1
|
Boon SS, Lee YC, Yip KL, Luk HY, Xiao C, Yim MK, Chen Z, Chan PKS. Interaction between Human Papillomavirus-Encoded E6 Protein and AurB Induces Cell Immortalization and Proliferation-A Potential Target of Intervention. Cancers (Basel) 2023; 15:cancers15092465. [PMID: 37173932 PMCID: PMC10177266 DOI: 10.3390/cancers15092465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
The human papillomavirus E6 and E7 oncoproteins interact with a different subset of host proteins, leading to dysregulation of the apoptotic, cell cycle, and signaling pathways. In this study, we identified, for the first time, that Aurora kinase B (AurB) is a bona fide interacting partner of E6. We systematically characterized the AurB-E6 complex formation and its consequences in carcinogenesis using a series of in vitro and cell-based assays. We also assessed the efficacy of Aurora kinase inhibitors in halting HPV-mediated carcinogenesis using in vitro and in vivo models. We showed that AurB activity was elevated in HPV-positive cells, and this correlated positively with the E6 protein level. E6 interacted directly with AurB in the nucleus or mitotic cells. A previously unidentified region of E6, located upstream of C-terminal E6-PBM, was important for AurB-E6 complex formation. AurB-E6 complex led to reduced AurB kinase activity. However, the AurB-E6 complex increased the hTERT protein level and its telomerase activity. On the other hand, AurB inhibition led to the inhibition of telomerase activity, cell proliferation, and tumor formation, even though this may occur in an HPV-independent manner. In summary, this study dissected the molecular mechanism of how E6 recruits AurB to induce cell immortalization and proliferation, leading to the eventual cancer development. Our findings revealed that the treatment of AZD1152 exerted a non-specific anti-tumor effect. Hence, a continuous effort to seek a specific and selective inhibitor that can halt HPV-mediated carcinogenesis should be warranted.
Collapse
Affiliation(s)
- Siaw Shi Boon
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Yin Ching Lee
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Ka Lai Yip
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Ho Yin Luk
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Chuanyun Xiao
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Man Kin Yim
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Zigui Chen
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Paul Kay Sheung Chan
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
2
|
Zhang Z, Han Z, Guo Y, Liu X, Gao Y, Zhang Y. Establishment of an Efficient Immortalization Strategy Using HMEJ-Based b TERT Insertion for Bovine Cells. Int J Mol Sci 2021; 22:ijms222212540. [PMID: 34830422 PMCID: PMC8622252 DOI: 10.3390/ijms222212540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/09/2022] Open
Abstract
Immortalized cell lines have been used in a wide range of applications in research on immune disorders and cellular metabolic regulation due to the stability and uniformity of their cellular characteristics. At present, the investigation into molecular functions and signaling pathways within bovine cells remains largely limited by the lack of immortalized model cells. Current methods for immortalizing bovine cells are mainly restricted to the ectopic expression of human telomerase reverse transcriptase (hTERT) through transient transfection or virus-mediated delivery, which have defects in efficiency and reliability. In this study, we identified bovine TERT (bTERT) as a novel potent biofactor for immortalizing bovine cells with great advantages over hTERT, and established an efficient and easily manipulated strategy for the immortalization of bovine primary cells. Through the homology-mediated end-joining-based insertion of bTERT at the ROSA26 locus, we successfully generated immortalized bovine fetal fibroblast cell lines with stable characteristics. The observed limitation of this strategy in immortalizing bovine bone marrow-derived macrophages was attributed to the post-translational modification of bTERT, causing inhibited nuclear localization and depressed activity of bTERT in this terminally differentiated cell. In summary, we constructed an innovative method to achieve the high-quality immortalization of bovine primary cells, thereby expanding the prospects for the future application of immortalized bovine model cell lines.
Collapse
Affiliation(s)
- Zihan Zhang
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (Z.Z.); (Z.H.); (Y.G.); (X.L.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Xianyang 712100, China
| | - Zhuo Han
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (Z.Z.); (Z.H.); (Y.G.); (X.L.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Xianyang 712100, China
| | - Ying Guo
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (Z.Z.); (Z.H.); (Y.G.); (X.L.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Xianyang 712100, China
| | - Xin Liu
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (Z.Z.); (Z.H.); (Y.G.); (X.L.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Xianyang 712100, China
| | - Yuanpeng Gao
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (Z.Z.); (Z.H.); (Y.G.); (X.L.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Xianyang 712100, China
- Correspondence: (Y.G.); (Y.Z.)
