1
|
Cao X, Fang L, Jiang Y, Zeng T, Bai S, Li S, Liu Y, Zhong W, Lu C, Yang H. Nanoscale octopus guiding telomere entanglement: An innovative strategy for inducing apoptosis in cancer cells. Biomaterials 2025; 313:122777. [PMID: 39222545 DOI: 10.1016/j.biomaterials.2024.122777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/02/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Telomere length plays a crucial role in cellular aging and the risk of diseases. Unlike normal cells, cancer cells can extend their own survival by maintaining telomere stability through telomere maintenance mechanism. Therefore, regulating the lengths of telomeres have emerged as a promising approach for anti-cancer treatment. In this study, we introduce a nanoscale octopus-like structure designed to induce physical entangling of telomere, thereby efficiently triggering telomere dysfunction. The nanoscale octopus, composed of eight-armed PEG (8-arm-PEG), are functionalized with cell penetrating peptide (TAT) to facilitate nuclear entry and are covalently bound to N-Methyl Mesoporphyrin IX (NMM) to target G-quadruplexes (G4s) present in telomeres. The multi-armed configuration of the nanoscale octopus enables targeted binding to multiple G4s, physically disrupting and entangling numerous telomeres, thereby triggering telomere dysfunction. Both in vitro and in vivo experiments indicate that the nanoscale octopus significantly inhibits cancer cell proliferation, induces apoptosis through telomere entanglement, and ultimately suppresses tumor growth. This research offers a novel perspective for the development of innovative anti-cancer interventions and provides potential therapeutic options for targeting telomeres.
Collapse
Affiliation(s)
- Xiuping Cao
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Liyang Fang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Yifan Jiang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Tao Zeng
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Shiyan Bai
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Shiqing Li
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Yana Liu
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Wukun Zhong
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Chunhua Lu
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China.
| | - Huanghao Yang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, People's Republic of China.
| |
Collapse
|
2
|
Lyu Y, Zhao H, Zeng G, Yang J, Shao Q, Wu H. Mapping the evolving trend of research on leukocyte telomere length: a text-mining study. Hum Genomics 2024; 18:117. [PMID: 39468654 PMCID: PMC11520877 DOI: 10.1186/s40246-024-00687-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Substantial evidence indicates that measuring leukocyte telomere length (LTL) is a useful tool that may be considered as a valuable biomarker of individual biological age, correlating with numerous chronic disorders. However, to date, there has been a lack of in-depth understanding regarding the current landscape and forthcoming developments in the LTL field. Therefore, this study aimed to utilize bibliometric methods to summarize the knowledge structure, current focus, and emerging directions in this field. METHOD Scientific publications on LTL spanning the period from 2000 to 2022 were acquired from the Web of Science Core Collection database. Several bibliometric tools including CiteSpace, VOSviewer, and an online website were utilized for bibliometric analysis. The primary evaluations encompassed investigating the major contributors and their collaborative relationships among countries/regions, institutions, and authors, conducting co-citation analyses of authors, journals, as well as reference, examining reference bursts, as well as performing co-occurrence analyses of keywords. RESULTS There are 1818 papers with 66,668 citations identified. Both the annual publication and citation counts on LTL exhibited significant upward trends. The United States emerged as the most prominent contributor, as evidenced by the greatest volume of papers and the highest H-index value. University of California San Francisco and Aviv A were identified as the most productive institution and author in this domain, respectively. Reference analysis revealed that longitudinal study and mendelian randomization study are the most concerned research method in this field recently. Keywords analysis showed that the most concerned diseases in LTL fields were aging, inflammation, cardiovascular diseases, endocrine diseases, neurological and psychiatric diseases, and cancers. In addition, the following research directions such as "COPD", "mendelian randomization", "adiposity", "colorectal cancer", "National Health and Nutrition Examination Survey (NHNES)", "telomerase reverse transcriptase", "pregnancy" have garnered increasing attention in recent times and hold the potential to evolve into research foci in the foreseeable future. CONCLUSION This is the first bibliometric study that provides comprehensive overview of LTL research. The findings of this study could become valuable references for investigators to explore and address the current and emerging challenges in LTL research.
Collapse
Affiliation(s)
- Yuanjun Lyu
- Department of Geriatric Respiratory and Sleep, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Hongjie Zhao
- Department of Oncology, Tianjin Medical University Baodi Hospital, Tianjin, China
| | - Guiping Zeng
- Department of Orthopaedic Surgery, Yangxin People's Hospital, Yangxin, 435200, Hubei, China
| | - Jia Yang
- Department of Orthopaedics, Jincheng General Hospital, Jincheng, 048006, Shanxi Province, China
| | - Qipeng Shao
- Department of Orthopaedics, Ganzhou People's Hospital, Ganzhou, China.
| | - Haiyang Wu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, China.
| |
Collapse
|
3
|
Chen Y, Chen H, Chen X, Zhang Y, Tu W, Mo J, Tian S, Sun C, Meng X, Wang Z, Yang J, Luo Y. Precision detection of hepatocellular carcinoma-associated telomerase RNA with SA@Comb-HCR nanosystem. Biosens Bioelectron 2024; 261:116496. [PMID: 38875865 DOI: 10.1016/j.bios.2024.116496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Accurate intracellular visualization of human telomerase RNA (hTR) is imperative for early diagnosis and treatment monitoring of hepatocellular carcinoma (HCC). While isothermal amplification-based DNA cascade strategies are promising, challenges persist in achieving great intake efficiency of detection probes within tumor cells and enhancing intracellular reaction efficiency. This study introduces a SA@Comb-HCR nanosystem, a highly effective approach for in situ hTR detection in HCC cells. Sodium alginate-coated liposomes ensures efficient nanoprobe delivery, which are then combined with proximity effect-inspired signal amplification. The coating of sodium alginate facilitates receptor-mediated endocytosis, prevents serum protein adhesion, and mitigates cationic liposome cytotoxicity. The designed Comb-like consolidated hairpin probe enhances the concentration of the local reactant, resulting in cascade amplification upon hTR activation. This technique achieves precision detection of intracellularly overexpressed hTR in HCC cells with a remarkable detection limit of 0.7 pM. This approach holds great promise for advancing targeted and sensitive early clinical diagnosis of HCC.
Collapse
Affiliation(s)
- Yi Chen
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Hengyi Chen
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Xiaohui Chen
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Yufang Zhang
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Wei Tu
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Jiaxi Mo
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, 646000, PR China
| | - Shanshan Tian
- Pre-hospital Emergency Department, Chongqing Emergency Medical Center, Chongqing University Central Hospital, No. 1 Health Road, Yuzhong District, Chongqing, 400014, PR China
| | - Chenyu Sun
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Xiaoqin Meng
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Zhizeng Wang
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China.
| | - Jichun Yang
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China.
| | - Yang Luo
- NHC Key Laboratory of Birth Defects and Reproductive Health, Center of Smart Laboratory and Molecular Medicine, Fuling Hospital, School of Medicine, Chongqing University, Chongqing, 400044, PR China; College of Life Science and Laboratory Medicine, Kunming Medical University, Kunming, Yunnan, 650050, PR China.
| |
Collapse
|
4
|
Ao Z, Xiao D, Wu J, Sun J, Liu H. CRL4DCAF4 E3 ligase-mediated degradation of MEN1 transcriptionally reactivates hTERT to sustain immortalization in colorectal cancer cells. Carcinogenesis 2024; 45:607-619. [PMID: 38573327 DOI: 10.1093/carcin/bgae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/08/2024] [Accepted: 04/03/2024] [Indexed: 04/05/2024] Open
Abstract
Telomerase reactivation is implicated in approximately 85% of human cancers, yet its underlying mechanism remains elusive. In this study, we elucidate that the cullin-RING ubiquitin ligase 4 (CRL4) complex drives the reactivation of human telomerase reverse transcriptase (hTERT) in colorectal cancer (CRC) by degrading the tumor suppressor, menin 1 (MEN1). Our data show that, in noncancerous intestinal epithelial cells, the transcription factor specificity protein 1 (Sp1) recruits both the histone acetyltransferase p300 and MEN1 to suppress hTERT expression, thus maintaining telomere shortness post-cell division. Inflammation-induced microenvironments trigger an activation of the CRL4DCAF4 E3 ligase, leading to MEN1 ubiquitination and degradation in CRC cells. This process nullifies MEN1's inhibitory action, reactivates hTERT expression at the transcriptional level, interrupts telomere shortening and spurs uncontrolled cellular proliferation. Notably, MEN1 overexpression in CRC cells partially counteracts these oncogenic phenotypes. NSC1517, an inhibitor of the CRL4DCAF4 complex identified through high-throughput screening from a plant-derived chemical pool, hinders MEN1 degradation, attenuates hTERT expression and suppresses tumor growth in mouse xenograft models. Collectively, our research elucidates the transcriptional mechanism driving hTERT reactivation in CRC. Targeting the CRL4DCAF4 E3 ligase emerges as a promising strategy to counteract cancer cell immortalization and curb tumor progression.
Collapse
Affiliation(s)
- Zhimin Ao
- Division of Surgery, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Dan Xiao
- Division of Surgery, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jing Wu
- Division of Surgery, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Ji Sun
- Division of Surgery, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Liu
- Division of Surgery, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Zuo H, Ru Y, Gao X, Chen H, Yan Y, Ma X, Liu X, Wang Y. Small Molecules Blocking the Assembly of TCAB1 and Telomerase Complexes: Lead Discovery and Biological Activity. ACS Med Chem Lett 2024; 15:1205-1212. [PMID: 39140071 PMCID: PMC11318020 DOI: 10.1021/acsmedchemlett.4c00310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024] Open
Abstract
The vast majority of tumor cells maintain the length of the telomeres through a telomerase-dependent maintenance mechanism, allowing for unlimited proliferation. TCAB1 is indispensable for the correct assembly of telomerase complexes and the delivery of telomerase to the telomere. Therefore, this study aimed to explore small molecules capable of interfering with the assembly of TCAB1 and the telomerase complex as novel efficient telomerase inhibitors. Through virtual screening, biological evaluation, and the confirmation of target engagement, the potential ligands of TCAB1 effectively inhibiting telomerase activity were discovered. Among them, compound 9 exhibited telomerase inhibitory activity at a two-digit nanomolar level (IC50 = 0.03 μM), which was dramatically enhanced in comparison with the previously reported telomerase inhibitors. This research, based on the blockage of telomerase assembly through disturbing TCAB1, provides a novel strategy and a potential target for telomerase inhibitor discovery.
