of an epigenetic cellular memory system that controls gene silencing via chromatin structure. However, recent reports suggest that they are also involved in controlling dynamics and plasticity of gene regulation, particularly during differentiation, by interacting with other components of the transcriptional apparatus. In this review, we discuss the role of PcG proteins in pluripotent ES cells and in well known mammalian cell differentiation systems including skeletal muscle, epidermal, neuronal differentiation. The emerging picture suggests that indeed, plasticity and not rigidity is a fundamental aspect of PcG physiology and cell memory function.
Collapse
Affiliation(s)
- Carolina Prezioso
- Dulbecco Telethon Institute, IRCCS Santa Lucia, Laboratory of Epigenetics and Genome Reprogramming, Rome, Italy
| | | |
Collapse
|
15
|
Eckert RL, Adhikary G, Rorke EA, Chew YC, Balasubramanian S. Polycomb group proteins are key regulators of keratinocyte function. J Invest Dermatol 2010; 131:295-301. [PMID: 21085188 DOI: 10.1038/jid.2010.318] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The Polycomb group (PcG) proteins are epigenetic suppressors of gene expression that function through modification of histones to change chromatin structure and modulate gene expression and cell behavior. Recent studies show that PcG proteins are expressed in epidermis, that their levels change during differentiation and in disease states, and that PcG expression is regulated by agents that influence cell proliferation and survival. The results indicate that PcG proteins regulate keratinocyte cell-cycle progression, apoptosis, senescence, and differentiation. These proteins are expressed in progenitor cells, in the basal layer, and in suprabasal keratinocytes, and the level, timing, and distribution of expression suggest that the PcG proteins have a central role in maintaining the balance between cell survival and death in multiple epidermal compartments. Additional studies indicate an important role in skin cancer progression.
Collapse
Affiliation(s)
- Richard L Eckert
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.
| | | | | | | | | |
Collapse
|
16
|
Kim RH, Lieberman MB, Lee R, Shin KH, Mehrazarin S, Oh JE, Park NH, Kang MK. Bmi-1 extends the life span of normal human oral keratinocytes by inhibiting the TGF-beta signaling. Exp Cell Res 2010; 316:2600-8. [PMID: 20630502 PMCID: PMC2924923 DOI: 10.1016/j.yexcr.2010.04.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Revised: 04/09/2010] [Accepted: 04/10/2010] [Indexed: 01/07/2023]
Abstract
We previously demonstrated that Bmi-1 extended the in vitro life span of normal human oral keratinocytes (NHOK). We now report that the prolonged life span of NHOK by Bmi-1 is, in part, due to inhibition of the TGF-beta signaling pathway. Serial subculture of NHOK resulted in replicative senescence and terminal differentiation and activation of TGF-beta signaling pathway. This was accompanied with enhanced intracellular and secreted TGF-beta1 levels, phosphorylation of Smad2/3, and increased expression of p15(INK4B) and p57(KIP2). An ectopic expression of Bmi-1 in NHOK (HOK/Bmi-1) decreased the level of intracellular and secreted TGF-beta1 induced dephosphorylation of Smad2/3, and diminished the level of p15(INK4B) and p57(KIP2). Moreover, Bmi-1 expression led to the inhibition of TGF-beta-responsive promoter activity in a dose-specific manner. Knockdown of Bmi-1 in rapidly proliferating HOK/Bmi-1 and cancer cells increased the level of phosphorylated Smad2/3, p15(INK4B), and p57(KIP2). In addition, an exposure of senescent NHOK to TGF-beta receptor I kinase inhibitor or anti-TGF-beta antibody resulted in enhanced replicative potential of cells. Taken together, these data suggest that Bmi-1 suppresses senescence of cells by inhibiting the TGF-beta signaling pathway in NHOK.
Collapse
Affiliation(s)
- Reuben H. Kim
- UCLA School of Dentistry, Los Angeles, CA 90095
- UCLA Dental Research Institute, Los Angeles, CA 90095
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095
| | | | - Rachel Lee
- UCLA School of Dentistry, Los Angeles, CA 90095
| | - Ki-Hyuk Shin
- UCLA School of Dentistry, Los Angeles, CA 90095
- UCLA Dental Research Institute, Los Angeles, CA 90095
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095
| | | | - Ju-Eun Oh
- UCLA School of Dentistry, Los Angeles, CA 90095
| | - No-Hee Park
- UCLA School of Dentistry, Los Angeles, CA 90095
- UCLA Dental Research Institute, Los Angeles, CA 90095
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095
- David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| | - Mo K. Kang
- UCLA School of Dentistry, Los Angeles, CA 90095
- UCLA Dental Research Institute, Los Angeles, CA 90095
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095
| |
Collapse
|
17
|
Yadav AK, Sahasrabuddhe AA, Dimri M, Bommi PV, Sainger R, Dimri GP. Deletion analysis of BMI1 oncoprotein identifies its negative regulatory domain. Mol Cancer 2010; 9:158. [PMID: 20569464 PMCID: PMC2900245 DOI: 10.1186/1476-4598-9-158] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 06/22/2010] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The polycomb group (PcG) protein BMI1 is an important regulator of development. Additionally, aberrant expression of BMI1 has been linked to cancer stem cell phenotype and oncogenesis. In particular, its overexpression has been found in several human malignancies including breast cancer. Despite its established role in stem cell maintenance, cancer and development, at present not much is known about the functional domains of BMI1 oncoprotein. In the present study, we carried out a deletion analysis of BMI1 to identify its negative regulatory domain. RESULTS We report that deletion of the C-terminal domain of BMI1, which is rich in proline-serine (PS) residues and previously described as PEST-like domain, increased the stability of BMI1, and promoted its pro-oncogenic activities in human mammary epithelial cells (HMECs). Specifically, overexpression of a PS region deleted mutant of BMI1 increased proliferation of HMECs and promoted an epithelial-mesenchymal transition (EMT) phenotype in the HMECs. Furthermore, when compared to the wild type BMI1, exogenous expression of the mutant BMI1 led to a significant downregulation of p16INK4a and an efficient bypass of cellular senescence in human diploid fibroblasts. CONCLUSIONS In summary, our data suggest that the PS domain of BMI1 is involved in its stability and that it negatively regulates function of BMI1 oncoprotein. Our results also suggest that the PS domain of BMI1 could be targeted for the treatment of proliferative disorders such as cancer and aging.
Collapse
Affiliation(s)
- Ajay K Yadav
- Department of Medicine, NorthShore University HealthSystem Research Institute, Evanston, IL 60201, USA
| | | | | | | | | | | |
Collapse
|
18
|
Zhang J, Sarge KD. Identification of a polymorphism in the RING finger of human Bmi-1 that causes its degradation by the ubiquitin-proteasome system. FEBS Lett 2009; 583:960-4. [PMID: 19233177 DOI: 10.1016/j.febslet.2009.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 02/11/2009] [Accepted: 02/15/2009] [Indexed: 02/02/2023]
Abstract
Bmi-1 is a polycomb protein that plays an important role in tumor cell development and maintaining stem cell populations of many cell lineages. Here we identify a polymorphism in human Bmi-1 that changes a cysteine within its RING domain to tyrosine. This C18Y polymorphism is associated with a significant decrease in Bmi-1 level and its elevated ubiquitination, suggesting that it is being destroyed by the ubiquitin-proteasome system. Consistent with this, treating cells with the proteasome inhibitor MG-132 significantly increases C18Y Bmi-1 levels. This is the first example of a polymorphism in Bmi-1 that reduces levels of this important protein.
