1
|
Yang R, Zhou Y, Zhang T, Wang S, Wang J, Cheng Y, Li H, Jiang W, Yang Z, Zhang X. The transcription factor HBP1 promotes ferroptosis in tumor cells by regulating the UHRF1-CDO1 axis. PLoS Biol 2023; 21:e3001862. [PMID: 37406020 DOI: 10.1371/journal.pbio.3001862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 05/31/2023] [Indexed: 07/07/2023] Open
Abstract
The induction of ferroptosis in tumor cells is one of the most important mechanisms by which tumor progression can be inhibited; however, the specific regulatory mechanism underlying ferroptosis remains unclear. In this study, we found that transcription factor HBP1 has a novel function of reducing the antioxidant capacity of tumor cells. We investigated the important role of HBP1 in ferroptosis. HBP1 down-regulates the protein levels of UHRF1 by inhibiting the expression of the UHRF1 gene at the transcriptional level. Reduced levels of UHRF1 have been shown to regulate the ferroptosis-related gene CDO1 by epigenetic mechanisms, thus up-regulating the level of CDO1 and increasing the sensitivity of hepatocellular carcinoma and cervical cancer cells to ferroptosis. On this basis, we constructed metal-polyphenol-network coated HBP1 nanoparticles by combining biological and nanotechnological. MPN-HBP1 nanoparticles entered tumor cells efficiently and innocuously, induced ferroptosis, and inhibited the malignant proliferation of tumors by regulating the HBP1-UHRF1-CDO1 axis. This study provides a new perspective for further research on the regulatory mechanism underlying ferroptosis and its potential role in tumor therapy.
Collapse
Affiliation(s)
- Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Yue Zhou
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Tongjia Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Shujie Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Jiyin Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Yuning Cheng
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Zhe Yang
- Department of pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| |
Collapse
|
2
|
Cheng Y, Yang R, Zhou Y, Wang J, Zhang T, Wang S, Li H, Jiang W, Zhang X. HBP1 inhibits the development of type 2 diabetes mellitus through transcriptional activation of the IGFBP1 gene. Aging (Albany NY) 2022; 14:8763-8782. [DOI: 10.18632/aging.204364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/21/2022] [Indexed: 11/22/2022]
Affiliation(s)
- Yuning Cheng
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Yue Zhou
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Jiyin Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Tongjia Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Shujie Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| |
Collapse
|
3
|
Wang J, Yang R, Cheng Y, Zhou Y, Zhang T, Wang S, Li H, Jiang W, Zhang X. Methylation of HBP1 by PRMT1 promotes tumor progression by regulating actin cytoskeleton remodeling. Oncogenesis 2022; 11:45. [PMID: 35941115 PMCID: PMC9360041 DOI: 10.1038/s41389-022-00421-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/09/2022] Open
Abstract
HBP1 is a sequence-specific transcription factor which generally considered as a crucial growth inhibitor. Posttranslational modification of HBP1 is vital for its function. In this study, we demonstrate that HBP1 is methylated at R378 by PRMT1, which decreases HBP1 protein stability by promoting its ubiquitination and proteasome-mediated degradation. PRMT1-mediated methylation of HBP1 alleviates the repressive effects of HBP1 on tumor metastasis and growth. GSN is identified as a novel target gene of HBP1. Methylation of HBP1 promotes actin cytoskeleton remodeling, glycolysis and tumor progression by downregulating GSN (a vital actin-binding protein) levels. The methylated HBP1-GSN axis is associated with the clinical outcomes of cancer patients. This investigation elucidates the mechanism of how methylated HBP1 facilitates actin cytoskeleton remodeling, thus attenuates its tumor-suppressive function and promotes tumor progression. Targeting methylated HBP1-GSN axis may provide a therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Jiyin Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yuning Cheng
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yue Zhou
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Tongjia Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Shujie Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China.
| |
Collapse
|
4
|
Chan CY, Chang CM, Chen YH, Sheu JJC, Lin TY, Huang CY. Regulatory role of transcription factor HBP1 in anticancer efficacy of EGFR inhibitor erlotinib in HNSCC. Head Neck 2020; 42:2958-2967. [PMID: 32677158 DOI: 10.1002/hed.26346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/13/2020] [Accepted: 06/09/2020] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) is often hyperactivated in head and neck squamous cell carcinoma (HNSCC); however, its downstream mediators are not fully identified. Here, we investigate the role of transcription factor HBP1 in the anticancer efficacy of EGFR inhibitor erlotinib in HNSCC. METHODS The effect of erlotinib and HBP1 on cell proliferation and invasion was examined by flow cytometric analysis and a Matrigel invasion assay, respectively. Oral tumor specimens were used to evaluate the association between the expression level of EGFR and HBP1, and metastatic potential. RESULTS Erlotinib caused cell growth arrest in the G1 phase and sluggish invasion with a concomitant increase in HBP1 and p27 expression. The erlotinib effect was attenuated upon HBP1 knockdown. Analysis of oral tumor specimens revealed that the low HBP1/high EGFR status can predict metastatic potential. CONCLUSIONS Our data support HBP1 as a crucial mediator of EGFR-targeting inhibitors in HNSCC.