Collapse
Affiliation(s)
- Haojie Zuo
- School
of Pharmacy, Anhui University of Chinese
Medicine, Hefei 230012, China
| | - Yiming Ru
- School
of Pharmacy, Anhui University of Chinese
Medicine, Hefei 230012, China
| | - Xiuxiu Gao
- School
of Pharmacy, Anhui University of Chinese
Medicine, Hefei 230012, China
| | - Hui Chen
- School
of Pharmacy, Anhui University of Chinese
Medicine, Hefei 230012, China
| | - Yaoyao Yan
- School
of Pharmacy, Anhui University of Chinese
Medicine, Hefei 230012, China
| | - Xiaodong Ma
- School
of Pharmacy, Anhui University of Chinese
Medicine, Hefei 230012, China
- Department
of Medicinal Chemistry, Anhui Academy of
Chinese Medicine, Hefei 230012, China
| | - Xinhua Liu
- School
of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yang Wang
- School
of Pharmacy, Anhui University of Chinese
Medicine, Hefei 230012, China
- Department
of Medicinal Chemistry, Anhui Academy of
Chinese Medicine, Hefei 230012, China
| |
Collapse
|
6
|
Boccardi V, Marano L. Aging, Cancer, and Inflammation: The Telomerase Connection. Int J Mol Sci 2024; 25:8542. [PMID: 39126110 PMCID: PMC11313618 DOI: 10.3390/ijms25158542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024] Open
Abstract
Understanding the complex dynamics of telomere biology is important in the strong link between aging and cancer. Telomeres, the protective caps at the end of chromosomes, are central players in this connection. While their gradual shortening due to replication limits tumors expansion by triggering DNA repair mechanisms, it also promotes oncogenic changes within chromosomes, thus sustaining tumorigenesis. The enzyme telomerase, responsible for maintaining telomere length, emerges as a central player in this context. Its expression in cancer cells facilitates the preservation of telomeres, allowing them to circumvent the growth-limiting effects of short telomeres. Interestingly, the influence of telomerase extends beyond telomere maintenance, as evidenced by its involvement in promoting cell growth through alternative pathways. In this context, inflammation accelerates telomere shortening, resulting in telomere dysfunction, while telomere elements also play a role in modulating the inflammatory response. The recognition of this interplay has promoted the development of novel therapeutic approaches centered around telomerase inhibition. This review provides a comprehensive overview of the field, emphasizing recent progress in knowledge and the implications in understanding of cancer biology.
Collapse
Affiliation(s)
- Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Luigi Marano
- Department of Medicine, Academy of Applied Medical and Social Sciences—AMiSNS: Akademia Medycznych I Spolecznych Nauk Stosowanych, 82-300 Elbląg, Poland;
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland
| |
Collapse
|
7
|
Shan L, Li Y, Ma Y, Yang Y, Wang J, Peng L, Wang W, Zhao F, Li W, Chen X. Hairpin DNA-Based Nanomaterials for Tumor Targeting and Synergistic Therapy. Int J Nanomedicine 2024; 19:5781-5792. [PMID: 38882546 PMCID: PMC11180469 DOI: 10.2147/ijn.s461774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/29/2024] [Indexed: 06/18/2024] Open
Abstract
Background While nanoplatform-based cancer theranostics have been researched and investigated for many years, enhancing antitumor efficacy and reducing toxic side effects is still an essential problem. Methods We exploited nanoparticle coordination between ferric (Fe2+) ions and telomerase-targeting hairpin DNA structures to encapsulate doxorubicin (DOX) and fabricated Fe2+-DNA@DOX nanoparticles (BDDF NPs). This work studied the NIR fluorescence imaging and pharmacokinetic studies targeting the ability and biodistribution of BDDF NPs. In vitro and vivo studies investigated the nano formula's toxicity, imaging, and synergistic therapeutic effects. Results The enhanced permeability and retention (EPR) effect and tumor targeting resulted in prolonged blood circulation times and high tumor accumulation. Significantly, BDDF NPs could reduce DOX-mediated cardiac toxicity by improving the antioxidation ability of cardiomyocytes based on the different telomerase activities and iron dependency in normal and tumor cells. The synergistic treatment efficacy is enhanced through Fe2+-mediated ferroptosis and the β-catenin/p53 pathway and improved the tumor inhibition rate. Conclusion Harpin DNA-based nanoplatforms demonstrated prolonged blood circulation, tumor drug accumulation via telomerase-targeting, and synergistic therapy to improve antitumor drug efficacy. Our work sheds new light on nanomaterials for future synergistic chemotherapy.
Collapse
Affiliation(s)
- Lingling Shan
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Yudie Li
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Yifan Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Yang Yang
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Jing Wang
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Lei Peng
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Weiwei Wang
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Fang Zhao
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Wanrong Li
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
8
|
Li Y, Tian X, Luo J, Bao T, Wang S, Wu X. Molecular mechanisms of aging and anti-aging strategies. Cell Commun Signal 2024; 22:285. [PMID: 38790068 PMCID: PMC11118732 DOI: 10.1186/s12964-024-01663-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Aging is a complex and multifaceted process involving a variety of interrelated molecular mechanisms and cellular systems. Phenotypically, the biological aging process is accompanied by a gradual loss of cellular function and the systemic deterioration of multiple tissues, resulting in susceptibility to aging-related diseases. Emerging evidence suggests that aging is closely associated with telomere attrition, DNA damage, mitochondrial dysfunction, loss of nicotinamide adenine dinucleotide levels, impaired macro-autophagy, stem cell exhaustion, inflammation, loss of protein balance, deregulated nutrient sensing, altered intercellular communication, and dysbiosis. These age-related changes may be alleviated by intervention strategies, such as calorie restriction, improved sleep quality, enhanced physical activity, and targeted longevity genes. In this review, we summarise the key historical progress in the exploration of important causes of aging and anti-aging strategies in recent decades, which provides a basis for further understanding of the reversibility of aging phenotypes, the application prospect of synthetic biotechnology in anti-aging therapy is also prospected.
Collapse
Affiliation(s)
- Yumeng Li
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Xutong Tian
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Juyue Luo
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Tongtong Bao
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Shujin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xin Wu
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China.
| |
Collapse
|
9
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Pang H, Peng Y, Zhang R, Gao Z, Lai X, Li D, Zhao X, Wang Y, Pei H, Qiao B, Ji Y, Wu Q. A triggered DNA nanomachine with enzyme-free for the rapid detection of telomerase activity in a one-step method. Anal Chim Acta 2024; 1299:342420. [PMID: 38499416 DOI: 10.1016/j.aca.2024.342420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/08/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Telomerase is considered a biomarker for the early diagnosis and clinical treatment of cancer. The rapid and sensitive detection of telomerase activity is crucial to biological research, clinical diagnosis, and drug development. However, the main obstacles facing the current telomerase activity assay are the cumbersome and time-consuming procedure, the easy degradation of the telomerase RNA template and the need for additional proteases. Therefore, it is necessary to construct a new method for the detection of telomerase activity with easy steps, efficient reaction and strong anti-interference ability. RESULTS Herein, an efficient, enzyme-free, economical, sensitive, fluorometric detection method for telomerase activity in one-step, named triggered-DNA (T-DNA) nanomachine, was created based on target-triggered DNAzyme-cleavage activity and catalytic molecular beacon (CMB). Telomerase served as a switch and extended few numbers of (TTAGGG)n repeat sequences to initiate the signal amplification in the T-DNA nanomachine, resulting in a strong fluorescent signal. The reaction was a one-step method with a shortened time of 1 h and a constant temperature of 37 °C, without the addition of any protease. It also sensitively distinguished telomerase activity in various cell lines. The T-DNA nanomachine offered a detection limit of 12 HeLa cells μL-1, 9 SK-Hep-1 cells μL-1 and 3 HuH-7 cells μL-1 with a linear correlation detection range of 0.39 × 102-6.25 × 102 HeLa cells μL-1 for telomerase activity. SIGNIFICANCE In conclusion, our study demonstrated that the triggered-DNA nanomachine fulfills the requirements for rapid detection of telomerase activity in one-step under isothermal and enzyme-free conditions with excellent specificity, and its simple and stable structure makes it ideal for complex systems. These findings indicated the application prospect of DNA nanomachines in clinical diagnostics and provided new insights into the field of DNA nanomachine-based bioanalysis.
Collapse
Affiliation(s)
- Huajie Pang
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China
| | - Yanan Peng
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China
| | - Rui Zhang
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China
| | - Zhijun Gao
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China
| | - Xiangde Lai
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China
| | - Dongxia Li
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China
| | - Xuan Zhao
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China
| | - Yuanyuan Wang
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China; Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, 571199, China
| | - Hua Pei
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China
| | - Bin Qiao
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China; Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, 571199, China.
| | - Yuxiang Ji
- The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, 571199, China.
| | - Qiang Wu
- The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; The Second Affiliated Hospital, School of Tropical Medicine, Hainan Medical University, Haikou, 570311, China; Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
11
|
Ahmad I, Ahmad S, Ahmad A, Zughaibi TA, Alhosin M, Tabrez S. Curcumin, its derivatives, and their nanoformulations: Revolutionizing cancer treatment. Cell Biochem Funct 2024; 42:e3911. [PMID: 38269517 DOI: 10.1002/cbf.3911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/27/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
Curcumin is a natural compound derived from turmeric and can target malignant tumor molecules involved in cancer propagation. It has potent antioxidant activity, but its effectiveness is limited due to poor absorption and rapid elimination from the body. Various curcumin derivatives have also shown anticancer potential in in-vitro and in-vivo models. Curcumin can target multiple signaling pathways involved in cancer development/progression or induce cancer cell death through apoptosis. In addition, curcumin and its derivatives could also enhance the effectiveness of conventional chemotherapy, radiation therapy and reduce their associated side effects. Lately, nanoparticle-based delivery systems are being developed/explored to overcome the challenges associated with curcumin's delivery, increasing its overall efficacy. The use of an imaging system to track these formulations could also give beneficial information about the bioavailability and distribution of the nano-curcumin complex. In conclusion, curcumin holds significant promise in the fight against cancer, especially in its nanoform, and could provide precise delivery to cancer cells without affecting normal healthy cells.
Collapse
Affiliation(s)
- Iftikhar Ahmad
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sameer Ahmad
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biotechnology & Genetics, Faculty of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ausaf Ahmad
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Torki A Zughaibi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmoud Alhosin
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
12
|
Guillon J, Le Borgne M, Milano V, Guédin-Beaurepaire A, Moreau S, Pinaud N, Ronga L, Savrimoutou S, Albenque-Rubio S, Marchivie M, Kalout H, Walker C, Chevallier L, Buré C, Largy E, Gabelica V, Mergny JL, Baylot V, Ferrer J, Idrissi Y, Chevret E, Cappellen D, Desplat V, Schelz Z, Zupkó I. New 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinazoline and 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinoline Derivatives: Synthesis and Biological Evaluation as Novel Anticancer Agents by Targeting G-Quadruplex. Pharmaceuticals (Basel) 2023; 17:30. [PMID: 38256866 PMCID: PMC10819771 DOI: 10.3390/ph17010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
The syntheses of novel 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinazolines 12 and 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinolines 13 are reported here in six steps starting from various halogeno-quinazoline-2,4-(1H,3H)-diones or substituted anilines. The antiproliferative activities of the products were determined in vitro against a panel of breast (MCF-7 and MDA-MB-231), human adherent cervical (HeLa and SiHa), and ovarian (A2780) cell lines. Disubstituted 6- and 7-phenyl-bis(3-dimethylaminopropyl)aminomethylphenyl-quinazolines 12b, 12f, and 12i displayed the most interesting antiproliferative activities against six human cancer cell lines. In the series of quinoline derivatives, 6-phenyl-bis(3-dimethylaminopropyl)aminomethylphenylquinoline 13a proved to be the most active. G-quadruplexes (G4) stacked non-canonical nucleic acid structures found in specific G-rich DNA, or RNA sequences in the human genome are considered as potential targets for the development of anticancer agents. Then, as small aza-organic heterocyclic derivatives are well known to target and stabilize G4 structures, their ability to bind G4 structures have been determined through FRET melting, circular dichroism, and native mass spectrometry assays. Finally, telomerase inhibition ability has been also assessed using the MCF-7 cell line.
