1
|
Murillo-Ortiz BO, García-Corrales K, Martínez-Garza S, Romero-Vázquez MJ, Agustín-Godínez E, Escareño-Gómez A, Silva-Guerrero DG, Mendoza-Ramírez S, Murguia-Perez M. Association of hTERT expression, Her2Neu, estrogen receptors, progesterone receptors, with telomere length before and at the end of treatment in breast cancer patients. Front Med (Lausanne) 2024; 11:1450147. [PMID: 39188883 PMCID: PMC11345256 DOI: 10.3389/fmed.2024.1450147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
Background Breast cancer shows significant clinical, morphologic, and molecular variation. Telomeres are nucleoprotein complexes composed of hexanucleotide repeat DNA sequence, TTAGGG, and numerous telomere-associated proteins. The maintenance of telomere length is carried out by a ribonucleoprotein called telomerase, which consists of two main components: a catalytic subunit called hTERT (human telomerase reverse transcriptase) and an RNA template called hTR (human telomerase RNA). The importance of evaluating hTERT expression lies in its potential therapeutic application, being an attractive target due to its almost non-existent expression in normal somatic cells. It is also expected that the anti-neoplastic effect would appear earlier in neoplastic cells with shorter telomeres. Additionally, a significant relationship has been observed between Her2-Neu overexpression and Her2-Neu positivity, which could suggest new combined therapies.The aim of this study was to detect the expression of hTERT, estrogen receptor (ER), progesterone receptor (PR), and HER2-Neu in neoplastic breast tissue embedded in paraffin before treatment and to investigate the relationship between them and with baseline and post-treatment telomere length, as well as with various clinicopathological parameters. Materials and methods A cross-sectional-correlational, 21 women diagnosed with breast cancer at the Oncology Service of the High Specialty Medical Unit No. 1 of Bajio of the Mexican Institute of Social Security. The study complies with the Helsinki Declaration and was approved by the Institutional Ethical Committee of the Mexican Institute of Social Security (R-2019-1001-127). A peripheral blood sample was obtained before oncological treatment and at the end of oncological treatment for the measurement of telomere length by extracting DNA from leukocytes, was performed by the quantitative polymerase chain reaction (PCR) method described by Cawthon. Tumor samples were collected from each patient at the oncology department for immunohistochemical determination of biomarker expression (ER, PR, Her2/neu) and hTERT. Results Of the 21 cases included in the study, the median age was 57.57 years. Eighteen cases were classified as invasive ductal carcinoma NOS (85.71%), 10 were histologic grade 2 (47.61%), 16 cases were hormone receptor positive (76.19%), 7 were Her2Neu positive (33.33%), and only 2 cases were triple negative (9.52%). Positive hTERT expression was detected in 11 cases (52.38%) and was negative in the remaining cases. A significant association was identified between hTERT-positive cases and Her2-Neu positive cases (p = 0.04). Baseline and post-treatment telomere lengths showed a significant difference using the non-parametric Wilcoxon t-test (p = 0.002). In hTERT-positive cases, there was significant telomere shortening at the end of oncological treatment (6.14 ± 1.54 vs. 4.75 ± 1.96 Kb, p = 0.007). Conclusion Positive hTERT immunostaining cases were associated with poor prognostic factors, such as Her2-Neu overexpression and post-treatment telomere shortening. In the future, hTERT immunostaining could be used to select patients for therapies with antagonistic effects on hTERT, as well as in the selection of more appropriate chemotherapy regimens for patients who express it.
Collapse
Affiliation(s)
- Blanca Olivia Murillo-Ortiz
- Unidad de Investigación en Epidemiología Clínica, OOAD Guanajuato, Instituto Mexicano del Seguro Social, León, Mexico
| | - Kenia García-Corrales
- Servicio de Anatomía Patológica, Hospital General de Zona No. 33, Instituto Mexicano del Seguro Social, Bahía de Banderas, Mexico
| | - Sandra Martínez-Garza
- Unidad de Investigación en Epidemiología Clínica, OOAD Guanajuato, Instituto Mexicano del Seguro Social, León, Mexico
| | - Marcos Javier Romero-Vázquez
- Unidad de Investigación en Epidemiología Clínica, OOAD Guanajuato, Instituto Mexicano del Seguro Social, León, Mexico
| | - Eduardo Agustín-Godínez
- Laboratorio de Anatomía Patológica e Inmunohistoquímica Especializada DIME, Hospital Médica Campestre, León, Mexico
| | - Andrea Escareño-Gómez
- Departamento de Patología Quirúrgica, UMAE Hospital de Especialidades No. 1, Centro Médico Nacional Bajío, Instituto Mexicano del Seguro Social, León, Mexico
| | | | | | - Mario Murguia-Perez
- Laboratorio de Anatomía Patológica e Inmunohistoquímica Especializada DIME, Hospital Médica Campestre, León, Mexico
- Departamento de Patología Quirúrgica, UMAE Hospital de Especialidades No. 1, Centro Médico Nacional Bajío, Instituto Mexicano del Seguro Social, León, Mexico
| |
Collapse
|
2
|
Fawzy RM, Abdel-Aziz AA, Bassiouny K, Fayed AM. Phytocompounds-based therapeutic approach: Investigating curcumin and green tea extracts on MCF-7 breast cancer cell line. J Genet Eng Biotechnol 2024; 22:100339. [PMID: 38494270 PMCID: PMC10980874 DOI: 10.1016/j.jgeb.2023.100339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
BACKGROUND Breast cancer (BC) has transcended lung cancer as the most common cancer in the world. Due to the disease's aggressiveness, rapid growth, and heterogeneity, it is crucial to investigate different therapeutic approaches for treatment. According to the World Health Organization (WHO), Plant-based therapeutics continue to be utilized as safe/non-toxic complementary or alternative treatments for cancer, even in developed countries, regardless of how cutting-edge conventional therapies are. Despite their low bioavailability, curcumin (CUR) and green tea (GT) represent safer therapeutic options. Due to their potent molecular-modulating properties on various cancer-related molecules and signaling pathways, they are considered gold-standard therapeutic agents and have been incorporated into the development of one or more therapeutic strategies of BC treatment. METHODS We investigated the modulatory role of CUR and GT extracts on significant multi molecular targets in MCF-7 BC cell line to assess their potential as BC multi-targeting agents. We analyzed the phytocompounds in GT leaves using High-performance liquid chromatography (HPLC) and Gas chromatography-mass spectrometry (GC-MS) techniques. The mRNA expression levels of Raf-1, Telomerase, Tumor necrosis factor alpha (TNF-α) and Interleukin-8 (IL-8) genes in MCF-7 cells were quantified using quantitative real-time PCR (qRT-PCR). The cytotoxicity of the extracts was assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and the released Lactate dehydrogenase (LDH), a valuable marker for identifying the programmed necrosis (necroptosis). Additionally, the concentrations of the necroptosis-related proinflammatory cytokines (TNF-α and IL-8) were measured using enzyme-linked immunosorbent assay (ELISA). RESULTS In contrast to the GT, the results showed the anticancer and cytotoxic properties of CUR against MCF-7 cells, with a relatively higher level of released LDH. The CUR extract downregulated the oncogenic Raf-1, suppressed the Telomerase and upregulated the TNF-α and IL-8 genes. Results from the ELISA showed a notable increase in IL-8 and TNF-α cytokines levels after CUR treatment, which culminated after 72 h. CONCLUSIONS Among both extracts, only CUR effectively modulated the understudy molecular targets, achieving multi-targeting anticancer activity against MCF-7 cells. Moreover, the applied dosage significantly increased levels of the proinflammatory cytokines, which represent a component of the cytokines-targeting-based therapeutic strategy. However, further investigations are recommended to validate this therapeutic approach.
Collapse
Affiliation(s)
- Radwa M Fawzy
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt.
| | - Amal A Abdel-Aziz
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Khalid Bassiouny
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Aysam M Fayed
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| |
Collapse
|
3
|
Kim TD, Gu R, Janknecht R. Methylation of the JMJD2B epigenetic regulator differentially affects its ability to coactivate the ETV1 and JUN transcription factors. INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 14:101-115. [PMID: 38213775 PMCID: PMC10776875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVES Jumonji C domain-containing (JMJD) 2B (JMJD2B) is a transcriptional cofactor and histone demethylase that is involved in prostate cancer formation. However, how its function is regulated by posttranslational modification has remained elusive. Hence, we examined if JMJD2B would be regulated by lysine methylation. METHODS Through in vitro methylation assays and Western blotting with methyl-lysine specific antibodies, we analyzed lysine methylation within JMJD2B. Identified methylated lysine residues were mutated to arginine residues and the respective impact on JMJD2B transcriptional activity measured with a reporter gene assay in human LNCaP prostate cancer cells. RESULTS We discovered that JMJD2B is methylated on up to six different lysine residues. Further, we identified the suppressor of variegation 3-9/enhancer of zeste/trithorax (SET) domain-containing protein 7/9 (SET7/9) as the methyltransferase being responsible for this posttranslational modification. Mutating the methylation sites in JMJD2B to arginine residues led to diminished coactivation of the Ju-nana (JUN) transcription factor, which is a known oncogenic protein in prostate tumors. In contrast, methylation of JMJD2B had no impact on its ability to coactivate another transcription factor associated with prostate cancer, the DNA-binding protein E26 transformation-specific (ETS) variant 1 (ETV1). Consistent with a potential joint action of JMJD2B, SET7/9 and JUN in prostate cancer, the expression of JMJD2B in human prostate tumors was positively correlated with both SET7/9 and JUN levels. CONCLUSIONS The identified SET7/9-mediated methylation of JMJD2B appears to impact its cooperation with selected interacting transcription factors in prostate cancer cells. Given the implicated roles of JMJD2B beyond prostate tumorigenesis, SET7/9-mediated methylation of JMJD2B possibly also influences the development of other cancers, while its impairment might have relevance for obesity or a global developmental delay that can be elicited by reduced JMJD2B activity.
Collapse
Affiliation(s)
- Tae-Dong Kim
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
| | - Ruicai Gu
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
- Department of Pathology, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
| |
Collapse
|
4
|
Gu R, Kim TD, Jiang H, Shin S, Oh S, Janknecht R. Methylation of the epigenetic JMJD2D protein by SET7/9 promotes prostate tumorigenesis. Front Oncol 2023; 13:1295613. [PMID: 38045004 PMCID: PMC10690936 DOI: 10.3389/fonc.2023.1295613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023] Open
Abstract
How the function of the JMJD2D epigenetic regulator is regulated or whether it plays a role in prostate cancer has remained elusive. We found that JMJD2D was overexpressed in prostate tumors, stimulated prostate cancer cell growth and became methylated by SET7/9 on K427. Mutation of this lysine residue in JMJD2D reduced the ability of DU145 prostate cancer cells to grow, invade and form tumors and elicited extensive transcriptomic changes. This included downregulation of CBLC, a ubiquitin ligase gene with hitherto unknown functions in prostate cancer, and upregulation of PLAGL1, a transcription factor with reported tumor suppressive characteristics in the prostate. Bioinformatic analyses indicated that CBLC expression was elevated in prostate tumors. Further, downregulation of CBLC largely phenocopied the effects of the K427 mutation on DU145 cells. In sum, these data have unveiled a novel mode of regulation of JMJD2D through lysine methylation, illustrated how this can affect oncogenic properties by influencing expression of the CBLC gene, and established a pro-tumorigenic role for CBLC in the prostate. A corollary is that JMJD2D and CBLC inhibitors could have therapeutic benefits in the treatment of prostate and possibly other cancers.