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (Z.Z.); (Z.H.); (Y.G.); (X.L.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Xianyang 712100, China
- Correspondence: (Y.G.); (Y.Z.)
| |
Collapse
|
3
|
Nobili S, Mannini A, Parenti A, Raggi C, Lapucci A, Chiorino G, Paccosi S, Di Gennaro P, Vezzosi V, Romagnoli P, Susini T, Coronnello M. Establishment and characterization of a new spontaneously immortalized ER -/PR -/HER2 + human breast cancer cell line, DHSF-BR16. Sci Rep 2021; 11:8340. [PMID: 33863935 PMCID: PMC8052418 DOI: 10.1038/s41598-021-87362-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/10/2021] [Indexed: 01/22/2023] Open
Abstract
Invasive ductal carcinoma (IDC) constitutes the most frequent malignant cancer endangering women’s health. In this study, a new spontaneously immortalized breast cancer cell line, DHSF-BR16 cells, was isolated from the primary IDC of a 74-years old female patient, treated with neoadjuvant chemotherapy and disease-free 5-years after adjuvant chemotherapy. Primary breast cancer tissue surgically removed was classified as ER−/PR−/HER2+, and the same phenotype was maintained by DHSF-BR16 cells. We examined DHSF-BR16 cell morphology and relevant biological and molecular markers, as well as their response to anticancer drugs commonly used for breast cancer treatment. MCF-7 cells were used for comparison purposes. The DHSF-BR16 cells showed the ability to form spheroids and migrate. Furthermore, DHSF-BR16 cells showed a mixed stemness phenotype (i.e. CD44+/CD24−/low), high levels of cytokeratin 7, moderate levels of cytokeratin 8 and 18, EpCAM and E-Cadh. Transcriptome analysis showed 2071 differentially expressed genes between DHSF-BR16 and MCF-7 cells (logFC > 2, p-adj < 0.01). Several genes were highly upregulated or downregulated in the new cell line (log2 scale fold change magnitude within − 9.6 to + 12.13). A spontaneous immortalization signature, mainly represented by extracellular exosomes-, plasma membrane- and endoplasmic reticulum membrane pathways (GO database) as well as by metabolic pathways (KEGG database) was observed in DHSF-BR16 cells. Also, these cells were more resistant to anthracyclines compared with MCF-7 cells. Overall, DHSF-BR16 cell line represents a relevant model useful to investigate cancer biology, to identify both novel prognostic and drug response predictive biomarkers as well as to assess new therapeutic strategies.
Collapse
Affiliation(s)
- Stefania Nobili
- Department of Health Science, Section of Clinical Pharmacology and Oncology, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy.,Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Antonella Mannini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Astrid Parenti
- Department of Health Science, Section of Clinical Pharmacology and Oncology, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy
| | - Chiara Raggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Andrea Lapucci
- Department of Health Science, Section of Clinical Pharmacology and Oncology, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy
| | | | - Sara Paccosi
- Department of Health Science, Section of Clinical Pharmacology and Oncology, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy
| | - Paola Di Gennaro
- Plastic and Reconstructive Surgery Unit - Regional Melanoma Referral Center - Tuscan Tumor Institute (ITT), Santa Maria Annunziata Hospital, Bagno a Ripoli, Florence, Italy
| | - Vania Vezzosi
- Department Organizational Structure (SOD) of Pathological Histology and Molecular Diagnostics, AOU Careggi, Florence, Italy
| | - Paolo Romagnoli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Tommaso Susini
- Department of Health Science, Section of Clinical Pharmacology and Oncology, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy
| | - Marcella Coronnello
- Department of Health Science, Section of Clinical Pharmacology and Oncology, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy.
| |
Collapse
|
4
|
Abbadie C, Pluquet O, Pourtier A. Epithelial cell senescence: an adaptive response to pre-carcinogenic stresses? Cell Mol Life Sci 2017; 74:4471-4509. [PMID: 28707011 PMCID: PMC11107641 DOI: 10.1007/s00018-017-2587-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 01/01/2023]
Abstract
Senescence is a cell state occurring in vitro and in vivo after successive replication cycles and/or upon exposition to various stressors. It is characterized by a strong cell cycle arrest associated with several molecular, metabolic and morphologic changes. The accumulation of senescent cells in tissues and organs with time plays a role in organismal aging and in several age-associated disorders and pathologies. Moreover, several therapeutic interventions are able to prematurely induce senescence. It is, therefore, tremendously important to characterize in-depth, the mechanisms by which senescence is induced, as well as the precise properties of senescent cells. For historical reasons, senescence is often studied with fibroblast models. Other cell types, however, much more relevant regarding the structure and function of vital organs and/or regarding pathologies, are regrettably often neglected. In this article, we will clarify what is known on senescence of epithelial cells and highlight what distinguishes it from, and what makes it like, replicative senescence of fibroblasts taken as a standard.