Collapse
Affiliation(s)
- Jean Guillon
- INSERM, CNRS, ARNA, U1212, UMR 5320, UFR des Sciences Pharmaceutiques, Univ. Bordeaux, F-33076 Bordeaux, France; (J.G.); (V.M.); (A.G.-B.); (S.M.); (S.S.); (S.A.-R.); (H.K.); (C.W.); (L.C.)
| | - Marc Le Borgne
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, Univ. Lyon, F-69373 Lyon, France
| | - Vittoria Milano
- INSERM, CNRS, ARNA, U1212, UMR 5320, UFR des Sciences Pharmaceutiques, Univ. Bordeaux, F-33076 Bordeaux, France; (J.G.); (V.M.); (A.G.-B.); (S.M.); (S.S.); (S.A.-R.); (H.K.); (C.W.); (L.C.)
| | - Aurore Guédin-Beaurepaire
- INSERM, CNRS, ARNA, U1212, UMR 5320, UFR des Sciences Pharmaceutiques, Univ. Bordeaux, F-33076 Bordeaux, France; (J.G.); (V.M.); (A.G.-B.); (S.M.); (S.S.); (S.A.-R.); (H.K.); (C.W.); (L.C.)
| | - Stéphane Moreau
- INSERM, CNRS, ARNA, U1212, UMR 5320, UFR des Sciences Pharmaceutiques, Univ. Bordeaux, F-33076 Bordeaux, France; (J.G.); (V.M.); (A.G.-B.); (S.M.); (S.S.); (S.A.-R.); (H.K.); (C.W.); (L.C.)
| | - Noël Pinaud
- ISM—CNRS UMR 5255, Univ. Bordeaux, F-33405 Talence, France;
| | - Luisa Ronga
- E2S UPPA, CNRS, IPREM, Université de Pau et des Pays de l’Adour, F-64053 Pau, France;
| | - Solène Savrimoutou
- INSERM, CNRS, ARNA, U1212, UMR 5320, UFR des Sciences Pharmaceutiques, Univ. Bordeaux, F-33076 Bordeaux, France; (J.G.); (V.M.); (A.G.-B.); (S.M.); (S.S.); (S.A.-R.); (H.K.); (C.W.); (L.C.)
| | - Sandra Albenque-Rubio
- INSERM, CNRS, ARNA, U1212, UMR 5320, UFR des Sciences Pharmaceutiques, Univ. Bordeaux, F-33076 Bordeaux, France; (J.G.); (V.M.); (A.G.-B.); (S.M.); (S.S.); (S.A.-R.); (H.K.); (C.W.); (L.C.)
| | | | - Haouraa Kalout
- INSERM, CNRS, ARNA, U1212, UMR 5320, UFR des Sciences Pharmaceutiques, Univ. Bordeaux, F-33076 Bordeaux, France; (J.G.); (V.M.); (A.G.-B.); (S.M.); (S.S.); (S.A.-R.); (H.K.); (C.W.); (L.C.)
| | - Charley Walker
- INSERM, CNRS, ARNA, U1212, UMR 5320, UFR des Sciences Pharmaceutiques, Univ. Bordeaux, F-33076 Bordeaux, France; (J.G.); (V.M.); (A.G.-B.); (S.M.); (S.S.); (S.A.-R.); (H.K.); (C.W.); (L.C.)
| | - Louise Chevallier
- INSERM, CNRS, ARNA, U1212, UMR 5320, UFR des Sciences Pharmaceutiques, Univ. Bordeaux, F-33076 Bordeaux, France; (J.G.); (V.M.); (A.G.-B.); (S.M.); (S.S.); (S.A.-R.); (H.K.); (C.W.); (L.C.)
| | - Corinne Buré
- CNRS, INSERM, IECB, US1, UAR 3033, Univ. Bordeaux, F-33600 Pessac, France;
| | - Eric Largy
- CNRS, INSERM, ARNA, UMR 5320, U1212, IECB, Univ. Bordeaux, F-33600 Pessac, France; (E.L.); (V.G.)
| | - Valérie Gabelica
- CNRS, INSERM, ARNA, UMR 5320, U1212, IECB, Univ. Bordeaux, F-33600 Pessac, France; (E.L.); (V.G.)
| | - Jean-Louis Mergny
- Ecole Polytechnique, Laboratoire d’Optique et Biosciences, CNRS, INSERM, Institut Polytechnique de Paris, F-91120 Palaiseau, France;
| | - Virginie Baylot
- Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, CNRS UMR7258, Inserm U1068, Univ. Aix Marseille, F-13009 Marseille, France;
| | - Jacky Ferrer
- INSERM UMR1312, BRIC, Bordeaux Institute of Oncology, Univ. Bordeaux, F-33076 Bordeaux, France; (J.F.); (Y.I.); (E.C.); (D.C.); (V.D.)
| | - Yamina Idrissi
- INSERM UMR1312, BRIC, Bordeaux Institute of Oncology, Univ. Bordeaux, F-33076 Bordeaux, France; (J.F.); (Y.I.); (E.C.); (D.C.); (V.D.)
| | - Edith Chevret
- INSERM UMR1312, BRIC, Bordeaux Institute of Oncology, Univ. Bordeaux, F-33076 Bordeaux, France; (J.F.); (Y.I.); (E.C.); (D.C.); (V.D.)
| | - David Cappellen
- INSERM UMR1312, BRIC, Bordeaux Institute of Oncology, Univ. Bordeaux, F-33076 Bordeaux, France; (J.F.); (Y.I.); (E.C.); (D.C.); (V.D.)
- Service Tumor Biology and Tumor Bank Laboratory, Groupe Hospitalier Bordeaux, CHU Bordeaux, F-33000 Bordeaux, France
| | - Vanessa Desplat
- INSERM UMR1312, BRIC, Bordeaux Institute of Oncology, Univ. Bordeaux, F-33076 Bordeaux, France; (J.F.); (Y.I.); (E.C.); (D.C.); (V.D.)
| | - Zsuzsanna Schelz
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, 6720 Szeged, Hungary;
| | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, 6720 Szeged, Hungary;
| |
Collapse
|
13
|
Ali JH, Walter M. Combining old and new concepts in targeting telomerase for cancer therapy: transient, immediate, complete and combinatory attack (TICCA). Cancer Cell Int 2023; 23:197. [PMID: 37679807 PMCID: PMC10483736 DOI: 10.1186/s12935-023-03041-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Telomerase can overcome replicative senescence by elongation of telomeres but is also a specific element in most cancer cells. It is expressed more vastly than any other tumor marker. Telomerase as a tumor target inducing replicative immortality can be overcome by only one other mechanism: alternative lengthening of telomeres (ALT). This limits the probability to develop resistance to treatments. Moreover, telomerase inhibition offers some degree of specificity with a low risk of toxicity in normal cells. Nevertheless, only one telomerase antagonist reached late preclinical studies. The underlying causes, the pitfalls of telomerase-based therapies, and future chances based on recent technical advancements are summarized in this review. Based on new findings and approaches, we propose a concept how long-term survival in telomerase-based cancer therapies can be significantly improved: the TICCA (Transient Immediate Complete and Combinatory Attack) strategy.
Collapse
Affiliation(s)
- Jaber Haj Ali
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Universitätsmedizin Rostock, Ernst-Heydemann-Straße 6, 18057, Rostock, Germany
| | - Michael Walter
- Institute of Clinical Chemistry and Laboratory Medicine, Universitätsmedizin Rostock, Ernst-Heydemann-Straße 6, 18057, Rostock, Germany.
| |
Collapse
|
14
|
Sorrenti V, Buriani A, Fortinguerra S, Davinelli S, Scapagnini G, Cassidy A, De Vivo I. Cell Survival, Death, and Proliferation in Senescent and Cancer Cells: the Role of (Poly)phenols. Adv Nutr 2023; 14:1111-1130. [PMID: 37271484 PMCID: PMC10509428 DOI: 10.1016/j.advnut.2023.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/06/2023] Open
Abstract
Cellular senescence has long been considered a permanent state of cell cycle arrest occurring in proliferating cells subject to different stressors, used as a cellular defense mechanism from acquiring potentially harmful genetic faults. However, recent studies highlight that senescent cells might also alter the local tissue environment and concur to chronic inflammation and cancer risk by secreting inflammatory and matrix remodeling factors, acquiring a senescence-associated secretory phenotype (SASP). Indeed, during aging and age-related diseases, senescent cells amass in mammalian tissues, likely contributing to the inevitable loss of tissue function as we age. Cellular senescence has thus become one potential target to tackle age-associated diseases as well as cancer development. One important aspect characterizing senescent cells is their telomere length. Telomeres shorten as a consequence of multiple cellular replications, gradually leading to permanent cell cycle arrest, known as replicative senescence. Interestingly, in the large majority of cancer cells, a senescence escape strategy is used and telomere length is maintained by telomerase, thus favoring cancer initiation and tumor survival. There is growing evidence showing how (poly)phenols can impact telomere maintenance through different molecular mechanisms depending on dose and cell phenotypes. Although normally, (poly)phenols maintain telomere length and support telomerase activity, in cancer cells this activity is negatively modulated, thus accelerating telomere attrition and promoting cancer cell death. Some (poly)phenols have also been shown to exert senolytic activity, thus suggesting both antiaging (directly eliminating senescent cells) and anticancer (indirectly, via SASP inhibition) potentials. In this review, we analyze selective (poly)phenol mechanisms in senescent and cancer cells to discriminate between in vitro and in vivo evidence and human applications considering (poly)phenol bioavailability, the influence of the gut microbiota, and their dose-response effects.
Collapse
Affiliation(s)
- Vincenzo Sorrenti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy; Maria Paola Belloni Center for Personalized Medicine, Padova, Italy.
| | | | | | - Sergio Davinelli
- Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy
| | - Aedin Cassidy
- Institute for Global Food Security, Queen's University Belfast, Belfast, Northern Ireland
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| |
Collapse
|
15
|
Yi M, Wang M, Xu Y, Cao Z, Ling Y, Zhang Z, Cao H. CRISPR-based m 6A modification and its potential applications in telomerase regulation. Front Cell Dev Biol 2023; 11:1200734. [PMID: 37519297 PMCID: PMC10382234 DOI: 10.3389/fcell.2023.1200734] [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: 04/05/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Telomerase determines cell lifespan by controlling chromosome stability and cell viability, m6A epigenetic modification plays an important role in the regulation of telomerase activity. Using CRISPR epigenome editing to analyze specific m6A modification sites in telomerase will provide an important tool for analyzing the molecular mechanism of m6A modification regulating telomerase activity. In this review, we clarified the relevant applications of CRISPR system, paid special attention to the regulation of m6A modification in stem cells and cancer cells based on CRISPR system, emphasized the regulation of m6A modification on telomerase activity, pointed out that m6A modification sites regulate telomerase activity, and discussed strategies based on telomerase activity and disease treatment, which are helpful to promote the research of anti-aging and tumor related diseases.