Collapse
Affiliation(s)
- Ruicai Gu
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Tae-Dong Kim
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Hanlin Jiang
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Sook Shin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, Oklahoma City, OK, United States
| | - Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, Oklahoma City, OK, United States
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, Oklahoma City, OK, United States
| |
Collapse
|
5
|
Galogre M, Rodin D, Pyatnitskiy M, Mackelprang M, Koman I. "A Review of HER2 overexpression and somatic mutations in cancers". Crit Rev Oncol Hematol 2023; 186:103997. [PMID: 37062337 DOI: 10.1016/j.critrevonc.2023.103997] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/14/2023] [Accepted: 04/13/2023] [Indexed: 04/18/2023] Open
Abstract
The Human Epidermal Growth Factor Receptor (HER) proteins family, which includes HER2, are membrane-bound receptors that activate many intracellular pathways associated with growth and development. When there are mutations in HER2, or when it becomes overexpressed, it can cause oncogenesis and offer differential prognosis and treatment across almost all cancer types. Both mutations in HER2 and its overexpression have distinct mechanisms by which they can cause these effects in cancers. This review outlines how HER2's normal pathway is altered in both overexpression and mutation and compiles all the well-known mechanisms by which HER2 can cause oncogenesis. Finally, this review briefly outlines how HER2 mutants and HER2 overexpression is detected, and how their detection can lead to different prognosis and treatment in cancers.
Collapse
Affiliation(s)
| | - Dmitry Rodin
- Institute of Personalised and Translational Medicine, Ariel University, Ariel, Israel Kiryat Hamada
| | - Mikhail Pyatnitskiy
- Institute of Biomedical Chemistry RAMS, Solianka st.,14, 109544, Moscow, Russia
| | | | - Igor Koman
- SmartOmica, Tērbatas iela 36 - 4, Latvia Rīga, LV-1011; Institute of Personalised and Translational Medicine, Ariel University, Ariel, Israel Kiryat Hamada
| |
Collapse
|
6
|
Sui Y, Jiang H, Kellogg CM, Oh S, Janknecht R. Promotion of colorectal cancer by transcription factor BHLHE40 involves upregulation of ADAM19 and KLF7. Front Oncol 2023; 13:1122238. [PMID: 36890812 PMCID: PMC9986587 DOI: 10.3389/fonc.2023.1122238] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
BHLHE40 is a transcription factor, whose role in colorectal cancer has remained elusive. We demonstrate that the BHLHE40 gene is upregulated in colorectal tumors. Transcription of BHLHE40 was jointly stimulated by the DNA-binding ETV1 protein and two associated histone demethylases, JMJD1A/KDM3A and JMJD2A/KDM4A, which were shown to also form complexes on their own and whose enzymatic activity was required for BHLHE40 upregulation. Chromatin immunoprecipitation assays revealed that ETV1, JMJD1A and JMJD2A interacted with several regions within the BHLHE40 gene promoter, suggesting that these three factors directly control BHLHE40 transcription. BHLHE40 downregulation suppressed both growth and clonogenic activity of human HCT116 colorectal cancer cells, strongly hinting at a pro-tumorigenic role of BHLHE40. Through RNA sequencing, the transcription factor KLF7 and the metalloproteinase ADAM19 were identified as putative BHLHE40 downstream effectors. Bioinformatic analyses showed that both KLF7 and ADAM19 are upregulated in colorectal tumors as well as associated with worse survival and their downregulation impaired HCT116 clonogenic activity. In addition, ADAM19, but not KLF7, downregulation reduced HCT116 cell growth. Overall, these data have revealed a ETV1/JMJD1A/JMJD2A→BHLHE40 axis that may stimulate colorectal tumorigenesis through upregulation of genes such as KLF7 and ADAM19, suggesting that targeting this axis represents a potential novel therapeutic avenue.
Collapse
Affiliation(s)
- Yuan Sui
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Hanlin Jiang
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Collyn M Kellogg
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Stephenson Cancer Center, Oklahoma City, OK, United States
| | - Ralf Janknecht
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Stephenson Cancer Center, Oklahoma City, OK, United States
| |
Collapse
|
7
|
Uno Y, Tanaka H, Miyakawa K, Akiyama N, Kamikokura Y, Yuzawa S, Kitada M, Takei H, Tanino M. Subcellular localization of hTERT in breast cancer: insights into its tumorigenesis and drug resistance mechanisms in HER2-immunopositive breast cancer. Hum Pathol 2022; 134:74-84. [PMID: 36549600 DOI: 10.1016/j.humpath.2022.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Human telomerase reverse transcriptase (hTERT) is highly expressed in various cancers, including breast cancer. Although telomere elongation is an essential role for hTERT, the nuclear export after oxdative stress has also been shown in several cancer cell lines and is associated with drug-resistance in vitro. As only a few reports focused on the subcellular localization of hTERT in clinical specimens, we performed immunohistochemistry (IHC) and analyzed the correlation between intracellular hTERT expression and the clinicopathological characteristics to identify the clinical significance of hTERT subcellular expression in breast cancers. 144 invasive breast cancers classified by IHC subtype without primary systemic therapy (PST), were selected from a surgical resection cohort and were immunostained for hTERT, p-STAT3, p-AKT and p-ERK. The nuclear and/or cytoplasmic staining intensity and proportion of hTERT were scored and compared with clinicopathological parameters. The nuclear hTERT expression was significantly correlated with HER2 expression (p = 0.00156), and the scores were significantly correlated with p-STAT3 and p-AKT expression scores (r = 0.532, p = 0.000587 and r = 0.345, p = 0.0339, respectively) in the HER2-immunopositive breast cancer including luminal-HER2 and HER2 subtypes. Furthermore, hTERT was expressed more in cytoplasm in the specimens after PST than those before PST, and the score tended to be negatively correlated with tumor shrinkage rate in HER2 subtype (r = -0.593, p = 0.0705). These results suggest that nuclear and/or cytoplasmic hTERT may play a different role before and after PST including the tumorigenesis and drug-resistance in breast cancer. Suppression of cytoplasmic hTERT expression may lead to more effective strategy for drug-resistant HER2 subtype in breast cancer.
Collapse
Affiliation(s)
- Yuji Uno
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Hokkaido, 0788510, Japan
| | - Hiroki Tanaka
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Hokkaido, 0788510, Japan
| | - Keita Miyakawa
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Hokkaido, 0788510, Japan
| | - Naoko Akiyama
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Hokkaido, 0788510, Japan
| | - Yuki Kamikokura
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Hokkaido, 0788510, Japan
| | - Sayaka Yuzawa
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Hokkaido, 0788510, Japan
| | - Masahiro Kitada
- Breast Center, Asahikawa Medical University Hospital, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Hokkaido, 0788510, Japan
| | - Hidehiro Takei
- Ochsner LSU Health Shreveport-Academic Medical Center, 1541 Kings Highway Shreveport, LA, 71103, USA
| | - Mishie Tanino
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Midorigaoka-Higashi 2-1-1-1, Asahikawa, Hokkaido, 0788510, Japan.
| |
Collapse
|
8
|
Akıncılar S, Chua J, Ng Q, Chan C, Eslami-S Z, Chen K, Low JL, Arumugam S, Aswad L, Chua C, Tan I, DasGupta R, Fullwood M, Tergaonkar V. Identification of mechanism of cancer-cell-specific reactivation of hTERT offers therapeutic opportunities for blocking telomerase specifically in human colorectal cancer. Nucleic Acids Res 2022; 51:1-16. [PMID: 35697349 PMCID: PMC9841410 DOI: 10.1093/nar/gkac479] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/18/2022] [Accepted: 05/26/2022] [Indexed: 01/29/2023] Open
Abstract
Transcriptional reactivation of hTERT is the limiting step in tumorigenesis. While mutations in hTERT promoter present in 19% of cancers are recognized as key drivers of hTERT reactivation, mechanisms by which wildtype hTERT (WT-hTERT) promoter is reactivated, in majority of human cancers, remain unknown. Using primary colorectal cancers (CRC) we identified Tert INTeracting region 2 (T-INT2), the critical chromatin region essential for reactivating WT-hTERT promoter in CRCs. Elevated β-catenin and JunD level in CRC facilitates chromatin interaction between hTERT promoter and T-INT2 that is necessary to turn on hTERTexpression. Pharmacological screens uncovered salinomycin, which inhibits JunD mediated hTERT-T-INT2 interaction that is required for the formation of a stable transcription complex on the hTERT promoter. Our results showed for the first time how known CRC alterations, such as APC, lead to WT-hTERT promoter reactivation during stepwise-tumorigenesis and provide a new perspective for developing cancer-specific drugs.
Collapse
Affiliation(s)
- Semih Can Akıncılar
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Joelle Yi Heng Chua
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Qin Feng Ng
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Claire Hian Tzer Chan
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Zahra Eslami-S
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Kaijing Chen
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Joo-Leng Low
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, 138672, Singapore
| | - Surendar Arumugam
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Luay Aswad
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Clarinda Chua
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 138672, Singapore,Department of Medical Oncology, National Cancer Centre Singapore, 169610, Singapore
| | - Iain Beehuat Tan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 138672, Singapore,Department of Medical Oncology, National Cancer Centre Singapore, 169610, Singapore
| | - Ramanuj DasGupta
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, 138672, Singapore
| | - Melissa Jane Fullwood
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore,School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Vinay Tergaonkar
- To whom correspondence should be addressed. Tel: +65 65869836; Fax: +65 67791117;
| |
Collapse
|
9
|
Zhou Z, Li Y, Xu H, Xie X, He Z, Lin S, Li R, Jin S, Cui J, Hu H, Liu F, Wu S, Ma W, Songyang Z. An inducible CRISPR/Cas9 screen identifies DTX2 as a transcriptional regulator of human telomerase. iScience 2022; 25:103813. [PMID: 35198878 PMCID: PMC8844827 DOI: 10.1016/j.isci.2022.103813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 01/12/2023] Open
Abstract
Most tumor cells reactivate telomerase to ensure unlimited proliferation, whereas the expression of human telomerase reverse transcriptase (hTERT) is tightly regulated and rate-limiting for telomerase activity maintenance. Several general transcription factors (TFs) have been found in regulating hTERT transcription; however, a systematic study is lacking. Here we performed an inducible CRISPR/Cas9 KO screen using an hTERT core promoter-driven reporter. We identified numerous positive regulators including an E3 ligase DTX2. In telomerase-positive cancer cells, DTX2 depletion downregulated hTERT transcription and telomerase activity, contributing to progressive telomere shortening, growth arrest, and increased apoptosis. Utilizing BioID, we characterized multiple TFs as DTX2 proximal proteins, among which NFIC functioned corporately with DTX2 in promoting hTERT transcription. Further analysis demonstrated that DTX2 mediated K63-linked ubiquitination of NFIC, which facilitated NFIC binding to the hTERT promoter and enhanced hTERT expression. These findings highlight a new hTERT regulatory pathway that may be exploited for potential cancer therapeutics. An inducible CRISPR/Cas9 screen identifies regulators for hTERT transcription DTX2 deficiency leads to telomere shortening and cell growth arrest DTX2 mediates ubiquitination on NFIC, stabilizing NFIC binding on hTERT promoter DTX2-NFIC functions corporately to promote hTERT transcription and tumorigenesis
Collapse
Affiliation(s)
- Zhifen Zhou
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Yujing Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Huimin Xu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Xiaowei Xie
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Zibin He
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Song Lin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Ruofei Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Hai Hu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Feng Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Su Wu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Corresponding author
| | - Wenbin Ma
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Corresponding author
| | - Zhou Songyang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
- Corresponding author
| |
Collapse
|
10
|
Segura-Bautista D, Maya-Nunez G, Aguilar-Rojas A, Huerta-Reyes M, Pérez-Solis MA. Contribution of Stemness-linked Transcription Regulators to the Progression of Breast Cancer. Curr Mol Med 2021; 22:766-778. [PMID: 34819003 DOI: 10.2174/1566524021666211124154803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/05/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022]
Abstract
Although there are currently several factors that allow measuring the risk of having breast cancer or predicting its progression, the underlying causes of this malignancy have remained unknown. Several molecular studies have described some mechanisms involved in the progress of breast cancer. These have helped in identifying new targets with therapeutic potential. However, despite the therapeutic strategies implemented from the advances achieved in breast cancer research, a large percentage of patients with breast cancer die due to the spread of malignant cells to other tissues or organs, such as bones and lungs. Therefore, determining the processes that promote the migration of malignant cells remains one of the greatest challenges for oncological research. Several research groups have reported evidence on how the dedifferentiation of tumor cells leads to the acquisition of stemness characteristics, such as invasion, metastasis, the capability to evade the immunological response, and resistance to several cytotoxic drugs. These phenotypic changes have been associated with a complex reprogramming of gene expression in tumor cells during the Epithelial-Mesenchymal Transition (EMT). Considering the determining role that the transcriptional regulation plays in the expression of the specific characteristics and attributes of breast cancer during ETM, in the present work, we reviewed and analyzed several transcriptional mechanisms that support the mesenchymal phenotype. In the same way, we established the importance of transcription factors with a therapeutic perspective in the progress of breast cancer.
