1
|
Ren X, Liu X, Zhang Q, Yang C, Xu Z. Simultaneous imaging of telomerase activity and protein tyrosine kinase 7 in living cells during epithelial-mesenchymal transformation via a near-infrared light-activatable nanoprobe. Talanta 2025; 282:126993. [PMID: 39383724 DOI: 10.1016/j.talanta.2024.126993] [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: 07/28/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
Exploring the relationship between key regulation molecules (such as telomerase and protein tyrosine kinase 7) during epithelial-mesenchymal transformation of cells is beneficial for studying malignant tumor metastasis. Fluorescence is usually used for real-time monitoring the distribution and expression of regulatory molecules in living cells. However, the recognition function of these classical nanoprobes is "always active" due to the absence of exogenous control, which leads to the amplification of both the background signal and the response signal, making it difficult to distinguish changes in biomolecule expression levels. To improve the fluorescence ratio between tumor and normal cells, we constructed near-infrared light-activatable nanoprobes by engineering the functional units of catalytic hairpin assembly and integrating upconversion luminescence nanoparticles. Under near-infrared light irradiation, the nanoparticles, serving as a near-infrared-to-ultraviolet light transducer, induced the photolysis of the photo-cleavable linkers sealed in hairpins. The recognition function of the nanoprobes can be controlled by near-infrared light, preventing them from recognizing the targets in non-irradiated regions. By employing the nanoprobes, we realized simultaneous imaging of two regulatory molecules in living cells and observed an increase in telomerase activity and a decrease in protein tyrosine kinase 7 expression during drug-induced epithelial-mesenchymal transformation. This work provides a promising method for revealing changes and relationships of regulatory molecules during tumor metastasis.
Collapse
Affiliation(s)
- Xiuyan Ren
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Xiaopeng Liu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Qi Zhang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Chunguang Yang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China.
| |
Collapse
|
2
|
Adámik M, Soldánová Z, Drotárová M, Brečková K, Petr M, Helma R, Jenner LP, Vorlíčková M, Sýkorová E, Brázdová M. Human ARMC6 binds in vitro to both cancer genes and telomeric RNA, favoring G-quadruplex structure recognition. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195050. [PMID: 39029558 DOI: 10.1016/j.bbagrm.2024.195050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/25/2024] [Accepted: 07/13/2024] [Indexed: 07/21/2024]
Abstract
Armadillo repeat-containing proteins (ARMCs) are a large family found throughout eukaryotes, which play prominent roles in cell adhesion, signaling and cytoskeletal regulation. The ARMC6 protein is highly conserved in primates, including humans, but to date does not have a clear function beyond initial hints of a link to cancer and telomerase activity. We report here in vitro experiments showing ARMC6 binding to DNA promoter sequences from several cancer-related genes (e.g., EGFR, VEGF and c-MYC), and also to the telomeric RNA repeat (TERRA). ARMC6 binding activity appears to recognize G-quadruplex motifs, which are being increasingly implicated as structure-based protein binding sites in chromosome maintenance and repair. In vivo investigation of ARMC6 function revealed that when this protein is overexpressed in human cell lines, there is different expression of genes connected with oncogenic pathways and those implicated in downstream non-canonical telomerase pathways (e.g., VEGF, hTERT, c-MYC, ESM1, MMP3). ARMC6 is already known to interact with human shelterin protein TRF2 and telomerase. The protein binds G-quadruplex structures and does so preferentially to RNA over DNA. As such, this protein may be an example of how a non-canonical nucleic acid structural motif allows mediation between gene regulation and telomeric chromatin rearrangement pathways.
Collapse
Affiliation(s)
- Matej Adámik
- Institute of Biophysics, Academy of Sciences of the Czech Republic v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Zuzana Soldánová
- Department of Molecular Pharmacy, Faculty of Pharmacy, Masaryk University, Palackého 1/3, 612 42 Brno, Czech Republic
| | - Magdaléna Drotárová
- Institute of Biophysics, Academy of Sciences of the Czech Republic v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Katarína Brečková
- Department of Molecular Pharmacy, Faculty of Pharmacy, Masaryk University, Palackého 1/3, 612 42 Brno, Czech Republic
| | - Marek Petr
- Institute of Biophysics, Academy of Sciences of the Czech Republic v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Robert Helma
- Institute of Biophysics, Academy of Sciences of the Czech Republic v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Leon P Jenner
- Institute of Biophysics, Academy of Sciences of the Czech Republic v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Michaela Vorlíčková
- Institute of Biophysics, Academy of Sciences of the Czech Republic v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Eva Sýkorová
- Institute of Biophysics, Academy of Sciences of the Czech Republic v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Marie Brázdová
- Institute of Biophysics, Academy of Sciences of the Czech Republic v.v.i., Královopolská 135, 612 00 Brno, Czech Republic; Department of Molecular Pharmacy, Faculty of Pharmacy, Masaryk University, Palackého 1/3, 612 42 Brno, Czech Republic.
| |
Collapse
|
3
|
Shen Y, Zhao M, Zhao P, Meng L, Zhang Y, Zhang G, Taishi Y, Sun L. Molecular mechanisms and therapeutic potential of lithium in Alzheimer's disease: repurposing an old class of drugs. Front Pharmacol 2024; 15:1408462. [PMID: 39055498 PMCID: PMC11269163 DOI: 10.3389/fphar.2024.1408462] [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: 03/28/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline and memory loss. Despite advances in understanding the pathophysiological mechanisms of AD, effective treatments remain scarce. Lithium salts, recognized as mood stabilizers in bipolar disorder, have been extensively studied for their neuroprotective effects. Several studies indicate that lithium may be a disease-modifying agent in the treatment of AD. Lithium's neuroprotective properties in AD by acting on multiple neuropathological targets, such as reducing amyloid deposition and tau phosphorylation, enhancing autophagy, neurogenesis, and synaptic plasticity, regulating cholinergic and glucose metabolism, inhibiting neuroinflammation, oxidative stress, and apoptosis, while preserving mitochondrial function. Clinical trials have demonstrated that lithium therapy can improve cognitive function in patients with AD. In particular, meta-analyses have shown that lithium may be a more effective and safer treatment than the recently FDA-approved aducanumab for improving cognitive function in patients with AD. The affordability and therapeutic efficacy of lithium have prompted a reassessment of its use. However, the use of lithium may lead to potential side effects and safety issues, which may limit its clinical application. Currently, several new lithium formulations are undergoing clinical trials to improve safety and efficacy. This review focuses on lithium's mechanism of action in treating AD, highlighting the latest advances in preclinical studies and clinical trials. It also explores the side effects of lithium therapy and coping strategies, offering a potential therapeutic strategy for patients with AD.
Collapse
Affiliation(s)
- Yanxin Shen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Meng Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Panpan Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Lingjie Meng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Yan Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Yezi Taishi
- Department of Cadre Ward, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
4
|
Szyposzynska A, Bielawska-Pohl A, Paprocka M, Bar J, Murawski M, Klimczak A. Comparative Analysis of Primary Ovarian Cancer Cells and Established Cell Lines as a New Tool for Studies on Ovarian Cancer Cell Complexity. Int J Mol Sci 2024; 25:5384. [PMID: 38791431 PMCID: PMC11121816 DOI: 10.3390/ijms25105384] [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: 04/09/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Primary cancer cells reflect the genetic background and phenotype of a tumor. Immortalized cells with higher proliferation activity have an advantage over primary cells. The aim of the study was to immortalize the primary ovarian cancer (OvCa) cells using the plasmid-carrying human telomerase reverse transcriptase (hTERT) gene and compare their phenotype and biological activity with the primary cells. The primary OvCa3 A and OvCa7 A cells were isolated from the ascitic fluid of two high-grade serous ovarian cancer patients and were characterized using immunocytochemical methods, flow cytometry, real-time RT-PCR, Western blot, metabolic activity, and migratory potential. Both immortalized ovarian cancer cell lines mirrored the phenotype of primary cancer cells, albeit with modifications. The OvCa3 A hTERT cells kept the mesenchymal stem cell phenotype of CD73/CD90/CD105-positivity and were CD133-negative, whereas the cell population of OvCa7 A hTERT lost CD73 expression, but almost 90% of cells expressed the CD133 characteristic for the CSCs phenotype. Immortalized OvCa cells differed in gene expression level with respect to Sox2 and Oct4, which was associated with stemness properties. The OvCa7 A hTERT cells showed higher metabolic and migratory activity and ALDH1 expression than the corresponding primary OvCa cells. Both primary and immortalized cell lines were able to form spheroids. The newly established unique immortalized cell line OvCa7 A hTERT, with the characteristic of a serous ovarian cancer malignancy feature, and with the accumulation of the p53, Pax8, and overexpression of the CD133 and CD44 molecules, may be a useful tool for research on therapeutic approaches, especially those targeting CSCs in ovarian cancer and in preclinical 2D and 3D models.