Collapse
Affiliation(s)
- Mingliang Yi
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
| | - Mingyue Wang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
| | - Yongjie Xu
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
| | - Zhikun Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
| | - Yinghui Ling
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zijun Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Hongguo Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
16
|
Zhao X, Luo D, Liu T, Zhang H, Xie Y, Kong W. BIBR1532 Affects Endometrial Cell Proliferation, Migration, and Invasion in Endometriosis via Telomerase Inhibition and MAPK Signaling. Gynecol Obstet Invest 2023; 88:226-239. [PMID: 37429261 DOI: 10.1159/000530460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 02/27/2023] [Indexed: 07/12/2023]
Abstract
OBJECTIVES The effect of telomerase inhibitor BIBR1532 on endometriotic cells was investigated to explore the inhibitory effect of targeting telomerase on endometriosis. DESIGN In vitro primary cell culture study. Participants/Materials: Primary endometrial cells derived from eutopic and ectopic endometrium in patients with endometriosis. SETTING The study was conducted in the university hospital. METHODS Paired eutopic and ectopic endometrial cells were collected from 6 patients from January 2018 to July 2021. A TRAP assay was performed to detect the telomerase activity of the cells. MTT, cell cycle, apoptosis, migration, and invasion assays were performed to study the inhibitory effect of BIBR1532. Enrichment analysis was performed to identify the key pathways involved in endometriosis progression and telomerase action. Then, Western blotting was used to investigate the expression of related proteins. RESULTS BIBR1532 treatment significantly inhibited the growth of eutopic and ectopic endometrial cells, with apoptosis and cell cycle signaling involved. Migration and invasion, important characteristics for the establishment of ectopic lesions, were also inhibited by BIBR1532. The MAPK signaling cascade, related to telomerase and endometriosis, was decreased in eutopic and ectopic endometrial stromal cells with the treatment of BIBR1532. LIMITATIONS The severe side effects of telomerase inhibitors might be the main obstacle to clinical application, so it is necessary to find better drug delivery methods in vivo. CONCLUSIONS The telomerase inhibitor BIBR1532 affects endometrial cell proliferation, migration, and invasion in endometriosis.
Collapse
Affiliation(s)
- Xiaoling Zhao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Dan Luo
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Tingting Liu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - He Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yunkai Xie
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| |
Collapse
|
17
|
Kohansal F, Mobed A, Aletaha N, Ghaseminasab K, Dolati S, Hasanzadeh M. Biosensing of telomerase antigen using sandwich type immunosensor based on poly(β-Cyclodextrin) decorated by Au@Pt nanoparticles: An innovative immune-platform toward early-stage identification of cancer. Microchem J 2023. [DOI: 10.1016/j.microc.2023.108649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
|
18
|
Luo Q, Liu P, Yu P, Qin T. Cancer Stem Cells are Actually Stem Cells with Disordered Differentiation: the Monophyletic Origin of Cancer. Stem Cell Rev Rep 2023; 19:827-838. [PMID: 36648606 PMCID: PMC10185654 DOI: 10.1007/s12015-023-10508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
Cancer stem cells (CSCs) play an important role in cancer development. Based on advancements in CSC research, we propose a monophyletic model of cancer. This model is based on the idea that CSCs are stem cells with disordered differentiation whose original purpose was to repair damaged tissues. Inflammatory responses and damage repair signals are crucial for the creation and maintenance of CSCs. Normal quiescent stem cells are activated by environmental stimulation, such as an inflammatory response, and undergo cell division and differentiation. In the initial stage of cancer development, stem cell differentiation leads to heteromorphism due to the accumulation of gene mutations, resulting in the development of metaplasia or precancerosis. In the second stage, accumulated mutations induce poor differentiation and lead to cancer development. The monophyletic model illustrates the evolution, biological behavior, and hallmarks of CSCs, proposes a concise understanding of the origin of cancer, and may encourage a novel therapeutic approach.
Collapse
Affiliation(s)
- Qiankun Luo
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China
| | - Pan Liu
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China
| | - Pengfei Yu
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China
| | - Tao Qin
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China.
| |
Collapse
|
19
|
Jibrin A, Uzairu A, Shallangwa GA, Abechi SE, Umar AB. In-silico profiling, design, molecular docking computation, and drug kinetic model evaluation of novel curcumin derivatives as potential anticancer agents. J INDIAN CHEM SOC 2023. [DOI: 10.1016/j.jics.2023.100979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
|
20
|
Telomerase: A prominent oncological target for development of chemotherapeutic agents. Eur J Med Chem 2023; 249:115121. [PMID: 36669398 DOI: 10.1016/j.ejmech.2023.115121] [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: 12/02/2022] [Revised: 12/26/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Telomerase is a ribonucleoprotein (RNP) responsible for the maintenance of chromosomal integrity by stabilizing telomere length. Telomerase is a widely expressed hallmark responsible for replicative immortality in 80-90% of malignant tumors. Cancer cells produce telomerase which prevents telomere shortening by adding telomeres sequences beyond Hayflick's limit; which enables them to divide uncontrollably. The activity of telomerase is relatively low in somatic cells and absent in normal cells, but the re-activation of this RNP in normal cells suppresses p53 activity which leads to the avoidance of senescence causing malignancy. Here, we have focused explicitly on various anti-telomerase therapies and telomerase-inhibiting molecules for the treatment of cancer. We have covered molecules that are reported in developmental, preclinical, and clinical trial stages as potent telomerase inhibitors. Apart from chemotherapy, we have also included details of immunotherapy, gene therapy, G-quadruplex stabilizers, and HSP-90 inhibitors. The purpose of this work is to discuss the challenges behind the development of novel telomerase inhibitors and to identify various perspectives for designing anti-telomerase compounds.
Collapse
|
21
|
Hussain T, Chai L, Wang Y, Zhang Q, Wang J, Shi W, Wang Q, Li M, Xie X. Activation of PPAR-γ prevents TERT-mediated pulmonary vascular remodeling in MCT-induced pulmonary hypertension. Heliyon 2023; 9:e14173. [PMID: 36938425 PMCID: PMC10015197 DOI: 10.1016/j.heliyon.2023.e14173] [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: 11/06/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Background It has been demonstrated that elevated telomerase reverse transcriptase (TERT) expression or activity is implicated in pulmonary hypertension (PH). In addition, activation of peroxisome-proliferator-activated receptor γ (PPAR-γ) has been found to prevent PH progression. However, the molecular mechanism responsible for the protective effect of PPAR-γ activation on TERT expression in the pathogenesis of PH remains unknown. This study was performed to address these issues. Methods Intraperitoneal injection of monocrotaline (MCT) was used to establish PH. BIBR1532 was applied to inhibit the activity of telomerase. The right ventricular systolic pressure (RVSP) and histological analysis were used to detect the development of PH. The protein levels of p-Akt, t-Akt, c-Myc and TERT were determined by western blotting. Pharmacological inhibition of TERT by BIBR1532 effectively suppressed RVSP, RVHI and the WT% in MCT-induced PH rats. Results Pharmacological inhibition of Akt/c-Myc pathway by LY294002 diminished TERT upregulation, RVSP, RVHI and WT% in MCT-PH rats. Activation of PPAR-γ by pioglitazone inhibited p-Akt and c-Myc expressions and further downregulated TERT, thus to reduced RVSP, RVHI and WT% in MCT-treated PH rats. Conclusions In conclusion, TERT upregulation contributes to PH development in MCT-treated rats. Activation of PPAR-γ prevents pulmonary arterial remodeling through Akt/c-Myc/TERT axis suppression.
Collapse
Affiliation(s)
- Tafseel Hussain
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
- Corresponding author. Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
22
|
Cheng L, Zhang S, Wang M, Lopez-Beltran A. Biological and clinical perspectives of TERT promoter mutation detection on bladder cancer diagnosis and management. Hum Pathol 2023; 133:56-75. [PMID: 35700749 DOI: 10.1016/j.humpath.2022.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/05/2022] [Indexed: 02/08/2023]
Abstract
The telomerase reverse transcriptase (TERT) promoter mutations are associated with increased TERT mRNA and TERT protein levels, telomerase activity, and shorter but stable telomere length. TERT promoter mutation is the most common mutation that occurs in approximately 60-80% of patients with bladder cancer. The TERT promoter mutations occur in a wide spectrum of urothelial lesions, including benign urothelial proliferation and tumor-like conditions, benign urothelial tumors, premalignant and putative precursor lesions, urothelial carcinoma and its variants, and nonurothelial malignancies. The prevalence and incidence of TERT promoter mutations in a total of 7259 cases from the urinary tract were systematically reviewed. Different platforms of TERT promoter mutation detection were presented. In this review, we also discussed the significance and clinical implications of TERT promoter mutation detection in urothelial tumorigenesis, surveillance and early detection, diagnosis, differential diagnosis, prognosis, prediction of treatment responses, and clinical outcome. Identification of TERT promoter mutations from urine or plasma cell-free DNA (liquid biopsy) will facilitate bladder cancer screening program and optimal clinical management. A better understanding of TERT promoter mutation and its pathway would open new therapeutic avenues for patients with bladder cancer.
Collapse
Affiliation(s)
- Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University and Lifespan Academic Medical Center, Providence, RI, 02903, USA.
| | - Shaobo Zhang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mingsheng Wang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Antonio Lopez-Beltran
- Department of Morphological Sciences, University of Cordoba Medical School, Cordoba, E-14004, Spain
| |
Collapse
|
23
|
Ropio J, Prochazkova-Carlotti M, Batista R, Pestana A, Chebly A, Ferrer J, Idrissi Y, Cappellen D, Durães C, Boaventura P, Vinagre J, Azzi-Martin L, Poglio S, Cabeçadas J, Campos MA, Beylot-Barry M, Sobrinho-Simões M, Merlio JP, Soares P, Chevret E. Spotlight on hTERT Complex Regulation in Cutaneous T-Cell Lymphomas. Genes (Basel) 2023; 14:439. [PMID: 36833366 PMCID: PMC9956048 DOI: 10.3390/genes14020439] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
As a major cancer hallmark, there is a sustained interest in understanding the telomerase contribution to carcinogenesis in order to therapeutically target this enzyme. This is particularly relevant in primary cutaneous T-cell lymphomas (CTCL), a malignancy showing telomerase dysregulation with few investigative data available. In CTCL, we examined the mechanisms involved in telomerase transcriptional activation and activity regulation. We analyzed 94 CTCL patients from a Franco-Portuguese cohort, as well as 8 cell lines, in comparison to 101 healthy controls. Our results showed that not only polymorphisms (SNPs) located at the promoter of human telomerase reverse transcriptase (hTERT) gene (rs2735940 and rs2853672) but also an SNP located within the coding region (rs2853676) could influence CTCL occurrence. Furthermore, our results sustained that the post-transcriptional regulation of hTERT contributes to CTCL lymphomagenesis. Indeed, CTCL cells present a different pattern of hTERT spliced transcripts distribution from the controls, mostly marked by an increase in the hTERT β+ variants proportion. This increase seems to be associated with CTCL development and progression. Through hTERT splicing transcriptome modulation with shRNAs, we observed that the decrease in the α-β+ transcript induced a decrease in the cell proliferation and tumorigenic capacities of T-MF cells in vitro. Taken together, our data highlight the major role of post-transcriptional mechanisms regulating telomerase non canonical functions in CTCL and suggest a new potential role for the α-β+ hTERT transcript variant.