Collapse
Affiliation(s)
- David Segura-Bautista
- Medical Research Unit in Reproductive Medicine, UMAE Hospital de Gineco Obstetricia no. 4 'Luis Castelazo-Ayala', Instituto Mexicano del Seguro Social, Mexico City. Mexico
| | - Guadalupe Maya-Nunez
- Medical Research Unit in Reproductive Medicine, UMAE Hospital de Gineco Obstetricia no. 4 'Luis Castelazo-Ayala', Instituto Mexicano del Seguro Social, Mexico City. Mexico
| | - Arturo Aguilar-Rojas
- Medical Research Unit in Reproductive Medicine, UMAE Hospital de Gineco Obstetricia no. 4 'Luis Castelazo-Ayala', Instituto Mexicano del Seguro Social, Mexico City. Mexico
| | - Maira Huerta-Reyes
- Medical Research Unit in Nephrological Diseases, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City. Mexico
| | - Marco Allan Pérez-Solis
- Medical Research Unit in Reproductive Medicine, UMAE Hospital de Gineco Obstetricia no. 4 'Luis Castelazo-Ayala', Instituto Mexicano del Seguro Social, Mexico City. Mexico
| |
Collapse
|
11
|
Oh S, Shin S, Janknecht R. Sumoylation of transcription factor ETV1 modulates its oncogenic potential in prostate cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2021; 14:795-810. [PMID: 34367411 PMCID: PMC8339722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/21/2021] [Indexed: 06/13/2023]
Abstract
The transcription factor ETS variant 1 (ETV1) is capable of promoting prostate tumorigenesis. We demonstrate that ETV1 can be posttranslationally modified by covalent attachment of small ubiquitin-like modifier 1 (SUMO1) onto four different lysine residues. In human embryonic kidney 293T cells, mutation of these sumoylation sites stimulated the transactivation potential of ETV1 at the matrix metalloproteinase 1 (MMP1), but not Yes-associated protein 1 gene promoter, while ETV1 protein stability and intracellular localization remained unchanged. In stark contrast, sumoylation-deficient ETV1 was repressed in its ability to stimulate the MMP1 promoter and to cooperate with a histone demethylase, JmjC domain-containing 2A (JMJD2A), in LNCaP prostate cancer cells. Mutation of sumoylation sites enhanced the ability of ETV1 to interact with the histone deacetylase (HDAC) 1, but had basically no impact on complex formation with HDAC3 or JMJD2A. Further, compared to non-sumoylated ETV1, its sumoylated forms were less able to bind to the transcription factor, SMAD family member 4. Lastly, in contrast to wild-type ETV1, sumoylation-deficient ETV1 repressed LNCaP cell growth. Altogether, these data suggest that sumoylation modulates ETV1 function in a cell type-specific manner, possibly by altering the spectrum of transcriptional cofactors being recruited. Notably, SUMO pathway components SUMO1, ubiquitin-like modifier activating enzyme 2 and ubiquitin conjugating enzyme 9 were upregulated in prostate tumors, implying that enhanced sumoylation indeed promotes ETV1's oncogenic activity during prostate cancer formation.
Collapse
Affiliation(s)
- Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
| | - Sook Shin
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
| |
Collapse
|
12
|
Oh S, Song H, Freeman WM, Shin S, Janknecht R. Cooperation between ETS transcription factor ETV1 and histone demethylase JMJD1A in colorectal cancer. Int J Oncol 2020; 57:1319-1332. [PMID: 33174020 PMCID: PMC7646594 DOI: 10.3892/ijo.2020.5133] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
ETS variant 1 (ETV1) is an oncogenic transcription factor. However, its role in colorectal cancer has remained understudied. The present study demonstrated that ETV1 downregulation led to reduced HCT116 colorectal cancer cell growth and clonogenic activity. Furthermore, the ETV1 mRNA levels were enhanced in colorectal tumors and were associated with disease severity. In addition, ETV1 directly bound to Jumonji C domain-containing (JMJD) 1A, a histone demethylase known to promote colon cancer. ETV1 and JMJD1A, but not a catalytically inactive mutant thereof, cooperated in inducing the matrix metalloproteinase (MMP)1 gene promoter that was similar to the cooperation between ETV1 and another histone demethylase, JMJD2A. RNA-sequencing revealed multiple potential ETV1 target genes in HCT116 cells, including the FOXQ1 and TBX6 transcription factor genes. Moreover, JMJD1A co-regulated FOXQ1 and other ETV1 target genes, but not TBX6, whereas JMJD2A downregulation had no impact on FOXQ1 as well as TBX6 transcription. Accordingly, the FOXQ1 gene promoter was stimulated by ETV1 and JMJD1A in a cooperative manner, and both ETV1 and JMJD1A bound to the FOXQ1 promoter. Notably, the overexpression of FOXQ1 partially reversed the growth inhibitory effects of ETV1 ablation on HCT116 cells, whereas TBX6 impaired HCT116 cell growth and may thereby dampen the oncogenic activity of ETV1. The latter also revealed for the first time, to the best of our knowledge, a potential tumor suppressive function of TBX6. Taken together, the present study uncovered a ETV1/JMJD1A-FOXQ1 axis that may drive colorectal tumorigenesis.
Collapse
Affiliation(s)
- Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hoogeun Song
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | - Sook Shin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
13
|
Bradfield A, Button L, Drury J, Green DC, Hill CJ, Hapangama DK. Investigating the Role of Telomere and Telomerase Associated Genes and Proteins in Endometrial Cancer. Methods Protoc 2020; 3:E63. [PMID: 32899298 PMCID: PMC7565490 DOI: 10.3390/mps3030063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/24/2020] [Accepted: 08/30/2020] [Indexed: 12/16/2022] Open
Abstract
Endometrial cancer (EC) is the commonest gynaecological malignancy. Current prognostic markers are inadequate to accurately predict patient survival, necessitating novel prognostic markers, to improve treatment strategies. Telomerase has a unique role within the endometrium, whilst aberrant telomerase activity is a hallmark of many cancers. The aim of the current in silico study is to investigate the role of telomere and telomerase associated genes and proteins (TTAGPs) in EC to identify potential prognostic markers and therapeutic targets. Analysis of RNA-seq data from The Cancer Genome Atlas identified differentially expressed genes (DEGs) in EC (568 TTAGPs out of 3467) and ascertained DEGs associated with histological subtypes, higher grade endometrioid tumours and late stage EC. Functional analysis demonstrated that DEGs were predominantly involved in cell cycle regulation, while the survival analysis identified 69 DEGs associated with prognosis. The protein-protein interaction network constructed facilitated the identification of hub genes, enriched transcription factor binding sites and drugs that may target the network. Thus, our in silico methods distinguished many critical genes associated with telomere maintenance that were previously unknown to contribute to EC carcinogenesis and prognosis, including NOP56, WFS1, ANAPC4 and TUBB4A. Probing the prognostic and therapeutic utility of these novel TTAGP markers will form an exciting basis for future research.
Collapse
Affiliation(s)
- Alice Bradfield
- Department of Women’s and Children’s Health, University of Liverpool, Crown St, Liverpool L69 7ZX, UK; (A.B.); (J.D.); (C.J.H.)
| | - Lucy Button
- Faculty of Health and Life Sciences, University of Liverpool, Brownlow Hill, Liverpool L69 7ZX, UK;
| | - Josephine Drury
- Department of Women’s and Children’s Health, University of Liverpool, Crown St, Liverpool L69 7ZX, UK; (A.B.); (J.D.); (C.J.H.)
| | - Daniel C. Green
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, UK;
| | - Christopher J. Hill
- Department of Women’s and Children’s Health, University of Liverpool, Crown St, Liverpool L69 7ZX, UK; (A.B.); (J.D.); (C.J.H.)
| | - Dharani K. Hapangama
- Department of Women’s and Children’s Health, University of Liverpool, Crown St, Liverpool L69 7ZX, UK; (A.B.); (J.D.); (C.J.H.)
- Liverpool Women’s NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool L8 7SS, UK
| |
Collapse
|
14
|
Aboelela S, Ashmawy A, Shaarawy S, El-Hefny M, Medhat A. Telomerase as a Possible Candidate Targeting Therapy in Different Breast Cancer Cell Lines. Asian Pac J Cancer Prev 2020; 21:2243-2250. [PMID: 32856851 PMCID: PMC7771952 DOI: 10.31557/apjcp.2020.21.8.2243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Indexed: 01/12/2023] Open
Abstract
Background: Telomerase activity is up regulated in most breast cancer subtypes but not in the adjacent normal tissues. Thus, it is a promising target for anticancer therapy. The present work investigated the effects of telomerase inhibition by siRNA on breast cancer cell lines and studied the feasibility of whether the combined effect of doxorubicin with siRNA treatment on breast cancer cells potentiates a rapid cellular response to the cytotoxic effect of chemotherapy. Methods: This study was performed on Luminal A (MCF-7), triple negative (MDA-MB-468), and HER-2/neu (SKBR-3) human breast cancer cell lines, wherein telomerase activity inhibition by hTERT siRNA and doxorubicin was detected by measuring telomerase activity using Telomeric Repeat Amplification Protocol (TRAP assay), assessing cell viability through MTT assay, and evaluating apoptosis through scanning electron microscopy (SEM) and through estimating caspase-3 and -8 activities using enzyme-linked immunosorbent assay (ELISA). Results: In the present study, hTERT siRNA effectively reduced telomerase activity and cell viability to more than 90% and 60%, respectively, in most breast cancer cell lines within 72 hours after transfection. The combination of hTERT siRNA and doxorubicin showed a cumulative effect compared with either treatment alone (P < 0.05). Meanwhile, SEM demonstrated apoptotic morphologic cell changes. Conclusion: Telomerase inhibition is a promising strategy for the effective treatment of breast cancer. When used in combination with doxorubicin, it could potentiate the cytotoxic effect of the drug on breast cancer cells.