Collapse
Affiliation(s)
- Jie Zhang
- Graduate Center for Toxicology, Chandler Medical Center, University of Kentucky, Lexington, KY 40536, USA
| | | |
Collapse
|
19
|
Miao D, Su H, He B, Gao J, Xia Q, Zhu M, Gu Z, Goltzman D, Karaplis AC. Severe growth retardation and early lethality in mice lacking the nuclear localization sequence and C-terminus of PTH-related protein. Proc Natl Acad Sci U S A 2008; 105:20309-14. [PMID: 19091948 PMCID: PMC2629292 DOI: 10.1073/pnas.0805690105] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Indexed: 12/25/2022] Open
Abstract
Parathyroid hormone (PTH) plays a central role in the regulation of serum calcium and phosphorus homeostasis, while parathyroid hormone-related protein (PTHrP) has important developmental roles. Both peptides signal through the same G protein-coupled receptor, the PTH/PTHrP or PTH type 1 receptor (PTH1R). PTHrP, normally a secreted protein, also contains a nuclear localization signal (NLS) that in vitro imparts functionality to the protein at the level of the nucleus. We investigated this functionality in vivo by introducing a premature termination codon in Pthrp in ES cells and generating mice that express PTHrP (1-84), a truncated form of the protein that is missing the NLS and the C-terminal region of the protein but can still signal through its cell surface receptor. Mice homozygous for the knock-in mutation (Pthrp KI) displayed retarded growth, early senescence, and malnutrition leading postnatally to their rapid demise. Decreased cellular proliferative capacity and increased apoptosis in multiple tissues including bone and bone marrow cells were associated with altered expression and subcellular distribution of the senescence-associated tumor suppressor proteins p16(INK4a) and p21 and the oncogenes Cyclin D, pRb, and Bmi-1. These findings provide in vivo experimental proof that substantiates the biologic relevance of the NLS and C-terminal portion of PTHrP, a polypeptide ligand that signals mainly via a cell surface G protein-coupled receptor.
Collapse
Affiliation(s)
- Dengshun Miao
- Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China; and
- Calcium Research Laboratory and Department of Medicine, McGill University Health Centre, McGill University, Montreal, Canada H3A 1A1
| | - Hanyi Su
- Division of Endocrinology, Department of Medicine and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Canada H3T 1E2
| | - Bin He
- Division of Endocrinology, Department of Medicine and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Canada H3T 1E2
| | - Jianjun Gao
- Calcium Research Laboratory and Department of Medicine, McGill University Health Centre, McGill University, Montreal, Canada H3A 1A1
| | - Qingwen Xia
- Calcium Research Laboratory and Department of Medicine, McGill University Health Centre, McGill University, Montreal, Canada H3A 1A1
| | - Min Zhu
- Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China; and
| | - Zhen Gu
- Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China; and
| | - David Goltzman
- Calcium Research Laboratory and Department of Medicine, McGill University Health Centre, McGill University, Montreal, Canada H3A 1A1
| | - Andrew C. Karaplis
- Division of Endocrinology, Department of Medicine and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Canada H3T 1E2
| |
Collapse
|
20
|
Epidermal growth factor receptor 1 (EGFR1) and its variant EGFRvIII regulate TATA-binding protein expression through distinct pathways. Mol Cell Biol 2008; 28:6483-95. [PMID: 18710943 DOI: 10.1128/mcb.00288-08] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) family regulates essential biological processes. Various epithelial tumors are linked to EGFR overexpression or expression of variant forms, such as the EGFR1 variant, EGFRvIII. Perturbations in expression of the transcription initiation factor, TATA-binding protein (TBP), alter cellular growth properties. Here we demonstrate that EGFR1 and EGFRvIII, but not HER2, induce TBP expression at a transcriptional level through distinct mechanisms. EGFR1 enhances the phosphorylation and function of Elk-1, recruiting it to the TBP promoter. In contrast, EGFRvIII robustly induces c-jun expression, stimulating recruitment of c-fos/c-jun to an overlapping AP-1 site. Enhancing c-jun expression alone induces TBP promoter activity through the AP-1 site. To determine the underlying mechanism for differences in Elk-1 function and c-jun expression by these receptors, we inhibited the internalization of EGFR1. Persistent EGFR1 cell surface occupancy mimics EGFRvIII-mediated effects on Elk-1 and c-jun and switches the requirement of Elk-1 to AP-1 for TBP promoter induction. Together, these studies define a new molecular mechanism for the regulation of TBP expression. In addition, we identify distinct molecular targets of EGFR1 and EGFRvIII and demonstrate the importance of receptor internalization in distinguishing their specific functions.
Collapse
|
21
|
Häyry V, Tanner M, Blom T, Tynninen O, Roselli A, Ollikainen M, Sariola H, Wartiovaara K, Nupponen NN. Copy number alterations of the polycomb gene BMI1 in gliomas. Acta Neuropathol 2008; 116:97-102. [PMID: 18427816 DOI: 10.1007/s00401-008-0376-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 04/01/2008] [Accepted: 04/04/2008] [Indexed: 01/08/2023]
Abstract
Gliomas are heterogeneous tumours that grow in an uninhibited fashion, and these brain tumour cells share numerous characteristics with neural stem cells. The BMI1 gene encodes a component of the polycomb protein complex regulating epigenetically gene activity via histone modification. It functions for instance during the development of the central nervous system and maturation of neural cells. BMI-1 protein expression is deregulated in several forms of cancer and gene amplification has been identified in mantle cell lymphomas. Since BMI1 is located at chromosome 10p, a region implicated frequently in brain tumourigenesis, we investigated the genetic status and the corresponding expression patterns of BMI1 in a series of 100 low- and high-grade primary and recurrent gliomas. Chromogenic in situ hybridisation (CISH) with probes directed against BMI1 at 10p13 and the centromere of chromosome 10 was used in the analyses. Of all gliomas, 59% demonstrated aberrant copy numbers of BMI1. Deletions of the BMI1 locus were found in most types of tumours, and in a univariate survival analysis these cases had poor prognosis. Increased copy numbers of the BMI1 locus (3-5 copies) were found in all histological types, especially in high-grade astrocytomas. No difference in prognosis between cases with normal copy numbers and cases with increased copy numbers could be observed. This data suggests that BMI1 gene is aberrant at the chromosomal level in a subset of gliomas, and possibly contributes to brain tumour pathogenesis.
Collapse
|
22
|
Zhang S, Li D, Li E, Li L, Wang J, Wang C, Lu J, Zhang X. Expression localization of Bmi1 in mice testis. Mol Cell Endocrinol 2008; 287:47-56. [PMID: 18359150 DOI: 10.1016/j.mce.2008.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 01/02/2008] [Accepted: 01/07/2008] [Indexed: 01/15/2023]
Abstract
B cell-specific Moloney murine leukaemia virus integration site 1 (Bmi1), known as the first functional member of PcG (Polycomb Group) family, is supposed to be a key regulator of stem cell self-proliferation. In this study, we investigated its expression in testis and its impact on spermatogonia proliferation for better understanding of its role in spermatogenesis. Results showed that Bmi1 was expressed in undifferentiated spermatogonia (A(s), A(pr) and A(al) spermatogonia). Overexpression of BMI1 could promote spermatogonia proliferation, while repression of endogenous Bmi1 by RNAi resulted in inhibition of the proliferation.
Collapse
Affiliation(s)
- Shiqing Zhang
- Institute of Basic Medical Sciences, Beijing 100850, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Tonini T, D'Andrilli G, Fucito A, Gaspa L, Bagella L. Importance of Ezh2 polycomb protein in tumorigenesis process interfering with the pathway of growth suppressive key elements. J Cell Physiol 2007; 214:295-300. [PMID: 17786943 DOI: 10.1002/jcp.21241] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
An understanding of the mechanisms that uncover the dynamic changes in the distribution of the chromatin modifying enzymes and regulatory proteins on their target loci could provide further insight into the phenomenon of malignant transformation. Based on the current available data, it seems more and more clear that an abnormal expression of Ezh2, a member of the Polycomb group (PcG) protein, may be involved in the tumorigenesis process, in addition, different studies identify Ezh2 as a potential marker that distinguish aggressive prostate and breast cancer from indolent one. Recent investigation show that ectopic expression of Ezh2 provides proliferative advantage to primary cells through interaction with the pathways of key elements that control cell growth arrest and differentiation, like members of the retinoblastoma (Rb) family. Here, we outline how these pathways converge and we review the recent advances on the molecular mechanisms that promote cell cycle progression through deregulation of Ezh2 protein level, providing novel links between cancer progression and chromatin remodeling machineries.