Collapse
Affiliation(s)
- Joana Ropio
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
- Institute of Biomedical Sciences of Abel Salazar, Porto University, 4050-313 Porto, Portugal
- Faculty of Veterinary Medicine, Lusófona University, 1749-024 Lisbon, Portugal
| | | | - Rui Batista
- Institute for Research and Innovation in Health (I3S), Porto University, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Cancer Biology Group, Porto University, 4200-465 Porto, Portugal
- Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
| | - Ana Pestana
- Institute for Research and Innovation in Health (I3S), Porto University, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Cancer Biology Group, Porto University, 4200-465 Porto, Portugal
- Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
| | - Alain Chebly
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
- Medical Genetics Unit, Faculty of Medicine, Saint Joseph University, Beirut 1104 2020, Lebanon
- Higher Institute of Public Health, Saint Joseph University, Beirut 1104 2020, Lebanon
| | - Jacky Ferrer
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
| | - Yamina Idrissi
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
| | - David Cappellen
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
- Tumor Bank and Tumor Biology Laboratory, Bordeaux University Hospital, 33075 Bordeaux, France
| | - Cecília Durães
- Institute for Research and Innovation in Health (I3S), Porto University, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Cancer Biology Group, Porto University, 4200-465 Porto, Portugal
| | - Paula Boaventura
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Cancer Biology Group, Porto University, 4200-465 Porto, Portugal
| | - João Vinagre
- Institute for Research and Innovation in Health (I3S), Porto University, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Cancer Biology Group, Porto University, 4200-465 Porto, Portugal
| | - Lamia Azzi-Martin
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
- UFR des Sciences Médicales, Bordeaux University, 33076 Bordeaux, France
| | - Sandrine Poglio
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
| | - José Cabeçadas
- Dermatology Departement, Instituto Português de Oncologia de Lisboa (IPO-L), 1099-023 Lisbon, Portugal
| | - Manuel António Campos
- Institute for Research and Innovation in Health (I3S), Porto University, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Cancer Biology Group, Porto University, 4200-465 Porto, Portugal
- Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
- Centro Hospitalar Vila Nova de Gaia/Espinho, E.P.E., Dermatology Departement, 4434-502 Vila Nova de Gaia, Portugal
| | - Marie Beylot-Barry
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
- Dermatology Department, Bordeaux University Hospital, 33075 Bordeaux, France
| | - Manuel Sobrinho-Simões
- Institute for Research and Innovation in Health (I3S), Porto University, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Cancer Biology Group, Porto University, 4200-465 Porto, Portugal
- Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
- Department of Pathology, Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
| | - Jean-Philippe Merlio
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
- Tumor Bank and Tumor Biology Laboratory, Bordeaux University Hospital, 33075 Bordeaux, France
| | - Paula Soares
- Institute for Research and Innovation in Health (I3S), Porto University, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Cancer Biology Group, Porto University, 4200-465 Porto, Portugal
- Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
- Department of Pathology, Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
| | - Edith Chevret
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, 33000 Bordeaux, France
| |
Collapse
|
24
|
Al-Karmalawy AA, Nafie MS, Shaldam MA, Elmaaty AA, Antar SA, El-Hamaky AA, Saleh MA, Elkamhawy A, Tawfik HO. Ligand-Based Design on the Dog-Bone-Shaped BIBR1532 Pharmacophoric Features and Synthesis of Novel Analogues as Promising Telomerase Inhibitors with In Vitro and In Vivo Evaluations. J Med Chem 2023; 66:777-792. [PMID: 36525642 DOI: 10.1021/acs.jmedchem.2c01668] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Telomerase is an outstanding biological target for cancer treatment. BIBR1532 is a non-nucleoside selective telomerase inhibitor; however, it experiences ineligible pharmacokinetics. Herein, we aimed to design new BIBR1532-based analogues as promising telomerase inhibitors. Therefore, two novel series of pyridazine-linked to cyclopenta[b]thiophene (8a-f) and tetrahydro-1-benzothiophene (9a-f) were synthesized. A quantitative real-time polymerase chain reaction was utilized to investigate the telomerase inhibitory activity of candidates. Notably, 8e and 9e exhibited the best inhibition profiles. Moreover, 8e showed strong antitumor effects against both MCF-7 and A549 cancer cell lines. The effects of 8e on the cell cycle and apoptosis were measured. Besides, 8e was evaluated for its in vivo antitumor activity using solid Ehrlich carcinoma. The reduction in both the tumor weight and volume was greater than doxorubicin. Also, molecular docking and ADME studies were performed. Finally, a SAR study was conducted to gain further insights into the different telomerase inhibition potentials upon variable structural modifications.
Collapse
Affiliation(s)
- Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt
| | - Mohamed S Nafie
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Ayman Abo Elmaaty
- Department of Medicinal Chemistry, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt
| | - Samar A Antar
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt.,Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia 24016, United States
| | - Anwar A El-Hamaky
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Mohamed A Saleh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, The United Arab Emirates.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed Elkamhawy
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea.,Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
25
|
Judasz E, Lisiak N, Kopczyński P, Taube M, Rubiś B. The Role of Telomerase in Breast Cancer's Response to Therapy. Int J Mol Sci 2022; 23:12844. [PMID: 36361634 PMCID: PMC9654063 DOI: 10.3390/ijms232112844] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/13/2022] [Accepted: 10/24/2022] [Indexed: 11/26/2023] Open
Abstract
Currently, breast cancer appears to be the most widespread cancer in the world and the most common cause of cancer deaths. This specific type of cancer affects women in both developed and developing countries. Prevention and early diagnosis are very important factors for good prognosis. A characteristic feature of cancer cells is the ability of unlimited cell division, which makes them immortal. Telomeres, which are shortened with each cell division in normal cells, are rebuilt in cancer cells by the enzyme telomerase, which is expressed in more than 85% of cancers (up to 100% of adenocarcinomas, including breast cancer). Telomerase may have different functions that are related to telomeres or unrelated. It has been shown that high activity of the enzyme in cancer cells is associated with poor cell sensitivity to therapies. Therefore, telomerase has become a potential target for cancer therapies. The low efficacy of therapies has resulted in the search for new combined and more effective therapeutic methods, including the involvement of telomerase inhibitors and telomerase-targeted immunotherapy.
Collapse
Affiliation(s)
- Eliza Judasz
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Natalia Lisiak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Przemysław Kopczyński
- Centre for Orthodontic Mini-Implants at the Department and Clinic of Maxillofacial Orthopedics and Orthodontics, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Magdalena Taube
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| |
Collapse
|
26
|
Akinnibosun OA, Maier MC, Eales J, Tomaszewski M, Charchar FJ. Telomere therapy for chronic kidney disease. Epigenomics 2022; 14:1039-1054. [PMID: 36177720 DOI: 10.2217/epi-2022-0073] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chronic kidney disease (CKD) is estimated to affect almost 10% of individuals worldwide and is one of the leading causes of morbidity and mortality. Renal fibrosis, a central pathway in CKD progression (irrespective of etiology), is associated with shortened or dysfunctional telomeres in animal studies. Telomeres are specialized nucleoprotein structures located at the chromosome end that maintain genomic integrity. The mechanisms of associations between telomere length and CKD have not yet been fully elucidated, however, CKD patients with shorter telomere length may have decreased renal function and a higher mortality rate. A plethora of ongoing research has focused on possible therapeutic applications of telomeres with the overall goal to preserve telomere length as a therapy to treat CKD.
Collapse
Affiliation(s)
| | - Michelle C Maier
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Victoria, Australia
| | - James Eales
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK.,Manchester Heart Centre and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Fadi J Charchar
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Victoria, Australia.,Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Shen Z, Zheng R, Yang H, Xing S, Jin X, Yan H, Zhu J, Mei Y, Lin F, Zheng X. G-quadruplex stabilizer Tetra-Pt(bpy) disrupts telomere maintenance and impairs FAK-mediated migration of telomerase-positive cells. Int J Biol Macromol 2022; 213:858-870. [PMID: 35697164 DOI: 10.1016/j.ijbiomac.2022.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/18/2022] [Accepted: 06/05/2022] [Indexed: 11/05/2022]
Abstract
G-quadruplex regulates a wide spectrum of biological processes, including telomere maintenance, DNA replication and transcription. The development of small molecules to selectively target G-quadruplex and their application remain hotspots in cancer therapy. Here, we explored the biological effect of G-quadruplexes stabilizer Tetra-Pt(bpy) in telomerase-positive cancer cells. Telomere maintenance was evaluated by telomerase repeat amplification protocol, chromosome orientation fluorescence in situ hybridization and telomere restriction fragment assays. We found that Tetra-Pt(bpy) accelerates telomere shortening through dual inhibition of telomerase activity and telomere sister chromatin exchanges mediated by telomeric G-quadruplexes. Consequently, Tetra-Pt(bpy)-treated cancer cells became enriched with extremely short telomeres and produced a strong telomeric DNA damage response following long-term treatment, leading to cell proliferation inhibition and senescence. Experimental evidence from RNA seq and cell migration-related assays showed that Tetra-Pt(bpy) decreased cell-matrix adhesion and inhibited the migration of non-senescent tumor cells. Mechanistically, Tetra-Pt(bpy) induced the formation of G-quadruplexes in focal adhesion kinase (FAK)-encoding gene PTK2, resulting in FAK transcription inhibition. Tetra-Pt(bpy) reduced xenograft tumor formation and inhibited tumor cell growth and migration in mice. This study further elucidates the function of G-quadruplexes in the human genome and reveals the potential of Tetra-Pt(bpy) as a novel chemotherapeutic agent for targeting telomerase-positive cancer cells.