Collapse
Affiliation(s)
- Salma Aboelela
- Electron Microscopy Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo Egypt
| | - Abeer Ashmawy
- Biochemistry unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Sabry Shaarawy
- Biochemistry unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohammed El-Hefny
- Biochemistry unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo Egypt. Department of Medical Genetics, Umm Al-Qura University, Alqunfudah, KSA
| | - Amina Medhat
- Biochemistry Department, Faculty of Science, Ain-Shams University, Cairo Egypt
| |
Collapse
|
15
|
Oh S, Shin S, Song H, Grande JP, Janknecht R. Relationship between ETS Transcription Factor ETV1 and TGF-β-regulated SMAD Proteins in Prostate Cancer. Sci Rep 2019; 9:8186. [PMID: 31160676 PMCID: PMC6546734 DOI: 10.1038/s41598-019-44685-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/22/2019] [Indexed: 01/12/2023] Open
Abstract
The ETS transcription factor ETV1 is frequently overexpressed in aggressive prostate cancer, which is one underlying cause of this disease. Accordingly, transgenic mice that prostate-specifically overexpress ETV1 develop prostatic intraepithelial neoplasia. However, progression to the adenocarcinoma stage is stifled in these mice, suggesting that inhibitory pathways possibly preclude ETV1 from exerting its full oncogenic potential. Here we provide evidence that TGF-β/SMAD signaling represents such an inhibitory pathway. First, we discovered that ETV1 forms complexes with SMAD4. Second, SMAD2, SMAD3 and SMAD4 overexpression impaired ETV1’s ability to stimulate gene transcription. Third, TGF-β1 inhibited ETV1-induced invasion by benign RWPE-1 prostate cells. Fourth, increased expression of SMAD3 and SMAD4 was observable in prostates of ETV1 transgenic mice. Conversely, we found that ETV1 may enhance TGF-β signaling in PC3 prostate cancer cells, revealing a different facet of the ETV1/TGF-β interplay. Altogether, these data provide more insights into the regulation and action of ETV1 and additionally suggest that TGF-β/SMAD signaling exerts its tumor suppressive activity, at least in part, by curtailing the oncogenic potential of ETV1 in prostatic lesions.
Collapse
Affiliation(s)
- Sangphil Oh
- University of Oklahoma Health Sciences Center, Department of Cell Biology, Oklahoma City, OK, 73104, USA.,Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
| | - Sook Shin
- University of Oklahoma Health Sciences Center, Department of Cell Biology, Oklahoma City, OK, 73104, USA.,Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
| | - Hoogeun Song
- University of Oklahoma Health Sciences Center, Department of Cell Biology, Oklahoma City, OK, 73104, USA
| | - Joseph P Grande
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN, 55905, USA
| | - Ralf Janknecht
- University of Oklahoma Health Sciences Center, Department of Cell Biology, Oklahoma City, OK, 73104, USA. .,Stephenson Cancer Center, Oklahoma City, OK, 73104, USA. .,University of Oklahoma Health Sciences Center, Department of Pathology, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
16
|
Eid W, Abdel-Rehim W. Genome-wide analysis of ETV1 targets: Insights into the role of ETV1 in tumor progression. J Cell Biochem 2019; 120:8983-8991. [PMID: 30629294 DOI: 10.1002/jcb.28169] [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: 09/17/2018] [Accepted: 11/08/2018] [Indexed: 12/15/2022]
Abstract
ETS variant 1 (ETV1) is a key player in metastatic progression in several types of human cancers, yet the direct target genes of ETV1 and the mechanisms by which ETV1 exerts its deleterious function remain largely elusive. Here, we performed large-scale mapping and analysis of target loci of ETV1 in the prostate cancer cells LNCaP using the DNA adenine methyltransferase identification technique, we identified close to 800 direct targets for ETV1. Expression analysis using quantitative reverse transcription polymerase chain reaction confirmed a positive regulation by ETV1 in most of the genes examined. Furthermore, gene and pathway analysis unraveled new signaling pathways and biological networks that interact with ETV1. Our findings cast light on genes and networks regulated by ETV1, it also opens new fronts for studying the role of ETV1 and its target genes in tumorigenesis.
Collapse
Affiliation(s)
- Wassim Eid
- Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria, Egypt
| | - Wafaa Abdel-Rehim
- Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria, Egypt
| |
Collapse
|
17
|
Li X, Oh S, Song H, Shin S, Zhang B, Freeman WM, Janknecht R. A potential common role of the Jumonji C domain-containing 1A histone demethylase and chromatin remodeler ATRX in promoting colon cancer. Oncol Lett 2018; 16:6652-6662. [PMID: 30405805 PMCID: PMC6202502 DOI: 10.3892/ol.2018.9487] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022] Open
Abstract
Jumonji C domain-containing 1A (JMJD1A) is a histone demethylase and epigenetic regulator that has been implicated in cancer development. In the current study, its mRNA and protein expression was analyzed in human colorectal tumors. It was demonstrated that JMJD1A levels were increased and correlated with a more aggressive phenotype. Downregulation of JMJD1A in human HCT116 colorectal cancer cells caused negligible growth defects, but robustly decreased clonogenic activity. Transcriptome analysis revealed that JMJD1A downregulation led to multiple changes in HCT116 cells, including inhibition of MYC- and MYCN-regulated pathways and stimulation of the TP53 tumor suppressor response. One gene identified to be stimulated by JMJD1A was α-thalassemia/mental retardation syndrome X-linked (ATRX), which encodes for a chromatin remodeler. The JMJD1A protein, but not a catalytically inactive mutant, activated the ATRX gene promoter and JMJD1A also affected levels of dimethylation on lysine 9 of histone H3. Similar to JMJD1A, ATRX was significantly overexpressed in human colorectal tumors and correlated with increased disease recurrence and lethality. Furthermore, ATRX downregulation in HCT116 cells reduced their growth and clonogenic activity. Accordingly, upregulation of ATRX may represent one mechanism by which JMJD1A promotes colorectal cancer. In addition, the data presented in this study suggest that the current notion of ATRX as a tumor suppressor is incomplete and that ATRX might context dependently also function as a tumor promoter.
Collapse
Affiliation(s)
- Xiaomeng Li
- Department of Endoscopy and Gastrointestinal Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA.,Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Hoogeun Song
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Sook Shin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA.,Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Bin Zhang
- Department of Endoscopy and Gastrointestinal Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Willard M Freeman
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA.,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA.,Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| |
Collapse
|
18
|
Ozturk MB, Li Y, Tergaonkar V. Current Insights to Regulation and Role of Telomerase in Human Diseases. Antioxidants (Basel) 2017; 6:antiox6010017. [PMID: 28264499 PMCID: PMC5384180 DOI: 10.3390/antiox6010017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/16/2017] [Accepted: 02/21/2017] [Indexed: 12/31/2022] Open
Abstract
The telomerase ribonucleoprotein complex has a pivotal role in regulating the proliferation and senescence of normal somatic cells as well as cancer cells. This complex is comprised mainly of telomerase reverse transcriptase (TERT), telomerase RNA component (TERC) and other associated proteins that function to elongate telomeres localized at the end of the chromosomes. While reactivation of telomerase is a major hallmark of most cancers, together with the synergistic activation of other oncogenic signals, deficiency in telomerase and telomeric proteins might lead to aging and senescence-associated disorders. Therefore, it is critically important to understand the canonical as well as non-canonical functions of telomerase through TERT to develop a therapeutic strategy against telomerase-related diseases. In this review, we shed light on the regulation and function of telomerase, and current therapeutic strategies against telomerase in cancer and age-related diseases.
Collapse
Affiliation(s)
- Mert Burak Ozturk
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore.
| | - Yinghui Li
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore.
| | - Vinay Tergaonkar
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore.
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide SA 5000, Australia.
| |
Collapse
|
19
|
Khattar E, Tergaonkar V. Transcriptional Regulation of Telomerase Reverse Transcriptase (TERT) by MYC. Front Cell Dev Biol 2017; 5:1. [PMID: 28184371 PMCID: PMC5266692 DOI: 10.3389/fcell.2017.00001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/11/2017] [Indexed: 12/22/2022] Open
Abstract
Telomerase elongates telomeres and is crucial for maintaining genomic stability. While stem cells and cancer cells display high telomerase activity, normal somatic cells lack telomerase activity primarily due to transcriptional repression of telomerase reverse transcriptase (TERT), the catalytic component of telomerase. Transcription factor binding, chromatin status as well as epigenetic modifications at the TERT promoter regulates TERT transcription. Myc is an important transcriptional regulator of TERT that directly controls its expression by promoter binding and associating with other transcription factors. In this review, we discuss the current understanding of the molecular mechanisms behind regulation of TERT transcription by Myc. We also discuss future perspectives in investigating the regulation of Myc at TERT promoter during cancer development.
Collapse
Affiliation(s)
- Ekta Khattar
- Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology, ASTAR Singapore, Singapore
| | - Vinay Tergaonkar
- Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology, ASTARSingapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore; Centre for Cancer Biology, University of South Australia and SA PathologyAdelaide, SA, Australia
| |
Collapse
|
20
|
Heidenreich B, Kumar R. TERT promoter mutations in telomere biology. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 771:15-31. [PMID: 28342451 DOI: 10.1016/j.mrrev.2016.11.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/10/2016] [Indexed: 02/07/2023]
Abstract
Telomere repeats at chromosomal ends, critical to genome integrity, are maintained through an elaborate network of proteins and pathways. Shelterin complex proteins shield telomeres from induction of DNA damage response to overcome end protection problem. A specialized ribonucleic protein, telomerase, maintains telomere homeostasis through repeat addition to counter intrinsic shortcomings of DNA replication that leads to gradual sequence shortening in successive mitoses. The biogenesis and recruitment of telomerase composed of telomerase reverse transcriptase (TERT) subunit and an RNA component, takes place through the intricate machinery that involves an elaborate number of molecules. The synthesis of telomeres remains a controlled and limited process. Inherited mutations in the molecules involved in the process directly or indirectly cause telomeropathies. Telomerase, while present in stem cells, is deactivated due to epigenetic silencing of the rate-limiting TERT upon differentiation in most of somatic cells with a few exceptions. However, in most of the cancer cells telomerase reactivation remains a ubiquitous process and constitutes one of the major hallmarks. Discovery of mutations within the core promoter of the TERT gene that create de novo binding sites for E-twenty-six (ETS) transcription factors provided a mechanism for cancer-specific telomerase reactivation. The TERT promoter mutations occur mainly in tumors from tissues with low rates of self-renewal. In melanoma, glioma, hepatocellular carcinoma, urothelial carcinoma and others, the promoter mutations have been shown to define subsets of patients with adverse disease outcomes, associate with increased transcription of TERT, telomerase reactivation and affect telomere length; in stem cells the mutations inhibit TERT silencing following differentiation into adult cells. The TERT promoter mutations cause an epigenetic switch on the mutant allele along with recruitment of pol II following the binding of GABPA/B1 complex that leads to mono-allelic expression. Thus, the TERT promoter mutations hold potential as biomarkers as well as future therapeutic targets.
Collapse
Affiliation(s)
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology; German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center, 69120 Heidelberg, Germany.
| |
Collapse
|
21
|
MacNeil DE, Bensoussan HJ, Autexier C. Telomerase Regulation from Beginning to the End. Genes (Basel) 2016; 7:genes7090064. [PMID: 27649246 PMCID: PMC5042394 DOI: 10.3390/genes7090064] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/25/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022] Open
Abstract
The vast body of literature regarding human telomere maintenance is a true testament to the importance of understanding telomere regulation in both normal and diseased states. In this review, our goal was simple: tell the telomerase story from the biogenesis of its parts to its maturity as a complex and function at its site of action, emphasizing new developments and how they contribute to the foundational knowledge of telomerase and telomere biology.
Collapse
Affiliation(s)
- Deanna Elise MacNeil
- Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte Ste-Catherine Road, Montréal, QC H3T 1E2, Canada.
- Room M-29, Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montréal, QC H3A 0C7, Canada.
| | - Hélène Jeanne Bensoussan
- Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte Ste-Catherine Road, Montréal, QC H3T 1E2, Canada.