Collapse
Affiliation(s)
- Agnieszka Szyposzynska
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.S.); (A.B.-P.)
| | - Aleksandra Bielawska-Pohl
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.S.); (A.B.-P.)
| | - Maria Paprocka
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.S.); (A.B.-P.)
| | - Julia Bar
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Marek Murawski
- 1st Department of Gynecology and Obstetrics, Wroclaw Medical University, 50-599 Wroclaw, Poland;
| | - Aleksandra Klimczak
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.S.); (A.B.-P.)
| |
Collapse
|
5
|
Yu M, Qin K, Fan J, Zhao G, Zhao P, Zeng W, Chen C, Wang A, Wang Y, Zhong J, Zhu Y, Wagstaff W, Haydon RC, Luu HH, Ho S, Lee MJ, Strelzow J, Reid RR, He TC. The evolving roles of Wnt signaling in stem cell proliferation and differentiation, the development of human diseases, and therapeutic opportunities. Genes Dis 2024; 11:101026. [PMID: 38292186 PMCID: PMC10825312 DOI: 10.1016/j.gendis.2023.04.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 03/18/2023] [Accepted: 04/12/2023] [Indexed: 02/01/2024] Open
Abstract
The evolutionarily conserved Wnt signaling pathway plays a central role in development and adult tissue homeostasis across species. Wnt proteins are secreted, lipid-modified signaling molecules that activate the canonical (β-catenin dependent) and non-canonical (β-catenin independent) Wnt signaling pathways. Cellular behaviors such as proliferation, differentiation, maturation, and proper body-axis specification are carried out by the canonical pathway, which is the best characterized of the known Wnt signaling paths. Wnt signaling has emerged as an important factor in stem cell biology and is known to affect the self-renewal of stem cells in various tissues. This includes but is not limited to embryonic, hematopoietic, mesenchymal, gut, neural, and epidermal stem cells. Wnt signaling has also been implicated in tumor cells that exhibit stem cell-like properties. Wnt signaling is crucial for bone formation and presents a potential target for the development of therapeutics for bone disorders. Not surprisingly, aberrant Wnt signaling is also associated with a wide variety of diseases, including cancer. Mutations of Wnt pathway members in cancer can lead to unchecked cell proliferation, epithelial-mesenchymal transition, and metastasis. Altogether, advances in the understanding of dysregulated Wnt signaling in disease have paved the way for the development of novel therapeutics that target components of the Wnt pathway. Beginning with a brief overview of the mechanisms of canonical and non-canonical Wnt, this review aims to summarize the current knowledge of Wnt signaling in stem cells, aberrations to the Wnt pathway associated with diseases, and novel therapeutics targeting the Wnt pathway in preclinical and clinical studies.
Collapse
Affiliation(s)
- Michael Yu
- School of Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Kevin Qin
- School of Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, The School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Guozhi Zhao
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Piao Zhao
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Zeng
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Neurology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523475, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Annie Wang
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yonghui Wang
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Clinical Laboratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai 200000, China
| | - Jiamin Zhong
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, The School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Sherwin Ho
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
6
|
Fahim SA, ElZohairy YA, Moustafa RI. Favipiravir, an antiviral drug, in combination with tamoxifen exerts synergistic effect in tamoxifen-resistant breast cancer cells via hTERT inhibition. Sci Rep 2024; 14:1844. [PMID: 38246945 PMCID: PMC10800350 DOI: 10.1038/s41598-024-51977-w] [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: 05/08/2022] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Tamoxifen (TAM) is one of the most successful treatments for breast cancer; however, TAM resistance continues to be a significant barrier. TAM resistance has been reported to be associated with increased expression of human telomerase reverse transcriptase (hTERT). This enzyme shares structural similarity with RNA-dependent RNA polymerase (RdRp) enzyme of RNA viruses, suggesting that RdRp inhibitors may also inhibit hTERT. Favipiravir (FAV) is an antiviral drug that inhibits RdRp of RNA viruses. Thus, we propose that FAV may also elicit an antitumor effect by suppressing hTERT. This study aimed to investigate the effect of FAV and TAM on TAM-resistant breast cancer (TAMR-1). The cell viabilities were determined. The levels of CDK1/ hTERT, in addition to regulators of hTERT-targeted signaling pathways were measured. Apoptosis, migration, and cell cycle distribution were also determined. Our data revealed that the combination of TAM and FAV suppressed cell proliferation synergistically (CI < 1) and resulted in a significant change in cell migration and apoptosis. Indeed, this was associated with reduced levels of hTERT and CDK1 and shift in the cell cycle distribution. Our findings suggest that the TAM/FAV combination exhibits synergistic effects against TAMR-1 human breast cancer cells by targeting hTERT.
Collapse
Affiliation(s)
- Sally A Fahim
- Department of Biochemistry, School of Pharmacy, Newgiza University (NGU), Newgiza, Km 22 Cairo-Alexandria Desert Road, 6th of October, P.O. Box 12577, Giza, Egypt.
| | - Yehia A ElZohairy
- School of Pharmacy, Newgiza University (NGU), Newgiza, Km 22 Cairo-Alexandria Desert Road, P.O. Box 12577, Giza, Egypt
| | - Rehab I Moustafa
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, Dokki, Giza, Egypt
- Microbiology Department, School of Pharmacy, Newgiza University (NGU), Newgiza, Km 22 Cairo-Alexandria Desert Road, P.O. Box 12577, Giza, Egypt
| |
Collapse
|
7
|
Nguyen QM, Dupré PF, Berchel M, Ghanem R, Jaffrès PA, d'Arbonneau F, Montier T. BSV163/DOPE-mediated TRAIL gene transfection acts synergistically with chemotherapy against cisplatin-resistant ovarian cancer. Chem Biol Drug Des 2024; 103:e14357. [PMID: 37731182 DOI: 10.1111/cbdd.14357] [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: 05/22/2023] [Revised: 08/04/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023]
Abstract
Ovarian cancer is the seventh most frequently diagnosed cancer among women worldwide. Most patients experience recurrence and succumb eventually to resistant disease, underscoring the need for an alternative treatment option. In the presented manuscript, we investigated the effect of the TRAIL-gene, transfected by an innovative bioinspired lipid vector BSV163/DOPE in the presence or absence of cisplatin, to fight against sensitive and resistant ovarian cancer. We showed that BSV163/DOPE can transfect ovarian cancer cell lines (Caov3, OVCAR3, and our new cisplatin-resistant, CR-Caov3) safely and efficiently. In addition, TRAIL-gene transfection in association with cisplatin inhibited cellular growth more efficiently (nearly 50% in Caov3 cells after the combined treatment, and 15% or 25% by each treatment alone, respectively) owing to an increase in apoptosis rate, caspases activity and TRAIL's death receptors expression. Most importantly, such synergistic effect was also observed in CR-Caov3 cells demonstrated by an apoptosis rate of 35% following the combined treatment in comparison with 17% after TRAIL-gene transfection or 6% after cisplatin exposition. These results suggest this combination may have potential application for sensitive as well as refractory ovarian cancer patients.
Collapse
Affiliation(s)
- Quoc Manh Nguyen
- Univ Brest, Inserm, EFS, UMR 1078, GGB, GTCA Team, "Gene Transfer and Combined therapeutic Approaches", Brest, France
| | - Pierre-François Dupré
- Univ Brest, Inserm, EFS, UMR 1078, GGB, GTCA Team, "Gene Transfer and Combined therapeutic Approaches", Brest, France
- CHU de Brest, Service de Chirurgie Gynécologique, Brest, France
| | | | - Rosy Ghanem
- Univ Brest, Inserm, EFS, UMR 1078, GGB, GTCA Team, "Gene Transfer and Combined therapeutic Approaches", Brest, France
- CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, Brest, France
| | | | - Frédérique d'Arbonneau
- Univ Brest, Inserm, EFS, UMR 1078, GGB, GTCA Team, "Gene Transfer and Combined therapeutic Approaches", Brest, France
- CHU de Brest, Service d'Odontologie, UFR d'Odontologie de Brest, Brest, France
| | - Tristan Montier
- Univ Brest, Inserm, EFS, UMR 1078, GGB, GTCA Team, "Gene Transfer and Combined therapeutic Approaches", Brest, France
- CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, Brest, France
- CHU de Brest, Centre de Référence des Maladies Rares "Maladies Neuromusculaires", Brest, France
| |
Collapse
|
8
|
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
|
9
|
Ye F, Dewanjee S, Li Y, Jha NK, Chen ZS, Kumar A, Vishakha, Behl T, Jha SK, Tang H. Advancements in clinical aspects of targeted therapy and immunotherapy in breast cancer. Mol Cancer 2023; 22:105. [PMID: 37415164 PMCID: PMC10324146 DOI: 10.1186/s12943-023-01805-y] [Citation(s) in RCA: 169] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023] Open
Abstract
Breast cancer is the second leading cause of death for women worldwide. The heterogeneity of this disease presents a big challenge in its therapeutic management. However, recent advances in molecular biology and immunology enable to develop highly targeted therapies for many forms of breast cancer. The primary objective of targeted therapy is to inhibit a specific target/molecule that supports tumor progression. Ak strain transforming, cyclin-dependent kinases, poly (ADP-ribose) polymerase, and different growth factors have emerged as potential therapeutic targets for specific breast cancer subtypes. Many targeted drugs are currently undergoing clinical trials, and some have already received the FDA approval as monotherapy or in combination with other drugs for the treatment of different forms of breast cancer. However, the targeted drugs have yet to achieve therapeutic promise against triple-negative breast cancer (TNBC). In this aspect, immune therapy has come up as a promising therapeutic approach specifically for TNBC patients. Different immunotherapeutic modalities including immune-checkpoint blockade, vaccination, and adoptive cell transfer have been extensively studied in the clinical setting of breast cancer, especially in TNBC patients. The FDA has already approved some immune-checkpoint blockers in combination with chemotherapeutic drugs to treat TNBC and several trials are ongoing. This review provides an overview of clinical developments and recent advancements in targeted therapies and immunotherapies for breast cancer treatment. The successes, challenges, and prospects were critically discussed to portray their profound prospects.
Collapse
Affiliation(s)
- Feng Ye
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Yuehua Li
- Department of Medical Oncology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, 11439, USA
| | - Ankush Kumar
- Pharmaceutical and Health Sciences, Career Point University, Hamirpur, Himachal Pradesh, India
| | - Vishakha
- Pharmaceutical and Health Sciences, Career Point University, Hamirpur, Himachal Pradesh, India
| | - Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, Uttarakhand, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India.
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India.
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India.