Collapse
Affiliation(s)
- Corinne Abbadie
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, 59000, Lille, France.
| | - Olivier Pluquet
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, 59000, Lille, France
| | - Albin Pourtier
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, 59000, Lille, France
| |
Collapse
|
5
|
Gleich A, Kaiser B, Schumann J, Fuhrmann H. Establishment and characterisation of a novel bovine SV40 large T-antigen-transduced foetal hepatocyte-derived cell line. In Vitro Cell Dev Biol Anim 2016; 52:662-72. [DOI: 10.1007/s11626-016-0018-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/16/2016] [Indexed: 12/12/2022]
|
6
|
Tang Z, Yu W, Zhang C, Zhao S, Yu Z, Xiao X, Tang R, Xuan Y, Yang W, Hao J, Xu T, Zhang Q, Huang W, Deng W, Guo W. CREB-binding protein regulates lung cancer growth by targeting MAPK and CPSF4 signaling pathway. Mol Oncol 2016; 10:317-29. [PMID: 26628108 PMCID: PMC5528962 DOI: 10.1016/j.molonc.2015.10.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/19/2015] [Indexed: 12/14/2022] Open
Abstract
CBP (CREB-binding protein) is a transcriptional co-activator which possesses HAT (histone acetyltransferases) activity and participates in many biological processes, including embryonic development, growth control and homeostasis. However, its roles and the underlying mechanisms in the regulation of carcinogenesis and tumor development remain largely unknown. Here we investigated the molecular mechanisms and potential targets of CBP involved in tumor growth and survival in lung cancer cells. Elevated expression of CBP was detected in lung cancer cells and tumor tissues compared to the normal lung cells and tissues. Knockdown of CBP by siRNA or inhibition of its HAT activity using specific chemical inhibitor effectively suppressed cell proliferation, migration and colony formation and induced apoptosis in lung cancer cells by inhibiting MAPK and activating cytochrome C/caspase-dependent signaling pathways. Co-immunoprecipitation and immunofluorescence analyses revealed the co-localization and interaction between CBP and CPSF4 (cleavage and polyadenylation specific factor 4) proteins in lung cancer cells. Knockdown of CPSF4 inhibited hTERT transcription and cell growth induced by CBP, and vice versa, demonstrating the synergetic effect of CBP and CPSF4 in the regulation of lung cancer cell growth and survival. Moreover, we found that high expression of both CBP and CPSF4 predicted a poor prognosis in the patients with lung adenocarcinomas. Collectively, our results indicate that CBP regulates lung cancer growth by targeting MAPK and CPSF4 signaling pathways.
Collapse
Affiliation(s)
- Zhipeng Tang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Changlin Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Shilei Zhao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhenlong Yu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiangsheng Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Ranran Tang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yang Xuan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Wenjing Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jiaojiao Hao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Tingting Xu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Qianyi Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Wenlin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China.
| | - Wei Guo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| |
Collapse
|
7
|
The human telomerase catalytic subunit and viral telomerase RNA reconstitute a functional telomerase complex in a cell-free system, but not in human cells. Cell Mol Biol Lett 2012; 17:598-615. [PMID: 22941205 PMCID: PMC6275662 DOI: 10.2478/s11658-012-0031-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 08/28/2012] [Indexed: 01/20/2023] Open
Abstract
The minimal vertebrate telomerase enzyme is composed of a protein component (telomerase reverse transcriptase, TERT) and an RNA component (telomerase RNA, TR). Expression of these two subunits is sufficient to reconstitute telomerase activity in vitro, while the formation of a holoenzyme comprising telomerase-associated proteins is necessary for proper telomere length maintenance. Previous reports demonstrated the high processivity of the human telomerase complex and the interspecies compatibility of human TERT (hTERT). In this study, we tested the function of the only known viral telomerase RNA subunit (vTR) in association with human telomerase, both in a cell-free system and in human cells. When vTR is assembled with hTERT in a cell-free environment, it is able to interact with hTERT and to reconstitute telomerase activity. However, in human cells, vTR does not reconstitute telomerase activity and could not be detected in the human telomerase complex, suggesting that vTR is not able to interact properly with the proteins constituting the human telomerase holoenzyme.
Collapse
|
8
|
The catalytic and the RNA subunits of human telomerase are required to immortalize equid primary fibroblasts. Chromosoma 2012; 121:475-88. [PMID: 22797876 PMCID: PMC3443485 DOI: 10.1007/s00412-012-0379-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Revised: 06/25/2012] [Accepted: 06/25/2012] [Indexed: 01/03/2023]
Abstract
Many human primary somatic cells can be immortalized by inducing telomerase activity through the exogenous expression of the human telomerase catalytic subunit (hTERT). This approach has been extended to the immortalization of cell lines from several mammals. Here, we show that hTERT expression is not sufficient to immortalize primary fibroblasts from three equid species, namely donkey, Burchelli’s zebra and Grevy’s zebra. In vitro analysis of a reconstituted telomerase composed by hTERT and an equid RNA component of telomerase (TERC) revealed a low activity of this enzyme compared to human telomerase, suggesting a low compatibility of equid and human telomerase subunits. This conclusion was also strengthened by comparison of human and equid TERC sequences, which revealed nucleotide differences in key regions for TERC and TERT interaction. We then succeeded in immortalizing equid fibroblasts by expressing hTERT and hTERC concomitantly. Expression of both human telomerase subunits led to telomerase activity and telomere elongation, indicating that human telomerase is compatible with the other equid telomerase subunits and proteins involved in telomere metabolism. The immortalization procedure described herein could be extended to primary cells from other mammals. The availability of immortal cells from endangered species could be particularly useful for obtaining new information on the organization and function of their genomes, which is relevant for their preservation.