Collapse
Affiliation(s)
- Chien-Yi Chan
- Department of Nutrition, China Medical University, Taichung, Taiwan, ROC.,Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, Taiwan, ROC
| | - Chin-Ming Chang
- Department of Nutrition, China Medical University, Taichung, Taiwan, ROC
| | - Yuan-Hong Chen
- Department of Nutrition, China Medical University, Taichung, Taiwan, ROC
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yatsen University, Kaohsiung, Taiwan, ROC
| | - Tzu-Yuan Lin
- Department of Nutrition, China Medical University, Taichung, Taiwan, ROC
| | - Chun-Yin Huang
- Department of Nutrition, China Medical University, Taichung, Taiwan, ROC
| |
Collapse
|
5
|
Chen H, Liu C, Liu Y, Li H, Cheng B. Transcription factor HBP1: A regulator of senescence and apoptosis of preadipocytes. Biochem Biophys Res Commun 2019; 517:216-220. [PMID: 31331641 DOI: 10.1016/j.bbrc.2019.07.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/17/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND /aim: HMG-box protein 1 (HBP1) plays an important role in the senescence and apoptosis of mammalian cells, but its role in chicken cells remains unclear. The aim of this study was to investigate the effects of HBP1 on senescence and apoptosis of chicken preadipocytes. METHODS The immortalized chicken preadipocyte cell line (ICP2) was used as a cell model. Chicken HBP1 knockout and overexpressing preadipocyte cell lines were established using CRISPR/Cas9 gene editing technology and lentiviral infection. Western blotting was used to detect the protein expression of HBP1 and senescence markers p16 and p53. Cell senescence was measured by Sa-β-Gal staining and apoptosis was detected by flow cytometry. RESULTS HBP1 was highly expressed in senescent ICP2 cells compared with young ICP2 cells. After the deletion of HBP1, the degree of senescence, the apoptosis rate and the protein expression levels of p16 and p53 were significantly reduced. After the overexpression of HBP1, the degree of senescence, the apoptosis rate and the protein expression levels of p16 and p53 were significantly increased. CONCLUSION HBP1 promotes the senescence and apoptosis of chicken preadipocytes.
Collapse
Affiliation(s)
- Hongyan Chen
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Chang Liu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Yumeng Liu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Hui Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Bohan Cheng
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China.
| |
Collapse
|
6
|
Bollaert E, de Rocca Serra A, Demoulin JB. The HMG box transcription factor HBP1: a cell cycle inhibitor at the crossroads of cancer signaling pathways. Cell Mol Life Sci 2019; 76:1529-1539. [PMID: 30683982 PMCID: PMC11105191 DOI: 10.1007/s00018-019-03012-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/20/2018] [Accepted: 01/15/2019] [Indexed: 12/19/2022]
Abstract
HMG box protein 1 (HBP1) is a transcription factor and a potent cell cycle inhibitor in normal and cancer cells. HBP1 activates or represses the expression of different cell cycle genes (such as CDKN2A, CDKN1A, and CCND1) through direct DNA binding, cofactor recruitment, chromatin remodeling, or neutralization of other transcription factors. Among these are LEF1, TCF4, and MYC in the WNT/beta-catenin pathway. HBP1 also contributes to oncogenic RAS-induced senescence and terminal cell differentiation. Collectively, these activities suggest a tumor suppressor function. However, HBP1 is not listed among frequently mutated cancer driver genes. Nevertheless, HBP1 expression is lower in several tumor types relative to matched normal tissues. Several micro-RNAs, such as miR-155, miR-17-92, and miR-29a, dampen HBP1 expression in cancer cells of various origins. The phosphatidylinositol-3 kinase (PI3K)/AKT pathway also inhibits HBP1 transcription by preventing FOXO binding to the HBP1 promoter. In addition, AKT directly phosphorylates HBP1, thereby inhibiting its transcriptional activity. Taken together, these findings place HBP1 at the center of a network of micro-RNAs and oncoproteins that control cell proliferation. In this review, we discuss our current understanding of HBP1 function in human physiology and diseases.
Collapse
Affiliation(s)
- Emeline Bollaert
- Université Catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200, Brussels, Belgium
| | - Audrey de Rocca Serra
- Université Catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200, Brussels, Belgium
| | - Jean-Baptiste Demoulin
- Université Catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200, Brussels, Belgium.