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
10
|
Yi W, Yang D, Xu Z, Chen Z, Xiao G, Qin L. Immortalization of mouse primary astrocytes. Gene 2023; 865:147327. [PMID: 36870428 DOI: 10.1016/j.gene.2023.147327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/08/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
In cell culture studies, immortalized primary cells have become a useful tool to investigate the molecular and cellular functions of different types of cells. Several immortalization agents, such as human telomerase reverse transcriptase (hTERT) and Simian Virus 40 (SV40) T antigens, are commonly used for primary cell immortalization. Astrocytes, as the most abundant glial cell type in the central nervous system, are promising therapeutical targets for many neuronal disorders, such as Alzheimer's disease and Parkinson's disease. Immortalized primary astrocytes can provide useful information for astrocytes biology, astrocytes-neuron interactions, glial interactions and astrocytes-associated neuronal diseases. In this study, we successfully purified primary astrocytes with immuno-panning method and examined the astrocyte functions after immortalization through both hTERT and SV40 Large-T antigens. As expected, both immortalized astrocytes presented unlimited lifespan and highly expressed multiple astrocyte-specific markers. However, SV40 Large-T antigen, but not hTERT, immortalized astrocytes displayed fast ATP-induced calcium wave in culture. Hence, SV40 Large-T antigen could be a better choice for primary astrocyte immortalization, which closely mimics the cell biology of primary astrocytes in culture. In summary, the purification and immortalization of primary astrocytes presented in this study can be used for studying astrocyte biology under physiological and pathological conditions.
Collapse
Affiliation(s)
- Weihong Yi
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China
| | - Dazhi Yang
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China
| | - Zhen Xu
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China
| | - Zecai Chen
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, 518055 Shenzhen, China.
| | - Lei Qin
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China.
| |
Collapse
|
11
|
Amin A, Morello M, Petrara MR, Rizzo B, Argenton F, De Rossi A, Giunco S. Short-Term TERT Inhibition Impairs Cellular Proliferation via a Telomere Length-Independent Mechanism and Can Be Exploited as a Potential Anticancer Approach. Cancers (Basel) 2023; 15:2673. [PMID: 37345011 DOI: 10.3390/cancers15102673] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/21/2023] [Accepted: 05/04/2023] [Indexed: 06/23/2023] Open
Abstract
Telomerase reverse transcriptase (TERT), the catalytic component of telomerase, may also contribute to carcinogenesis via telomere-length independent mechanisms. Our previous in vitro and in vivo studies demonstrated that short-term telomerase inhibition by BIBR1532 impairs cell proliferation without affecting telomere length. Here, we show that the impaired cell cycle progression following short-term TERT inhibition by BIBR1532 in in vitro models of B-cell lymphoproliferative disorders, i.e., Epstein-Barr virus (EBV)-immortalized lymphoblastoid cell lines (LCLs), and B-cell malignancies, i.e., Burkitt's lymphoma (BL) cell lines, is characterized by a significant reduction in NF-κB p65 nuclear levels leading to the downregulation of its target gene MYC. MYC downregulation was associated with increased expression and nuclear localization of P21, thus promoting its cell cycle inhibitory function. Consistently, treatment with BIBR1532 in wild-type zebrafish embryos significantly decreased Myc and increased p21 expression. The combination of BIBR1532 with antineoplastic drugs (cyclophosphamide or fludarabine) significantly reduced xenografted cells' proliferation rate compared to monotherapy in the zebrafish xenograft model. Overall, these findings indicate that short-term inhibition of TERT impairs cell growth through the downregulation of MYC via NF-κB signalling and supports the use of TERT inhibitors in combination with antineoplastic drugs as an efficient anticancer strategy.
Collapse
Affiliation(s)
- Aamir Amin
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, 35128 Padova, Italy
| | - Marzia Morello
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Maria Raffaella Petrara
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, 35128 Padova, Italy
| | - Beatrice Rizzo
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | | | - Anita De Rossi
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, 35128 Padova, Italy
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Silvia Giunco
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, 35128 Padova, Italy
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| |
Collapse
|
12
|
da Mota THA, Camargo R, Biojone ER, Guimarães AFR, Pittella-Silva F, de Oliveira DM. The Relevance of Telomerase and Telomere-Associated Proteins in B-Acute Lymphoblastic Leukemia. Genes (Basel) 2023; 14:genes14030691. [PMID: 36980962 PMCID: PMC10048576 DOI: 10.3390/genes14030691] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Telomeres and telomerase are closely linked to uncontrolled cellular proliferation, immortalization and carcinogenesis. Telomerase has been largely studied in the context of cancer, including leukemias. Deregulation of human telomerase gene hTERT is a well-established step in leukemia development. B-acute lymphoblastic leukemia (B-ALL) recovery rates exceed 90% in children; however, the relapse rate is around 20% among treated patients, and 10% of these are still incurable. This review highlights the biological and clinical relevance of telomerase for B-ALL and the implications of its canonical and non-canonical action on signaling pathways in the context of disease and treatment. The physiological role of telomerase in lymphocytes makes the study of its biomarker potential a great challenge. Nevertheless, many works have demonstrated that high telomerase activity or hTERT expression, as well as short telomeres, correlate with poor prognosis in B-ALL. Telomerase and related proteins have been proven to be promising pharmacological targets. Likewise, combined therapy with telomerase inhibitors may turn out to be an alternative strategy for B-ALL.
Collapse
Affiliation(s)
- Tales Henrique Andrade da Mota
- Laboratory of Molecular Pathology of Cancer, University of Brasilia, Brasilia 70910-900, Brazil
- Laboratory of Molecular Analysis, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, Brazil
- Correspondence:
| | - Ricardo Camargo
- Brasília Children’s Hospital José Alencar, Brasilia 70684-831, Brazil
| | | | - Ana Flávia Reis Guimarães
- Laboratory of Molecular Analysis, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, Brazil
| | - Fabio Pittella-Silva
- Laboratory of Molecular Pathology of Cancer, University of Brasilia, Brasilia 70910-900, Brazil
| | | |
Collapse
|
13
|
Themoteo RM, De Paula VJR, Rocha NKR, Brentani H, Forlenza OV. Lithium Prevents Telomere Shortening in Cortical Neurons in Amyloid-Beta Induced Toxicity. NEUROSCI 2023; 4:1-8. [PMID: 39484296 PMCID: PMC11523687 DOI: 10.3390/neurosci4010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND There is consistent evidence of the potential benefits of lithium attenuating mechanisms of neurodegeneration, including those related to the pathophysiology of Alzheimer's disease (AD), and facilitating neurotrophic and protective responses, including maintenance of telomere length. The aim was to investigate the protective effect of the pre-treatment with lithium on amyloid-beta (Aβ)-induced toxicity and telomere length in neurons. METHODS Cortical neurons were treated with lithium chloride at therapeutic and subtherapeutic concentrations (2 mM, 0.2 mM and 0.02 mM) for seven days. Amyloid toxicity was induced 24 h before the end of lithium treatment. RESULTS Lithium resulted in 120% (2 mM), 180% (0.2 mM) and 140% (0.02 mM) increments in telomere length as compared to untreated controls. Incubation with Aβ1-42 was associated with significant reductions in MTT uptake (33%) and telomere length (83%) as compared to controls. CONCLUSIONS Lithium prevented loss of culture viability and telomere shortening in neuronal cultures challenged with Aβ fibrils.
Collapse
Affiliation(s)
- Rafael M. Themoteo
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| | - Vanessa J. R. De Paula
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
- Laboratory of Psychobiology (LIM-23), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| | - Nicole K. R. Rocha
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
- Laboratory of Psychobiology (LIM-23), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| | - Helena Brentani
- Laboratory of Psychobiology (LIM-23), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| | - Orestes V. Forlenza
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| |
Collapse
|
14
|
Hasanau TN, Pisarev EP, Kisil OV, Zvereva ME. The TERT Promoter: A Key Player in the Fight for Cancer Cell Immortality. BIOCHEMISTRY (MOSCOW) 2023; 88:S21-S38. [PMID: 37069112 DOI: 10.1134/s000629792314002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The review describes the role of telomeres and telomerase in tumor progression, as well as various mechanisms of the activation of telomerase reverse transcriptase (TERT) expression in CNS tumors and other cancers. The main mechanism of TERT activation involves acquisition of somatic mutations by the TERT gene promoter (TERTp). The article presents information on the TERTp structure and transcription factors directly interacting with TERTp and regulating its transcription. The prospects of using the mutational status of TERTp as a prognostic marker of CNS malignancies and other tumors with a common profile of TERTp mutations are discussed.
Collapse
Affiliation(s)
- Tsimur N Hasanau
- Natural Compounds Department, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Eduard P Pisarev
- Natural Compounds Department, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Olga V Kisil
- Gause Institute of New Antibiotics, Moscow, 119021, Russia
| | - Maria E Zvereva
- Natural Compounds Department, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
15
|
Villarinho NJ, Vasconcelos FDC, Mazzoccoli L, da Silva Robaina MC, Pessoa LS, Siqueira PET, Maia RC, de Oliveira DM, Leite de Sampaio E Spohr TC, Lopes GF. Effects of long-term exposure to MST-312 on lung cancer cells tumorigenesis: Role of SHH/GLI-1 axis. Cell Biol Int 2022; 46:1468-1479. [PMID: 35811464 DOI: 10.1002/cbin.11843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 01/26/2022] [Accepted: 04/25/2022] [Indexed: 11/12/2022]
Abstract
Replicative immortality is a key feature of cancer cells and it is maintained by the expression of telomerase, a promising target of novel therapies. Long-term telomerase inhibition can induce resistance, but the mechanisms underlying this process remain unclear. The Sonic hedgehog pathway (SHH) is an embryogenic pathway involved in tumorigenesis and modulates the transcription of telomerase. We evaluated the effects of long-term treatment of the telomerase inhibitor MST-312 in morphology, proliferation, resistance, and in the SHH pathway molecules expression levels in lung cancer cells. Cells treated for 12 weeks with MST-312 showed changes in morphology, such as spindle-shaped cells, and a shift in the distribution of F-ACTIN from cortical to diffuse. Treatment also significantly reduced cells' efficiency to form spheroids and their clonogenic potential, independently of the cell cycle and telomeric DNA content. Moreover, GLI-1 expression levels were significantly reduced after 12 weeks of MST-312 treatment, indicating a possible inhibition of this signaling axis in the SHH pathway, without hindering NANOG and OCT4 expression. Here, we described a novel implication of long-term treatment with MST-312 functionally and molecularly, shedding new light on the molecular mechanisms of this drug in vitro.