- Room M-29, Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montréal, QC H3A 0C7, Canada.
| | - Chantal Autexier
- Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte Ste-Catherine Road, Montréal, QC H3T 1E2, Canada.
- Room M-29, Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montréal, QC H3A 0C7, Canada.
- Department of Experimental Medicine, McGill University, 1110 Pins Avenue West, Room 101, Montréal, QC H3A 1A3, Canada.
| |
Collapse
|
22
|
Transcription Regulation of the Human Telomerase Reverse Transcriptase (hTERT) Gene. Genes (Basel) 2016; 7:genes7080050. [PMID: 27548225 PMCID: PMC4999838 DOI: 10.3390/genes7080050] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/23/2016] [Accepted: 08/01/2016] [Indexed: 12/11/2022] Open
Abstract
Embryonic stem cells and induced pluripotent stem cells have the ability to maintain their telomere length via expression of an enzymatic complex called telomerase. Similarly, more than 85%–90% of cancer cells are found to upregulate the expression of telomerase, conferring them with the potential to proliferate indefinitely. Telomerase Reverse Transcriptase (TERT), the catalytic subunit of telomerase holoenzyme, is the rate-limiting factor in reconstituting telomerase activity in vivo. To date, the expression and function of the human Telomerase Reverse Transcriptase (hTERT) gene are known to be regulated at various molecular levels (including genetic, mRNA, protein and subcellular localization) by a number of diverse factors. Among these means of regulation, transcription modulation is the most important, as evident in its tight regulation in cancer cell survival as well as pluripotent stem cell maintenance and differentiation. Here, we discuss how hTERT gene transcription is regulated, mainly focusing on the contribution of trans-acting factors such as transcription factors and epigenetic modifiers, as well as genetic alterations in hTERT proximal promoter.
Collapse
|
23
|
Kim TD, Oh S, Lightfoot SA, Shin S, Wren JD, Janknecht R. Upregulation of PSMD10 caused by the JMJD2A histone demethylase. Int J Clin Exp Med 2016; 9:10123-10134. [PMID: 28883898 PMCID: PMC5584593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
PSMD10, also known as gankyrin, is associated with the proteasome and has been shown to be an oncoprotein in the liver. Here, we report that PSMD10 expression is stimulated by the histone demethylase JMJD2A/KDM4A and its interaction partner, the ETV1 transcription factor, in LNCaP prostate cancer cells. Global analysis of expression patterns revealed that PSMD10 mRNA levels are positively correlated with those of both JMJD2A and ETV1. In human prostate tumors, PSMD10 is highly overexpressed at the protein level and correlates with JMJD2A overexpression; further, PSMD10 expression is enhanced in the prostates of transgenic JMJD2A mice. Moreover, PSMD10 is particularly overexpressed in high Gleason score prostate tumors. Downregulation of PSMD10 in LNCaP prostate cancer cells impaired their growth, indicating that PSMD10 may exert a pro-oncogenic function in the prostate. Lastly, we observed that PSMD10 expression is correlated to YAP1, a component of the Hippo signaling pathway and whose gene promoter is regulated by JMJD2A, and that PSMD10 can cooperate with YAP1 in stimulating LNCaP cell growth. Altogether, these data indicate that PSMD10 is a novel downstream effector of JMJD2A and suggest that inhibition of the JMJD2A histone demethylase by small molecule drugs may be effective to curtail the oncogenic activity of PSMD10 in various PSMD10-overexpressing tumors.
Collapse
Affiliation(s)
- Tae-Dong Kim
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Stan A Lightfoot
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Sook Shin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jonathan D Wren
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, Oklahoma City, OK 73104, USA
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
24
|
Oztas E, Kara H, Kara ZP, Aydogan MU, Uras C, Ozhan G. Association Between Human Telomerase Reverse Transcriptase Gene Variations and Risk of Developing Breast Cancer. Genet Test Mol Biomarkers 2016; 20:459-64. [PMID: 27336831 DOI: 10.1089/gtmb.2015.0339] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Despite a reduction in the number of deaths from cancers made possible by the development of early detection tests, improvements in treatment, changes in the age distribution of the population, and changes of personal behaviors as a result of awareness, breast cancer remains a major health problem worldwide. Breast cancer is the most common cancer and second leading cause of cancer death in women. Several genetic and environmental factors are known to be involved in breast cancer pathogenesis, but its exact etiology is complicated and is not clearly identified. The structure and integrity of telomeres are pivotal for genome stability, and telomere length is maintained by the expression of the telomerase enzyme. The human telomerase reverse transcriptase (hTERT) gene is a principal functional subunit of the telomerase. Several recent studies have provided evidence that hTERT gene variants may have an important role in cancer development. METHODS Three hTERT variants (rs2736100, rs2736098, and rs2853669) were genotyped for 107 breast cancer patients and 110 healthy controls to determine their effect on breast cancer susceptibility. RESULTS It was observed that hTERT rs2736098 was associated with breast cancer risk (odds ratio [OR] = 1.88; p = 0.034), while rs2736100 and rs2853669 did not significantly differ between the groups. CONCLUSIONS These findings are the first description of hTERT allele distributions in the Turkish population and may contribute to our understanding of breast cancer development. Nevertheless, further large-scale population studies are needed to understand the role of the hTERT polymorphisms and haplotypes in the development of breast cancer.
Collapse
Affiliation(s)
- Ezgi Oztas
- 1 Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University , Istanbul, Turkey
| | - Halil Kara
- 2 Department of General Surgery, Faculty of Medicine, Acibadem University , Istanbul, Turkey
| | - Zeliha Pala Kara
- 3 Department of Pharmacology, Faculty of Pharmacy, Istanbul University , Istanbul, Turkey
| | - Manolya Uras Aydogan
- 4 Department of Physiology, Cerrahpasa Medical Faculty, Istanbul University , Istanbul, Turkey
| | - Cihan Uras
- 2 Department of General Surgery, Faculty of Medicine, Acibadem University , Istanbul, Turkey
| | - Gul Ozhan
- 1 Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University , Istanbul, Turkey
| |
Collapse
|
25
|
Kim TD, Shin S, Janknecht R. ETS transcription factor ERG cooperates with histone demethylase KDM4A. Oncol Rep 2016; 35:3679-88. [PMID: 27109047 PMCID: PMC4869937 DOI: 10.3892/or.2016.4747] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/11/2016] [Indexed: 12/20/2022] Open
Abstract
ERG (ETS-related gene) is a member of the ETS (erythroblast transformation-specific) family of transcription factors. Overexpression of the ERG transcription factor is observed in half of all prostate tumors and is an underlying cause of this disease. However, the mechanisms involved in the functions of ERG are still not fully understood. In the present study, we showed that ERG can directly bind to KDM4A (also known as JMJD2A), a histone demethylase that particularly demethylates lysine 9 on histone H3. ERG and KDM4A cooperated in upregulating the promoter of Yes-associated protein 1 (YAP1), a downstream effector in the Hippo signaling pathway and crucial growth regulator. Multiple ERG binding sites within the human YAP1 gene promoter were identified and their impact on transcription was determined through mutational analysis. Furthermore, we found that ERG expression reduced histone H3 lysine 9 trimethylation at the YAP1 gene promoter, consistent with its epigenetic regulation through the ERG interaction partner, KDM4A. Finally, downregulation of YAP1 phenocopied the growth-retarding effect of ERG or KDM4A depletion in human VCaP prostate cancer cells. Collectively, these results elucidated a novel mechanism - ERG promotes prostate tumorigenesis together with KDM4A through the upregulation of YAP1. A corollary is that KDM4A as well as YAP1 inhibitors may prove beneficial for the therapy of ERG-overexpressing prostate tumors.
Collapse
Affiliation(s)
- Tae-Dong Kim
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Sook Shin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
26
|
Kulić A, Plavetić ND, Gamulin S, Jakić-Razumović J, Vrbanec D, Sirotković-Skerlev M. Telomerase activity in breast cancer patients: association with poor prognosis and more aggressive phenotype. Med Oncol 2016; 33:23. [PMID: 26833480 DOI: 10.1007/s12032-016-0736-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 01/18/2016] [Indexed: 11/28/2022]
Abstract
Telomerase expression is an important mechanism of tumor unlimited replicative potential. The aim of this study was to evaluate prognostic impact of telomerase activity in breast cancer patients and to correlate telomerase activity with established prognostic factors. We analyzed tissue of 102 malignant breast lesions and 20 healthy breast tissues. Telomerase activity was determined by telomeric repeat amplification protocol assay. Telomerase activity was present in 77 (75.49 %) of 102 breast cancers. Telomerase activity in breast cancers was statistically significantly higher in comparison with the activity in normal breast tissue. The levels of telomerase activity were significantly positively correlated with tumor size, axillary nodal status, histological grade, HER-2/neu protein expression in tumor tissue and expression of the nuclear antigen Ki-67. A statistically significant negative correlation was found between the presence of ER and telomerase activity. There was no correlation between telomerase activity and concentration of PR or the age of patients. Kaplan-Meier analysis showed that patients with higher telomerase activity had significantly shorter 10-year disease-free survival (p < 0.0001) and 10-year overall survival (p < 0.0001) than those with lower telomerase activity. These results were confirmed by logistic regression analysis. Our results support the prognostic role of telomerase activity and its relationship with the more aggressive phenotype of breast cancer.
Collapse
Affiliation(s)
- Ana Kulić
- Department of Oncology, Division of Pathophysiology and Experimental Oncology, University Hospital Center Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia
| | - Natalija Dedić Plavetić
- Department of Oncology, Division of Medical Oncology, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | | | - Jasminka Jakić-Razumović
- Department of Pathology, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Damir Vrbanec
- Department of Oncology, Division of Medical Oncology, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Maja Sirotković-Skerlev
- Department of Oncology, Division of Pathophysiology and Experimental Oncology, University Hospital Center Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia. .,School of Medicine, University of Zagreb, Zagreb, Croatia.
| |
Collapse
|
27
|
Heeg S. Variations in telomere maintenance and the role of telomerase inhibition in gastrointestinal cancer. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2015; 8:171-80. [PMID: 26675332 PMCID: PMC4675635 DOI: 10.2147/pgpm.s52808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Immortalization is an important step toward the malignant transformation of human cells and is critically dependent upon telomere maintenance. There are two known mechanisms to maintain human telomeres. The process of telomere maintenance is either mediated through activation of the enzyme telomerase or through an alternative mechanism of telomere lengthening called ALT. While 85% of all human tumors show reactivation of telomerase, the remaining 15% are able to maintain telomeres via ALT. The therapeutic potential of telomerase inhibitors is currently investigated in a variety of human cancers. Gastrointestinal tumors are highly dependent on telomerase as a mechanism of telomere maintenance, rendering telomeres as well as telomerase potential targets for cancer therapy. This article focuses on the molecular mechanisms of telomere biology and telomerase activation in gastrointestinal cancers and reviews strategies of telomerase inhibition and their potential therapeutic use in these tumor entities.
Collapse
Affiliation(s)
- Steffen Heeg
- Department of Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
28
|
Li Y, Tong Y, Wong YH. Regulatory functions of Nm23-H2 in tumorigenesis: insights from biochemical to clinical perspectives. Naunyn Schmiedebergs Arch Pharmacol 2014; 388:243-56. [PMID: 25413836 DOI: 10.1007/s00210-014-1066-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 11/07/2014] [Indexed: 12/12/2022]
Abstract
Substantial effort has been directed at elucidating the functions of the products of the Nm23 tumor metastasis suppressor genes over the past two decades, with the ultimate goal of exploring their translational potentials in changing cancer patients' outcomes. Much attention has been focused on the better-known Nm23-H1, but despite having high sequence similarity, Nm23-H2 functions differently in many aspects. Besides acting as a metastasis suppressor, compelling data suggest that Nm23-H2 may modulate various tumor-associated biological events to enhance tumorigenesis in human solid tumors and hematological malignancies. Linkage to tumorigenesis may occur through the ability of Nm23-H2 to regulate transcription, cell proliferation, apoptosis, differentiation, and telomerase activity. In this review, we examine the linkages of Nm23-H2 to tumorigenesis in terms of its biochemical and structural properties and discuss its potential role in various tumor-associated events.