Collapse
|
9
|
Babizhayev MA, Vishnyakova KS, Yegorov YE. Telomere-dependent senescent phenotype of lens epithelial cells as a biological marker of aging and cataractogenesis: the role of oxidative stress intensity and specific mechanism of phospholipid hydroperoxide toxicity in lens and aqueous. Fundam Clin Pharmacol 2011; 25:139-62. [DOI: 10.1111/j.1472-8206.2010.00829.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
10
|
Zhao JQ, Xie SS, Liu WB, Xiao YM, Zeng XM, Deng M, Gong L, Liu JP, Chen PC, Zhou J, Hu XH, Lv JH, Yu XQ, Wang D, Li C, Peng YL, Liao GP, Liu Y, Li DWC. Molecular Cloning of the Genes Encoding the PR55/Bβ/δ Regulatory Subunits for PP-2A and Analysis of Their Functions in Regulating Development of Goldfish, Carassius auratus. GENE REGULATION AND SYSTEMS BIOLOGY 2010; 4:135-48. [PMID: 21245947 PMCID: PMC3020040 DOI: 10.4137/grsb.s6065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The protein phosphatase-2A (PP-2A), one of the major phosphatases in eukaryotes, is a heterotrimer, consisting of a scaffold A subunit, a catalytic C subunit and a regulatory B subunit. Previous studies have shown that besides regulating specific PP-2A activity, various B subunits encoded by more than 16 different genes, may have other functions. To explore the possible roles of the regulatory subunits of PP-2A in vertebrate development, we have cloned the PR55/B family regulatory subunits: β and δ, analyzed their tissue specific and developmental expression patterns in Goldfish ( Carassius auratus). Our results revealed that the full-length cDNA for PR55/Bβ consists of 1940 bp with an open reading frame of 1332 nucleotides coding for a deduced protein of 443 amino acids. The full length PR55/Bδ cDNA is 2163 bp containing an open reading frame of 1347 nucleotides encoding a deduced protein of 448 amino acids. The two isoforms of PR55/B display high levels of sequence identity with their counterparts in other species. The PR55/Bβ mRNA and protein are detected in brain and heart. In contrast, the PR55/Bδ is expressed in all 9 tissues examined at both mRNA and protein levels. During development of goldfish, the mRNAs for PR55/Bβ and PR55/Bδ show distinct patterns. At the protein level, PR55/Bδ is expressed at all developmental stages examined, suggesting its important role in regulating goldfish development. Expression of the PR55/Bδ anti-sense RNA leads to significant downregulation of PR55/Bδ proteins and caused severe abnormality in goldfish trunk and eye development. Together, our results suggested that PR55/Bδ plays an important role in governing normal trunk and eye formation during goldfish development.
Collapse
Affiliation(s)
- Jun-Qiong Zhao
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Si-Si Xie
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Wen-Bin Liu
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Ya-Mei Xiao
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Xiao-Ming Zeng
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Mi Deng
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
| | - Lili Gong
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
| | - Jin-Ping Liu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
| | - Pei-Chao Chen
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Jie Zhou
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Xiao-Hui Hu
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Jia-Han Lv
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Xiang-Qian Yu
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Dao Wang
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Chi Li
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Yun-Lei Peng
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Gao-Peng Liao
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Yun Liu
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - David Wan-Cheng Li
- Key Laboratory of Protein Chemistry and Developmental Biology of Educational Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
| |
Collapse
|
11
|
Cassar L, Li H, Jiang FX, Liu JP. TGF-beta induces telomerase-dependent pancreatic tumor cell cycle arrest. Mol Cell Endocrinol 2010; 320:97-105. [PMID: 20138964 DOI: 10.1016/j.mce.2010.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 01/27/2010] [Accepted: 02/02/2010] [Indexed: 12/21/2022]
Abstract
Recent studies suggest that transforming growth factor beta (TGF-beta) inhibits telomerase activity by repression of the telomerase reverse transcriptase (TERT) gene. In this report, we show that TGF-beta induces TERT repression-dependent apoptosis in pancreatic tumor, vascular smooth muscle, and cervical cancer cell cultures. TGF-beta activates Smad3 signaling, induces TERT gene repression and results in G1/S phase cell cycle arrest and apoptosis. TERT over-expression stimulates the G1/S phase transition and alienates TGF-beta-induced cell cycle arrest and apoptosis. Our data suggest that telomere maintenance is a limiting factor of the transition of the cell cycle. TGF-beta triggers cell cycle arrest and death by a mechanism involving telomerase deregulation of telomere maintenance.