Collapse
Affiliation(s)
- Zhe Shen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou 325035, Zhejiang, China
| | - Ruiling Zheng
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou 325035, Zhejiang, China
| | - Huamao Yang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou 325035, Zhejiang, China
| | - Sunhui Xing
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou 325035, Zhejiang, China
| | - Xiangxiang Jin
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou 325035, Zhejiang, China
| | - Hao Yan
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou 325035, Zhejiang, China
| | - Jufan Zhu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yanan Mei
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou 325035, Zhejiang, China
| | - Feng Lin
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; Department of Gynecology, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| | - Xiaohui Zheng
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou 325035, Zhejiang, China.
| |
Collapse
|
28
|
He T, Hao Z, Lin M, Xin Z, Chen Y, Ouyang W, Yang Q, Chen X, Zhou H, Zhang W, Wu P, Xu F. Oncolytic adenovirus promotes vascular normalization and nonclassical tertiary lymphoid structure formation through STING-mediated DC activation. Oncoimmunology 2022; 11:2093054. [PMID: 35800155 PMCID: PMC9255224 DOI: 10.1080/2162402x.2022.2093054] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Inducing a full antitumor immune response in the tumor microenvironment (TME) is essential for successful cancer immunotherapy. Here, we report that an oncolytic adenovirus carrying mIL-15 (Ad-IL15) can effectively induce antitumor immune response and inhibit tumor growth in a mouse model of cancer. We found that Ad-IL15 facilitated the activation and infiltration of immune cells, including dendritic cells (DCs), T cells and natural killer (NK) cells, in the TME. Unexpectedly, we observed that Ad-IL15 also induced vascular normalization and tertiary lymphoid structure formation in the TME. Moreover, we demonstrated these Ad-IL15-induced changes in the TME were depended on the Ad-IL15-induced activation of the STING-TBK1-IRF3 pathway in DCs. Taken together, our findings suggest that Ad-IL15 is a candidate for cancer immunotherapy that promotes immune cell activation and infiltration, tumor vascular normalization and tertiary lymphoid structure formation in the TME.
Collapse
Affiliation(s)
- Teng He
- Department of Infectious Diseases, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhixing Hao
- Department of Thoracic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mingjie Lin
- Department of Thoracic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhongwei Xin
- Department of Thoracic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yongyuan Chen
- Department of Thoracic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Ouyang
- Department of Infectious Diseases, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qi Yang
- Department of Emergency, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoke Chen
- Department of Thoracic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hui Zhou
- Department of Infectious Diseases, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wanying Zhang
- Department of Infectious Diseases, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Pin Wu
- Department of Thoracic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Feng Xu
- Department of Infectious Diseases, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
29
|
MAPK/ERK-CBP-RFPL-3 Mediates Adipose-Derived Stem Cell-Induced Tumor Growth in Breast Cancer Cells by Activating Telomerase Reverse Transcriptase Expression. Stem Cells Int 2022; 2022:8540535. [PMID: 35711680 PMCID: PMC9197637 DOI: 10.1155/2022/8540535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/16/2022] [Accepted: 04/29/2022] [Indexed: 11/26/2022] Open
Abstract
Adipose-derived stem cells (ASCs) improve the self-renewal and survival of fat grafts in breast reconstruction after oncology surgery. However, ASCs have also been found to enhance breast cancer growth, and its role in tumor proliferation remains largely elusive. Here, we explored a novel mechanism that mediates hTERT reactivation during ASC-induced tumor growth in breast cancer cells. In this study, we found the proliferative ability of breast cancer cells markedly increased with ASC coculture. To explore the molecular mechanism, we treated cells with anibody/inhibitor and found that the activation of MEK-ERK pathway was triggered in breast cancer cells by SCF secreted from ASCs, leading to the nuclear recruitment of CBP. As a coactivator of hTERT, CBP subsequently coordinated with RFPL-3 upregulated hTERT transcription and telomerase activity. The inhibition of CBP and RFPL-3 abrogated the activation of hTERT transcription and the promotion of proliferation in breast cancer cells with cocultured ASCs in vitro and in vivo. Collectively, our study findings indicated that CBP coordination with RFPL-3 promotes ASC-induced breast cancer cell proliferation by anchoring to the hTERT promoter and upregulating telomerase activity, which is activated by the MAPK/ERK pathway.
Collapse
|
30
|
Mc Leer A, Foll M, Brevet M, Antoine M, Novello S, Mondet J, Cadranel J, Girard N, Giaj Levra M, Demontrond P, Audigier-Valette C, Letouzé E, Lantuéjoul S, Fernandez-Cuesta L, Moro-Sibilot D. Detection of acquired TERT amplification in addition to predisposing p53 and Rb pathways alterations in EGFR-mutant lung adenocarcinomas transformed into small-cell lung cancers. Lung Cancer 2022; 167:98-106. [PMID: 35183375 DOI: 10.1016/j.lungcan.2022.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Among the different mechanisms of acquired resistance to EGFR tyrosine kinase inhibitors (TKIs) reported in EGFR-mutated lung adenocarcinoma (EGFR-LUAD) patients, histological transformation into small cell carcinoma (SCLC) occurs in 3-14% of resistant cases, regardless of the generation of EGFR-TKI. In recent studies, bi-allelic inactivation of TP53 and RB1 has been identified in a vast majority of transformed SCLCs. However, the molecular mechanisms driving this histologic transformation remain largely unknown, mainly due to the rarity of samples. PATIENTS AND METHODS Out of an initial cohort of 64 patients, tumor tissues of adequate quality and quantity for whole exome sequencing (WES) analysis were available for nine tumors for six patients: paired pre- and post-SCLC transformation samples for three Patients and post-SCLC transformation samples for three other patients. RESULTS Mutational analyses showed concurrent TP53 mutations and Rb pathway alterations in five of the six patients analyzed, confirming their suggested role as predisposing genetic alterations to SCLC transformation. In addition, TERT amplification was detected in four of the six patients and found to be an event acquired during SCLC transformation. Clonal history evolution analyses from the paired LUAD/SCLC samples showed different evolution patterns. In two patients, a large proportion of mutations were present in the most recent common ancestor cell of the initial LUAD and the transformed SCLC clones, whereas in the third patient, few clonal mutations were common between the LUAD and SCLC samples and the ancestor clone that lead to SCLC was present at low frequency in the initial LUAD. CONCLUSION Despite varied clinical presentations and clonal history evolution patterns, in addition to p53 and Rb pathways alterations, TERT amplification emerged as another common genetic mechanism of EGFR-LUAD to SCLC transformation in our cohort, and could represent a candidate therapeutic target in this subset of SCLC tumors.
Collapse
Affiliation(s)
- Anne Mc Leer
- Université Grenoble Alpes, Grenoble, France; Service d'Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France; UGA/INSERM U1209/CNRS 5309-Institute for Advanced Biosciences - Université Grenoble Alpes, Grenoble, France.
| | - Matthieu Foll
- International Agency for Research on Cancer (IARC-WHO), Section of Genetics, Lyon, France
| | - Marie Brevet
- Hospices Civils de Lyon, Institut de pathologie Multisite, Site Est, Bron and Université Claude Bernard Lyon 1, Lyon, France
| | - Martine Antoine
- Service d'Anatomie et Cytologie Pathologique, APHP, Hôpital Tenon, 75020 Paris, France and Theranoscan CRC#4 and Curamus Sorbonne Université, Paris, France
| | - Silvia Novello
- Department of Oncology, AOU San Luigi-Orbassano, University of Turin, Italy
| | - Julie Mondet
- Université Grenoble Alpes, Grenoble, France; Service d'Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France; UGA/INSERM U1209/CNRS 5309-Institute for Advanced Biosciences - Université Grenoble Alpes, Grenoble, France
| | - Jacques Cadranel
- Service de Pneumologie et Oncologie thoracique, APHP, Hôpital Tenon and GRC#4 Theranoscan and Curamus Sorbonne Université, Paris, France
| | | | - Matteo Giaj Levra
- Clinique Hospitalo-Universitaire de Pneumologie Physiologie, Pôle Thorax et Vaisseaux, CHU Grenoble Alpes, Grenoble, France
| | | | | | - Eric Letouzé
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Université Paris Nord, Functional Genomics of Solid Tumors Laboratory, Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France
| | - Sylvie Lantuéjoul
- Université Grenoble Alpes, Grenoble, France; Cancer Research Center Lyon, Centre Léon Bérard, Lyon, France
| | | | - Denis Moro-Sibilot
- Université Grenoble Alpes, Grenoble, France; UGA/INSERM U1209/CNRS 5309-Institute for Advanced Biosciences - Université Grenoble Alpes, Grenoble, France; Clinique Hospitalo-Universitaire de Pneumologie Physiologie, Pôle Thorax et Vaisseaux, CHU Grenoble Alpes, Grenoble, France
| |
Collapse
|
31
|
Abstract
Spontaneous tumors in dogs share several environmental, epidemiologic, biologic, clinical and molecular features with a wide variety of human cancers, making this companion animal an attractive model. Nuclear factor kappa B (NF-kB) transcription factor overactivation is common in several human cancers, and there is evidence that similar signaling aberrations also occur in canine cancers including lymphoma, leukemia, hemangiosarcoma, mammary cancer, melanoma, glioma, and prostate cancer. This review provides an overview of NF-kB signaling biology, both in health and in cancer development. It also summarizes available evidence of aberrant NF-kB signaling in canine cancer, and reviews antineoplastic compounds that have been shown to inhibit NF-kB activity used in various types of canine cancers. Available data suggest that dogs may be an excellent model for human cancers that have overactivation of NF-kB.
Collapse
|
32
|
Tawfik HO, El-Hamaky AA, El-Bastawissy EA, Shcherbakov KA, Veselovsky AV, Gladilina YA, Zhdanov DD, El-Hamamsy MH. New Genetic Bomb Trigger: Design, Synthesis, Molecular Dynamics Simulation, and Biological Evaluation of Novel BIBR1532-Related Analogs Targeting Telomerase against Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2022; 15:ph15040481. [PMID: 35455478 PMCID: PMC9025901 DOI: 10.3390/ph15040481] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Telomeres serve a critical function in cell replication and proliferation at every stage of the cell cycle. Telomerase is a ribonucleoprotein, responsible for maintaining the telomere length and chromosomal integrity of frequently dividing cells. Although it is silenced in most human somatic cells, telomere restoration occurs in cancer cells because of telomerase activation or alternative telomere lengthening. The telomerase enzyme is a universal anticancer target that is expressed in 85–95% of cancers. BIBR1532 is a selective non-nucleoside potent telomerase inhibitor that acts by direct noncompetitive inhibition. Relying on its structural features, three different series were designed, and 30 novel compounds were synthesized and biologically evaluated as telomerase inhibitors using a telomeric repeat amplification protocol (TRAP) assay. Target compounds 29a, 36b, and 39b reported the greatest inhibitory effect on telomerase enzyme with IC50 values of 1.7, 0.3, and 2.0 μM, respectively, while BIBR1532 displayed IC50 = 0.2 μM. Compounds 29a, 36b, and 39b were subsequently tested using a living-cell TRAP assay and were able to penetrate the cell membrane and inhibit telomerase inside living cancer cells. Compound 36b was tested for cytotoxicity against 60 cancer cell lines using the NCI (USA) procedure, and the % growth was minimally impacted, indicating telomerase enzyme selectivity. To investigate the interaction of compound 36b with the telomerase allosteric binding site, molecular docking and molecular dynamics simulations were used.
Collapse
Affiliation(s)
- Haytham O. Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (A.A.E.-H.); (E.A.E.-B.); (M.H.E.-H.)