Collapse
Affiliation(s)
- Tiziana Tonini
- Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
24
|
Lee K, Adhikary G, Balasubramanian S, Gopalakrishnan R, McCormick T, Dimri GP, Eckert RL, Rorke EA. Expression of Bmi-1 in epidermis enhances cell survival by altering cell cycle regulatory protein expression and inhibiting apoptosis. J Invest Dermatol 2007; 128:9-17. [PMID: 17625597 PMCID: PMC3043461 DOI: 10.1038/sj.jid.5700949] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The polycomb group (PcG) genes are epigenetic suppressors of gene expression that play an important role in development. In this study, we examine the role of Bmi-1 (B-cell-specific Moloney murine leukemia virus integration site 1) as a regulator of human epidermal keratinocyte survival. We identify Bmi-1 mRNA and protein expression in epidermis and in cultured human keratinocytes. Bmi-1 is located in the nucleus in cultured keratinocytes, and in epidermis it is expressed in the basal and suprabasal layers. Adenovirus-delivered Bmi-1 promotes keratinocyte survival and protects keratinocytes from stress agent-mediated cell death. This is associated with increased levels of cyclin D1 and selected cyclin-dependent kinases, and reduced caspase activity and poly(ADP-ribose) polymerase (PARP) cleavage. Bmi-1 may be involved in the maintenance of disease state, as Bmi-1 levels are elevated in transformed keratinocytes, skin tumors, and psoriasis. The presence of Bmi-1 in suprabasal non-proliferative cells of the epidermis and within a high percentage of cells within skin tumors suggests a non-stem cell pro-survival role for Bmi-1 in this tissue. Based on the suprabasal distribution of Bmi-1 in epidermis, we propose that Bmi-1 may promote maintenance of suprabasal keratinocyte survival to prevent premature death during differentiation. Such a function would help assure proper formation of the stratified epidermis.
Collapse
Affiliation(s)
- Kathy Lee
- Departments of Physiology and Biophysics, Case Western Reserve School of Medicine, Cleveland, Ohio, USA
| | - Gautam Adhikary
- Departments of Physiology and Biophysics, Case Western Reserve School of Medicine, Cleveland, Ohio, USA
| | | | - Ramamurthy Gopalakrishnan
- Departments of Physiology and Biophysics, Case Western Reserve School of Medicine, Cleveland, Ohio, USA
| | - Thomas McCormick
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Goberdhan P. Dimri
- Department of Medicine, Division of Cancer Biology, Evanston, Illinois, USA
| | - Richard L. Eckert
- Departments of Physiology and Biophysics, Case Western Reserve School of Medicine, Cleveland, Ohio, USA
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Oncology, Case Western Reserve University School Medicine, Cleveland, Ohio, USA
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Ellen A. Rorke
- Department of Environmental Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
25
|
Wiederschain D, Chen L, Johnson B, Bettano K, Jackson D, Taraszka J, Wang YK, Jones MD, Morrissey M, Deeds J, Mosher R, Fordjour P, Lengauer C, Benson JD. Contribution of polycomb homologues Bmi-1 and Mel-18 to medulloblastoma pathogenesis. Mol Cell Biol 2007; 27:4968-79. [PMID: 17452456 PMCID: PMC1951487 DOI: 10.1128/mcb.02244-06] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Bmi-1 and Mel-18 are structural homologues that belong to the Polycomb group of transcriptional regulators and are believed to stably maintain repression of gene expression by altering the state of chromatin at specific promoters. While a number of clinical and experimental observations have implicated Bmi-1 in human tumorigenesis, the role of Mel-18 in cancer cell growth has not been investigated. We report here that short hairpin RNA-mediated knockdown of either Bmi-1 or Mel-18 in human medulloblastoma DAOY cells results in the inhibition of proliferation, loss of clonogenic survival, anchorage-independent growth, and suppression of tumor formation in nude mice. Furthermore, overexpression of both Bmi-1 and Mel-18 significantly increases the clonogenic survival of Rat1 fibroblasts. In contrast, stable downregulation of Bmi-1 or Mel-18 alone does not affect the growth of normal human WI38 fibroblasts. Proteomics-based characterization of Bmi-1 and Mel-18 protein complexes isolated from cancer cells revealed substantial similarities in their respective compositions. Finally, gene expression analysis identified a number of cancer-relevant pathways that may be controlled by Bmi-1 and Mel-18 and also showed that these Polycomb proteins regulate a set of common gene targets. Taken together, these results suggest that Bmi-1 and Mel-18 may have overlapping functions in cancer cell growth.
Collapse
Affiliation(s)
- Dmitri Wiederschain
- Oncology Research, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Haga K, Ohno SI, Yugawa T, Narisawa-Saito M, Fujita M, Sakamoto M, Galloway DA, Kiyono T. Efficient immortalization of primary human cells by p16INK4a-specific short hairpin RNA or Bmi-1, combined with introduction of hTERT. Cancer Sci 2007; 98:147-54. [PMID: 17233832 PMCID: PMC11158394 DOI: 10.1111/j.1349-7006.2006.00373.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Activation of telomerase is sufficient for immortalization of some types of human cells but additional factors may also be essential. It has been proposed that stress imposed by inadequate culture conditions induces senescence due to accumulation of p16(INK4a). Here, we present evidence that many human cell types undergo senescence by activation of the p16(INK4a)/Rb pathway, and that introduction of Bmi-1 can inhibit p16(INK4a) expression and extend the life span of human epithelial cells derived from skin, mammary gland and lung. Introduction of p16(INK4a)-specific short hairpin RNA, as well as Bmi-1, suppressed p16(INK4a) expression in human mammary epithelial cells without promoter methylation, and extended their life span. Subsequent introduction of hTERT, the telomerase catalytic subunit, into cells with low p16(INK4a) levels resulted in efficient immortalization of three cell types without crisis or growth arrest. The majority of the human mammary epithelial cells thus immortalized showed almost normal ploidy as judged by G-banding and spectral karyotyping analysis. Our data suggest that inhibition of p16(INK4a) and introduction of hTERT can immortalize many human cell types with little chromosomal instability.
Collapse
Affiliation(s)
- Kei Haga
- Virology Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuoku 104-0045, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Ben-Saadon R, Zaaroor D, Ziv T, Ciechanover A. The Polycomb Protein Ring1B Generates Self Atypical Mixed Ubiquitin Chains Required for Its In Vitro Histone H2A Ligase Activity. Mol Cell 2006; 24:701-711. [PMID: 17157253 DOI: 10.1016/j.molcel.2006.10.022] [Citation(s) in RCA: 191] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2006] [Revised: 08/31/2006] [Accepted: 10/12/2006] [Indexed: 11/23/2022]
Abstract
Polycomb complexes mediate gene silencing, in part by modifying histones. Ring1B and Bmi1 are RING finger proteins that are members of the Polycomb repressive complex 1 (PRC1). Ring1B is an E3 that mediates its own polyubiquitination and monoubiquitination of histone H2A. In contrast, Bmi1 has no self-ubiquitinating activity. We show that unlike other RING finger proteins that are believed to mediate their own ubiquitination and degradation, Ring1B and Bmi1 are degraded by an exogenous E3, independent of their RING domain. The RING domains of both proteins mediate their association and subsequent stabilization. Consistent with the nonproteolytic self-ligase activity of Ring1B, it generates atypical mixed K6-, K27-, and K48-based polyubiquitin chains, which require the presence of all these lysine residues on the same ubiquitin molecule. The modification is required for Ring1B ability to monoubiquitinate H2A in vitro, unraveling an as yet undescribed mechanism for ligase activation via noncanonical self-ubiquitination.