Collapse
Affiliation(s)
- Yuanjun Li
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | | | | |
Collapse
|
29
|
Recent advances in the structural molecular biology of Ets transcription factors: interactions, interfaces and inhibition. Biochem Soc Trans 2014; 42:130-8. [PMID: 24450640 PMCID: PMC3901394 DOI: 10.1042/bst20130227] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The Ets family of eukaryotic transcription factors is based around the conserved Ets DNA-binding domain. Although their DNA-binding selectivity is biochemically and structurally well characterized, structures of homodimeric and ternary complexes point to Ets domains functioning as versatile protein-interaction modules. In the present paper, we review the progress made over the last decade to elucidate the structural mechanisms involved in modulation of DNA binding and protein partner selection during dimerization. We see that Ets domains, although conserved around a core architecture, have evolved to utilize a variety of interaction surfaces and binding mechanisms, reflecting Ets domains as dynamic interfaces for both DNA and protein interaction. Furthermore, we discuss recent advances in drug development for inhibition of Ets factors, and the roles structural biology can play in their future.
Collapse
|
30
|
Kim TD, Fuchs JR, Schwartz E, Abdelhamid D, Etter J, Berry WL, Li C, Ihnat MA, Li PK, Janknecht R. Pro-growth role of the JMJD2C histone demethylase in HCT-116 colon cancer cells and identification of curcuminoids as JMJD2 inhibitors. Am J Transl Res 2014; 6:236-247. [PMID: 24936217 PMCID: PMC4058306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 03/17/2014] [Indexed: 06/03/2023]
Abstract
Colon tumors are a major cause of cancer death, yet their molecular intricacies are not fully understood. We demonstrate that the histone demethylases JMJD2A, JMJD2B and JMJD2C are overexpressed in colon cancer cell lines, whereas another related protein, JMJD2D, is not. Interestingly, despite their high homology, the intracellular localization of JMJD2A-C is different in colon and other cancer cells, with JMJD2A being present comparably in the cytoplasm and nucleus, JMJD2B more prevalent in the nucleus and JMJD2C strongly associated with chromatin. This suggests that each of these three proteins performs different, non-redundant functions. Moreover, we show that JMJD2C (also called KDM4C) forms complexes with β-catenin, an oncoprotein whose overexpression is crucial for the development of most colonic tumors. In addition, JMJD2C downregulation reduced both growth and clonogenic capacity of HCT-116 colon cancer cells. Further, JMJD2C was required for efficient expression of the growth stimulatory proteins FRA1 and cyclin D1 as well as the survival factor BCL2. Lastly, we identified derivatives of curcumin as in vitro inhibitors of JMJD2 enzymes, suggesting that these curcuminoids could be useful for decreasing JMJD2 activity in vivo. In conclusion, our data highlight that overexpression of JMJD2C confers a pro-growth effect on colon cancer cells and, therefore, its inhibition by curcuminoids or other small molecules could be beneficial as an adjuvant therapy for colon cancer patients.
Collapse
Affiliation(s)
- Tae-Dong Kim
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - James R Fuchs
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State UniversityColumbus, OH 43210, USA
| | - Eric Schwartz
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State UniversityColumbus, OH 43210, USA
| | - Dalia Abdelhamid
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State UniversityColumbus, OH 43210, USA
| | - Jonathan Etter
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State UniversityColumbus, OH 43210, USA
| | - William L Berry
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Chenglong Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State UniversityColumbus, OH 43210, USA
| | - Michael A Ihnat
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
- Peggy and Charles Stephenson Cancer CenterOklahoma City, OK 73104, USA
| | - Pui-Kai Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State UniversityColumbus, OH 43210, USA
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
- Peggy and Charles Stephenson Cancer CenterOklahoma City, OK 73104, USA
| |
Collapse
|
31
|
Heidenreich B, Rachakonda PS, Hemminki K, Kumar R. TERT promoter mutations in cancer development. Curr Opin Genet Dev 2013; 24:30-7. [PMID: 24657534 DOI: 10.1016/j.gde.2013.11.005] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/18/2013] [Accepted: 11/03/2013] [Indexed: 12/20/2022]
Abstract
Human telomerase reverse transcriptase (TERT) encodes a rate-limiting catalytic subunit of telomerase that maintains genomic integrity. TERT expression is mostly repressed in somatic cells with exception of proliferative cells in self-renewing tissues and cancer. Immortality associated with cancer cells has been attributed to telomerase over-expression. The precise mechanism behind the TERT activation in cancers has mostly remained unknown. The newly described germline and recurrent somatic mutations in melanoma and other cancers in the TERT promoter that create de novo E-twenty six/ternary complex factors (Ets/TCF) binding sites, provide an insight into the possible cause of tumor-specific increased TERT expression. In this review we discuss the discovery and possible implications of the TERT promoter mutations in melanoma and other cancers.
Collapse
Affiliation(s)
- Barbara Heidenreich
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - P Sivaramakrishna Rachakonda
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany; Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany.
| |
Collapse
|
32
|
Holysz H, Lipinska N, Paszel-Jaworska A, Rubis B. Telomerase as a useful target in cancer fighting-the breast cancer case. Tumour Biol 2013; 34:1371-80. [PMID: 23558965 PMCID: PMC3661921 DOI: 10.1007/s13277-013-0757-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 03/20/2013] [Indexed: 12/17/2022] Open
Abstract
Telomerase was initially considered as a relevant factor distinguishing cancer from normal cells. During detailed studies, it appeared that its expression and activity is not only limited to cancer cells however, but in this particular cells, the telomerase is much more abundant. Thus, it has become a very promising target for an anticancer therapy. It was revealed in many studies that regulation of telomerase is a multifactorial process in mammalian cells, involving regulation of expression of telomerase subunits coding genes, post-translational protein–protein interactions, and protein phosphorylation. Numerous proto-oncogenes and tumor suppressor genes are engaged in this mechanism, and the complexity of telomerase control is studied in the context of tumor development as well as aging. Additionally, since numerous studies reveal a correlation between short telomeres and increased genome instability or cell mortality, the telomerase control appears to be one of the crucial factors to study in order to improve the cancer diagnostics and therapy or prevention. Interestingly, almost 100 % of adenocarcinoma, including breast cancer cells, expresses telomerase which makes it a good target for telomerase-related therapy. Additionally, telomerase is also supposed to be associated with drug resistance. Thus, targeting the enzyme might result in attenuation of this phenomenon. Moreover, since stem cells existence was reported, it must be considered whether targeting telomerase can bring some serious side effects and result in stem cells viability or their regenerative potential decrease. Thus, we review some molecular mechanisms engaged in therapy based on targeting telomerase in breast cancer cells.
Collapse
Affiliation(s)
- Hanna Holysz
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| | | | | | | |
Collapse
|
33
|
Xie L, Gazin C, Park SM, Zhu LJ, Debily MA, Kittler ELW, Zapp ML, Lapointe D, Gobeil S, Virbasius CM, Green MR. A synthetic interaction screen identifies factors selectively required for proliferation and TERT transcription in p53-deficient human cancer cells. PLoS Genet 2012; 8:e1003151. [PMID: 23284306 PMCID: PMC3527276 DOI: 10.1371/journal.pgen.1003151] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 10/23/2012] [Indexed: 01/01/2023] Open
Abstract
Numerous genetic and epigenetic alterations render cancer cells selectively dependent on specific genes and regulatory pathways, and represent potential vulnerabilities that can be therapeutically exploited. Here we describe an RNA interference (RNAi)–based synthetic interaction screen to identify genes preferentially required for proliferation of p53-deficient (p53−) human cancer cells. We find that compared to p53-competent (p53+) human cancer cell lines, diverse p53− human cancer cell lines are preferentially sensitive to loss of the transcription factor ETV1 and the DNA damage kinase ATR. In p53− cells, RNAi–mediated knockdown of ETV1 or ATR results in decreased expression of the telomerase catalytic subunit TERT leading to growth arrest, which can be reversed by ectopic TERT expression. Chromatin immunoprecipitation analysis reveals that ETV1 binds to a region downstream of the TERT transcriptional start-site in p53− but not p53+ cells. We find that the role of ATR is to phosphorylate and thereby stabilize ETV1. Our collective results identify a regulatory pathway involving ETV1, ATR, and TERT that is preferentially important for proliferation of diverse p53− cancer cells. The conversion of a normal cell into a cancer cell involves activating genes that promote cancer growth (oncogenes) and/or inactivating genes that normally act to inhibit cancer growth (tumor suppressor genes). The tumor suppressor gene p53 is the most frequently mutated gene in human cancers, being inactivated in approximately half of all tumors. In addition, loss of p53 function is often associated with increased resistance to chemotherapy and/or poor survival. For these reasons, the selective destruction of p53-deficient (p53−) tumors has remained one of the most important goals and challenges of cancer therapy. One strategy for destroying p53− tumors is to inactivate genes that are preferentially required for the growth or survival of p53− cells. Here we carry out a large-scale genetic screen to identify a cellular pathway that is preferentially required for growth of p53− cancer cells.
Collapse
Affiliation(s)
- Li Xie
- Howard Hughes Medical Institute, Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Claude Gazin
- Howard Hughes Medical Institute, Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- CEA/DSV/iRCM/LEFG, Genopole G2, Evry, France
- INSERM U967 and Université Paris Diderot, Evry, France
- * E-mail: (CG); (MRG)
| | - Sung Mi Park
- Howard Hughes Medical Institute, Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Lihua J. Zhu
- Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Marie-anne Debily
- CEA/DSV/iRCM/LEFG, Genopole G2, Evry, France
- Université d'Evry Val d'Essonne, Evry, France
| | - Ellen L. W. Kittler
- Program in Molecular Medicine and Center for AIDS Research, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Maria L. Zapp
- Program in Molecular Medicine and Center for AIDS Research, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - David Lapointe
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Stephane Gobeil
- Howard Hughes Medical Institute, Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ching-Man Virbasius
- Howard Hughes Medical Institute, Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Michael R. Green
- Howard Hughes Medical Institute, Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (CG); (MRG)
| |
Collapse
|
34
|
Kumar M, Witt B, Knippschild U, Koch S, Meena JK, Heinlein C, Weise JM, Krepulat F, Kuchenbauer F, Iben S, Rudolph KL, Deppert W, Günes C. CEBP factors regulate telomerase reverse transcriptase promoter activity in whey acidic protein-T mice during mammary carcinogenesis. Int J Cancer 2012; 132:2032-43. [PMID: 23023397 DOI: 10.1002/ijc.27880] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/12/2012] [Indexed: 01/17/2023]
Abstract
Telomerase is activated in the majority of invasive breast cancers, but the time point of telomerase activation during mammary carcinogenesis is not clear. We have recently presented a transgenic mouse model to study human telomerase reverse transcriptase (TERT) gene expression in vivo (hTERTp-lacZ). In the present study, hTERTp-lacZxWAP-T bitransgenic mice were generated to analyze the mechanisms responsible for human and mouse TERT upregulation during tumor progression in vivo. We found that telomerase activity and TERT expression were consistently upregulated in SV40-induced invasive mammary tumors compared to normal and hyperplastic tissues and ductal carcinoma in situ (DCIS). Human and mouse TERT genes are regulated similarly in the breast tissue, involving the CEBP transcription factors. Loss of CEBP-α and induction of CEBP-β expression correlated well with the activation of TERT expression in mouse mammary tumors. Transfection of CEBP-α into human or murine cells resulted in TERT repression, whereas knockdown of CEBP-α in primary human mammary epithelial cells resulted in reactivation of endogenous TERT expression and telomerase activity. Conversely, ectopic expression of CEBP-β activated endogenous TERT gene expression. Moreover, ChIP and EMSA experiments revealed binding of CEBP-α and CEBP-β to human TERT-promoter. This is the first evidence indicating that CEBP-α and CEBP-β are involved in TERT gene regulation during carcinogenesis.