- Correspondence: (H.O.T.); (D.D.Z.)
| | - Anwar A. El-Hamaky
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (A.A.E.-H.); (E.A.E.-B.); (M.H.E.-H.)
| | - Eman A. El-Bastawissy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (A.A.E.-H.); (E.A.E.-B.); (M.H.E.-H.)
| | - Kirill A. Shcherbakov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (K.A.S.); (A.V.V.); (Y.A.G.)
| | - Alexander V. Veselovsky
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (K.A.S.); (A.V.V.); (Y.A.G.)
| | - Yulia A. Gladilina
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (K.A.S.); (A.V.V.); (Y.A.G.)
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (K.A.S.); (A.V.V.); (Y.A.G.)
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya St. 6, 117198 Moscow, Russia
- Correspondence: (H.O.T.); (D.D.Z.)
| | - Mervat H. El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (A.A.E.-H.); (E.A.E.-B.); (M.H.E.-H.)
| |
Collapse
|
33
|
Targeting visualization of malignant tumor based on the alteration of DWI signal generated by hTERT promoter–driven AQP1 overexpression. Eur J Nucl Med Mol Imaging 2022; 49:2310-2322. [DOI: 10.1007/s00259-022-05684-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/09/2022] [Indexed: 02/07/2023]
|
34
|
Pal S, Fatma K, Ravichandiran V, Dash J. Triazolyl Dibenzo[ a,c]phenazines Stabilize Telomeric G-quadruplex and Inhibit Telomerase. ASIAN J ORG CHEM 2021; 10:2921-2926. [PMID: 37823002 PMCID: PMC7614908 DOI: 10.1002/ajoc.202100468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Indexed: 11/10/2022]
Abstract
We herein report the synthesis and biophysical evaluation of triazolyl dibenzo[a,c]phenazine derivatives as a novel class of G-quadruplex ligands. The aromatic core facilitates π-π interaction and the flexible, protonatable side chains interact with the phosphate backbone of DNA via electrostatic interactions. Förster resonance energy transfer (FRET) melting assay and isothermal titration calorimetry (ITC) studies suggest that these ligands show binding preference for the hTELO G-quadruplex over G-quadruplexes found in the promoter region of various oncogenes and duplex DNA. The in vitro telomeric repeat amplification protocol (Q-TRAP) assay reveals that these ligands reduce telomerase activity in cancer cells.
Collapse
Affiliation(s)
- Sarmistha Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata-700032, India
- Department of Medicinal Chemistry, NIPER-KOLKATA, Chunilal Bhawan (Adjacent to BCPL), 168, Maniktala Main Road P.O. Bengal Chemicals, P.S. Phoolbagan, Kolkata – 700054, West Bengal
| | - Khushnood Fatma
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata-700032, India
| | - Velayutham Ravichandiran
- Department of Medicinal Chemistry, NIPER-KOLKATA, Chunilal Bhawan (Adjacent to BCPL), 168, Maniktala Main Road P.O. Bengal Chemicals, P.S. Phoolbagan, Kolkata – 700054, West Bengal
| | - Jyotirmayee Dash
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata-700032, India
| |
Collapse
|
35
|
Zhao J, Du G, Sun X. Tumor Antigen-Based Nanovaccines for Cancer Immunotherapy: A Review. J Biomed Nanotechnol 2021; 17:2099-2113. [PMID: 34906272 DOI: 10.1166/jbn.2021.3178] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
As an important means of tumor immunotherapy, tumor vaccines have achieved exciting results in the past few decades. However, there are still many obstacles that hinder tumor vaccines from achieving maximum efficacy, including lack of tumor antigens, low antigen immunogenicity and poor delivery efficiency. To overcome these challenges, researchers have developed and investigated various new types of tumor antigens with higher antigenic specificity and broader antigen spectrum, such as tumor-specific peptide antigens, tumor lysates, tumor cell membrane, tumor associated exosomes, etc. At the same time, different nanoparticulate delivery platforms have been developed to increase the immunogenicity of the tumor antigens, for example by increasing their targeting efficiency of antigen-presenting cells and lymph nodes, and by co-delivering antigens with adjuvants. In this review, we summarized different types of the tumor antigens that have been reported, and introduced several nanovaccine strategies for increasing the immunogenicity of tumor antigens. The review of recent progress in these fields may provide reference for the follow-up studies of tumor antigen-based cancer immunotherapy.
Collapse
Affiliation(s)
- Jiaxuan Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
36
|
Dai C, Sun B, Guan S, Wang W, Liu H, Li Y, Zhang J, Kang J. Evolution of a refractory prolactin-secreting pituitary adenoma into a pituitary carcinoma: report of a challenging case and literature review. BMC Endocr Disord 2021; 21:217. [PMID: 34715828 PMCID: PMC8555299 DOI: 10.1186/s12902-021-00874-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 10/15/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Pituitary carcinomas (PCs), defined as distant metastases of pituitary neoplasms, are very rare malignancies. Because the clinical presentation of PCs is variable, early diagnosis and management remain challenging. PCs are always refractory to comprehensive treatments, and patients with PCs have extremely poor prognoses. CASE PRESENTATION We describe one case of a prolactin-secreting pituitary adenoma (PA) refractory to conventional therapy that evolved into a PC with intraspinal metastasis. A 34-year-old female was diagnosed with an invasive prolactin-secreting PA in 2009 and was unresponsive to medical treatment with bromocriptine. The tumor was gross totally removed via transsphenoidal surgery (TSS). However, the patient experienced multiple tumor recurrences or regrowth despite comprehensive treatments, including medical therapy, two gamma knife radiosurgeries (GKSs), and four frontal craniotomies. In 2016, she was found to have an intradural extramedullary mass at the level of the fourth lumbar vertebra. The intraspinal lesion was completely resected and was confirmed as a metastatic PC based on histomorphology and immunohistochemical staining. The literature on the diagnosis, molecular pathogenesis, treatment, and prognosis of patients with prolactin-secreting PCs was reviewed. CONCLUSION PCs are very rare neoplasms with variable clinical features and poor prognosis. Most PCs usually arise from aggressive PAs refractory to conventional therapy. There is no reliable marker to identify aggressive PAs with a risk for progression to PCs; thus, it is difficult to diagnose these PCs early until the presence of metastatic lesions. It is still very challenging to manage patients with PCs due to a lack of standardized protocols for diagnosis and treatment. Establishing molecular biomarkers and the pathobiology of PCs could help in the early identification of aggressive PAs most likely to evolve into PCs.
Collapse
Affiliation(s)
- Congxin Dai
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Bowen Sun
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Shusen Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100730, China
| | - Wei Wang
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Honggang Liu
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yong Li
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jialiang Zhang
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jun Kang
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
37
|
Roque CR, Sampaio LR, Ito MN, Pinto DV, Caminha JSR, Nunes PIG, Raposo RS, Santos FA, Windmöller CC, Crespo-Lopez ME, Alvarez-Leite JI, Oriá RB, Pinheiro RF. Methylmercury chronic exposure affects the expression of DNA single-strand break repair genes, induces oxidative stress, and chromosomal abnormalities in young dyslipidemic APOE knockout mice. Toxicology 2021; 464:152992. [PMID: 34670124 DOI: 10.1016/j.tox.2021.152992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 10/01/2021] [Accepted: 10/12/2021] [Indexed: 12/31/2022]
Abstract
Mercury (Hg) is one of the most toxic environmental pollutants, especially when methylated, forming methylmercury (MeHg). MeHg affects DNA repair, increases oxidative stress, and predisposes to cancer. MeHg neurotoxicity is well-known, but recently MeHg-associated cardiovascular effects were recognized. This study evaluated circulating lipids, oxidative stress, and genotoxicity after MeHg-chronic exposure (20 mg/L in drinking water) in C57BL/6J wild-type and APOE knockout (ko) mice, the latter, being spontaneously dyslipidemic. Experimental mice were assigned to four groups: non-intoxicated and MeHg-intoxicated wild-type mice and non-intoxicated and MeHg-intoxicated APOE ko mice. Plasma levels of triglycerides, total cholesterol (TC), HDL, and LDL were analyzed. Liver lipid peroxidation and splenic gene expression of xeroderma pigmentosum complementation groups A, C, D, and G (XPA, XPC, XPD, and XPG), X-ray repair cross-complementing protein 1 (XRCC1), and telomerase reverse transcriptase (TERT) were measured. Fur Hg levels confirmed chronic MeHg intoxication. MeHg exposure raises TC levels both in wild-type and APOE ko mice. HDL and LDL-cholesterol levels were increased only in the MeHg-challenged APOE ko mice. MeHg increased liver lipid peroxidation, regardless of the genetic background. Unintoxicated APOE ko mice showed higher expression of TERT than all other groups. APOE deficiency increases XPA expression, regardless of MeHg intoxication. Furthermore, MeHg-intoxicated mice had more cytogenetic abnormalities, effect which was independent of APOE deficiency. More studies are needed to dissect the interactions between circulating lipids, MeHg intoxication, and DNA-repair pathways even at young age, interactions that likely play critical roles in cell senescence and the risk for chronic disorders later in life.
Collapse
Affiliation(s)
- Cássia R Roque
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Letícia R Sampaio
- Cancer Cytogenomics Laboratory, Drug Research, and Development Center, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Mayumi N Ito
- Cancer Cytogenomics Laboratory, Drug Research, and Development Center, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Daniel V Pinto
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Juan S R Caminha
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Paulo I G Nunes
- Natural Products Laboratory, Biomedicine Center, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Ramon S Raposo
- Experimental Biology core, Health Sciences, University of Fortaleza, Fortaleza, CE, Brazil
| | - Flávia A Santos
- Natural Products Laboratory, Biomedicine Center, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Cláudia C Windmöller
- Laboratory of Atherosclerosis and Nutritional Biochemistry, Department of Biochemistry and Immunology, ICB, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Para, Belém, PA, Brazil
| | - Jacqueline I Alvarez-Leite
- Laboratory of Atherosclerosis and Nutritional Biochemistry, Department of Biochemistry and Immunology, ICB, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Reinaldo B Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
| | - Ronald F Pinheiro
- Cancer Cytogenomics Laboratory, Drug Research, and Development Center, Federal University of Ceara, Fortaleza, CE, Brazil
| |
Collapse
|
38
|
Plyasova AA, Berrino E, Khan II, Veselovsky AV, Pokrovsky VS, Angeli A, Ferraroni M, Supuran CT, Pokrovskaya MV, Alexandrova SS, Gladilina YA, Sokolov NN, Hilal A, Carta F, Zhdanov DD. Mechanisms of the Antiproliferative and Antitumor Activity of Novel Telomerase-Carbonic Anhydrase Dual-Hybrid Inhibitors. J Med Chem 2021; 64:11432-11444. [PMID: 34283610 DOI: 10.1021/acs.jmedchem.1c00756] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Human (h) telomerase (TL; EC 2.7.7.49) plays a key role in sustaining cancer cells by means of elongating telomeric repeats at the 3' ends of chromosomes. Since TL-inhibitor (TI) stand-alone cancer therapy has been proven to be remarkably challenging, a polypharmacological approach represents a valid alternative. Here we consider a series of compounds able to inhibit both hTL and the tumor-associated carbonic anhydrases (CAs; EC 4.2.1.1) IX and XII. Compounds 7 and 9 suppressed hTL activity in both cell lysates and human colon cancer cell lines, and prolonged incubation with either 7 or 9 resulted in telomere shortening, cell cycle arrest, replicative senescence, and apoptosis. Enzyme kinetics showed that 7 and 9 are mixed-type inhibitors of the binding of DNA primers and deoxynucleoside triphosphate (dNTP) to the TL catalytic subunit hTERT, which is in agreement with docking experiments. Compound 9 showed antitumor activity in Colo-205 mouse xenografts and suppressed telomerase activity by telomere reduction.