Collapse
Affiliation(s)
- Ronen Ben-Saadon
- Cancer and Vascular Biology Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Efron Street, Bat Galim, Haifa 31096, Israel
| | - Daphna Zaaroor
- Cancer and Vascular Biology Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Efron Street, Bat Galim, Haifa 31096, Israel
| | - Tamar Ziv
- Faculty of Biology, Technion-Israel Institute of Technology, Efron Street, Bat Galim, Haifa 31096, Israel
| | - Aaron Ciechanover
- Cancer and Vascular Biology Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Efron Street, Bat Galim, Haifa 31096, Israel.
| |
Collapse
|
28
|
Maurelli R, Zambruno G, Guerra L, Abbruzzese C, Dimri G, Gellini M, Bondanza S, Dellambra E. Inactivation of p16INK4a (inhibitor of cyclin-dependent kinase 4A) immortalizes primary human keratinocytes by maintaining cells in the stem cell compartment. FASEB J 2006; 20:1516-8. [PMID: 16754749 DOI: 10.1096/fj.05-4480fje] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Replicative senescence of human keratinocytes is determined by a progressive decline of clonogenic and dividing cells, and its timing is controlled by clonal evolution (i.e., the transition from stem cells to transient amplifying and postmitotic cells). Progressive increase of p16INK4a (inhibitor of cyclin-dependent kinase 4A) expression has been shown to correlate with keratinocyte clonal evolution. Thus, the aim of our study is to understand whether p16INK4a accumulation is a triggering mechanism of epidermal clonal evolution or a secondary event. We show that inactivation of p16INK4a, by an antisense strategy, allows primary human keratinocytes to escape replicative senescence. Specifically, p16INK4a inactivation alone blocks clonal evolution and maintains keratinocytes in the stem cell compartment. Antisense excision is followed by keratinocyte senescence, confirming that persistent p16INK4a inactivation is required for maintenance of clonal evolution block. Immortalization is accompanied by resumption of B-Cell Specific Moloney murine leukemia virus site 1 (Bmi-1) expression and telomerase activity, hallmarks of tissue regenerative capacity. In turn, Bmi-1 expression is necessary to maintain the impairment of clonal evolution induced by p16INK4a inactivation. Finally, p16INK4a down-regulation in transient amplifying keratinocytes does not affect clonal evolution, and cells undergo senescence. Thus, p16INK4a inactivation appears to selectively prevent clonal conversion in cells endowed with a high proliferative potential. These data indicate that p16INK4a regulates keratinocyte clonal evolution and that inactivation of p16INK4a in epidermal stem cells is necessary for maintaining stemness.
Collapse
Affiliation(s)
- Riccardo Maurelli
- Laboratory of Tissue Engineering and Cutaneous Physiopathology, I.D.I.-IRCCS, Istituto Dermopatico dell'Immacolata, Via dei Castelli Romani, 83/85, Pomezia (Roma), 00040 Italy.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Liu L, Andrews LG, Tollefsbol TO. Loss of the human polycomb group protein BMI1 promotes cancer-specific cell death. Oncogene 2006; 25:4370-5. [PMID: 16501599 PMCID: PMC2423216 DOI: 10.1038/sj.onc.1209454] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The polycomb group protein BMI1 has been shown to support normal stem cell proliferation via its putative stem cell factor function, but it is not known if BMI1 may also act as a cancer stem cell factor to promote cancer development. To determine the role of human BMI1 in cancer growth and survival, we performed a loss-of-function analysis of BMI1 by RNA interference (RNAi) in both normal and malignant human cells. Our results indicate that BMI1 is crucial for the short-term survival of cancer cells but not of normal cells. We also demonstrated that loss of BMI1 was more effective in suppressing cancer cell growth than retinoid-treatment, and surviving cancer cells showed significantly reduced tumorigenicity. The cancer-specific growth retardation was mediated by an increased level of apoptosis and a delayed cell cycle progression due to the loss of BMI1. By comparison, BMI1 deficiency caused only a moderate inhibition of the cell cycle progression in normal lung cells. In both normal and cancer cells, the loss of BMI1 led to an upregulation of INK4A-ARF, but with no significant effect on the level of telomerase gene expression, suggesting that other BMI1-cooperative factors in addition to INK4A-ARF activation may be involved in the BMI1-dependent cancer-specific growth retardation. Thus, human BMI1 is critical for the short-term survival of cancer cells, and inhibition of BMI1 has minimal effect on the survival of normal cells. These findings provide a foundation for developing a cancer-specific therapy targeting BMI1.
Collapse
Affiliation(s)
- L Liu
- Department of Biology, University of Alabama at Birmingham, 35294, USA.
| | | | | |
Collapse
|
30
|
King IFG, Emmons RB, Francis NJ, Wild B, Müller J, Kingston RE, Wu CT. Analysis of a polycomb group protein defines regions that link repressive activity on nucleosomal templates to in vivo function. Mol Cell Biol 2005; 25:6578-91. [PMID: 16024794 PMCID: PMC1190323 DOI: 10.1128/mcb.25.15.6578-6591.2005] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Polycomb group (PcG) genes propagate patterns of transcriptional repression throughout development. The products of several such genes are part of Polycomb repressive complex 1 (PRC1), which inhibits chromatin remodeling and transcription in vitro. Genetic and biochemical studies suggest the product of the Posterior sex combs (Psc) gene plays a central role in both PcG-mediated gene repression in vivo and PRC1 activity in vitro. To dissect the relationship between the in vivo and in vitro activities of Psc, we identified the lesions associated with 11 genetically characterized Psc mutations and asked how the corresponding mutant proteins affect Psc activity on nucleosomal templates in vitro. Analysis of both single-mutant Psc proteins and recombinant complexes containing mutant protein revealed that Psc encodes at least two functions, complex formation and the inhibition of remodeling and transcription, which require different regions of the protein. There is an excellent correlation between the in vivo phenotypes of mutant Psc alleles and the structure and in vitro activities of the corresponding proteins, suggesting that the in vitro activities of PRC1 reflect essential functions of Psc in vivo.
Collapse
Affiliation(s)
- Ian F G King
- Massachusetts General Hospital, Dept. of Molecular Biology, Welman 10, 50 Blossom St., Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Wang S, Robertson GP, Zhu J. A novel human homologue of Drosophila polycomblike gene is up-regulated in multiple cancers. Gene 2005; 343:69-78. [PMID: 15563832 DOI: 10.1016/j.gene.2004.09.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2004] [Revised: 09/03/2004] [Accepted: 09/10/2004] [Indexed: 10/26/2022]
Abstract
Polycomb group (PcG) proteins function to maintain the stable epigenetic repression of homeotic genes and other important developmental and cell cycle regulatory genes. Such maintenance establishes a form of cellular memory for its identity or state of differentiation. Accumulating evidence indicates that perturbation of this transcriptional memory may be required for tumor progression and may represent a hallmark of cancer. We have identified a novel gene encoding a human homologue of the Drosophila polycomblike protein, hPCL3. Through alternative polyadenylation and/or splicing, the gene encodes two nuclear proteins, hPCL3S and hPCL3L. Both proteins repressed transcription upon recruitment to the proximity of an HSV-tk promoter by a Gal4 DNA binding domain. Interestingly, the products of the hPCL3 gene, particularly the short form, hPCL3S, are markedly overexpressed in many types of cancers, including colon, skin, lung, rectal, cervical, uterus, and liver cancers. This increase in expression correlated with tumor progression. Both hPCL3S and hPCL3L messages were increased dramatically in most cell lines derived from various stages of melanoma and glioma tumor progression. Thus, our data link PcG deregulation to the progression of multiple cancers and may have important implications for unraveling the mechanisms of tumor progression.