Collapse
Affiliation(s)
- Lucy Cassar
- Department of Immunology, Monash University, Central Clinical School, AMREP, Commercial Road, Melbourne, Victoria 3004, Australia.
| | | | | | | |
Collapse
|
12
|
Neelima PS, Rao Rekha M, Rama S, Rao AJ. Effect of human telomerase reverse transcriptase transfection on differentiation in BeWo choriocarcinoma cells. Reprod Biomed Online 2009; 18:838-49. [PMID: 19490790 DOI: 10.1016/s1472-6483(10)60035-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Arrest of proliferation is one of the prerequisites for differentiation of cytotrophoblasts into syncytiotrophoblasts, and thus during differentiation telomerase activity, as well as human telomerase reverse transcriptase (hTERT) expression, is down-regulated. Considering this, it is of interest to investigate whether syncytium formation can be delayed by prolonging the expression of telomerase in cytotrophoblasts. BeWo cells were transfected with pLPC-hTERT retroviral vector and the reverse transcription-polymerase chain reaction analysis for hTERT mRNA concentrations in the transfected cells revealed a several-fold increase in hTERT mRNA compared with the cells transfected with empty vector, and this confirmed that the transfection was successful. An increase in the proliferation, as assessed by bromodeoxyuridine incorporation assay, as well as an increase in mRNA and protein concentration of various cyclins and proliferating cell nuclear antigen, was noticed. The effect of hTERT transfection was also assessed after the addition of forskolin to induce differentiation and it was observed that cell-cell fusion was delayed and differentiation did not occur in hTERT-transfected cells. However, the effects seen were only transient as stable transfection was not possible and the cells were undergoing apoptosis after 72 h, which suggested that apart from hTERT other factors might be important for immortalization of BeWo cells.
Collapse
Affiliation(s)
- P S Neelima
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | | | | | |
Collapse
|
13
|
Deville L, Hillion J, Ségal-Bendirdjian E. Telomerase regulation in hematological cancers: a matter of stemness? Biochim Biophys Acta Mol Basis Dis 2009; 1792:229-39. [PMID: 19419697 DOI: 10.1016/j.bbadis.2009.01.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 01/30/2009] [Accepted: 01/30/2009] [Indexed: 01/02/2023]
Abstract
Human telomerase is a nuclear ribonucleoprotein enzyme complex that catalyzes the synthesis and extension of telomeric DNA. This enzyme is highly expressed and active in most malignant tumors while it is usually not or transiently detectable in normal somatic cells, suggesting that it plays an important role in cellular immortalization and tumorigenesis. As most leukemic cells are generally telomerase-positive and have often shortened telomeres, our understanding of how telomerase is deregulated in these diseases could help to define novel therapies targeting the telomere/telomerase complex. Nonetheless, considering that normal hematopoietic stem cells and some of their progeny do express a functional telomerase, it is tempting to consider such an activity in leukemias as a sustained stemness feature and important to understand how telomere length and telomerase activity are regulated in the various forms of leukemias.
Collapse
Affiliation(s)
- Laure Deville
- INSERM UMR-S 685, Institut d'Hématologie, Hôpital Saint-Louis, 75475 Paris cedex 10, France
| | | | | |
Collapse
|
14
|
Gossypol suppresses telomerase activity in human leukemia cells via regulating hTERT. FEBS Lett 2008; 582:3367-73. [DOI: 10.1016/j.febslet.2008.08.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Revised: 08/15/2008] [Accepted: 08/25/2008] [Indexed: 11/23/2022]
|
15
|
Li H, Liu JP. Mechanisms of action of TGF-beta in cancer: evidence for Smad3 as a repressor of the hTERT gene. Ann N Y Acad Sci 2007; 1114:56-68. [PMID: 17934056 DOI: 10.1196/annals.1396.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Transforming growth factor-beta (TGF-beta) induces cell differentiation and suppresses cell proliferation, but the mechanisms underlying the actions of TGF-beta remain to be fully elucidated. Recent studies suggest that TGF-beta suppresses neoplastic cell development by employing Smad3 protein to repress the gene of human telomerase reverse transcriptase (hTERT). In human breast cancer cells, TGF-beta induces rapid phosphorylation and subsequent entry of Smad3 into the nucleus. In the nucleus, Smad3 binds to the hTERT gene promoter directly and inhibits hTERT gene transcription activity. By interacting with c-myc, Smad3 also represses the c-myc gene. Thus, TGF-beta prevents continuous cell proliferation by switching off telomerase activity through Smad3 repression of the hTERT gene and the action of c-myc. Modulating the interface between Smad3 and the hTERT gene, and the potential feedback loop from telomeres to Smad3 via Smurf2, may represent a novel approach to regulate cell lifespan of proliferation.
Collapse
Affiliation(s)
- He Li
- Department of Immunology, AMREP, Monash Medical School, Commercial Road, Prahran, Victoria 3181, Australia.
| | | |
Collapse
|
16
|
Nicholson IP, Gault EA, Foote CG, Nasir L, Bennett D. Human telomerase reverse transcriptase (hTERT) extends the lifespan of canine chondrocytes in vitro without inducing neoplastic transformation. Vet J 2007; 174:570-6. [PMID: 17827040 DOI: 10.1016/j.tvjl.2007.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Revised: 07/05/2007] [Accepted: 07/06/2007] [Indexed: 11/17/2022]
Abstract
To determine if the exogenous expression of the human telomerase reverse transcriptase (hTERT) protein can extend the in vitro lifespan of chondrocytes from normal and osteoarthritic canine donors, articular chondrocytes were harvested and expanded initially in monolayer culture. Cells were transfected with pCIneo or pCIneo-hTERT and selected using G418. Transfectants were cultured either in monolayer or alginate beads and telomerase activity, replicative lifespan and the tumourogenic potential of the transfected cells were assessed. hTERT expression in canine chondrocytes prolonged the replicative lifespan of these cells but did not permit growth in low serum conditions or promote the formation of foci in anchorage independence assays. In addition, hTERT expression resulted in the down-regulation of MMP-1. This suggests that hTERT may represent a tool for the generation of tissue engineered chondrocytes suitable for autologous re-implantation into the affected areas of osteoarthritic joints.