Collapse
Affiliation(s)
- Nicolas Jones Villarinho
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer IECPN, Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Medicina (Anatomia Patológica), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavia da Cunha Vasconcelos
- Laboratório de Hemato-oncologia Celular e Molecular, Coordenação de Pesquisa, Instituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciano Mazzoccoli
- Laboratório de Hemato-oncologia Celular e Molecular, Coordenação de Pesquisa, Instituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcela Cristina da Silva Robaina
- Laboratório de Hemato-oncologia Celular e Molecular, Coordenação de Pesquisa, Instituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Santos Pessoa
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer IECPN, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pablo Enrique Torres Siqueira
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer IECPN, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel Ciuvalschi Maia
- Laboratório de Hemato-oncologia Celular e Molecular, Coordenação de Pesquisa, Instituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego Madureira de Oliveira
- Departamento de Bases Biológicas da Saúde, Universidade Federal de Brasília-Campus Ceilândia, Brasilia, Brazil
| | - Tania Cristina Leite de Sampaio E Spohr
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer IECPN, Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Medicina (Anatomia Patológica), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giselle Faria Lopes
- Programa de Pós-graduação em Medicina (Anatomia Patológica), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Hemato-oncologia Celular e Molecular, Coordenação de Pesquisa, Instituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brazil.,Divisão de Bioprodutos, Departamento de Biotecnologia Marinha, Instituto de Estudos do Mar Almirante Paulo Moreira (IEAPM), Arraial do Cabo, Brazil
| |
Collapse
|
16
|
Mota JIS, Silva-Júnior RMP, Martins CS, Bueno AC, Wildemberg LE, Antunes XLDS, Ozaki JGO, Coeli-Lacchini FB, Garcia-Peral C, Oliveira AER, Santos AC, Moreira AC, Machado HR, Dos Santos MV, Colli LM, Gadelha MR, Antonini SRR, de Castro M. Telomere length and Wnt/β-catenin pathway in adamantinomatous craniopharyngiomas. Eur J Endocrinol 2022; 187:219-230. [PMID: 35584004 DOI: 10.1530/eje-21-1269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/18/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVES To evaluate how telomere length behaves in adamantinomtous craniopharyngioma (aCP) and if it contributes to the pathogenesis of aCPs with and without CTNNB1 mutations. DESIGN Retrospective cross-sectional study enrolling 42 aCP patients from 2 tertiary institutions. METHODS Clinicopathological features were retrieved from the patient's charts. Fresh frozen tumors were used for RNA and DNA analyses. Telomere length was evaluated by qPCR (T/S ratio). Somatic mutations in TERT promoter (TERTp) and CTNNB1 were detected by Sanger and/or whole-exome sequencing. We performed RNA-Seq to identify differentially expressed genes in aCPs presenting with shorter or longer telomere lengths. RESULTS Mutations in CTNNB1 were detected in 29 (69%) tumors. There was higher frequency of CTNNB1 mutations in aCPs from patients diagnosed under the age of 15 years (85% vs 15%; P = 0.04) and a trend to recurrent disease (76% vs 24%; P = 0.1). No mutation was detected in the TERTp region. The telomeres were shorter in CTNNB1-mutated aCPs (0.441, IQR: 0.297-0.597vs 0.607, IQR: 0.445-0.778; P = 0.04), but it was neither associated with clinicopathological features nor with recurrence. RNAseq identified a total of 387 differentially expressed genes, generating two clusters, being one enriched for short telomeres and CTNNB1-mutated aCPs. CONCLUSIONS: CTNNB1 mutations are more frequent in children and adolescents and appear to associate with progressive disease. CTNNB1-mutated aCPs have shorter telomeres, demonstrating a relationship between the Wnt/β-catenin pathway and telomere biology in the pathogenesis of aCPs.
Collapse
Affiliation(s)
- Jose Italo Soares Mota
- Department of Internal Medicine of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | - Clarissa Silva Martins
- Department of Internal Medicine of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Ana Carolina Bueno
- Department of Pediatrics of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Luiz Eduardo Wildemberg
- Neuroendocrinology Research Center/Endocrinology Section, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ximene Lima da Silva Antunes
- Neuroendocrinology Research Center/Endocrinology Section, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge Guilherme Okanobo Ozaki
- Department of Internal Medicine of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | - Carlos Garcia-Peral
- Institute of Neuroscience of Castilla y León, University of Salamanca, Salamanca, Spain
| | - Antonio Edson Rocha Oliveira
- Department of Internal Medicine of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Antônio Carlos Santos
- Department of Medical Imaging, Hematology and Oncology of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Ayrton Custodio Moreira
- Department of Internal Medicine of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Helio Rubens Machado
- Department of Surgery and Anatomy of Ribeirao Preto Medical School, Hematology and Oncology of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Marcelo Volpon Dos Santos
- Department of Surgery and Anatomy of Ribeirao Preto Medical School, Hematology and Oncology of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Leandro M Colli
- Department of Medical Imaging, Hematology and Oncology of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Monica R Gadelha
- Neuroendocrinology Research Center/Endocrinology Section, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sonir Roberto R Antonini
- Department of Pediatrics of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Margaret de Castro
- Department of Internal Medicine of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
17
|
Akıncılar S, Chua J, Ng Q, Chan C, Eslami-S Z, Chen K, Low JL, Arumugam S, Aswad L, Chua C, Tan I, DasGupta R, Fullwood M, Tergaonkar V. Identification of mechanism of cancer-cell-specific reactivation of hTERT offers therapeutic opportunities for blocking telomerase specifically in human colorectal cancer. Nucleic Acids Res 2022; 51:1-16. [PMID: 35697349 PMCID: PMC9841410 DOI: 10.1093/nar/gkac479] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/18/2022] [Accepted: 05/26/2022] [Indexed: 01/29/2023] Open
Abstract
Transcriptional reactivation of hTERT is the limiting step in tumorigenesis. While mutations in hTERT promoter present in 19% of cancers are recognized as key drivers of hTERT reactivation, mechanisms by which wildtype hTERT (WT-hTERT) promoter is reactivated, in majority of human cancers, remain unknown. Using primary colorectal cancers (CRC) we identified Tert INTeracting region 2 (T-INT2), the critical chromatin region essential for reactivating WT-hTERT promoter in CRCs. Elevated β-catenin and JunD level in CRC facilitates chromatin interaction between hTERT promoter and T-INT2 that is necessary to turn on hTERTexpression. Pharmacological screens uncovered salinomycin, which inhibits JunD mediated hTERT-T-INT2 interaction that is required for the formation of a stable transcription complex on the hTERT promoter. Our results showed for the first time how known CRC alterations, such as APC, lead to WT-hTERT promoter reactivation during stepwise-tumorigenesis and provide a new perspective for developing cancer-specific drugs.
Collapse
Affiliation(s)
- Semih Can Akıncılar
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Joelle Yi Heng Chua
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Qin Feng Ng
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Claire Hian Tzer Chan
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Zahra Eslami-S
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Kaijing Chen
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Joo-Leng Low
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, 138672, Singapore
| | - Surendar Arumugam
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Luay Aswad
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Clarinda Chua
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 138672, Singapore,Department of Medical Oncology, National Cancer Centre Singapore, 169610, Singapore
| | - Iain Beehuat Tan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 138672, Singapore,Department of Medical Oncology, National Cancer Centre Singapore, 169610, Singapore
| | - Ramanuj DasGupta
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, 138672, Singapore
| | - Melissa Jane Fullwood
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore,School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Vinay Tergaonkar
- To whom correspondence should be addressed. Tel: +65 65869836; Fax: +65 67791117;
| |
Collapse
|
18
|
Fernandez RJ, Gardner ZJG, Slovik KJ, Liberti DC, Estep KN, Yang W, Chen Q, Santini GT, Perez JV, Root S, Bhatia R, Tobias JW, Babu A, Morley MP, Frank DB, Morrisey EE, Lengner CJ, Johnson FB. GSK3 inhibition rescues growth and telomere dysfunction in dyskeratosis congenita iPSC-derived type II alveolar epithelial cells. eLife 2022; 11:64430. [PMID: 35559731 PMCID: PMC9200405 DOI: 10.7554/elife.64430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/11/2022] [Indexed: 11/27/2022] Open
Abstract
Dyskeratosis congenita (DC) is a rare genetic disorder characterized by deficiencies in telomere maintenance leading to very short telomeres and the premature onset of certain age-related diseases, including pulmonary fibrosis (PF). PF is thought to derive from epithelial failure, particularly that of type II alveolar epithelial (AT2) cells, which are highly dependent on Wnt signaling during development and adult regeneration. We use human induced pluripotent stem cell-derived AT2 (iAT2) cells to model how short telomeres affect AT2 cells. Cultured DC mutant iAT2 cells accumulate shortened, uncapped telomeres and manifest defects in the growth of alveolospheres, hallmarks of senescence, and apparent defects in Wnt signaling. The GSK3 inhibitor, CHIR99021, which mimics the output of canonical Wnt signaling, enhances telomerase activity and rescues the defects. These findings support further investigation of Wnt agonists as potential therapies for DC-related pathologies.