Collapse
Affiliation(s)
- Shuwen Wang
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | |
Collapse
|
32
|
Gong Y, Wang X, Liu J, Shi L, Yin B, Peng X, Qiang B, Yuan J. NSPc1, a mainly nuclear localized protein of novel PcG family members, has a transcription repression activity related to its PKC phosphorylation site at S183. FEBS Lett 2005; 579:115-21. [PMID: 15620699 DOI: 10.1016/j.febslet.2004.11.056] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2004] [Revised: 11/15/2004] [Accepted: 11/17/2004] [Indexed: 02/07/2023]
Abstract
Nervous system polycomb 1 (NSPc1) shares high homology with vertebrate PcG proteins Mel-18 and Bmi-1. The mRNA of NSPc1 is highly expressed in the developmental nervous system [Mech. Dev. 102 (2001) 219-222]. However, the functional characterization of NSPc1 protein is not clear. In the present study, using Western blotting technique, we aimed to describe the distributions of NSPc1 protein in rat tissues and cell lines. The subcellular localization of NSPc1 was examined in HeLa and SH-SY5Ycell lines, and its transcriptional repression activity was examined in COS-7 cell line. We found that the NSPc1 protein was localized mainly in the nucleus. NSPc1 remarkably repressed the transcription. Most interestingly, both the C-terminal of NSPc1 and two phosphorylation sites in the C-terminal, especially the PKC phosphorylation site at S183, were important in mediating transcription repression. Taken together, results from our study suggest that NSPc1, as a typical PcG family member, has powerful transcriptional repression ability, which may be related to the PKC signaling pathway.
Collapse
Affiliation(s)
- Yanhua Gong
- Institute of Basic Medical Sciences, National Laboratory of Medical Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, National Human Genome Center, Beijing 100005, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Tonini T, Bagella L, D'Andrilli G, Claudio PP, Giordano A. Ezh2 reduces the ability of HDAC1-dependent pRb2/p130 transcriptional repression of cyclin A. Oncogene 2004; 23:4930-7. [PMID: 15077161 DOI: 10.1038/sj.onc.1207608] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The polycomb group (PcG) proteins are known to be involved in maintaining the silenced state of several developmentally regulated genes. Enhancer of zeste homolog 2 (Ezh2), a member of this large protein family, has also been shown to be deregulated in different tumor types and its role, both as a potential primary effector and as a mediator of tumorigenesis, has become a subject of increased interest. We observed that Ezh2 binds to pRb2/p130, a member of the retinoblastoma family; as such, we were led to consider the possible ability of Ezh2 to modulate cell cycle progression. Both Ezh2 and pRb2/p130 repress gene expression by recruiting histone deacetylase (HDAC1), which decreases DNA accessibility for activating transcription factors. Additionally, we observed that Ezh2 interacts with the C-terminal region of pRb2/p130, essential for interaction with HDAC1. We show that Ezh2 is able to reverse pRb2/p130-HDAC1-mediated repression of the cyclin A promoter. This indicates a functional role of this complex in regulating cyclin A expression, known to be crucial in mediating cell cycle advancement. We also detected a significant decrease in the retention of HDAC1 activity associated with pRb2/p130 when Ezh2 was overexpressed. Finally, electromobility shift assays (EMSA) demonstrated that overexpression of Ezh2 caused the abrogation of the pRb2/p130-HDAC1 complex on the cyclin A promoter. These data, taken together, suggest that Ezh2 competes with HDAC1 in binding to pRb2/p130, disrupting their occupancy on the cyclin A promoter. In this study, we propose a new mechanism for the functional inactivation of pRb2/p130 that ultimately contributes to cell cycle progression and malignant transformation.
Collapse
Affiliation(s)
- Tiziana Tonini
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | | | | | | | | |
Collapse
|
34
|
Smith KS, Chanda SK, Lingbeek M, Ross DT, Botstein D, van Lohuizen M, Cleary ML. Bmi-1 Regulation of INK4A-ARF Is a Downstream Requirement for Transformation of Hematopoietic Progenitors by E2a-Pbx1. Mol Cell 2003; 12:393-400. [PMID: 14536079 DOI: 10.1016/s1097-2765(03)00277-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Loss-of-function alterations of INK4A are commonly observed in lymphoid malignancies, but are consistently absent in pre-B cell leukemias induced by the chimeric oncoprotein E2a-Pbx1 created by t(1;19) chromosomal translocations. We report here that experimental induction of E2a-Pbx1 enhances expression of BMI-1, a lymphoid oncogene whose product functions as a transcriptional repressor of the INK4A-ARF tumor suppressor locus. Bmi-1-deficient hematopoietic progenitors are resistant to transformation by E2a-Pbx1; however, the requirement for Bmi-1 is alleviated in cells deficient for both Bmi-1 and INK4A-ARF. Furthermore, the adverse effects of E2a-Pbx1 on pre-B cell survival and differentiation are partially bypassed by forced expression of p16(Ink4a). These results link E2a-Pbx1 with Bmi-1 on an oncogenic pathway that is likely to play a role in the pathogenesis of human lymphoid leukemias through downregulation of the INK4A-ARF gene.
Collapse
Affiliation(s)
- Kevin S Smith
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Itahana K, Zou Y, Itahana Y, Martinez JL, Beausejour C, Jacobs JJL, Van Lohuizen M, Band V, Campisi J, Dimri GP. Control of the replicative life span of human fibroblasts by p16 and the polycomb protein Bmi-1. Mol Cell Biol 2003; 23:389-401. [PMID: 12482990 PMCID: PMC140680 DOI: 10.1128/mcb.23.1.389-401.2003] [Citation(s) in RCA: 322] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The polycomb protein Bmi-1 represses the INK4a locus, which encodes the tumor suppressors p16 and p14(ARF). Here we report that Bmi-1 is downregulated when WI-38 human fibroblasts undergo replicative senescence, but not quiescence, and extends replicative life span when overexpressed. Life span extension by Bmi-1 required the pRb, but not p53, tumor suppressor protein. Deletion analysis showed that the RING finger and helix-turn-helix domains of Bmi-1 were required for life span extension and suppression of p16. Furthermore, a RING finger deletion mutant exhibited dominant negative activity, inducing p16 and premature senescence. Interestingly, presenescent cultures of some, but not all, human fibroblasts contained growth-arrested cells expressing high levels of p16 and apparently arrested by a p53- and telomere-independent mechanism. Bmi-1 selectively extended the life span of these cultures. Low O(2) concentrations had no effect on p16 levels or life span extension by Bmi-1 but reduced expression of the p53 target, p21. We propose that some human fibroblast strains are more sensitive to stress-induced senescence and have both p16-dependent and p53/telomere-dependent pathways of senescence. Our data suggest that Bmi-1 extends the replicative life span of human fibroblasts by suppressing the p16-dependent senescence pathway.
Collapse
Affiliation(s)
- Koji Itahana
- Life Sciences Division, Lawrence Berkeley National Laboratory, California 94720, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Akasaka T, Takahashi N, Suzuki M, Koseki H, Bodmer R, Koga H. MBLR, a new RING finger protein resembling mammalian Polycomb gene products, is regulated by cell cycle-dependent phosphorylation. Genes Cells 2002; 7:835-50. [PMID: 12167161 DOI: 10.1046/j.1365-2443.2002.00565.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The RING finger proteins function in a variety of fundamental cellular processes. The products of some members of the Polycomb group (PcG) bear ring finger domains and are defined as a subclass of RING finger proteins. Among them are Drosophila posterior sex combs and suppressor 2 of zeste, whose RING fingers are conserved in vertebrate PcG proteins Mel18 and Bmi1. RESULTS We have identified a new mammalian RING finger protein, termed MBLR due to its structural similarity to Mel18 and Bmi1 (Mel18 and Bmi1-like RING finger protein). MBLR interacts with some PcG proteins: in vitro biochemical data support the idea of a direct interaction of MBLR's RING finger domain with Ring1B, which is highly homologous to one of the mammalian PcG genes, Ring1A. We also show that MBLR acts as a transcriptional repressor in transiently transfected cells, as is the case for other PcG proteins. Immunocytochemical analysis reveals that MBLR protein is localized in a fine-grained distribution throughout the nucleoplasm in interphase cultured cells and in a fainter diffuse cytoplasmic distribution in mitotic cells. In addition, we find that serine 32 of MBLR is specifically phosphorylated during mitosis, most likely by CDK7, a component of the basal transcriptional machinery. CONCLUSION Similarities to previously defined PcG proteins suggest that MBLR should be included in the same subclass of RING finger proteins as Mel18 and Bmi1. Although the biological relevance of the cell cycle-related phosphorylation remains to be demonstrated, serine 32 phosphorylation could nevertheless be functionally important.