Collapse
Affiliation(s)
- Mukesh Kumar
- Institute of Molecular Medicine and Max-Planck Research Group on Stem Cell Aging, Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Shin S, Oh S, An S, Janknecht R. ETS variant 1 regulates matrix metalloproteinase-7 transcription in LNCaP prostate cancer cells. Oncol Rep 2012; 29:306-14. [PMID: 23076342 DOI: 10.3892/or.2012.2079] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 08/30/2012] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer is characterized by the recurrent translocation of ETS transcription factors, including ETS variant 1 (ETV1) [also known as ETS-related 81 (ER81)]. Transgenic ETV1 mice develop prostatic intraepithelial neoplasia, yet the mechanisms by which ETV1 exerts its deleterious function remain largely unexplored. In this study, we demonstrated that ETV1 is capable of binding to the matrix metalloproteinase-7 (MMP-7) gene promoter both in vitro and in vivo. ETV1 stimulated the activity of the MMP-7 promoter, which was suppressed upon mutation of two ETV1 binding sites located within 200 base pairs upstream of the MMP-7 transcription start site. ETV1 overexpression in human LNCaP prostate cancer cells induced endogenous MMP-7 gene transcription, whereas ETV1 downregulation had the opposite effect. While MMP-7 overexpression did not influence LNCaP cell proliferation, it increased cell migration, which may be important during later stages of tumorigenesis. Finally, MMP-7 mRNA was significantly overexpressed in human prostate tumors compared to normal tissue. Together, these results showed that MMP-7 is a bona fide ETV1 target gene, implicating that MMP-7 upregulation is partially responsible for the oncogenic effects of ETV1 in the prostate.
Collapse
Affiliation(s)
- Sook Shin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | |
Collapse
|
36
|
Kim TD, Shin S, Berry WL, Oh S, Janknecht R. The JMJD2A demethylase regulates apoptosis and proliferation in colon cancer cells. J Cell Biochem 2012; 113:1368-76. [PMID: 22134899 DOI: 10.1002/jcb.24009] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
JMJD2A is a transcriptional cofactor and enzyme that catalyzes demethylation of histone H3 lysines 9 and 36 and is overexpressed in human tumors, but its role in oncogenesis remains unclear. Here, we show that JMJD2A interacts with the tumor suppressor p53 both in vitro and in HCT116 colon cancer cells. Chromatin immunoprecipitation assays demonstrated that JMJD2A was recruited together with p53 to the promoter of the p21 cell cycle inhibitor upon stimulation with the DNA damaging agent, adriamycin. Downregulation of JMJD2A resulted in increased expression of p21 and of the pro-apoptotic Puma protein, whereas levels of the anti-apoptotic Bcl-2 protein were decreased. Furthermore, JMJD2A knock-down led to reduced HCT116, DLD-1 and HT-29 colon cancer cell proliferation, while overexpression of JMJD2A enhanced HCT116 proliferation in low serum media. Finally, JMJD2A depletion induced apoptosis in HCT116 cells and this effect was less pronounced in the absence of p53. Collectively, these data indicate that JMJD2A is a novel promoter of colon cancer cell proliferation and survival, which mediates its effects in p53-dependent and -independent ways. JMJD2A may therefore be a valid target to sensitize tumor cells to chemotherapy-induced cell death and growth suppression.
Collapse
Affiliation(s)
- Tae-Dong Kim
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | |
Collapse
|
37
|
Zhang YL, Hua YW, Zhang ZD, Ma F, Xu JL. HER-2 amplification and overexpression in primary tumors and paired lymph node metastases of gastric cancer. Shijie Huaren Xiaohua Zazhi 2012; 20:2485-2490. [DOI: 10.11569/wcjd.v20.i26.2485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression and amplification status of HER-2 in primary gastric cancer and lymph node metastases and to analyze their association with clinicopathologic features of gastric cancer.
METHODS: The expression and amplification status of HER-2 in primary tumors (n = 180) and metastatic lymph nodes (n = 112) of gastric cancer were detected by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH). Their association with clinicopathologic features of gastric cancer was analyzed using SPSS17.0.
RESULTS: The positive rate of HER-2 expression in primary tumors and paired lymph node metastases was 13.9% and 20.5%, respectively, with no significant difference between the two groups (P > 0.05). In primary tumors and metastatic lymph nodes, the concordance between HER-2 positivity by IHC and FISH was 84.0% and 82.6%, respectively. There were 6 cases with discordant HER-2 results between primary tumors and matched lymph node metastases, of which 3 were negative for HER-2 overexpression and 2 were negative for HER-2 amplification in primary lesions but showed positive conversion in metastatic lymph nodes. No cases showed negative conversion during the metastatic process. The positive rate of HER-2 was significantly associated with clinical stage, Lauren type, lymph node metastasis, depth of invasion, and degree of differentiation (all P < 0.05).
CONCLUSION: There was a high concordance between IHC and FISH results. HER-2 overexpression is correlated with invasion and metastasis of gastric cancer. HER-2 overexpression and amplification in metastatic sites have heterogeneity compared to primary tumors. HER-2 status should be assessed in metastatic sites if primary tumors are tested negative for HER-2.
Collapse
|
38
|
Shoda J, Ishige K, Sugiyama H, Kawamoto T. Biliary tract carcinoma: clinical perspectives on molecular targeting strategies for therapeutic options. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2012; 19:342-53. [DOI: 10.1007/s00534-012-0520-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Junichi Shoda
- Field of Basic Sports Medicine, Sports Medicine; University of Tsukuba Graduate School of Comprehensive Human Sciences; Tsukuba-shi Ibaraki Japan
- Department of Gastroenterology, Institute of Clinical Medicine; University of Tsukuba Graduate School of Comprehensive Human Sciences; Tsukuba-shi Ibaraki Japan
| | - Kazunori Ishige
- Department of Gastroenterology, Institute of Clinical Medicine; University of Tsukuba Graduate School of Comprehensive Human Sciences; Tsukuba-shi Ibaraki Japan
| | - Hiroaki Sugiyama
- Department of Gastroenterology, Institute of Clinical Medicine; University of Tsukuba Graduate School of Comprehensive Human Sciences; Tsukuba-shi Ibaraki Japan
| | - Toru Kawamoto
- Department of Surgery, Institute of Gastroenterology; Tokyo Women's Medical University; Shinjuku-ku Tokyo Japan
| |
Collapse
|
39
|
Chen Y, Zou H, Yang LY, Li Y, Wang L, Hao Y, Yang JL. ER81-shRNA Inhibits Growth of Triple-negative Human Breast Cancer Cell Line MDA-MB-231 In Vivo and in Vitro. Asian Pac J Cancer Prev 2012; 13:2385-92. [DOI: 10.7314/apjcp.2012.13.5.2385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
40
|
DiTacchio L, Bowles J, Shin S, Lim DS, Koopman P, Janknecht R. Transcription factors ER71/ETV2 and SOX9 participate in a positive feedback loop in fetal and adult mouse testis. J Biol Chem 2012; 287:23657-66. [PMID: 22613723 DOI: 10.1074/jbc.m111.320101] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
ER71, also known as ETV2, is an ETS transcription factor that is expressed during embryogenesis and in adult testes. We show that Er71 transcription can be up-regulated by SRY, the key determinant of male differentiation. Accordingly, SRY bound to and activated the Er71 promoter, and mutation of a putative SRY binding site abolished this promoter activation. In turn, ER71 was able to bind to the promoter of Sox9, the primary target of SRY and a critical transcription factor for maintenance of the Sertoli cell phenotype. Mutation of the ER71 binding site in the Sox9 promoter suppressed ER71-dependent up-regulation of Sox9 transcription, and a dominant-negative ER71 molecule severely reduced Sox9 transcription in a Sertoli cell line. Conversely, SOX9 bound the Er71 promoter in vivo and Sox9 down-regulation reduced Er71 transcript levels. Together, these data suggest a mechanism by which SRY induces Sox9 and Er71 transcription early in testis differentiation, whereas ER71 and SOX9 participate in an autoregulatory loop to sustain each other's expression after Sry expression has subsided in mice. Thereby, ER71 and SOX9 may affect late testis development as well as the function of the adult male gonad.
Collapse
|
41
|
Regulation of tumor suppressor p53 and HCT116 cell physiology by histone demethylase JMJD2D/KDM4D. PLoS One 2012; 7:e34618. [PMID: 22514644 PMCID: PMC3326058 DOI: 10.1371/journal.pone.0034618] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 03/02/2012] [Indexed: 12/12/2022] Open
Abstract
JMJD2D, also known as KDM4D, is a histone demethylase that removes methyl moieties from lysine 9 on histone 3 and from lysine 26 on histone 1.4. Here, we demonstrate that JMJD2D forms a complex with the p53 tumor suppressor in vivo and interacts with the DNA binding domain of p53 in vitro. A luciferase reporter plasmid driven by the promoter of p21, a cell cycle inhibitor and prominent target gene of p53, was synergistically activated by p53 and JMJD2D, which was dependent on JMJD2D catalytic activity. Likewise, overexpression of JMJD2D induced p21 expression in U2OS osteosarcoma cells in the absence and presence of adriamycin, an agent that induces DNA damage. Furthermore, downregulation of JMJD2D inhibited cell proliferation in wild-type and even more so in p53−/− HCT116 colon cancer cells, suggesting that JMJD2D is a pro-proliferative molecule. JMJD2D depletion also induced more strongly apoptosis in p53−/− compared to wild-type HCT116 cells. Collectively, our results demonstrate that JMJD2D can stimulate cell proliferation and survival, suggesting that its inhibition may be helpful in the fight against cancer. Furthermore, our data imply that activation of p53 may represent a mechanism by which the pro-oncogenic functions of JMJD2D become dampened.
Collapse
|
42
|
Design, synthesis and evaluation of telomerase inhibitory, hTERT repressing, and anti-proliferation activities of symmetrical 1,8-disubstituted amidoanthraquinones. Eur J Med Chem 2012; 50:102-12. [DOI: 10.1016/j.ejmech.2012.01.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 01/19/2012] [Accepted: 01/22/2012] [Indexed: 11/22/2022]
|
43
|
Abstract
Telomeres are stretches of repeated DNA sequences located at the ends of chromosomes that are necessary to prevent loss of gene-coding DNA regions during replication. Telomerase – the enzyme responsible for immortalising cancer cells through the addition of telomeric repeats – is active in ~90% of human cancers. Telomerase activity is inhibited by various phytochemicals such as isoprenoids, genistein, curcumin, epigallocatechin-3-gallate, resveratrol and others. Human TERT (telomerase reverse transcriptase – the rate-limiting component of telomerase), heat shock protein 90, Akt, p70 S6 kinase (S6K) and mammalian target of rapamycin (mTOR) form a physical and functional complex with one another. The inclusion of Akt, mTOR and S6K in the TERT complex is compelling evidence to support mTOR-mediated control of telomerase activity. This review will define the role of mTOR, the master regulator of protein translation, in telomerase regulation and provide additional insights into the numerous ways in which telomerase activity is hindered by phytochemicals.