Collapse
Affiliation(s)
- Iain P Nicholson
- Institute of Comparative Medicine, Faculty of Veterinary Medicine, University of Glasgow, Garscube Estate, Bearsden Road, Glasgow, UK G61 1QH, UK
| | | | | | | | | |
Collapse
|
17
|
Liu WR, Lu L, Rosenberg DS, Procaccini PSA, Mustoe TA. Synergistic activation of extracellular signal-regulated kinase in human dermal fibroblasts by human telomerase reverse transcriptase and transforming growth factor-beta1. J Surg Res 2007; 143:415-21. [PMID: 17662305 DOI: 10.1016/j.jss.2007.02.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Revised: 02/08/2007] [Accepted: 02/20/2007] [Indexed: 01/04/2023]
Abstract
BACKGROUND Human telomerase reverse transcriptase (hTERT) is primarily known for its ability to elongate telomeres for maintaining chromosomal integrity and delaying cellular senescence. Recently, hTERT has emerged as having a role in promoting cellular proliferation that is independent of telomere elongation. How hTERT elicits this novel function is a fundamental question in cell biology. Understanding this question may have therapeutic implications in regenerative medicine for patients with damaged organs or tissues, cardiovascular disorders, stroke, ischemic chronic wounds, and other ischemia-reperfusion injuries. Toward this end, we treated hTERT-transfected human dermal fibroblasts (HDFs) with transforming growth factor (TGF)-beta1 and investigated the activation of extracellular signal-regulated kinase (ERK) 1/2, vital mediators of cell proliferation. MATERIALS AND METHODS Primary HDFs were transfected with either recombinant adenovirus expressing hTERT (Ad-hTERT) or control adenovirus (Ad-NULL) and subsequently treated with TGF-beta1 (2 pg/mL). ERK 1/2 activation was determined by Western blotting using an antibody recognizing only activated ERK 1/2 that is dually phosphorylated at Thr(202) and Tyr(204). TGF-beta1, TGFbeta-RI, TGFbeta-RII, and Col1 A1 mRNA levels were analyzed by real-time PCR. RESULTS Ad-hTERT-transfected HDFs showed more than 7-fold up-regulation of phospho-ERK 1/2 over Ad-NULL-transfected HDFs upon TGF-beta1 treatment. The synergistic ERK 1/2 activation in Ad-hTERT-transfected HDFs occurred as early as 10 min and was sustained for at least 30 min after TGF-beta1 treatment. There were no statistically significant differences in TGF-beta1, TGFbeta-RI, TGFbeta-RII, and Col1 A1 mRNA levels between HDFs that were transfected with Ad-hTERT and those that were transfected with Ad-NULL after TGF-beta1 treatment. CONCLUSIONS hTERT and extremely low concentrations of TGF-beta1 (2 pg/mL) synergistically activate ERK 1/2 in HDFs by a mechanism that is independent of the autocrine TGF-beta1 loop.
Collapse
Affiliation(s)
- W Robert Liu
- Wound Healing Research Laboratory, Division of Plastic and Reconstructive Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | |
Collapse
|
18
|
Yan Q, Liu WB, Qin J, Liu J, Chen HG, Huang X, Chen L, Sun S, Deng M, Gong L, Li Y, Zhang L, Liu Y, Feng H, Xiao Y, Liu Y, Li DWC. Protein Phosphatase-1 Modulates the Function of Pax-6, a Transcription Factor Controlling Brain and Eye Development. J Biol Chem 2007; 282:13954-65. [PMID: 17374606 DOI: 10.1074/jbc.m611476200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Pax-6 is an evolutionarily conserved transcription factor and acts high up in the regulatory hierarchy controlling eye and brain development in humans, mice, zebrafish, and Drosophila. Previous studies have shown that Pax-6 is a phosphoprotein, and its phosphorylation by ERK, p38, and homeodomain-interacting protein kinase 2 greatly enhances its transactivation activity. However, the protein phosphatases responsible for the dephosphorylation of Pax-6 remain unknown. Here, we present both in vitro and in vivo evidence to show that protein serine/threonine phosphatase-1 is a major phosphatase that directly dephosphorylates Pax-6. First, purified protein phosphatase-1 directly dephosphorylates Pax-6 in vitro. Second, immunoprecipitation-linked Western blot revealed that both protein phosphatase-1alpha and protein phosphatase-1beta interact with Pax-6. Third, overexpression of protein phosphatase-1 in human lens epithelial cells leads to dephosphorylation of Pax-6. Finally, inhibition of protein phosphatase-1 activity by calyculin A or knockdown of protein phosphatase-1alpha and protein phosphatase-1beta by RNA interference leads to enhanced phosphorylation of Pax-6. Moreover, our results also demonstrate that dephosphorylation of Pax-6 by protein phosphatase-1 significantly modulates its function in regulating expression of both exogenous and endogenous genes. These results demonstrate that protein phosphatase 1 acts as a major phosphatase to dephosphorylate Pax-6 and modulate its function.