Collapse
Affiliation(s)
- Takeshi Akasaka
- Department of Cellular, Molecular and Developmental Biology, University of Michigan, 830 N University, Ann Arbor, MI 48109-1048, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Jacobs JJL, van Lohuizen M. Polycomb repression: from cellular memory to cellular proliferation and cancer. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1602:151-61. [PMID: 12020801 DOI: 10.1016/s0304-419x(02)00052-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The transcriptional repressors of the Polycomb group (PcG), together with the counteracting Trithorax group (TrxG) proteins, establish a form of cellular memory by regulating gene expression in a heritable fashion at the level of chromatin. This cellular memory function is required for a correct cell fate/behavior, which is not only crucial during development for the generation of a correct body plan but also later in life to prevent cellular transformation. Here, we summarize the rapidly accumulating data that implicate several mammalian PcG members in the control of cellular proliferation and tumorigenesis.
Collapse
Affiliation(s)
- Jacqueline J L Jacobs
- Department of Molecular Genetics, The Netherlands Cancer Institute, H5, Plesmanlaan 121, Amsterdam, The Netherlands
| | | |
Collapse
|
38
|
Dai KS, Liew CC. A novel human striated muscle RING zinc finger protein, SMRZ, interacts with SMT3b via its RING domain. J Biol Chem 2001; 276:23992-9. [PMID: 11283016 DOI: 10.1074/jbc.m011208200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The RING domain is a conserved zinc finger motif, which serves as a protein-protein interaction interface. Searches of a human heart expressed sequence tag data base for genes encoding the RING domain identified a novel cDNA, named striated muscle RING zinc finger protein (SMRZ). The SMRZ cDNA is 1.9 kilobase pairs in length and encodes a polypeptide of 288 amino acid residues; analysis of the peptide sequence demonstrated an N-terminal RING domain. Fluorescence in situ hybridization localized SMRZ to chromosome 1p33-34. Northern blots demonstrated that SMRZ is expressed exclusively in striated muscle. In the cardiovascular system, SMRZ is more highly expressed in the fetal heart than in the adult heart (slightly higher expression in the ventricle than in the atrium), suggesting that SMRZ is developmentally regulated. SMRZ was found to interact with SMT3b, a ubiquitin-like protein, through the SMRZ-RING domain. This interaction was abolished by mutagenesis of conserved RING domain residues. Transient transfection of SMRZ into C2C12 myoblasts showed localization of SMRZ to the nucleus. These data suggest that SMRZ may play an important role in striated muscle cell embryonic development and perhaps in cell cycle regulation.
Collapse
Affiliation(s)
- K S Dai
- Institute of Medical Science and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5G 1L5, Canada
| | | |
Collapse
|
39
|
Nakakura EK, Watkins DN, Schuebel KE, Sriuranpong V, Borges MW, Nelkin BD, Ball DW. Mammalian Scratch: a neural-specific Snail family transcriptional repressor. Proc Natl Acad Sci U S A 2001; 98:4010-5. [PMID: 11274425 PMCID: PMC31170 DOI: 10.1073/pnas.051014098] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Members of the Snail family of zinc finger transcription factors are known to play critical roles in neurogenesis in invertebrates, but none of these factors has been linked to vertebrate neuronal differentiation. We report the isolation of a gene encoding a mammalian Snail family member that is restricted to the nervous system. Human and murine Scratch (Scrt) share 81% and 69% identity to Drosophila Scrt and the Caenorhabditis elegans neuronal antiapoptotic protein, CES-1, respectively, across the five zinc finger domain. Expression of mammalian Scrt is predominantly confined to the brain and spinal cord, appearing in newly differentiating, postmitotic neurons and persisting into postnatal life. Additional expression is seen in the retina and, significantly, in neuroendocrine (NE) cells of the lung. In a parallel fashion, we detect hScrt expression in lung cancers with NE features, especially small cell lung cancer. hScrt shares the capacity of other Snail family members to bind to E-box enhancer motifs, which are targets of basic helix--loop--helix (bHLH) transcription factors. We show that hScrt directly antagonizes the function of heterodimers of the proneural bHLH protein achaete-scute homolog-1 and E12, leading to active transcriptional repression at E-box motifs. Thus, Scrt has the potential to function in newly differentiating, postmitotic neurons and in cancers with NE features by modulating the action of bHLH transcription factors critical for neuronal differentiation.
Collapse
Affiliation(s)
- E K Nakakura
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Bárdos JI, Saurin AJ, Tissot C, Duprez E, Freemont PS. HPC3 is a new human polycomb orthologue that interacts and associates with RING1 and Bmi1 and has transcriptional repression properties. J Biol Chem 2000; 275:28785-92. [PMID: 10825164 DOI: 10.1074/jbc.m001835200] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Polycomb group (PcG) proteins were first described in Drosophila as factors responsible for maintaining the transcriptionally repressed state of Hox/homeotic genes in a stable and heritable manner throughout development. A growing number of vertebrate genes related to the Drosophila PcG proteins have recently been identified, including two Polycomb orthologues, Pc2 and M33. PcG proteins form multiprotein complexes, termed PcG bodies, that are thought to repress transcription by altering chromatin structure. Here we report the identification and characterization of HPC3 (human Polycomb 3), a novel PcG protein isolated in a yeast two-hybrid screen using human RING1 as bait. HPC3 shows strong sequence similarity to Drosophila Pc and also to vertebrate Pc2 and M33, particularly within the chromodomain and C-box. Previous studies indicate that M33 and human Pc2 (HPC2) can interact with RING1, and we show here that HPC3 also binds to RING1. This interaction is dependent upon the HPC3 C-box but, only partially on the RING finger of RING1. In contrast to HPC2, HPC3 interactions with RING1 are only observed in vivo with covalently modified forms of RING1. HPC3 also colocalizes with other PcG proteins in human PcG bodies. Consistent with its role as a PcG member, HPC3 is able to act as a long range transcriptional silencer when targeted to a reporter gene by a heterologous DNA-binding domain. Taken together, these data suggest that HPC3 is part of a large multiprotein complex that also contains other PcG proteins and is involved in repression of transcriptional activity.
Collapse
Affiliation(s)
- J I Bárdos
- Molecular Structure and Function Laboratory, Imperial Cancer Research Fund, P.O. Box 123, London WC2A 3PX, United Kingdom
| | | | | | | | | |
Collapse
|
41
|
Johnson SA, Mandavia N, Wang HD, Johnson DL. Transcriptional regulation of the TATA-binding protein by Ras cellular signaling. Mol Cell Biol 2000; 20:5000-9. [PMID: 10866657 PMCID: PMC85950 DOI: 10.1128/mcb.20.14.5000-5009.2000] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Our previous studies have demonstrated that the level of the central transcription factor TATA-binding protein (TBP) is increased in cells expressing the hepatitis B virus (HBV) X protein through the activation of the Ras signaling pathway, which serves to enhance both RNA polymerase I and III promoter activities. To understand the mechanism by which TBP is regulated, we have investigated whether enhanced expression is modulated at the transcriptional level. Nuclear run-on assays revealed that the HBV X protein increases the number of active transcription complexes on the TBP gene. In transient-transfection assays with both transformed and primary hepatocytes, the human TBP promoter was shown to be induced by expression of the HBV X protein in a Ras-dependent manner, requiring both Ral guanine nucleotide dissociation stimulator (RalGDS) and Raf signaling. Transient overexpression of TBP did not affect TBP promoter activity. To further delineate the downstream Ras-mediated events contributing to TBP promoter regulation in primary rat hepatocytes, the best-characterized Ras effectors, Raf, phosphoinositide 3-kinase (PI-3 kinase), and RalGDS, were examined. Activation of either Raf or RalGDS, but not that of PI-3 kinase, was sufficient to induce TBP promoter activity. Both Raf- and RalGDS-mediated induction required the activation of mitogen-activated protein kinase kinase (MEK). In addition, another distinct Ras-activated pathway, which does not require MEK activation, appears to induce TBP promoter activity. Analysis of the DNA sequence requirement within the TBP promoter responsible for these regulatory events defined three distinct regions that modulate the abilities of Raf, RalGDS, and the Ras-dependent, MEK-independent pathways to regulate human TBP promoter activity. Together, these results provide new evidence that TBP can be regulated at the transcriptional level and identify three distinct Ras-activated pathways that modulate this central eukaryotic transcription factor.