| |
Collapse
|
7
|
Chan CY, Lin TY, Sheu JJC, Wu WC, Huang CY. Matrix metalloproteinase-13 is a target gene of high-mobility group box-containing protein 1 in modulating oral cancer cell invasion. J Cell Physiol 2018; 234:4375-4384. [PMID: 30191992 DOI: 10.1002/jcp.27223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/17/2018] [Indexed: 01/11/2023]
Abstract
Transcription factor high-mobility group box-containing protein 1 (HBP1) may function as a tumor suppressor in various types of cancer. In a previous study, we demonstrated that HBP1 suppressed cell invasion in oral cancer. To further understand the underlying mechanism, the current study is aimed at investigating how HBP1 exerts its antimetastatic potential in oral cancer. In a cell model, ectopic expression of HBP1 potently suppressed epithelial-mesenchymal transition, cellular migration, and invasion; conversely, HBP1 knockdown promoted these malignant phenotypes. The matrix metalloproteinase (MMP) family is highly implicated in tumor metastasis. Therefore, we examined the effect of HBP1 on the activation of the MMP members, MMP-2, -9, and -13 that are highly associated with the aggressiveness of oral cancer. Ectopic expression of HBP1 resulted in a mild reduction in the expression and activity of MMP-2 and -9, yet it had a potent inhibitory effect on MMP-13. In contrast, HBP1 knockdown strongly enhanced the activation of MMP-13. Further, we demonstrated that MMP-13 is a target of HBP1 transcription repression as evidenced by the identification of an HBP1 binding site in the cis proximal region of the MMP-13 promoter. More important, MMP-13 knockdown significantly alleviated HBP1 small interfering RNA-mediated promotion in cell invasion. Analysis of oral tumor specimens revealed that the low HBP1 (<0.3-fold)/high MMP-13 (>3-fold) status was associated with metastatic potential. All told, our study provides evidence supporting the idea that the HBP1-MMP-13 axis is a key regulator of the aggressiveness in oral cancer.
Collapse
Affiliation(s)
- Chien-Yi Chan
- Department of Nutrition and Health Sciences, Chang Jung Christian University, Taiwan, China.,Department of Nutrition, China Medical University, Taiwan, China
| | - Tzu-Yuan Lin
- Department of Nutrition, China Medical University, Taiwan, China
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yatsen University, Taiwan, China.,Department of Health and Nutrition Biotechnology, Asia University, Taiwan, China
| | - Wen-Chieh Wu
- Department of Nutrition, China Medical University, Taiwan, China
| | - Chun-Yin Huang
- Department of Nutrition, China Medical University, Taiwan, China
| |
Collapse
|
8
|
Chan CY, Huang SY, Sheu JJC, Roth MM, Chou IT, Lien CH, Lee MF, Huang CY. Transcription factor HBP1 is a direct anti-cancer target of transcription factor FOXO1 in invasive oral cancer. Oncotarget 2017; 8:14537-14548. [PMID: 28099936 PMCID: PMC5362424 DOI: 10.18632/oncotarget.14653] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/07/2017] [Indexed: 12/23/2022] Open
Abstract
Either FOXO1 or HBP1 transcription factor is a downstream effector of the PI3K/Akt pathway and associated with tumorigenesis. However, the relationship between FOXO1 and HBP1 in oral cancer remains unclear. Analysis of 30 oral tumor specimens revealed that mean mRNA levels of both FOXO1 and HBP1 in non-invasive and invasive oral tumors were found to be significantly lower than that of the control tissues, and the status of low FOXO1 and HBP1 (< 0.3 fold of the control) was associated with invasiveness of oral tumors. To investigate if HBP1 is a direct transcription target of FOXO1, we searched potential FOXO1 binding sites in the HBP1 promoter using the MAPPER Search Engine, and two putative FOXO1 binding sites located in the HBP1 promoter –132 to –125 bp and –343 to –336 bp were predicted. These binding sites were then confirmed by both reporter gene assays and the in cellulo ChIP assay. In addition, Akt activity manipulated by PI3K inhibitor LY294002 or Akt mutants was shown to negatively affect FOXO1-mediated HBP1 promoter activation and gene expression. Last, the biological significance of the FOXO1-HBP1 axis in oral cancer malignancy was evaluated in cell growth, colony formation, and invasiveness. The results indicated that HBP1 knockdown potently promoted malignant phenotypes of oral cancer and the suppressive effect of FOXO1 on cell growth, colony formation, and invasion was alleviated upon HBP1 knockdown in invasive oral cancer cells. Taken together, our data provide evidence for HBP1 as a direct downstream target of FOXO1 in oral cancer malignancy.
Collapse
Affiliation(s)
- Chien-Yi Chan
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yatsen University, Kaohsiung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | | | - I-Tai Chou
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Chia-Hsien Lien
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Ming-Fen Lee
- Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Chun-Yin Huang
- Department of Nutrition, China Medical University, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
9
|
Chen Y, Pan K, Wang P, Cao Z, Wang W, Wang S, Hu N, Xue J, Li H, Jiang W, Li G, Zhang X. HBP1-mediated Regulation of p21 Protein through the Mdm2/p53 and TCF4/EZH2 Pathways and Its Impact on Cell Senescence and Tumorigenesis. J Biol Chem 2016; 291:12688-12705. [PMID: 27129219 PMCID: PMC4933444 DOI: 10.1074/jbc.m116.714147] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Indexed: 01/09/2023] Open
Abstract
The activity of the CDK inhibitor p21 is associated with diverse biological activities, including cell proliferation, senescence, and tumorigenesis. However, the mechanisms governing transcription of p21 need to be extensively studied. In this study, we demonstrate that the high-mobility group box-containing protein 1 (HBP1) transcription factor is a novel activator of p21 that works as part of a complex mechanism during senescence and tumorigenesis. We found that HBP1 activates the p21 gene through enhancing p53 stability by inhibiting Mdm2-mediated ubiquitination of p53, a well known positive regulator of p21. HBP1 was also found to enhance p21 transcription by inhibiting Wnt/β-catenin signaling. We identified histone methyltransferase EZH2, the catalytic subunit of polycomb repressive complex 2, as a target of Wnt/β-catenin signaling. HBP1-mediated repression of EZH2 through Wnt/β-catenin signaling decreased the level of trimethylation of histone H3 at lysine 27 of overall and specific histone on the p21 promoter, resulting in p21 transactivation. Although intricate, the reciprocal partnership of HBP1 and p21 has exceptional importance. HBP1-mediated elevation of p21 through the Mdm2/p53 and TCF4/EZH2 pathways contributes to both cellular senescence and tumor inhibition. Together, our results suggest that the HBP1 transcription factor orchestrates a complex regulation of key genes during cellular senescence and tumorigenesis with an impact on protein ubiquitination and overall histone methylation state.