Collapse
Affiliation(s)
- Rafael Jesus Fernandez
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Zachary J G Gardner
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Katherine J Slovik
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Derek C Liberti
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Katrina N Estep
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Wenli Yang
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Qijun Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Garrett T Santini
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Javier V Perez
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Sarah Root
- College of Arts and Sciences and Vagelos Scholars Program, University of Pennsylvania, Philadelphia, United States
| | - Ranvir Bhatia
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - John W Tobias
- Penn Genomic Analysis Core, University of Pennsylvania, Philadelphia, United States
| | - Apoorva Babu
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - Michael P Morley
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - David B Frank
- Penn-CHOP Lung Biology Institute, Children's Hospital of Philadelphia, Philadelphia, United States
| | - Edward E Morrisey
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Christopher J Lengner
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, United States
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
19
|
Yuan H, Wu Y, Wang J, Qin X, Huang Y, Yan L, Fana Y, Zedenius J, Juhlin CC, Larsson C, Lui WO, Xu D. Synergistic effects of telomerase reverse transcriptase and regulator of telomere elongation helicase 1 on aggressiveness and outcomes in adrenocortical carcinoma. Biomed Pharmacother 2022; 149:112796. [PMID: 35279598 DOI: 10.1016/j.biopha.2022.112796] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is one of the deadliest endocrine malignancies and telomere maintenance by activated telomerase is critically required for ACC development and progression. Because telomerase reverse transcriptase (TERT) and regulator of telomere elongation helicase 1 (RTEL1) play key roles in telomere homeostasis, we determined their effect on ACC pathogenesis and outcomes. Analyses of TCGA and GEO datasets showed significantly higher expression of RTEL1 but not TERT in ACC tumors, compared to their benign or normal counterparts. Furthermore, gains/amplifications of both TERT and RTEL1 genes were widespread in ACC tumors and their expression correlated with their gene copy numbers. Higher expression of either TERT or RTEL1 was associated with shorter overall and progression-free survival (OS and PFS) in the TCGA ACC patient cohort, and higher levels of both TERT and RTEL1 mRNA predicted the shortest patient OS and PFS. However, multivariate analyses showed that only RTEL1 independently predicted patient OS and PFS. Gene set enrichment analysis further showed enrichments of wnt/β-catenin, MYC, glycolysis, MTOR, and DNA repair signaling pathways in ACC tumors expressing high TERT and RTEL1 mRNA levels. Taken together, TERT and RTEL1 promote ACC aggressiveness synergistically and may serve as prognostic factors and therapeutic targets for ACC.
Collapse
Affiliation(s)
- Huiyang Yuan
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yujiao Wu
- Department of Medicine, Division of Hematology, Bioclinicum and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm SE-171 64, Sweden
| | - Jing Wang
- Department of Urologic Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Xin Qin
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yongsheng Huang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lei Yan
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Yidong Fana
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Department of Medicine, Division of Hematology, Bioclinicum and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm SE-171 64, Sweden; Department of Urologic Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China; Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm SE-171 64, Sweden; Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital Solna, Stockholm SE-171 64, Sweden; Department of Pathology and Cancer Diagnostics, Karolinska University Hospital Solna, Stockholm SE-171 64, Sweden; Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital Solna, Bioclinicum, Stockholm SE-171 64, Sweden.
| | - Jan Zedenius
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm SE-171 64, Sweden; Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital Solna, Stockholm SE-171 64, Sweden
| | - C Christofer Juhlin
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital Solna, Stockholm SE-171 64, Sweden; Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital Solna, Bioclinicum, Stockholm SE-171 64, Sweden
| | - Catharina Larsson
- Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital Solna, Bioclinicum, Stockholm SE-171 64, Sweden
| | - Weng-Onn Lui
- Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital Solna, Bioclinicum, Stockholm SE-171 64, Sweden
| | - Dawei Xu
- Department of Medicine, Division of Hematology, Bioclinicum and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm SE-171 64, Sweden.
| |
Collapse
|
20
|
Polho GB, Cardillo GM, Kerr DS, Chile T, Gattaz WF, Forlenza OV, Brentani HP, De-Paula VJ. Antipsychotics preserve telomere length in peripheral blood mononuclear cells after acute oxidative stress injury. Neural Regen Res 2022; 17:1156-1160. [PMID: 34558545 PMCID: PMC8552857 DOI: 10.4103/1673-5374.324852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/04/2021] [Accepted: 05/12/2021] [Indexed: 11/04/2022] Open
Abstract
Antipsychotics may prolong or retain telomere length, affect mitochondrial function, and then affect the metabolism of nerve cells. To validate the hypothesis that antipsychotics can prolong telomere length after oxidative stress injury, leukocytes from healthy volunteers were extracted using Ficoll-Histopaque density gradient. The mononuclear cells layer was resuspended in cell culture medium. Oxidative stress was induced with hydrogen peroxide in cultured leukocytes. Four days later, leukocytes were treated with aripiprazole, haloperidol or clozapine for 7 days. Real-time PCR revealed that treatments with aripiprazole and haloperidol increased the telomere length by 23% and 20% in peripheral blood mononuclear cells after acute oxidative stress injury. These results suggest that haloperidol and aripiprazole can reduce the damage to telomeres induced by oxidative stress. The experiment procedure was approved by the Ethics Committee of Faculty of Medicine of the University of São Paulo (FMUSP/CAAE approval No. 52622616.8.0000.0065).
Collapse
Affiliation(s)
- Gabriel B. Polho
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brasil
| | - Giancarlo M. Cardillo
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brasil
| | - Daniel S. Kerr
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brasil
| | - Thais Chile
- Laboratório de Psicobiologia (LIM-23), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brasil
| | - Wagner F. Gattaz
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brasil
| | - Orestes V. Forlenza
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brasil
| | - Helena P. Brentani
- Laboratório de Psicobiologia (LIM-23), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brasil
| | - Vanessa J. De-Paula
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brasil
- Laboratório de Psicobiologia (LIM-23), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brasil
| |
Collapse
|
21
|
Rafat A, Dizaji Asl K, Mazloumi Z, Movassaghpour AA, Farahzadi R, Nejati B, Nozad Charoudeh H. Telomerase-based therapies in haematological malignancies. Cell Biochem Funct 2022; 40:199-212. [PMID: 35103334 DOI: 10.1002/cbf.3687] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/10/2022] [Indexed: 02/02/2023]
Abstract
Telomeres are specialized genetic structures present at the end of all eukaryotic linear chromosomes. They progressively get shortened after each cell division due to end replication problems. Telomere shortening (TS) and chromosomal instability cause apoptosis and massive cell death. Following oncogene activation and inactivation of tumour suppressor genes, cells acquire mechanisms such as telomerase expression and alternative lengthening of telomeres to maintain telomere length (TL) and prevent initiation of cellular senescence or apoptosis. Significant TS, telomerase activation and alteration in expression of telomere-associated proteins are frequent features of different haematological malignancies that reflect on the progression, response to therapy and recurrence of these diseases. Telomerase is a ribonucleoprotein enzyme that has a pivotal role in maintaining the TL. However, telomerase activity in most somatic cells is insufficient to prevent TS. In 85-90% of tumour cells, the critically short telomeric length is maintained by telomerase activation. Thus, overexpression of telomerase in most tumour cells is a potential target for cancer therapy. In this review, alteration of telomeres, telomerase and telomere-associated proteins in different haematological malignancies and related telomerase-based therapies are discussed.
Collapse
Affiliation(s)
- Ali Rafat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Dizaji Asl
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Mazloumi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Babak Nejati
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
22
|
Xiang Y, Liang C, Li Q, Chen Q, Zhou Y, Zheng X, Zhou D, Wang Z, Wang G, Cao W. Chicken telomerase reverse transcriptase promotes the tumorigenicity of avian leukosis virus subgroup J by regulating the Wnt/β-catenin signaling pathway. Vet Res 2022; 53:100. [PMID: 36461084 PMCID: PMC9717515 DOI: 10.1186/s13567-022-01120-2] [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: 07/25/2022] [Accepted: 10/24/2022] [Indexed: 12/03/2022] Open
Abstract
This research aimed to analyze the regulatory effect of chicken telomerase reverse transcriptase (chTERT) on the Wnt/β-catenin signaling pathway and its effect on the tumorigenicity of avian leukosis virus subgroup J (ALV-J) through in vivo experiments. The chTERT eukaryotic expression plasmid and its recombinant lentivirus particles were constructed for in vivo transfection of chTERT to analyze the effect of chTERT continuously overexpressed in chickens on the tumorigenicity of ALV-J. During 156 days of the artificial ALV-J tumor-inducing process, 7 solid tumors developed in 3 chickens in the chTERT-overexpression group (n = 26*2) and no tumors developed in the control group (n = 26*2). Another 18 tumors induced by ALV-J were confirmed and collected from breeding poultry farms. And we confirmed that chTERT was significantly highly expressed in ALV-J tumors. The ELISA data suggested that the protein levels of β-catenin and c-Myc in the chicken plasma of the chTERT-overexpressing group with ALV-J infected were consistently and significantly higher than those of the control group. Compared with that of the tumor-adjacent tissues, the activity of the Wnt/β-catenin signaling pathway and expression of the c-Myc was significantly increased in ALV-J tumors. And the percentage of apoptosis in ALV-J tumors significantly lower than that in tumor-adjacent tissues. Immunohistochemistry, Western blot and RT-qPCR suggested that the replication level of ALV-J in tumors was significantly higher than that in tumor-adjacent tissues. This study suggests that chTERT plays a critical role in the tumorigenicity of ALV-J by enhancing the Wnt/β-catenin signaling pathway, which will contribute to further elucidating the tumor-inducing mechanism of ALV-J.