Collapse
|
44
|
Oh S, Shin S, Janknecht R. ETV1, 4 and 5: an oncogenic subfamily of ETS transcription factors. Biochim Biophys Acta Rev Cancer 2012; 1826:1-12. [PMID: 22425584 DOI: 10.1016/j.bbcan.2012.02.002] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 02/26/2012] [Accepted: 02/27/2012] [Indexed: 12/30/2022]
Abstract
The homologous ETV1, ETV4 and ETV5 proteins form the PEA3 subfamily of ETS transcription factors. In Ewing tumors, chromosomal translocations affecting ETV1 or ETV4 are an underlying cause of carcinogenesis. Likewise, chromosomal rearrangements of the ETV1, ETV4 or ETV5 gene occur in prostate tumors and are thought to be one of the major driving forces in the genesis of prostate cancer. In addition, these three ETS proteins are implicated in melanomas, breast and other types of cancer. Complex posttranslational modifications govern the activity of PEA3 factors, which can promote cell proliferation, motility and invasion. Here, we review evidence for a role of ETV1, 4 and 5 as oncoproteins and describe modes of their action. Modulation of their activation or interaction with cofactors as well as inhibiting crucial target gene products may ultimately be exploited to treat various cancers that are dependent on the PEA3 group of ETS transcription factors.
Collapse
Affiliation(s)
- Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | |
Collapse
|
45
|
Matsuo T, Shimose S, Kubo T, Fujimori J, Yasunaga Y, Sugita T, Ochi M. Correlation between p38 mitogen-activated protein kinase and human telomerase reverse transcriptase in sarcomas. J Exp Clin Cancer Res 2012; 31:5. [PMID: 22243975 PMCID: PMC3296589 DOI: 10.1186/1756-9966-31-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 01/16/2012] [Indexed: 11/27/2022] Open
Abstract
Background One of the major components of telomerase is the human telomerase reverse transcriptase (hTERT) as the catalytic protein. hTERT mRNA expression are reported to be associated with prognosis and tumor progression in several sarcomas. However, there is no clear understanding of the mechanisms of hTERT in human sarcomas. Recent studies have suggested that signals transmitted through p38 mitogen-activated protein kinase (MAPK) can increase or decrease hTERT transcription in human cells. The purpose of this study was to analyse the correlation between p38 MAPK and hTERT in sarcoma samples. Methods We investigated 36 soft tissue malignant fibrous histiocytomas (MFH), 24 liposarcomas (LS) and 9 bone MFH samples for hTERT and p38 MAPK expression. Quantitative detection of hTERT and p38 MAPK was performed by RT-PCR. Results There was a significant positive correlation between the values of hTERT and p38 MAPK in all samples (r = 0.445, p = 0.0001), soft tissue MFH (r = 0.352, p = 0.0352), LS (r = 0.704, p = 0.0001) and bone MFH samples (r = 0.802, p = 0.0093). Patients who had a higher than average expression of p38 MAPK had a significantly worse prognosis than other patients (p = 0.0036). Conclusions p38 MAPK may play a role in up-regulation of hTERT, and therefore, p38 MAPK may be a useful marker in the assessment of hTERT and patients' prognosis in sarcomas.
Collapse
Affiliation(s)
- Toshihiro Matsuo
- Department of Orthopaedic Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center: 3-1, Aoyamacho, Kure, Hiroshima, 7370023 Japan.
| | | | | | | | | | | | | |
Collapse
|
46
|
Synthesis, antiproliferative activities and telomerase inhibition evaluation of novel asymmetrical 1,2-disubstituted amidoanthraquinone derivatives. Eur J Med Chem 2012; 47:323-36. [DOI: 10.1016/j.ejmech.2011.10.059] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 10/26/2011] [Accepted: 10/31/2011] [Indexed: 11/24/2022]
|
47
|
Prescott JD, Poczobutt JM, Tentler JJ, Walker DM, Gutierrez-Hartmann A. Mapping of ESE-1 subdomains required to initiate mammary epithelial cell transformation via a cytoplasmic mechanism. Mol Cancer 2011; 10:103. [PMID: 21871131 PMCID: PMC3183030 DOI: 10.1186/1476-4598-10-103] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Accepted: 08/28/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The ETS family transcription factor ESE-1 is often overexpressed in human breast cancer. ESE-1 initiates transformation of MCF-12A cells via a non-transcriptional, cytoplasmic process that is mediated by a unique 40-amino acid serine and aspartic acid rich (SAR) subdomain, whereas, ESE-1's nuclear transcriptional property is required to maintain the transformed phenotype of MCF7, ZR-75-1 and T47D breast cancer cells. RESULTS To map the minimal functional nuclear localization (NLS) and nuclear export (NES) signals, we fused in-frame putative NLS and NES motifs between GFP and the SAR domain. Using these GFP constructs as reporters of subcellular localization, we mapped a single NLS to six basic amino acids (242 HGKRRR 247) in the AT-hook and two CRM1-dependent NES motifs, one to the pointed domain (NES1: 102 LCNCALEELRL 112) and another to the DNA binding domain (DBD), (NES2: 275 LWEFIRDILI 284). Moreover, analysis of a putative NLS located in the DBD (316 GQKKKNSN 323) by a similar GFP-SAR reporter or by internal deletion of the DBD, revealed this sequence to lack NLS activity. To assess the role of NES2 in regulating ESE-1 subcellular localization and subsequent transformation potency, we site-specifically mutagenized NES2, within full-length GFP-ESE-1 and GFP-NES2-SAR reporter constructs. These studies show that site-specific mutation of NES2 completely abrogates ESE-1 transforming activity. Furthermore, we show that exclusive cytoplasmic targeting of the SAR domain is sufficient to initiate transformation, and we report that an intact SAR domain is required, since block mutagenesis reveals that an intact SAR domain is necessary to maintain its full transforming potency. Finally, using a monoclonal antibody targeting the SAR domain, we demonstrate that the SAR domain contains a region accessible for protein - protein interactions. CONCLUSIONS These data highlight that ESE-1 contains NLS and NES signals that play a critical role in regulating its subcellular localization and function, and that an intact SAR domain mediates MEC transformation exclusively in the cytoplasm, via a novel nontranscriptional mechanism, whereby the SAR motif is accessible for ligand and/or protein interactions. These findings are significant, since they provide novel molecular insights into the functions of ETS transcription factors in mammary cell transformation.
Collapse
Affiliation(s)
- Jason D Prescott
- Medical Scientist Training Program, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | |
Collapse
|
48
|
Papanikolaou V, Athanassiou E, Dubos S, Dimou I, Papathanasiou I, Kitsiou-Tzeli S, Kappas C, Tsezou A. hTERT regulation by NF-κB and c-myc in irradiated HER2-positive breast cancer cells. Int J Radiat Biol 2011; 87:609-21. [PMID: 21627565 DOI: 10.3109/09553002.2011.572112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE Telomerase activity (TA), frequently observed in cancer, compensates for telomere shortening thus preventing cell senescence and conferring resistance to therapy. In the present study, we investigated the expression of human telomerase reverse transcriptase (hTERT) and TA and their regulation, as well as apoptotic rates and correlation with the presence of human epidermal growth factor receptor 2 (HER2), in irradiated tumour-derived breast cancer cells. MATERIALS AND METHODS In 50 breast cancer tissue samples hTERT mRNA expression and TA were correlated with cell features (HER2, Estrogen and Progesterone Receptor status). Cells from six samples were then irradiated with 10 and 20 Gy; apoptotic rates were measured by flow cytometry, hTERT mRNA expression by real-time polymerase chain reaction and TA by telomeric repeat amplification protocol assay, at 24-144 h post-irradiation. Chromatin immunoprecipitation was performed to investigate hTERT and cellular-myelocytomatosis (c-myc) promoters' activity. HER2 gene knockdown was performed using small interfering RNA technology. RESULTS hTERT/TA were found increased only in irradiated HER2-positive cells, which were found to be more radioresistant, while HER2 knockdown led to hTERT/TA downregulation. HER2 was found to mediate hTERT expression through activation of Nuclear Factor-kappa B (NF-κB) and c-myc. CONCLUSIONS The present study suggests that following irradiation, HER2 receptor activates hTERT/telomerase, increasing the breast cancer cells' survival potential, through sequential induction of transcription factors NF-κΒ and c-myc.
Collapse
|
49
|
Gladych M, Wojtyla A, Rubis B. Human telomerase expression regulation. Biochem Cell Biol 2011; 89:359-76. [DOI: 10.1139/o11-037] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Since telomerase has been recognized as a relevant factor distinguishing cancer cells from normal cells, it has become a very promising target for anti-cancer therapy. A correlation between short telomere length and increased mortality was revealed in many studies. The telomerase expression/activity appears to be one of the most crucial factors to study to improve cancer therapy and prevention. However, this multisubunit enzymatic complex can be regulated at various levels. Thus, several strategies have been proposed to control telomerase in cancer cells such as anti-sense technology against TR and TERT, ribozymes against TERT, anti-estrogens, progesterone, vitamin D, retinoic acid, quadruplex stabilizers, telomere and telomerase targeting agents, modulation of interaction with other proteins involved in the regulation of telomerase and telomeres, etc. However, the transcription control of key telomerase subunits seems to play the crucial role in whole complexes activity and cancer cells immortality. Thus, the research of telomerase regulation can bring significant insight into the knowledge concerning stem cells metabolism but also ageing. This review summarizes the current state of knowledge of numerous telomerase regulation mechanisms at the transcription level in human that might become attractive anti-cancer therapy targets.
Collapse
Affiliation(s)
- Marta Gladych
- Poznan University of Medical Sciences, Department of Clinical Chemistry and Molecular Diagnostics, Przybyszewskiego 49 St., 60-355 Poznan, Poland
| | - Aneta Wojtyla
- Poznan University of Medical Sciences, Department of Clinical Chemistry and Molecular Diagnostics, Przybyszewskiego 49 St., 60-355 Poznan, Poland
| | - Blazej Rubis
- Poznan University of Medical Sciences, Department of Clinical Chemistry and Molecular Diagnostics, Przybyszewskiego 49 St., 60-355 Poznan, Poland
| |
Collapse
|
50
|
Horikawa I, Michishita E, Barrett JC. Regulation of hTERT transcription: a target of cellular and viral mechanisms for immortalization and carcinogenesis. Cytotechnology 2011; 45:23-32. [PMID: 19003240 DOI: 10.1007/s10616-004-5122-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2004] [Accepted: 09/21/2004] [Indexed: 01/30/2023] Open
Abstract
A hallmark of human cancer cells is immortal cell growth, which is associated with telomere maintenance by telomerase. The transcriptional regulation of the human telomerase reverse transcriptase (hTERT) gene is a major mechanism that negatively and positively controls telomerase activity in normal and cancer cells, respectively. A growing body of data suggests that various cellular and viral factors and pathways involved in cell senescence, immortalization and carcinogenesis act on the hTERT promoter. The activity of the hTERT promoter is regulated, either directly or through signaling pathways, by oncogene products (e.g., Myc and Ets families) and tumor suppressor proteins (e.g., BRCA1). Endogenous factors involved in the physiological repression of the hTERT gene have also been revealed by chromosome transfer experiments. The integration of viral genomes in the hTERT locus can lead to hTERT activation and telomerase induction. Here, we summarize these findings and pay special attention to recent findings with relevance to the endogenous regulatory mechanisms of hTERT transcription.
Collapse
Affiliation(s)
- Izumi Horikawa
- Laboratory of Biosystems and Cancer, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 37, Room 5046, Bethesda, MD, 20892, USA,
| | | | | |
Collapse
|