Collapse
Affiliation(s)
- Yifan Chen
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Kewu Pan
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Pingzhang Wang
- the Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhengyi Cao
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Weibin Wang
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Shuya Wang
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Ningguang Hu
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Junhui Xue
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Hui Li
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Wei Jiang
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Gang Li
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Xiaowei Zhang
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and.
| |
Collapse
|
10
|
Lee MF, Hsieh NT, Huang CY, Li CI. AllTrans-Retinoic Acid Mediates MED28/HMG Box-Containing Protein 1 (HBP1)/β-Catenin Signaling in Human Colorectal Cancer Cells. J Cell Physiol 2015; 231:1796-803. [DOI: 10.1002/jcp.25285] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/09/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Ming-Fen Lee
- Department of Nutrition and Health Sciences; Chang Jung Christian University; Tainan Taiwan, R.O.C
| | - Nien-Tsu Hsieh
- Department of Nutrition; China Medical University; Taichung Taiwan, R.O.C
| | - Chun-Yin Huang
- Department of Nutrition; China Medical University; Taichung Taiwan, R.O.C
| | - Chun-I Li
- Department of Nutrition and Health Sciences; Chang Jung Christian University; Tainan Taiwan, R.O.C
| |
Collapse
|
11
|
The expression of the tumour suppressor HBP1 is down-regulated by growth factors via the PI3K/PKB/FOXO pathway. Biochem J 2014; 460:25-34. [PMID: 24762137 DOI: 10.1042/bj20131467] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Growth factors inactivate the FOXO (forkhead box O) transcription factors through PI3K (phosphoinositide 3-kinase) and PKB (protein kinase B). By comparing microarray data from multiple model systems, we identified HBP1 (high-mobility group-box protein 1) as a novel downstream target of this pathway. HBP1 mRNA was down-regulated by PDGF (platelet-derived growth factor), FGF (fibroblast growth factor), PI3K and PKB, whereas it was up-regulated by FOXO factors. This observation was confirmed in human and murine fibroblasts as well as in cell lines derived from leukaemia, breast adenocarcinoma and colon carcinoma. Bioinformatics analysis led to the identification of a conserved consensus FOXO-binding site in the HBP1 promoter. By luciferase activity assay and ChIP, we demonstrated that FOXO bound to this site and regulated the HBP1 promoter activity in a PI3K-dependent manner. Silencing of HBP1 by shRNA increased the proliferation of human fibroblasts in response to growth factors, suggesting that HBP1 limits cell growth. Finally, by analysing a transcriptomics dataset from The Cancer Genome Atlas, we observed that HBP1 expression was lower in breast tumours that had lost FOXO expression. In conclusion, HBP1 is a novel target of the PI3K/FOXO pathway and controls cell proliferation in response to growth factors.
Collapse
|
12
|
HBP1-mediated transcriptional regulation of DNA methyltransferase 1 and its impact on cell senescence. Mol Cell Biol 2012; 33:887-903. [PMID: 23249948 DOI: 10.1128/mcb.00637-12] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The activity of DNA methyltransferase 1 (DNMT1) is associated with diverse biological activities, including cell proliferation, senescence, and cancer development. In this study, we demonstrated that the HMG box-containing protein 1 (HBP1) transcription factor is a new repressor of DNMT1 in a complex mechanism during senescence. The DNMT1 gene contains an HBP1-binding site at bp -115 to -134 from the transcriptional start site. HBP1 repressed the endogenous DNMT1 gene through sequence-specific binding, resulting in both gene-specific (e.g., p16(INK4)) and global DNA hypomethylation changes. The HBP1-mediated repression by DNMT1 contributed to replicative and premature senescence, the latter of which could be induced by Ras and HBP1 itself. A detailed investigation unexpectedly revealed that HBP1 has dual and complex transcriptional functions, both of which contribute to premature senescence. HBP1 both repressed the DNMT1 gene and activated the p16 gene in premature senescence. The opposite transcriptional functions proceeded through different DNA sequences and differential protein acetylation. While intricate, the reciprocal partnership between HBP1 and DNMT1 has exceptional importance, since its abrogation compromises senescence and promotes tumorigenesis. Together, our results suggest that the HBP1 transcription factor orchestrates a complex regulation of key genes during cellular senescence, with an impact on overall DNA methylation state.