Collapse
Affiliation(s)
- Anna A Plyasova
- Institute of Biomedical Chemistry, Pogodinskaya Street 10/8, Moscow 119121, Russia
| | - Emanuela Berrino
- Dipartimento di Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy
| | - Irina I Khan
- Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya Street 6, Moscow 117198, Russia.,N.N. Blokhin Cancer Research Center, Kashirskoe Shosse 24, Moscow 115478, Russia
| | | | - Vadim S Pokrovsky
- Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya Street 6, Moscow 117198, Russia.,N.N. Blokhin Cancer Research Center, Kashirskoe Shosse 24, Moscow 115478, Russia
| | - Andrea Angeli
- Dipartimento di Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy
| | - Marta Ferraroni
- Dipartimento di Chimica "Ugo Schiff", Università degli Studi di Firenze, Via della Lastruccia 3-13, 50019 Florence, Italy
| | - Claudiu T Supuran
- Dipartimento di Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy
| | - Marina V Pokrovskaya
- Institute of Biomedical Chemistry, Pogodinskaya Street 10/8, Moscow 119121, Russia
| | | | - Yulia A Gladilina
- Institute of Biomedical Chemistry, Pogodinskaya Street 10/8, Moscow 119121, Russia
| | - Nikolay N Sokolov
- Institute of Biomedical Chemistry, Pogodinskaya Street 10/8, Moscow 119121, Russia
| | - Abdullah Hilal
- Institute of Biomedical Chemistry, Pogodinskaya Street 10/8, Moscow 119121, Russia
| | - Fabrizio Carta
- Dipartimento di Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy
| | - Dmitry D Zhdanov
- Institute of Biomedical Chemistry, Pogodinskaya Street 10/8, Moscow 119121, Russia
| |
Collapse
|
39
|
Bhari VK, Kumar D, Kumar S, Mishra R. Shelterin complex gene: Prognosis and therapeutic vulnerability in cancer. Biochem Biophys Rep 2021; 26:100937. [PMID: 33553693 PMCID: PMC7859307 DOI: 10.1016/j.bbrep.2021.100937] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/08/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Telomere encompasses a (TTAGGG)n tandem repeats, and its dysfunction has emerged as the epicenter of driving carcinogenesis by promoting genetic instability. Indeed, they play an essential role in stabilizing chromosomes and therefore protecting them from end-to-end fusion and DNA degradation. Telomere length homeostasis is regulated by several key players including shelterin complex genes, telomerase, and various other regulators. Targeting these regulatory players can be a good approach to combat cancer as telomere length is increasingly correlated with cancer initiation and progression. In this review, we have aimed to describe the telomere length regulator's role in prognostic significance and important drug targets in breast cancer. Moreover, we also assessed alteration in telomeric function by various telomere length regulators and compares this to the regulatory mechanisms that can be associated with clinical biomarkers in cancer. Using publicly available software we summarized mutational and CpG island prediction analysis of the TERT gene breast cancer patient database. Studies have reported that the TERT gene has prognostic significance in breast cancer progression however mechanistic approaches are not defined yet. Interestingly, we reported using the UCSC Xena web-based tool, we confirmed a positive correlation of shelterin complex genes TERF1 and TERF2 in recurrent free survival, indicating the critical role of these genes in breast cancer prognosis. Moreover, the epigenetic landscape of DNA damage repair genes in different breast cancer subtypes also being analyzed using the UCSC Xena database. Together, these datasets provide a comprehensive resource for shelterin complex gene profiles and define epigenetic landscapes of DNA damage repair genes which reveals the key role of shelterin complex genes in breast cancer with the potential to identify novel and actionable targets for treatment.
Collapse
Affiliation(s)
- Vikas Kumar Bhari
- Department of Biosciences, Manipal University Jaipur, Rajasthan, India
| | - Durgesh Kumar
- Department of Physiology, Government Medical College, Kannauj, Uttar Pradesh, India
| | - Surendra Kumar
- Department of Neurology, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Rajeev Mishra
- Department of Biosciences, Manipal University Jaipur, Rajasthan, India
| |
Collapse
|
40
|
Plyasova AA, Zhdanov DD. Alternative Splicing of Human Telomerase Reverse Transcriptase (hTERT) and Its Implications in Physiological and Pathological Processes. Biomedicines 2021; 9:526. [PMID: 34065134 PMCID: PMC8150890 DOI: 10.3390/biomedicines9050526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
Alternative splicing (AS) of human telomerase catalytic subunit (hTERT, human telomerase reverse transcriptase) pre-mRNA strongly regulates telomerase activity. Several proteins can regulate AS in a cell type-specific manner and determine the functions of cells. In addition to being involved in telomerase activity regulation, AS provides cells with different splice variants that may have alternative biological activities. The modulation of telomerase activity through the induction of hTERT AS is involved in the development of different cancer types and embryos, and the differentiation of stem cells. Regulatory T cells may suppress the proliferation of target human and murine T and B lymphocytes and NK cells in a contact-independent manner involving activation of TERT AS. This review focuses on the mechanism of regulation of hTERT pre-mRNA AS and the involvement of splice variants in physiological and pathological processes.
Collapse
Affiliation(s)
| | - Dmitry D. Zhdanov
- Institute of Biomedical Chemistry, Pogodinskaya st 10/8, 119121 Moscow, Russia;
| |
Collapse
|
41
|
TERT Promoter Alterations in Glioblastoma: A Systematic Review. Cancers (Basel) 2021; 13:cancers13051147. [PMID: 33800183 PMCID: PMC7962450 DOI: 10.3390/cancers13051147] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Glioblastoma is the most common malignant primary brain tumor in adults. Glioblastoma accounts for 2 to 3 cases per 100,000 persons in North America and Europe. Glioblastoma classification is now based on histopathological and molecular features including isocitrate dehydrogenase (IDH) mutations. At the end of the 2000s, genome-wide sequencing of glioblastoma identified recurrent somatic genetic alterations involved in oncogenesis. Among them, the alterations in the promoter region of the telomerase reverse transcriptase (TERTp) gene are highly recurrent and occur in 70% to 80% of all glioblastomas, including glioblastoma IDH wild type and glioblastoma IDH mutated. This review focuses on recent advances related to physiopathological mechanisms, diagnosis, and clinical implications. Abstract Glioblastoma, the most frequent and aggressive primary malignant tumor, often presents with alterations in the telomerase reverse transcriptase promoter. Telomerase is responsible for the maintenance of telomere length to avoid cell death. Telomere lengthening is required for cancer cell survival and has led to the investigation of telomerase activity as a potential mechanism that enables cancer growth. The aim of this systematic review is to provide an overview of the available data concerning TERT alterations and glioblastoma in terms of incidence, physiopathological understanding, and potential therapeutic implications.
Collapse
|
42
|
[Are telomeres and telomerase still relevant targets in oncology?]. Bull Cancer 2020; 108:30-38. [PMID: 33256968 DOI: 10.1016/j.bulcan.2020.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
|
43
|
Kamal S, Junaid M, Ejaz A, Bibi I, Akash MSH, Rehman K. The secrets of telomerase: Retrospective analysis and future prospects. Life Sci 2020; 257:118115. [PMID: 32698073 DOI: 10.1016/j.lfs.2020.118115] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/06/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022]
Abstract
Telomerase plays a significant role to maintain and regulate the telomere length, cellular immortality and senescence by the addition of guanine-rich repetitive sequences. Chronic inflammation or oxidative stress-induced infection downregulates TERT gene modifying telomerase activity thus contributing to the early steps of gastric carcinogenesis process. Furthermore, telomere-telomerase system performs fundamental role in the pathogenesis and progression of diabetes mellitus as well as in its vascular intricacy. The cessation of cell proliferation in cultured cells by inhibiting the telomerase activity of transformed cells renders the rationale for culling of telomerase as a target therapy for the treatment of metabolic disorders and various types of cancers. In this article, we have briefly described the role of immune system and malignant cells in the expression of telomerase with critical analysis on the gaps and potential for future studies. The key findings regarding the secrets of the telomerase summarized in this article will help in future treatment modalities for the prevention of various types of cancers and metabolic disorders notably diabetes mellitus.
Collapse
Affiliation(s)
- Shagufta Kamal
- Department of Biochemistry, Government College University Faisalabad, Pakistan
| | - Muhammad Junaid
- Department of Biochemistry, Government College University Faisalabad, Pakistan
| | - Arslan Ejaz
- Department of Biochemistry, Government College University Faisalabad, Pakistan
| | - Ismat Bibi
- Department of Chemistry, Islamia University, Bahawalpur, Pakistan
| | | | - Kanwal Rehman
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan.
| |
Collapse
|
44
|
Eckburg A, Dein J, Berei J, Schrank Z, Puri N. Oligonucleotides and microRNAs Targeting Telomerase Subunits in Cancer Therapy. Cancers (Basel) 2020; 12:E2337. [PMID: 32825005 PMCID: PMC7565511 DOI: 10.3390/cancers12092337] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022] Open
Abstract
Telomerase provides cancer cells with replicative immortality, and its overexpression serves as a near-universal marker of cancer. Anti-cancer therapeutics targeting telomerase have garnered interest as possible alternatives to chemotherapy and radiotherapy. Oligonucleotide-based therapies that inhibit telomerase through direct or indirect modulation of its subunits, human telomerase reverse transcriptase (hTERT) and human telomerase RNA gene (hTERC), are a unique and diverse subclass of telomerase inhibitors which hold clinical promise. MicroRNAs that play a role in the upregulation or downregulation of hTERT and respective progression or attenuation of cancer development have been effectively targeted to reduce telomerase activity in various cancer types. Tumor suppressor miRNAs, such as miRNA-512-5p, miRNA-138, and miRNA-128, and oncogenic miRNAs, such as miRNA-19b, miRNA-346, and miRNA-21, have displayed preclinical promise as potential hTERT-based therapeutic targets. Antisense oligonucleotides like GRN163L and T-oligos have also been shown to uniquely target the telomerase subunits and have become popular in the design of novel cancer therapies. Finally, studies suggest that G-quadruplex stabilizers, such as Telomestatin, preserve telomeric oligonucleotide architecture, thus inhibiting hTERC binding to the telomere. This review aims to provide an adept understanding of the conceptual foundation and current state of therapeutics utilizing oligonucleotides to target the telomerase subunits, including the advantages and drawbacks of each of these approaches.
Collapse
Affiliation(s)
| | | | | | | | - Neelu Puri
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, USA; (A.E.); (J.D.); (J.B.); (Z.S.)
| |
Collapse
|