Collapse
Affiliation(s)
- Yong Xiang
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Canxin Liang
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Qingbo Li
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Qinxi Chen
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Yang Zhou
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Xiaoxue Zheng
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Di Zhou
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Zepeng Wang
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Guyao Wang
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Weisheng Cao
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China ,grid.20561.300000 0000 9546 5767Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, 510642 China ,grid.20561.300000 0000 9546 5767National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, 510642 China ,grid.20561.300000 0000 9546 5767Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642 China ,grid.20561.300000 0000 9546 5767Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642 China
| |
Collapse
|
23
|
S L, A S, Dv S, Bs R, R S, Sharaf S, Sa A, G R. Comparative differential cytotoxicity of clinically used SERMs in human cancer lines of different origin and its predictive molecular docking studies of key target genes involved in cancer progression and treatment responses. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 3:100080. [PMID: 35059624 PMCID: PMC8760488 DOI: 10.1016/j.crphar.2021.100080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
SERMS like Tamoxifene, 5-hydroxy tamoxifene, raloxifene and endoxifene has been used for the treatment of hormonal imbalances and dependent cancers owing to their action via Estrogen receptors as in the treatment of estrogen sensitive breast cancers. Due to the adverse side effects, modifications and development of the existing or newer SERMS has always been of immense interest. Ormeloxifene, a SERM molecule manufactured by HLL Lifecare Ltd, India as birth control under the trade names Saheli, Novex, and Novex-DS which is also investigated against mastalgia, fibro-adenoma and abnormal uterine bleeding. Anti-cancer effects have been reported in estrogen dependent and independent cancers which shows its wide scope to be implemented in cancer therapy. Current investigation is a comprehensive effort to find the cytotoxic potential of Ormeloxifene in comparison with clinically used four SERMS in twenty six cancer cell lines of different origin using Adriamycin as positive control. Also the computational studies pertaining to selected target/ligand with respect to tumor progression, development, treatment responses and apoptosis. The studies proved effective cytotoxicity of Ormeloxifene on cancer cell lines with lower TGI, GI50 and LC50 values which are significantly comparable. Also the in silico studies proved that the docking score of the compound suggests the interaction of the compound which could tightly regulate key target genes controlling cancer like ER, EGFR kinase, EGFR-cSRC, HDAC-2, PARP-1 and BRAF. This study brings out the superior efficacy of Ormeloxifene compared to other SERMS with proven safety profile to be repositioned as an anti-cancer drug to treat diverse cancer types.
Collapse
Affiliation(s)
- Lakshmi S
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Shanitha A
- Dept. of Computational Biology & Bioinformatics, University of Kerala, Thiruvananthapuram, Pincode-695581, India
| | - Shiny Dv
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Rahul Bs
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Saikant R
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Shehna Sharaf
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Abi Sa
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Rajmohan G
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| |
Collapse
|
24
|
Zhu Z, Tran H, Mathahs MM, Fink BD, Albert JA, Moninger TO, Meier JL, Li M, Schmidt WN. Zinc protoporphyrin binding to telomerase complexes and inhibition of telomerase activity. Pharmacol Res Perspect 2021; 9:e00882. [PMID: 34747573 PMCID: PMC8573827 DOI: 10.1002/prp2.882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022] Open
Abstract
Zinc protoporphyrin (ZnPP), a naturally occurring metalloprotoporphyrin (MPP), is currently under development as a chemotherapeutic agent although its mechanism is unclear. When tested against other MPPs, ZnPP was the most effective DNA synthesis and cellular proliferation inhibitor while promoting apoptosis in telomerase positive but not telomerase negative cells. Concurrently, ZnPP down-regulated telomerase expression and was the best overall inhibitor of telomerase activity in intact cells and cellular extracts with IC50 and EC50 values of ca 2.5 and 6 µM, respectively. The natural fluorescence properties of ZnPP enabled direct imaging in cellular fractions using non-denaturing agarose gel electrophoresis, western blots, and confocal fluorescence microscopy. ZnPP localized to large cellular complexes (>600 kD) that contained telomerase and dysskerin as confirmed with immunocomplex mobility shift, immunoprecipitation, and immunoblot analyses. Confocal fluorescence studies showed that ZnPP co-localized with telomerase reverse transcriptase (TERT) and telomeres in the nucleus of synchronized S-phase cells. ZnPP also co-localized with TERT in the perinuclear regions of log phase cells but did not co-localize with telomeres on the ends of metaphase chromosomes, a site known to be devoid of telomerase complexes. Overall, these results suggest that ZnPP does not bind to telomeric sequences per se, but alternatively, interacts with other structural components of the telomerase complex to inhibit telomerase activity. In conclusion, ZnPP actively interferes with telomerase activity in neoplastic cells, thus promoting pro-apoptotic and anti-proliferative properties. These data support further development of natural or synthetic protoporphyrins for use as chemotherapeutic agents to augment current treatment protocols for neoplastic disease.
Collapse
Affiliation(s)
- Zhaowen Zhu
- Department of Internal Medicine and Research ServiceVeterans Affairs Medical CenterIowa CityIowaUSA
- Department of Internal MedicineRoy G. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Huy Tran
- Department of Internal MedicineRoy G. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Meleah M. Mathahs
- Department of Internal Medicine and Research ServiceVeterans Affairs Medical CenterIowa CityIowaUSA
| | - Brian D. Fink
- Department of Internal Medicine and Research ServiceVeterans Affairs Medical CenterIowa CityIowaUSA
| | - John A. Albert
- Department of Internal Medicine and Research ServiceVeterans Affairs Medical CenterIowa CityIowaUSA
| | - Thomas O. Moninger
- Central Microscopy Research Facility Roy G. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Jeffery L. Meier
- Department of Internal Medicine and Research ServiceVeterans Affairs Medical CenterIowa CityIowaUSA
- Department of Internal MedicineRoy G. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Ming Li
- Department of Internal Medicine and Research ServiceVeterans Affairs Medical CenterIowa CityIowaUSA
| | - Warren N. Schmidt
- Department of Internal Medicine and Research ServiceVeterans Affairs Medical CenterIowa CityIowaUSA
- Department of Internal MedicineRoy G. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
25
|
Wnt signaling and Hedgehog expression in basal cell carcinoma. EUROPEAN JOURNAL OF PLASTIC SURGERY 2021. [DOI: 10.1007/s00238-021-01920-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
Nishi Y, Aoki T, Shimizu T, Sato S, Matsumoto T, Shiraki T, Sakuraoka Y, Mori S, Iso Y, Ishizuka M, Kubota K. Significance of cytoplasmic expression of telomerase reverse transcriptase in patients with hepatocellular carcinoma undergoing liver resection. Mol Clin Oncol 2021; 15:244. [PMID: 34650811 PMCID: PMC8506680 DOI: 10.3892/mco.2021.2406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/13/2021] [Indexed: 12/04/2022] Open
Abstract
Telomerase reverse transcriptase (TERT) is reportedly expressed in various types of cancer. However, to the best of our knowledge, the significance of subcellular expression of TERT in hepatocellular carcinoma (HCC) has not been evaluated in detail. The present study evaluated TERT expression in resected HCC tumor tissues using immunohistochemistry. TERT expression was assessed in both the cytoplasm and the nucleus of HCC cells. The associations between TERT expression and clinical characteristics, including expression levels of DNA-dependent protein kinase catalytic unit (DNA-PKcs) and 8-hydroxyganosine (8-OHdG), were investigated. Among the 135 HCCs, TERT expression was positive only in the cytoplasm in 86 tumors (63.7%), was positive only in the nucleus in 3 tumors (2.2%), was positive in both the cytoplasm and the nucleus in 5 tumors (3.7%) and was negative in 41 tumors (30.4%). Similar results were confirmed using another antibody for TERT. Cytoplasmic TERT expression was markedly associated with hepatitis B surface antigen, poor tumor differentiation, and expression levels of DNA-PKcs and 8-OHdG. However, TERT expression in the cytoplasm or in the nucleus was not significantly associated with the overall or recurrence-free survival periods. In conclusion, TERT was mainly expressed in the cytoplasm of HCC tissues. Cytoplasmic TERT expression was closely associated with hepatitis B virus-related HCC and DNA-PKcs expression, as well as oxidative stress.
Collapse
Affiliation(s)
- Yusuke Nishi
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Taku Aoki
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Takayuki Shimizu
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Shun Sato
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Takatsugu Matsumoto
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Takayuki Shiraki
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Yuhki Sakuraoka
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Shozo Mori
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Yukihiro Iso
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Mitsuru Ishizuka
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Keiichi Kubota
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| |
Collapse
|
27
|
TRIM28 is a transcriptional activator of the mutant TERT promoter in human bladder cancer. Proc Natl Acad Sci U S A 2021; 118:2102423118. [PMID: 34518220 DOI: 10.1073/pnas.2102423118] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 12/20/2022] Open
Abstract
Bladder cancer (BC) has a 70% telomerase reverse transcriptase (TERT or hTERT in humans) promoter mutation prevalence, commonly at -124 base pairs, and this is associated with increased hTERT expression and poor patient prognosis. We inserted a green fluorescent protein (GFP) tag in the mutant hTERT promoter allele to create BC cells expressing an hTERT-GFP fusion protein. These cells were used in a fluorescence-activated cell sorting-based pooled CRISPR-Cas9 Kinome knockout genetic screen to identify tripartite motif containing 28 (TRIM28) and TRIM24 as regulators of hTERT expression. TRIM28 activates, while TRIM24 suppresses, hTERT transcription from the mutated promoter allele. TRIM28 is recruited to the mutant promoter where it interacts with TRIM24, which inhibits its activity. Phosphorylation of TRIM28 through the mTOR complex 1 (mTORC1) releases it from TRIM24 and induces hTERT transcription. TRIM28 expression promotes in vitro and in vivo BC cell growth and stratifies BC patient outcome. mTORC1 inhibition with rapamycin analog Ridaforolimus suppresses TRIM28 phosphorylation, hTERT expression, and cell viability. This study may lead to hTERT-directed cancer therapies with reduced effects on normal progenitor cells.