Collapse
Affiliation(s)
- Qin Yan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Dairkee SH, Nicolau M, Sayeed A, Champion S, Ji Y, Moore DH, Yong B, Meng Z, Jeffrey SS. Oxidative stress pathways highlighted in tumor cell immortalization: association with breast cancer outcome. Oncogene 2007; 26:6269-79. [PMID: 17471242 DOI: 10.1038/sj.onc.1210452] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
An improved understanding of cell immortalization and its manifestation in clinical tumors could facilitate novel therapeutic approaches. However, only rare tumor cells, which maintain telomerase expression in vitro, immortalize spontaneously. By expression-profiling analyses of limited-life primary breast tumor cultures pre- and post-hTERT transduction, and spontaneously immortalized breast cancer cell lines, we identified a common signature characteristic of tumor cell immortalization. A predominant feature of this immortalization signature (ImmSig) was the significant overexpression of oxidoreductase genes. In contrast to epithelial cells derived from low histologic grade primary tumors, which required hTERT transduction for the acquisition of ImmSig, spontaneously immortalizing high-grade tumor cultures displayed similar molecular changes independent of exogenous hTERT. Silencing the hTERT gene reversed ImmSig expression, increased cellular reactive oxygen species levels, altered mitochondrial membrane potential and induced apoptotic and proliferation changes in immortalized cells. In clinical breast cancer samples, cell-proliferation-pathway genes were significantly associated with ImmSig. In these cases, ImmSig expression itself was inversely correlated with patient survival (P=0), and was particularly relevant to the outcome of estrogen receptor-positive tumors. Our data support the notion that ImmSig assists in surmounting normal barriers related to oxidative and replicative stress response. Targeting a subset of aggressive breast cancers by reversing ImmSig components could be a practical therapeutic strategy.
Collapse
Affiliation(s)
- S H Dairkee
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Constitutive activation of the mitogen-activated protein (MAP) kinase signaling pathway by oncogenic stimulation is widespread in human cancers. With the recently demonstrated links between MAP kinase, histone phosphorylation, gene transcription factors, and hTERT gene promoter activity, abnormal MAP kinase activity is likely to be one of the essential forces that impact on hTERT gene transcription in transformed human cells. Several proteins have been implicated as playing important roles in MAP kinase signaling to hTERT gene, including Ets and activator protein-1 (AP-1). Inhibition of these signaling mechanisms may have a consequential effect on hTERT gene expression and telomerase activity. In this study, we brief the current progress and strategy in molecular targeting to the interface between MAP kinase and hTERT gene promoter in cancer.
Collapse
Affiliation(s)
- Dakang Xu
- Department of Immunology, Monash Medical School, Melbourne, Australia
| | | | | |
Collapse
|
21
|
Abstract
Transforming growth factor beta (TGF-beta) carries out tumor suppressor activity in epithelial and lymphoid cells, whereas telomerase is required for most cancers. Although the molecular mechanisms by which TGF-beta acts as a tumor suppressor are yet to be fully established, a link between TGFb and its tumor suppressor activity by telomerase has been suggested. Recently, we have noted a novel mode of action for TGF-beta through which human telomerase reverse transcriptase (hTERT) gene is repressed in immortal and neoplastic cells, confirming that one of the mechanisms underlying TGF-beta suppression of tumor growth may be through inhibiting hTERT gene transcription. Moreover, the inhibition of hTERT gene by TGF-beta suggests a cis action of the TGF-beta signaling molecule Smad3 on hTERT promoter directly. This article examines our current understanding and investigation of TGF-beta regulation of telomerase activity, and presents a model in which Smad3 participates in regulating hTERT gene transcription by acting as a repressor directly. Engineering the interface between Smad3 and hTERT gene may lead to a new strategy to inhibit telomerase activity in cancer.