Collapse
|
13
|
Pérez-Montarelo D, Hudson NJ, Fernández AI, Ramayo-Caldas Y, Dalrymple BP, Reverter A. Porcine tissue-specific regulatory networks derived from meta-analysis of the transcriptome. PLoS One 2012; 7:e46159. [PMID: 23049964 PMCID: PMC3458843 DOI: 10.1371/journal.pone.0046159] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 08/28/2012] [Indexed: 11/19/2022] Open
Abstract
The processes that drive tissue identity and differentiation remain unclear for most tissue types. So are the gene networks and transcription factors (TF) responsible for the differential structure and function of each particular tissue, and this is particularly true for non model species with incomplete genomic resources. To better understand the regulation of genes responsible for tissue identity in pigs, we have inferred regulatory networks from a meta-analysis of 20 gene expression studies spanning 480 Porcine Affymetrix chips for 134 experimental conditions on 27 distinct tissues. We developed a mixed-model normalization approach with a covariance structure that accommodated the disparity in the origin of the individual studies, and obtained the normalized expression of 12,320 genes across the 27 tissues. Using this resource, we constructed a network, based on the co-expression patterns of 1,072 TF and 1,232 tissue specific genes. The resulting network is consistent with the known biology of tissue development. Within the network, genes clustered by tissue and tissues clustered by site of embryonic origin. These clusters were significantly enriched for genes annotated in key relevant biological processes and confirm gene functions and interactions from the literature. We implemented a Regulatory Impact Factor (RIF) metric to identify the key regulators in skeletal muscle and tissues from the central nervous systems. The normalization of the meta-analysis, the inference of the gene co-expression network and the RIF metric, operated synergistically towards a successful search for tissue-specific regulators. Novel among these findings are evidence suggesting a novel key role of ERCC3 as a muscle regulator. Together, our results recapitulate the known biology behind tissue specificity and provide new valuable insights in a less studied but valuable model species.
Collapse
Affiliation(s)
- Dafne Pérez-Montarelo
- Computational and Systems Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Brisbane, Queensland, Australia
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Nicholas J. Hudson
- Computational and Systems Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Brisbane, Queensland, Australia
| | - Ana I. Fernández
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Yuliaxis Ramayo-Caldas
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Brian P. Dalrymple
- Computational and Systems Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Brisbane, Queensland, Australia
| | - Antonio Reverter
- Computational and Systems Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
14
|
Lee MF, Chan CY, Hung HC, Chou IT, Yee AS, Huang CY. N-acetylcysteine (NAC) inhibits cell growth by mediating the EGFR/Akt/HMG box-containing protein 1 (HBP1) signaling pathway in invasive oral cancer. Oral Oncol 2012; 49:129-35. [PMID: 22944050 DOI: 10.1016/j.oraloncology.2012.08.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 07/31/2012] [Accepted: 08/04/2012] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Overexpression of the epidermal growth factor (EGF) receptor (EGFR) gene in the squamous cell carcinomas of the head and neck (SCCHN) is often associated with inauspicious prognosis and poor survival. N-acetylcysteine (NAC), a compound from some vegetables and allium species, appears anti-tumorigenesis, but the underlying mechanism is unclear. The objective of this study is to investigate the role of NAC in EGFR-overexpressing oral cancer. MATERIALS AND METHODS Both HSC-3 and SCC-4 human tongue squamous carcinoma cell lines and an HSC-3 xenograft mouse model were used to test the anti-growth efficacy of NAC in vitro and in vivo, respectively. RESULTS NAC treatment suppressed cell growth, with concomitantly increased expression of HMG box-containing protein 1 (HBP1), a transcription suppressor, and decreased EGFR/Akt activation, in EGFR-overexpressing HSC-3 oral cancer cells. HBP1 knockdown attenuated the growth arrest and apoptosis induced by NAC. Lastly, NAC and AG1478, an EGFR inhibitor, additively suppressed colony formation in HSC-3 cells. CONCLUSION Taken together, our data indicate that NAC exerts its growth-inhibitory function through modulating EGFR/Akt signaling and HBP1 expression in EGFR-overexpressing oral cancer.