Collapse
Affiliation(s)
- S A Johnson
- Departments of Molecular Pharmacology and Biochemistry, Norris Comprehensive Cancer Center, University of Southern California School of Pharmacy and Keck School of Medicine, Los Angeles, California 90089-9121, USA
| | | | | | | |
Collapse
|
42
|
Kinnon S, Fulton R. Nucleotide sequence of the feline Bmi-1 coding region. DNA SEQUENCE : THE JOURNAL OF DNA SEQUENCING AND MAPPING 2000; 10:335-8. [PMID: 10727088 DOI: 10.3109/10425179909033960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The bmi-1 gene was discovered as a clonal integration site of the Moloney murine leukaemia virus in B-cell lymphomas. The Bmi-1 protein contains the RING finger motif and is homologous to two Drosophila proteins known to be part of a multimeric protein complex involved in repressing gene transcription. A similar role for the highly conserved Bmi-1 protein in mammalian cells has been suggested. The coding regions for the mouse and human genes are known and are 92% homologous. This study involved PCR amplification, cloning, and sequencing of the 980bp feline bmi-1 coding region which was shown to be 92% and 97% homologous to the mouse and human genes respectively. From the open reading frame the feline protein is 326 amino acids in length and is 99% homologous to the human protein and 97% homologous to the mouse protein. This data is consistent with the closer relationship between the feline and human genomes and provides another experimental system in which to analyse Bmi-1 function.
Collapse
Affiliation(s)
- S Kinnon
- School of Biological and Biomedical Sciences, Glasgow Caledonian University, Scotland
| | | |
Collapse
|
43
|
Geiman DE, Ton-That H, Johnson JM, Yang VW. Transactivation and growth suppression by the gut-enriched Krüppel-like factor (Krüppel-like factor 4) are dependent on acidic amino acid residues and protein-protein interaction. Nucleic Acids Res 2000; 28:1106-13. [PMID: 10666450 PMCID: PMC102607 DOI: 10.1093/nar/28.5.1106] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Gut-enriched Krüppel-like factor (GKLF or KLF4) is a pleiotropic (activating and repressive) transcription factor. This study characterizes the mechanisms of transactivation by GKLF. Using a GAL4 fusion assay, the activating domain of murine GKLF was localized to the 109 amino acid residues in the N-terminus. Site-directed mutagenesis showed that two adjacent clusters of acidic residues within this region are responsible for the activating effect. Transactivation by GKLF involves intermolecular interactions as demonstrated by the ability of wild-type, but not mutated, GKLF to compete with the N-terminal activation domain. In addition, wild-type adenovirus E1A, but not a mutated E1A that failed to bind p300/CBP, inhibited transactivation by the N-terminal 109 amino acids of GKLF, suggesting that p300/CBP are GKLF's interacting partners. A physical interaction between GKLF and CBP was demonstrated by glutathione- S -transferase pull-down and by in vivo co-immuno-precipitation experiments. We also showed that the two acidic amino acid clusters are essential for this interaction, since GKLF with mutations in these residues failed to co-immunoprecipitate with CBP. Importantly, the same mutations abrogated the ability of GKLF to suppress cell growth as determined by a colony suppression assay. These studies therefore provide plausible evidence for a structural and functional correlation between the transactivating and growth-suppressing effects of GKLF.
Collapse
Affiliation(s)
- D E Geiman
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
44
|
Satijn DP, Otte AP. Polycomb group protein complexes: do different complexes regulate distinct target genes? BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1447:1-16. [PMID: 10500238 DOI: 10.1016/s0167-4781(99)00130-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- D P Satijn
- E.C. Slater Instituut, University of Amsterdam, Plantage Muidergracht 12, 1018 TV, Amsterdam, The Netherlands
| | | |
Collapse
|
45
|
García E, Marcos-Gutiérrez C, del Mar Lorente M, Moreno JC, Vidal M. RYBP, a new repressor protein that interacts with components of the mammalian Polycomb complex, and with the transcription factor YY1. EMBO J 1999; 18:3404-18. [PMID: 10369680 PMCID: PMC1171420 DOI: 10.1093/emboj/18.12.3404] [Citation(s) in RCA: 192] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The products of the Polycomb group (PcG) of genes are necessary for the maintenance of transcriptional repression of a number of important developmental genes, including the homeotic genes. A two-hybrid screen was used to search for putative new members of the PcG of genes in mammals. We have identified a new Zn finger protein, RYBP, which interacts directly with both Ring1 proteins (Ring1A and Ring1B) and with M33, two mutually interacting sets of proteins of the mammalian Polycomb complex. Ring1 binds RYBP and M33 through the same C-terminal domain, whereas the RYBP-M33 interaction takes place through an M33 domain not involved in Ring1 binding. RYBP also interacts directly with YY1, a transcription factor partially related to the product of the Drosophila pleiohomeotic gene. In addition, we show here that RYBP acts as a transcriptional repressor in transiently transfected cells. Finally, RYBP shows a dynamic expression pattern during embryogenesis which initially overlaps partially that of Ring1A in the central nervous system, and later becomes ubiquitous. Taken together, these data suggest that RYBP may play a relevant role in PcG function in mammals.
Collapse
Affiliation(s)
- E García
- Centro de Investigaciones Biológicas, Department of Developmental and Cell Biology, Velázquez 144, 28006 Madrid, Spain
| | | | | | | | | |
Collapse
|
46
|
Satijn DP, Otte AP. RING1 interacts with multiple Polycomb-group proteins and displays tumorigenic activity. Mol Cell Biol 1999; 19:57-68. [PMID: 9858531 PMCID: PMC83865 DOI: 10.1128/mcb.19.1.57] [Citation(s) in RCA: 102] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Polycomb-group (PcG) proteins form large multimeric protein complexes that are involved in maintaining the transcriptionally repressive state of genes. Previously, we reported that RING1 interacts with vertebrate Polycomb (Pc) homologs and is associated with or is part of a human PcG complex. However, very little is known about the role of RING1 as a component of the PcG complex. Here we undertake a detailed characterization of RING1 protein-protein interactions. By using directed two-hybrid and in vitro protein-protein analyses, we demonstrate that RING1, besides interacting with the human Pc homolog HPC2, can also interact with itself and with the vertebrate PcG protein BMI1. Distinct domains in the RING1 protein are involved in the self-association and in the interaction with BMI1. Further, we find that the BMI1 protein can also interact with itself. To better understand the role of RING1 in regulating gene expression, we overexpressed the protein in mammalian cells and analyzed differences in gene expression levels. This analysis shows that overexpression of RING1 strongly represses En-2, a mammalian homolog of the well-characterized Drosophila PcG target gene engrailed. Furthermore, RING1 overexpression results in enhanced expression of the proto-oncogenes c-jun and c-fos. The changes in expression levels of these proto-oncogenes are accompanied by cellular transformation, as judged by anchorage-independent growth and the induction of tumors in athymic mice. Our data demonstrate that RING1 interacts with multiple human PcG proteins, indicating an important role for RING1 in the PcG complex. Further, deregulation of RING1 expression leads to oncogenic transformation by deregulation of the expression levels of certain oncogenes.