Collapse
Affiliation(s)
- He Li
- Department of Immunology, Molecular Signaling Laboratory, Monash University, Melbourne, Australia.
| | | | | | | |
Collapse
|
22
|
Li H, Xu D, Li J, Berndt MC, Liu JP. Transforming growth factor beta suppresses human telomerase reverse transcriptase (hTERT) by Smad3 interactions with c-Myc and the hTERT gene. J Biol Chem 2006; 281:25588-600. [PMID: 16785237 DOI: 10.1074/jbc.m602381200] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Telomerase underpins stem cell renewal and proliferation and is required for most neoplasia. Recent studies suggest that hormones and growth factors play physiological roles in regulating telomerase activity. In this report we show a rapid repression of the telomerase reverse transcriptase (TERT) gene by transforming growth factor beta (TGF-beta) in normal and neoplastic cells by a mechanism depending on the intracellular signaling protein Smad3. In human breast cancer cells TGF-beta induces rapid entry of Smad3 into the nucleus where it binds to the TERT gene promoter and represses TERT gene transcription. Silencing Smad3 gene expression or genetically deleting the Smad3 gene disrupts TGF-beta repression of TERT gene expression. Expression of the Smad3 antagonist, Smad7, also interrupts TGF-beta-mediated Smad3-induced repression of the TERT gene. Mutational analysis identified the Smad3 site on the TERT gene promoter, mediating TERT repression. In response to TGF-beta, Smad3 binds to c-Myc; knocking down c-Myc, Smad3 does not bind to the TERT gene, suggesting that c-Myc recruits Smad3 to the TERT promoter. Thus, TGF-beta negatively regulates telomerase activity via Smad3 interactions with c-Myc and the TERT gene promoter. Modifying the interaction between Smad3 and TERT gene may, thus, lead to novel strategies to regulate telomerase.
Collapse
Affiliation(s)
- He Li
- Department of Immunology and Monash Institute of Medical Research, Monash University, Melbourne, Victoria 3181, Australia
| | | | | | | | | |
Collapse
|
23
|
Geserick C, Tejera A, González-Suárez E, Klatt P, Blasco MA. Expression of mTert in primary murine cells links the growth-promoting effects of telomerase to transforming growth factor-β signaling. Oncogene 2006; 25:4310-9. [PMID: 16501597 DOI: 10.1038/sj.onc.1209465] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Here, we show that ectopic expression of the catalytic subunit of mouse telomerase (mTert) confers a growth advantage to primary murine embryonic fibroblasts (MEFs), which have very long telomeres, as well as facilitates their spontaneous immortalization and increases their colony-forming capacity upon activation of oncogenes. We demonstrate that these telomere length-independent growth-promoting effects of mTert overexpression require catalytically active mTert, as well as the formation of mTert/Terc complexes. The gene expression profile of mTert-overexpressing MEFs indicates that telomerase enhances growth in these cells through the repression of growth-inhibiting genes of the transforming growth factor-beta (TGF-beta) signaling network. We functionally validate this result by showing that mTert abrogates the growth-inhibitory effect of TGF-beta in MEFs, thus demonstrating that telomerase increments the proliferative potential of primary mouse embryonic fibroblasts by targeting the TGF-beta pathway.
Collapse
Affiliation(s)
- C Geserick
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Center (CNIO), Melchor Fernández Almagro, Madrid, Spain
| | | | | | | | | |
Collapse
|
24
|
Li DWC, Liu JP, Mao YW, Xiang H, Wang J, Ma WY, Dong Z, Pike HM, Brown RE, Reed JC. Calcium-activated RAF/MEK/ERK signaling pathway mediates p53-dependent apoptosis and is abrogated by alpha B-crystallin through inhibition of RAS activation. Mol Biol Cell 2005; 16:4437-53. [PMID: 16000378 PMCID: PMC1196350 DOI: 10.1091/mbc.e05-01-0010] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Revised: 05/16/2005] [Accepted: 06/27/2005] [Indexed: 12/20/2022] Open
Abstract
The ocular lens is the only organ that does not develop spontaneous tumor. The molecular mechanism for this phenomenon remains unknown. Through examination of the signaling pathways mediating stress-induced apoptosis, here we presented evidence to show that different from most other tissues in which the extracellular signal-regulated kinases (ERKs) pathway is generally implicated in mediation of survival signals activated by different factors, the RAF/MEK/ERK signaling pathway alone plays a key role in stress-activated apoptosis of lens epithelial cells. Treatment of N/N1003A cells with calcimycin, a calcium mobilizer, activates the RAF/MEK/ERK pathway through RAS, which is indispensable for the induced apoptosis because inhibition of this pathway by either pharmacological drug or dominant negative mutants greatly attenuates the induced apoptosis. Calcimycin also activates p38 kinase and JNK2, which are not involved in calcium-induced apoptosis. Downstream of ERK activation, p53 is essential. Activation of RAF/MEK/ERK pathway by calcimycin leads to distinct up-regulation of p53. Moreover, overexpression of p53 enhances calcimycin-induced apoptosis, whereas inhibition of p53 expression attenuates calcimycin-induced apoptosis. Up-regulation of p53 directly promotes Bax expression, which changes the integrity of mitochondria, leading to release of cytochrome c, activation of caspase-3 and eventually execution of apoptosis. Overexpression of alphaB-crystallin, a member of the small heat-shock protein family, blocks activation of RAS to inhibit ERK1/2 activation, and greatly attenuates calcimycin-induced apoptosis. Together, our results provide 1) a partial explanation for the lack of spontaneous tumor in the lens, 2) a novel signaling pathway for calcium-induced apoptosis, and 3) a novel antiapoptotic mechanism for alphaB-crystallin.
Collapse
|