Collapse
Affiliation(s)
- Ming-Fen Lee
- Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
15
|
Chen YC, Zhang XW, Niu XH, Xin DQ, Zhao WP, Na YQ, Mao ZB. Macrophage migration inhibitory factor is a direct target of HBP1-mediated transcriptional repression that is overexpressed in prostate cancer. Oncogene 2010; 29:3067-78. [PMID: 20383199 DOI: 10.1038/onc.2010.97] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a well-described proinflammatory mediator. MIF overexpression has been observed in many tumors and is implicated in oncogenic transformation and tumor progression. However, the molecular mechanisms responsible for regulating MIF expression remain poorly understood. In this study, we showed that the transcriptional repressor HBP1 (HMG box-containing protein 1) negatively regulates MIF expression. We first identified a large high-affinity HBP1 DNA-binding element at positions -811 to -792 from the transcriptional start site within the MIF promoter by computer analysis. Reporter analyses showed that this element was required for HBP1-mediated transcriptional repression. Furthermore, HBP1 associated with the MIF promoter in vivo and repressed endogenous MIF gene expression. Consistent with HBP1-mediated repression of MIF, low levels of HBP1 expression were associated with high levels of MIF expression in prostate cancer samples. Importantly, HBP1-mediated repression of MIF inhibited tumorigenic growth and invasion, and the repressive effect of HBP1 on tumorigenic growth and invasion could be partially rescued by the addition of recombinant MIF to the culture medium. Finally, prostate tumor samples with low HBP1 and high MIF expression were associated with a significant decrease in relapse-free survival. Taken together, these results indicated that HBP1 directly inhibited MIF gene transcription, and suggested that the loss of HBP1 expression or activity may contribute to the upregulation of MIF expression in prostate tumor tissue.
Collapse
Affiliation(s)
- Y C Chen
- The Department of Urology, Peking University First Hospital and the Institute of Urology, Peking University, Beijing, PR China
| | | | | | | | | | | | | |
Collapse
|
16
|
Fan LM, Teng L, Li JM. Knockout of p47 phox uncovers a critical role of p40 phox in reactive oxygen species production in microvascular endothelial cells. Arterioscler Thromb Vasc Biol 2009; 29:1651-6. [PMID: 19608974 DOI: 10.1161/atvbaha.109.191502] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE p40(phox) is an important regulatory subunit of NADPH oxidase, but its role in endothelial reactive oxygen species (ROS) production remains unknown. METHODS AND RESULTS Using coronary microvascular endothelial cells isolated from wild-type and p47(phox) knockout mice, we found that knockout of p47(phox) increased the level of p40(phox) expression, whereas depletion of p40(phox) in wild-type cells increased p47(phox) expression. In both cases, the basal ROS production (without agonist stimulation) was well preserved. Double knockout of p40(phox) and p47(phox) dramatically reduced (approximately 65%) ROS production and cells started to die. The transcriptional regulation of p40(phox) and p47(phox) expressions involves HBP1. p40(phox) was prephosphorylated in resting cells. PMA stimulation induced p40(phox) swift dephosphorylation (within 1 minute) in parallel with the start of p47(phox) phosphorylation. p40(phox) was then rephosphorylated, and this was accompanied with an increase in ROS production. Depletion of p40(phox) resulted in approximately 67% loss in agonist-induced ROS production despite the presence of p47(phox). These were further supported by experiments on mouse aortas stimulated with angiotensin II. CONCLUSIONS p40(phox) is prephosphorylated in resting endothelial cells and can compensate p47(phox) in keeping basal ROS production. Dephosphorylation of p40(phox) is a prerequisite for agonist-induced p47(phox) phosphorylation, and p40(phox) through its dynamic dephosphorylation and rephosphorylation is involved in the regulation of agonist-induced ROS production.
Collapse
Affiliation(s)
- Lampson M Fan
- Faculty of Health and Medical Sciences, AY Building, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | | | | |
Collapse
|
17
|
Yao CJ, Works K, Romagnoli PA, Austin GE. Effects of overexpression of HBP1 upon growth and differentiation of leukemic myeloid cells. Leukemia 2005; 19:1958-68. [PMID: 16179914 DOI: 10.1038/sj.leu.2403918] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
HMG-box containing protein 1 (HBP1) is a member of the high mobility group (HMG) of chromosomal proteins. Since HBP1 exhibits tumor-suppressor activity in nonmyeloid tissues, we examined the effects of ectopic overexpression of HBP1 upon the growth and differentiation of myeloid cells. We prepared transient and stable transfectants of the myeloblast cell line K562, which overexpress HBP1 mRNA and protein. HBP1 transfectants displayed slower growth in cell culture and reduced colony formation in soft agar, retardation of S-phase progression, reduced expression of cyclin D1 and D3 mRNAs and increased expression of p21 mRNA. HBP1 transfectants also underwent increased apoptosis, as demonstrated by morphology and binding of Annexin V. Fas ligand mRNA levels were increased in HBP1 transfectants, suggesting involvement of the Fas/Fas ligand pathway. HBP1 overexpression enhanced differentiation of K562 cells towards erythroid and megakaryocyte lineages, as evidenced by increased hemoglobin and CD41a expression. Overexpression of HBP1 modulated mRNA levels for myeloid-specific transcription factors C/EBPalpha, c-Myb, c-Myc, and JunB, as well as lineage-specific transcription factors PU.1, GATA-1, and RUNX1. These findings suggest that in myeloid cells HBP1 may serve as a tumor suppressor and a general differentiation inducer and may synergize with chemical differentiating agents to enhance lineage-specific differentiation.