Collapse
Affiliation(s)
- D P Satijn
- E. C. Slater Instituut, BioCentrum Amsterdam, University of Amsterdam, 1018 TV Amsterdam, The Netherlands
| | | |
Collapse
|
47
|
Tatematsu K, Tokunaga C, Nakagawa N, Tanizawa K, Kuroda S, Kikkawa U. Transcriptional activity of RBCK1 protein (RBCC protein interacting with PKC 1): requirement of RING-finger and B-Box motifs and regulation by protein kinases. Biochem Biophys Res Commun 1998; 247:392-6. [PMID: 9642138 DOI: 10.1006/bbrc.1998.8795] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The RBCK1 protein was recently identified as a protein kinase C-interacting protein with a new type of RBCC (RING-B-Box-Coiled-coil) region, possessing both DNA-binding and transcriptional activities unlike other proteins in the RBCC protein family (Tokunaga et al. Biochem. Biophys. Res. Commun. 244, 353-359, 1998). To identify protein motifs in the RBCC region of RBCK1 essential for the transcriptional activity, RBCK1 mutant proteins have been constructed and analyzed by using the GAL4 chimeric transcription regulator system. We have found that both of the RING-finger and the B-Box motifs are indispensable for the transcriptional activity of RBCK1. This is the first observation that these protein motifs of the RBCC protein family play a crucial role in transcriptional activation. In addition, we have examined the effect of co-expression of several protein kinases on the transcriptional activity of RBCK1. Protein kinase A (PKA) was found to enhance the activity by about eightfold, whereas both ERK (extracellular signal-regulated kinase) activator kinase 1 (MEK1) and MEK kinase 1 (MEKK1) significantly repressed the activity. Because RBCC proteins are presumed to act as a proto-oncoprotein, these results suggest that the RBCK1 protein is involved in the intracellular signaling cascades along with PKA, MEK1, and MEKK1 and mediates cell growth and differentiation.
Collapse
Affiliation(s)
- K Tatematsu
- Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Alkema MJ, Jacobs J, Voncken JW, Jenkins NA, Copeland NG, Satijn DP, Otte AP, Berns A, van Lohuizen M. MPc2, a new murine homolog of the Drosophila polycomb protein is a member of the mouse polycomb transcriptional repressor complex. J Mol Biol 1997; 273:993-1003. [PMID: 9367786 DOI: 10.1006/jmbi.1997.1372] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The evolutionarily conserved polycomb and trithorax-group genes are required to maintain stable expression patterns of homeotic genes and other target genes throughout development. Here, we report the cloning and characterization of a novel mouse polycomb homolog, MPc2, in addition to the previously described M33 polycomb gene. Co-immunoprecipitations and subnuclear co-localization studies show that MPc2 interacts with the mouse polycomb-group oncoprotein Bmi1 and is a new member of the mouse polycomb multiprotein complex. Gal4DB-MPc2 or -M33 fusion proteins mediate a five- to tenfold repression of stably integrated reporter constructs carrying GAL4 binding sites, demonstrating that these proteins are transcriptional repressors. The MPc2 gene is localized on chromosome 11, in close proximity to the classical mouse mutations tail short (Ts) and rabo torcido (Rbt). Ts and Rbt hemizygous mice display anemia and transformations of the axial skeleton reminiscent of phenotypes observed in mice with mutated polycomb or trithorax-group genes, suggesting that MPc2 is a candidate gene for Ts and Rbt.
Collapse
Affiliation(s)
- M J Alkema
- The Netherlands Cancer Institute and Department of Biochemistry, University of Amsterdam, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Satijn DP, Olson DJ, van der Vlag J, Hamer KM, Lambrechts C, Masselink H, Gunster MJ, Sewalt RG, van Driel R, Otte AP. Interference with the expression of a novel human polycomb protein, hPc2, results in cellular transformation and apoptosis. Mol Cell Biol 1997; 17:6076-86. [PMID: 9315667 PMCID: PMC232457 DOI: 10.1128/mcb.17.10.6076] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Polycomb (Pc) is involved in the stable and heritable repression of homeotic gene activity during Drosophila development. Here, we report the identification of a novel human Pc homolog, hPc2. This gene is more closely related to a Xenopus Pc homolog, XPc, than to a previously described human Pc homolog, CBX2 (hPc1). However, the hPc2 and CBX2/hPc1 proteins colocalize in interphase nuclei of human U-2 OS osteosarcoma cells, suggesting that the proteins are part of a common protein complex. To study the functions of the novel human Pc homolog, we generated a mutant protein, delta hPc2, which lacks an evolutionarily conserved C-terminal domain. This C-terminal domain is important for hPc2 function, since the delta hPc2 mutant protein which lacks the C-terminal domain is unable to repress gene activity. Expression of the delta hPc2 protein, but not of the wild-type hPc2 protein, results in cellular transformation of mammalian cell lines as judged by phenotypic changes, altered marker gene expression, and anchorage-independent growth. Specifically in delta hPc2-transformed cells, the expression of the c-myc proto-oncogene is strongly enhanced and serum deprivation results in apoptosis. In contrast, overexpression of the wild-type hPc2 protein results in decreased c-myc expression. Our data suggest that hPc2 is a repressor of proto-oncogene activity and that interference with hPc2 function can lead to derepression of proto-oncogene transcription and subsequently to cellular transformation.
Collapse
MESH Headings
- 3T3 Cells
- Amino Acid Sequence
- Animals
- Apoptosis/genetics
- Base Sequence
- Cell Nucleus/chemistry
- Cell Transformation, Neoplastic/genetics
- Cloning, Molecular
- Gene Expression Regulation, Neoplastic/genetics
- Genes, myc/genetics
- Humans
- Ligases
- Mammary Neoplasms, Experimental
- Mice
- Molecular Sequence Data
- Organ Specificity
- Osteosarcoma/chemistry
- Polycomb Repressive Complex 1
- Polycomb-Group Proteins
- Proto-Oncogene Mas
- RNA, Messenger/analysis
- RNA, Neoplasm/analysis
- Rats
- Repressor Proteins/analysis
- Repressor Proteins/genetics
- Repressor Proteins/physiology
- Sequence Analysis, DNA
- Sequence Deletion
- Sequence Homology, Amino Acid
- Sequence Homology, Nucleic Acid
- Tumor Cells, Cultured
- Ubiquitin-Protein Ligases
Collapse
Affiliation(s)
- D P Satijn
- E. C. Slater Instituut, University of Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Schoorlemmer J, Marcos-Gutiérrez C, Were F, Martínez R, García E, Satijn DP, Otte AP, Vidal M. Ring1A is a transcriptional repressor that interacts with the Polycomb-M33 protein and is expressed at rhombomere boundaries in the mouse hindbrain. EMBO J 1997; 16:5930-42. [PMID: 9312051 PMCID: PMC1170224 DOI: 10.1093/emboj/16.19.5930] [Citation(s) in RCA: 127] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In Drosophila, the products of the Polycomb group (Pc-G) of genes act as chromatin-associated multimeric protein complexes that repress expression of homeotic genes. Vertebrate Pc-G homologues have been identified, but the nature of the complexes they form and the mechanisms of their action are largely unknown. The Polycomb homologue M33 is implicated in mesoderm patterning in the mouse and here we show that it acts as a transcriptional repressor in transiently transfected cells. Furthermore, we have identified two murine proteins, Ring1A and Ring1B, that interact directly with the repressor domain of M33. Ring1A and Ring1B display blocks of similarity throughout their sequences, including an N-terminal RING finger domain. However, the interaction with M33 occurs through a region at the C-terminus. Ring1A represses transcription through sequences not involved in M33 binding. Ring1A protein co-localizes in nuclear domains with M33 and other Pc-G homologues, such as Bmi1. The expression of Ring1A at early stages of development is restricted to the neural tube, whereas M33 is expressed ubiquitously. Within the neural tube, Ring1A RNA is located at the rhombomere boundaries of the hindbrain. Taken together, these data suggest that Ring1A may contribute to a tissue-specific function of Pc-G-protein complexes during mammalian development.
Collapse
Affiliation(s)
- J Schoorlemmer
- Centro de Investigaciones Biológicas, Department of Developmental and Cell Biology, Velázquez 144, 28006 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|