Collapse
|
28
|
Kotiyal S, Evason KJ. Exploring the Interplay of Telomerase Reverse Transcriptase and β-Catenin in Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13164202. [PMID: 34439356 PMCID: PMC8393605 DOI: 10.3390/cancers13164202] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Liver cancer is one of the deadliest human cancers. Two of the most common molecular aberrations in liver cancer are: (1) activating mutations in the gene encoding β-catenin (CTNNB1); and (2) promoter mutations in telomerase reverse transcriptase (TERT). Here, we review recent findings regarding the interplay between TERT and β-catenin in order to better understand their role in liver cancer. Abstract Hepatocellular carcinoma (HCC) is one of the deadliest human cancers. Activating mutations in the telomerase reverse transcriptase (TERT) promoter (TERTp) and CTNNB1 gene encoding β-catenin are widespread in HCC (~50% and ~30%, respectively). TERTp mutations are predicted to increase TERT transcription and telomerase activity. This review focuses on exploring the role of TERT and β-catenin in HCC and the current findings regarding their interplay. TERT can have contradictory effects on tumorigenesis via both its canonical and non-canonical functions. As a critical regulator of proliferation and differentiation in progenitor and stem cells, activated β-catenin drives HCC; however, inhibiting endogenous β-catenin can also have pro-tumor effects. Clinical studies revealed a significant concordance between TERTp and CTNNB1 mutations in HCC. In stem cells, TERT acts as a co-factor in β-catenin transcriptional complexes driving the expression of WNT/β-catenin target genes, and β-catenin can bind to the TERTp to drive its transcription. A few studies have examined potential interactions between TERT and β-catenin in HCC in vivo, and their results suggest that the coexpression of these two genes promotes hepatocarcinogenesis. Further studies are required with vertebrate models to better understand how TERT and β-catenin influence hepatocarcinogenesis.
Collapse
|
29
|
Xiang Y, Yu Y, Li Q, Jiang Z, Li J, Liang C, Chen J, Li Y, Chen X, Cao W. Mutual regulation between chicken telomerase reverse transcriptase and the Wnt/β-catenin signalling pathway inhibits apoptosis and promotes the replication of ALV-J in LMH cells. Vet Res 2021; 52:110. [PMID: 34412690 PMCID: PMC8375160 DOI: 10.1186/s13567-021-00979-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/10/2021] [Indexed: 12/14/2022] Open
Abstract
This study aimed to explore the mutual regulation between chicken telomerase reverse transcriptase (chTERT) and the Wnt/β-catenin signalling pathway and its effects on cell growth and avian leukosis virus subgroup J (ALV-J) replication in LMH cells. First, LMH cells stably overexpressing the chTERT gene (LMH-chTERT cells) and corresponding control cells (LMH-NC cells) were successfully constructed with a lentiviral vector expression system. The results showed that chTERT upregulated the expression of β-catenin, Cyclin D1, TCF4 and c-Myc. chTERT expression level and telomerase activity were increased when cells were treated with LiCl. When the cells were treated with ICG001 or IWP-2, the activity of the Wnt/β-catenin signalling pathway was significantly inhibited, and chTERT expression and telomerase activity were also inhibited. However, when the β-catenin gene was knocked down by small interfering RNA (siRNA), the changes in chTERT expression and telomerase activity were consistent with those in cells treated with ICG001 or IWP-2. These results indicated that chTERT and the Wnt/β-catenin signalling pathway can be mutually regulated. Subsequently, we found that chTERT not only shortened the cell cycle to promote proliferation but also inhibited apoptosis by downregulating the expression of Caspase 3, Caspase 9 and BAX; upregulating BCL-2 and BCL-X expression; and promoting autophagy. Moreover, chTERT significantly enhanced the migration ability of LMH cells, upregulated the protein and mRNA expression of ALV-J and increased the virus titre. ALV-J replication promoted chTERT expression and telomerase activity.
Collapse
Affiliation(s)
- Yong Xiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yun Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qingbo Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zeng Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jinqun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Canxin Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoyan Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weisheng Cao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China. .,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, 510642, China. .,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China. .,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, 510642, China. .,Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
30
|
Mini-review: The anti-aging effects of lithium in bipolar disorder. Neurosci Lett 2021; 759:136051. [PMID: 34139318 DOI: 10.1016/j.neulet.2021.136051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 11/22/2022]
Abstract
The medical use of lithium has grown since its initial introduction in the 1800s as a treatment for gout. Today, the divalent cation remains as the pharmacological gold standard in treatment of bipolar disorder (BD) with strong mood stabilizing effects. Lithium has demonstrated efficacy in the treatment of acute affective episodes, in the reduction of affective episode recurrence, and in significantly decreasing the risk of suicide in patients. BD has been consistently associated with clinical signs of accelerated aging, including increased rates of age-related diseases such as cardiovascular diseases, malignancies, and diabetes mellitus. This clinical scenario parallels accelerated aging mechanisms observed on a molecular basis, with studies reporting shortened telomeres, increased oxidative stress, and accelerated epigenetic aging in patients with BD compared to controls. Lithium has proved useful as a potential agent in slowing down this accelerated aging process in BD, potentially reversing effects induced by the disorder. This mini-review summarizes findings of anti-aging mechanisms associated with lithium use and provides a discussion of the clinical implications and perspectives of this evolving field. Despite many promising results, more studies are warranted in order to elucidate the exact mechanism by which lithium may act as an anti-aging agent and the extent to which these mechanisms are relevant to its mood stabilizing properties in BD.
Collapse
|
31
|
Subasri M, Shooshtari P, Watson AJ, Betts DH. Analysis of TERT Isoforms across TCGA, GTEx and CCLE Datasets. Cancers (Basel) 2021; 13:cancers13081853. [PMID: 33924498 PMCID: PMC8070023 DOI: 10.3390/cancers13081853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/01/2021] [Accepted: 04/08/2021] [Indexed: 12/14/2022] Open
Abstract
Reactivation of the multi-subunit ribonucleoprotein telomerase is the primary telomere maintenance mechanism in cancer, but it is rate-limited by the enzymatic component, telomerase reverse transcriptase (TERT). While regulatory in nature, TERT alternative splice variant/isoform regulation and functions are not fully elucidated and are further complicated by their highly diverse expression and nature. Our primary objective was to characterize TERT isoform expression across 7887 neoplastic and 2099 normal tissue samples using The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression Project (GTEx), respectively. We confirmed the global overexpression and splicing shift towards full-length TERT in neoplastic tissue. Stratifying by tissue type we found uncharacteristic TERT expression in normal brain tissue subtypes. Stratifying by tumor-specific subtypes, we detailed TERT expression differences potentially regulated by subtype-specific molecular characteristics. Focusing on β-deletion splicing regulation, we found the NOVA1 trans-acting factor to mediate alternative splicing in a cancer-dependent manner. Of relevance to future tissue-specific studies, we clustered cancer cell lines with tumors from related origin based on TERT isoform expression patterns. Taken together, our work has reinforced the need for tissue and tumour-specific TERT investigations, provided avenues to do so, and brought to light the current technical limitations of bioinformatic analyses of TERT isoform expression.
Collapse
Affiliation(s)
- Mathushan Subasri
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON N6A 5C1, Canada; (M.S.); (A.J.W.)
| | - Parisa Shooshtari
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada;
- Department of Pathology and Laboratory Medicine, The University of Western Ontario, London, ON N6A 5C1, Canada
- Department of Computer Science, The University of Western Ontario, London, ON N6A 5C1, Canada
- The Children’s Health Research Institute—Lawson Health Research Institute, London, ON N6C 2R5, Canada
| | - Andrew J. Watson
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON N6A 5C1, Canada; (M.S.); (A.J.W.)
- The Children’s Health Research Institute—Lawson Health Research Institute, London, ON N6C 2R5, Canada
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, ON N6A 5C1, Canada
| | - Dean H. Betts
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON N6A 5C1, Canada; (M.S.); (A.J.W.)
- The Children’s Health Research Institute—Lawson Health Research Institute, London, ON N6C 2R5, Canada
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, ON N6A 5C1, Canada
- Correspondence: ; Tel.: +1-519-661-2111 (ext. 83786)
| |
Collapse
|
32
|
Anmella G, Fico G, Lotfaliany M, Hidalgo-Mazzei D, Soto-Angona Ó, Giménez-Palomo A, Amoretti S, Murru A, Radua J, Solanes A, Pacchiarotti I, Verdolini N, Cowdery S, Dodd S, Williams LJ, Mohebbi M, Carvalho AF, Kessing LV, Vieta E, Berk M. Risk of cancer in bipolar disorder and the potential role of lithium: International collaborative systematic review and meta-analyses. Neurosci Biobehav Rev 2021; 126:529-541. [PMID: 33831461 DOI: 10.1016/j.neubiorev.2021.03.034] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 12/16/2022]
Abstract
We examined bipolar disorder (BD) as a risk factor for developing cancer and the role of lithium on cancer incidence. We conducted two systematic review and meta-analyses of population-based studies providing data on these associations. We screened articles indexed in MEDLINE, Scopus, Embase, and PsycINFO up to August 2020. The first random-effects meta-analysis, based on 4,910,661 individuals from nine studies estimated an increased risk of cancer of any kind [RR = 1.24 (1.05-1.46); p < 0.01], especially breast cancer [RR = 1.33 (1.15-1.55); p < 0.01] in BD. The second random-effects meta-analysis, based on 2,606,187 individuals from five studies did not show increased risk of cancer in people with BD using lithium, and even suggested a small protective effect both in overall [RR = 0.94 (0.72-1.22); p = 0.66] and urinary cancer [RR = 0.93 (0.75-1.14); p = 0.48] although these findings did not reach statistical significance. The current evidence highlights that cancer risk is increased in individuals with BD, particularly breast cancer in women. Lithium may have a potential protective effect on cancer, including urinary cancer. The role of lithium as a mainstay of treatment for BD is reinforced by this study.