Collapse
Affiliation(s)
- C J Yao
- Department of Pathology and Laboratory Medicine, Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | | | | | | |
Collapse
|
18
|
Yee AS, Paulson EK, McDevitt MA, Rieger-Christ K, Summerhayes I, Berasi SP, Kim J, Huang CY, Zhang X. The HBP1 transcriptional repressor and the p38 MAP kinase: unlikely partners in G1 regulation and tumor suppression. Gene 2004; 336:1-13. [PMID: 15225871 DOI: 10.1016/j.gene.2004.04.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Revised: 03/03/2004] [Accepted: 04/05/2004] [Indexed: 02/07/2023]
Abstract
Mechanisms that inhibit cell cycle progression and establish growth arrest are fundamental to tumor suppression and to normal cell differentiation. A complete understanding of these mechanisms should provide new diagnostic and therapeutic targets for future clinical applications related to cancer-specific pathways. This review will focus on the HMG-box protein 1 (HBP1) transcriptional repressor and its roles in cell cycle progression and tumor suppression. The work of several labs now suggests a new pathway for inhibiting G1 progression with exciting possible implications for tumor suppression. Our recent work suggests that the two previously unassociated proteins-the HBP1 transcription factor and the p38 MAP kinase pathway-may now participate together in a G1 regulatory network. Several recent papers collectively highlight an unexpected role and connection of the p38 MAP kinase-signaling pathway in cell cycle control, senescence, and tumor suppression. Together, these initially divergent observations may provide clues into a new tumor suppressive network and spur further investigations that may contribute to new diagnostic and therapeutic targets for cancer.
Collapse
Affiliation(s)
- Amy S Yee
- Department of Biochemistry, Tufts University School of Medicine, Boston, MA 02111, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Berasi SP, Xiu M, Yee AS, Paulson KE. HBP1 repression of the p47phox gene: cell cycle regulation via the NADPH oxidase. Mol Cell Biol 2004; 24:3011-24. [PMID: 15024088 PMCID: PMC371097 DOI: 10.1128/mcb.24.7.3011-3024.2004] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2003] [Revised: 10/08/2003] [Accepted: 12/30/2003] [Indexed: 11/20/2022] Open
Abstract
Several studies have linked the production of reactive oxygen species (ROS) by the NADPH oxidase to cellular growth control. In many cases, activation of the NADPH oxidase and subsequent ROS generation is required for growth factor signaling and mitogenesis in nonimmune cells. In this study, we demonstrate that the transcriptional repressor HBP1 (HMG box-containing protein 1) regulates the gene for the p47phox regulatory subunit of the NADPH oxidase. HBP1 represses growth regulatory genes (e.g., N-Myc, c-Myc, and cyclin D1) and is an inhibitor of G(1) progression. The promoter of the p47phox gene contains six tandem high-affinity HBP1 DNA-binding elements at positions -1243 to -1318 bp from the transcriptional start site which were required for repression. Furthermore, HBP1 repressed the expression of the endogenous p47phox gene through sequence-specific binding. With HBP1 expression and the subsequent reduction in p47phox gene expression, intracellular superoxide production was correspondingly reduced. Using both the wild type and a dominant-negative mutant of HBP1, we demonstrated that the repression of superoxide production through the NADPH oxidase contributed to the observed cell cycle inhibition by HBP1. Together, these results indicate that HBP1 may contribute to the regulation of NADPH oxidase-dependent superoxide production through transcriptional repression of the p47phox gene. This study defines a transcriptional mechanism for regulating intracellular ROS levels and has implications in cell cycle regulation.
Collapse
Affiliation(s)
- Stephen P Berasi
- Department of Biochemistry, Tufts University School of Medicine, School of Nutrition, Boston, Massachusetts 02111, USA
| | | | | | | |
Collapse
|
20
|
Chen YW, Allen MD, Veprintsev DB, Löwe J, Bycroft M. The Structure of the AXH Domain of Spinocerebellar Ataxin-1. J Biol Chem 2004; 279:3758-65. [PMID: 14583607 DOI: 10.1074/jbc.m309817200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Spinocerebellar ataxia type 1 is a late-onset neurodegenerative disease caused by the expansion of a CAG triplet repeat in the SCA1 gene. This results in the lengthening of a polyglutamine tract in the gene product ataxin-1. This produces a toxic gain of function that results in specific neuronal death. A region in ataxin-1, the AXH domain, exhibits significant sequence similarity to the transcription factor HBP1. This region of the protein has been implicated in RNA binding and self-association. We have determined the crystal structure of the AXH domain of ataxin-1. The AXH domain is dimeric and contains an OB-fold, a structural motif found in many oligonucleotide-binding proteins, supporting its proposed role in RNA binding. By structure comparison with other proteins that contain an OB-fold, a putative RNA-binding site has been identified. We also identified a cluster of charged surface residues that are well conserved among AXH domains. These residues may constitute a second ligand-binding surface, suggesting that all AXH domains interact with a common yet unidentified partner.
Collapse
Affiliation(s)
- Yu Wai Chen
- Centre for Protein Engineering, Medical Research Council Centre, Cambridge, UK
| | | | | | | | | |
Collapse
|
21
|
Xiu M, Kim J, Sampson E, Huang CY, Davis RJ, Paulson KE, Yee AS. The transcriptional repressor HBP1 is a target of the p38 mitogen-activated protein kinase pathway in cell cycle regulation. Mol Cell Biol 2003; 23:8890-901. [PMID: 14612426 PMCID: PMC262665 DOI: 10.1128/mcb.23.23.8890-8901.2003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The p38 mitogen-activated protein (MAP) kinase signaling pathway participates in both apoptosis and G1 arrest. In contrast to the established role in apoptosis, the documented induction of G1 arrest by activation of the p38 MAP kinase pathway has attracted recent attention with reports of substrates that are linked to cell cycle regulation. Here, we identify the high-mobility group box protein HBP1 transcriptional repressor as a new substrate for p38 MAP kinase. Our previous work had shown that HBP1 inhibits G1 progression in cell and animal models, and thus indicated that HBP1 could be a relevant substrate for p38 MAP kinase in cell cycle regulation. In the present work, a p38 MAP kinase docking site (amino acids [aa] 81 to 125) and a p38 MAP kinase phosphorylation site (serine 401) were identified in the HBP1 protein. Furthermore, the docking and phosphorylation sites on HBP1 were specific for p38 MAP kinase. In defining the role of p38 MAP kinase regulation, the inhibition of p38 MAP kinase activity was shown to decrease HBP1 protein levels by triggering protein instability, as manifested by a decrease in protein half-life. Consistently, a decrease in protein levels was accompanied by a decrease in overall DNA binding activity. A mutation of the p38 MAP kinase phosphorylation site at aa 401 [(S-A)401HBP1] also triggered HBP1 protein instability. While protein stability was compromised by mutation, the specific activities of (S-A)401HBP1 and of wild-type HBP1 appeared comparable for transcriptional repression. This comparison of transcription-specific activity highlighted that p38 MAP kinase regulated HBP1 protein levels but not the intrinsic activity for DNA binding or for transcriptional repression. Finally, p38 MAP kinase-mediated regulation of the HBP1 protein also contributed to the regulation of G1 progression. Together, our work supports a molecular framework in which p38 MAP kinase activity contributes to cell cycle inhibition by increasing HBP1 and other G1 inhibitory factors by regulating protein stability.
Collapse
Affiliation(s)
- Mei Xiu
- Department of Biochemistry, School of Medicine, Tufts University, Boston, Massachusetts 02111, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Sampson EM, Haque ZK, Ku MC, Tevosian SG, Albanese C, Pestell RG, Paulson K, Yee AS. Negative regulation of the Wnt-beta-catenin pathway by the transcriptional repressor HBP1. EMBO J 2001; 20:4500-11. [PMID: 11500377 PMCID: PMC125566 DOI: 10.1093/emboj/20.16.4500] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In certain cancers, constitutive Wnt signaling results from mutation in one or more pathway components. The result is the accumulation and nuclear localization of beta-catenin, which interacts with the lymphoid enhancer factor-1 (LEF)/T-cell factor (TCF) family of HMG-box transcription factors, which activate important growth regulatory genes, including cyclin D1 and c-myc. As exemplified by APC and axin, the negative regulation of beta-catenin is important for tumor suppression. Another potential mode of negative regulation is transcriptional repression of cyclin D1 and other Wnt target genes. In mammals, the transcriptional repressors in the Wnt pathway are not well defined. We have previously identified HBP1 as an HMG-box repressor and a cell cycle inhibitor. Here, we show that HBP1 is a repressor of the cyclin D1 gene and inhibits the Wnt signaling pathway. The inhibition of Wnt signaling and growth requires a common domain of HBP1. The apparent mechanism is an inhibition of TCF/LEF DNA binding through a physical interaction with HBP1. These data suggest that the suppression of Wnt signaling by HBP1 may be a mechanism to prevent inappropriate proliferation.
Collapse
Affiliation(s)
- Ellen M. Sampson
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 and The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Present address: Department of Genetics, Dartmouth University, Hanover, NH 03755, USA Corresponding author e-mail:
| | - Zaffar K. Haque
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 and The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Present address: Department of Genetics, Dartmouth University, Hanover, NH 03755, USA Corresponding author e-mail:
| | - Man-Ching Ku
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 and The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Present address: Department of Genetics, Dartmouth University, Hanover, NH 03755, USA Corresponding author e-mail:
| | - Sergei G. Tevosian
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 and The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Present address: Department of Genetics, Dartmouth University, Hanover, NH 03755, USA Corresponding author e-mail:
| | - Chris Albanese
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 and The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Present address: Department of Genetics, Dartmouth University, Hanover, NH 03755, USA Corresponding author e-mail:
| | - Richard G. Pestell
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 and The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Present address: Department of Genetics, Dartmouth University, Hanover, NH 03755, USA Corresponding author e-mail:
| | - K.Eric Paulson
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 and The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Present address: Department of Genetics, Dartmouth University, Hanover, NH 03755, USA Corresponding author e-mail:
| | - Amy S. Yee
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 and The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Present address: Department of Genetics, Dartmouth University, Hanover, NH 03755, USA Corresponding author e-mail:
| |
Collapse
|