Collapse
Affiliation(s)
- Gerard Anmella
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Giovanna Fico
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Mojtaba Lotfaliany
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Faculty of Health, Biostatistics Unit, Geelong, Australia
| | - Diego Hidalgo-Mazzei
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Óscar Soto-Angona
- Department of Psychiatry, Vall d'Hebron University Hospital, Barcelona, Catalonia, Spain
| | - Anna Giménez-Palomo
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Silvia Amoretti
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Andrea Murru
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Joaquim Radua
- Imaging of Mood- and Anxiety-Related Disorders (IMARD) group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERSAM, Barcelona, Spain
| | - Aleix Solanes
- Imaging of Mood- and Anxiety-Related Disorders (IMARD) group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERSAM, Barcelona, Spain
| | - Isabella Pacchiarotti
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Norma Verdolini
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Stephanie Cowdery
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Seetal Dodd
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, and the Department of Psychiatry, The University of Melbourne, Parkville, Australia
| | - Lana J Williams
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Mohammadreza Mohebbi
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Faculty of Health, Biostatistics Unit, Geelong, Australia
| | - Andre F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction & Mental Health (CAMH), Toronto, ON, Canada
| | - Lars Vedel Kessing
- Copenhagen Affective Disorder Research Center (CADIC), Psychiatric Center Copenhagen, Copenhagen, Denmark; Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Eduard Vieta
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Michael Berk
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, and the Department of Psychiatry, The University of Melbourne, Parkville, Australia; Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
33
|
Behrooz AB, Syahir A. Could We Address the Interplay Between CD133, Wnt/β-Catenin, and TERT Signaling Pathways as a Potential Target for Glioblastoma Therapy? Front Oncol 2021; 11:642719. [PMID: 33869033 PMCID: PMC8047678 DOI: 10.3389/fonc.2021.642719] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/15/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal forms of primary brain tumors. Glioblastoma stem cells (GSCs) play an undeniable role in tumor development by activating multiple signaling pathways such as Wnt/β-catenin and PI3K/AKT/mTOR that facilitate brain tumor formation. CD133, a transmembrane glycoprotein, has been used to classify cancer stem cells (CSCs) in GBM. The therapeutic value of CD133 is a biomarker of the CSC in multiple cancers. It also leads to growth and recurrence of the tumor. More recent findings have confirmed the association of telomerase/TERT with Wnt/β-catenin and the PI3K/AKT/mTOR signaling pathways. Advance studies have shown that crosstalk between CD133, Wnt/β-catenin, and telomerase/TERT can facilitate GBM stemness and lead to therapeutic resistance. Mechanistic insight into signaling mechanisms downstream of surface biomarkers has been revolutionized by facilitating targeting of tumor-specific molecular deregulation. This review also addresses the importance of interplay between CD133, Wnt/β-catenin and TERT signaling pathways in GSCs and outlines the future therapeutic goals for glioblastoma treatment.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Malaysia
| | - Amir Syahir
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Malaysia.,MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
34
|
Chen D, Huang L, Zhou H, Zhang Y. Combining IL-10 and Oncolytic Adenovirus Demonstrates Enhanced Antitumor Efficacy Through CD8 + T Cells. Front Immunol 2021; 12:615089. [PMID: 33717103 PMCID: PMC7952747 DOI: 10.3389/fimmu.2021.615089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/11/2021] [Indexed: 11/27/2022] Open
Abstract
Oncolytic viruses are of growing importance in cancer therapeutics since they combine direct oncolytic effect and the stimulation of antitumor immunity. Emerging evidences showed that the function of oncolytic viruses is dependent on immune response in tumor microenvironment, and the modulation of immunity could influence their efficacy. Here we combined the interleukin 10 (IL-10) and oncolytic adenovirus Ad-hTERT to treat lung cancer and explored the underlying mechanism under combination therapy. Lewis lung carcinoma (LLC) and B16F10 tumor-bearing immunocompetent C57BL/6 mice that received Ad-hTERT or IL-10 alone showed mild antitumor effect, while the combination therapy shrink tumor bulks and prolonged survival remarkably. In addition, IL-10 didn’t show direct influence on tumor cell viability or Ad-hTERT mediated tumor cell lysis in vitro. To further explore the influence of combination therapy mediated antitumor capacity, we eliminated CD8+ T, CD4+ T or natural killer (NK) cells in LLC and B16F10-bearing C57BL/6 mice, and found that CD8+ T cells were critical mediator in the combination therapy. The combination therapy induced intensive infiltration of CD8+ T cells in tumors, increased tumor-specific IFN-γ secretion by CD8+ T cells. The long-term tumor-specific immune memory induced by the combination therapy rejected rechallenge by respective tumor cell lines. This study demonstrated that the therapy combining IL-10 and Ad-hTERT augmented antitumor efficacy which was CD8+ T cells dependent. Our findings paved the way to combine cytokines and oncolytic viruses to enhance antitumor immunotherapy in treating cancer.
Collapse
Affiliation(s)
- Duo Chen
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Luyu Huang
- Division of Thoracic Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Haiyu Zhou
- Division of Thoracic Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuhui Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
35
|
GV1001 interacts with androgen receptor to inhibit prostate cell proliferation in benign prostatic hyperplasia by regulating expression of molecules related to epithelial-mesenchymal transition. Aging (Albany NY) 2021; 13:3202-3217. [PMID: 33539321 PMCID: PMC7906190 DOI: 10.18632/aging.202242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022]
Abstract
Prostate cell proliferation, driven by testosterone, is a major characteristic of benign prostatic hyperplasia (BPH). GV1001, a human telomerase reverse transcriptase catalytic subunit, is an injectable formulation used as a cancer vaccine. It functions as a cell penetrating peptide to regulate cell proliferation. Here, we found that GV1001 effectively suppressed proliferation of prostatic stromal myofibroblasts (WPMY-1) and prostatic epithelial cells (RWPE-1 and WPE-NA22) treated with dihydrotestosterone. Also, GV1001 bound to androgen receptors (ARs) in the cytosol of stromal and epithelial cells. In an experimental animal model implanted with an infusion pump for spontaneous and continuous release of testosterone, revealed that GV1001 reduced prostatic hypertrophy and inhibited the cell proliferation and the expression of Ki67, proliferating cell nuclear antigen, and prostate specific antigen. In addition, GV1001 prevented fibrosis of the prostate by downregulating expression of prostatic epithelial-mesenchymal transition (EMT)-related proteins such as transforming growth factor (TGF)-β, Snail, Slug, N-cadherin, and Vimentin, and by up-regulating E-cadherin. Taken together, these results suggest that GV1001, which suppresses TGF-β-mediated EMT by outcompeting testosterone for binding to AR, is a potential therapeutic drug for BPH accompanied by prostatic fibrosis.
Collapse
|
36
|
Abstract
Cancer cells accumulate iron to supplement their aberrant growth and metabolism. Depleting cells of iron by iron chelators has been shown to be selectively cytotoxic to cancer cells in vitro and in vivo. Iron chelators are effective at combating a range of cancers including those which are difficult to treat such as androgen insensitive prostate cancer and cancer stem cells. This review will evaluate the impact of iron chelation on cancer cell survival and the underlying mechanisms of action. A plethora of studies have shown iron chelators can reverse some of the major hallmarks and enabling characteristics of cancer. Iron chelators inhibit signalling pathways that drive proliferation, migration and metastasis as well as return tumour suppressive signalling. In addition to this, iron chelators stimulate apoptotic and ER stress signalling pathways inducing cell death even in cells lacking a functional p53 gene. Iron chelators can sensitise cancer cells to PARP inhibitors through mimicking BRCAness; a feature of cancers trademark genomic instability. Iron chelators target cancer cell metabolism, attenuating oxidative phosphorylation and glycolysis. Moreover, iron chelators may reverse the major characteristics of oncogenic transformation. Iron chelation therefore represent a promising selective mode of cancer therapy.
Collapse
|
37
|
Jebaraj BMC, Stilgenbauer S. Telomere Dysfunction in Chronic Lymphocytic Leukemia. Front Oncol 2021; 10:612665. [PMID: 33520723 PMCID: PMC7844343 DOI: 10.3389/fonc.2020.612665] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Telomeres are nucleprotein structures that cap the chromosomal ends, conferring genomic stability. Alterations in telomere maintenance and function are associated with tumorigenesis. In chronic lymphocytic leukemia (CLL), telomere length is an independent prognostic factor and short telomeres are associated with adverse outcome. Though telomere length associations have been suggested to be only a passive reflection of the cell's replication history, here, based on published findings, we suggest a more dynamic role of telomere dysfunction in shaping the disease course. Different members of the shelterin complex, which form the telomere structure have deregulated expression and POT1 is recurrently mutated in about 3.5% of CLL. In addition, cases with short telomeres have higher telomerase (TERT) expression and activity. TERT activation and shelterin deregulation thus may be pivotal in maintaining the minimal telomere length necessary to sustain survival and proliferation of CLL cells. On the other hand, activation of DNA damage response and repair signaling at dysfunctional telomeres coupled with checkpoint deregulation, leads to terminal fusions and genomic complexity. In summary, multiple components of the telomere system are affected and they play an important role in CLL pathogenesis, progression, and clonal evolution. However, processes leading to shelterin deregulation as well as cell intrinsic and microenvironmental factors underlying TERT activation are poorly understood. The present review comprehensively summarizes the complex interplay of telomere dysfunction in CLL and underline the mechanisms that are yet to be deciphered.
Collapse
Affiliation(s)
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
- Klinik für Innere Medizin I, Universitätsklinikum des Saarlandes, Homburg, Germany
| |
Collapse
|
38
|
Hu Y, Ciminieri C, Hu Q, Lehmann M, Königshoff M, Gosens R. WNT Signalling in Lung Physiology and Pathology. Handb Exp Pharmacol 2021; 269:305-336. [PMID: 34463851 DOI: 10.1007/164_2021_521] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The main physiological function of the lung is gas exchange, mediated at the interface between the alveoli and the pulmonary microcapillary network and facilitated by conducting airway structures that regulate the transport of these gases from and to the alveoli. Exposure to microbial and environmental factors such as allergens, viruses, air pollution, and smoke contributes to the development of chronic lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and lung cancer. Respiratory diseases as a cluster are the commonest cause of chronic disease and of hospitalization in children and are among the three most common causes of morbidity and mortality in the adult population worldwide. Many of these chronic respiratory diseases are associated with inflammation and structural remodelling of the airways and/or alveolar tissues. They can often only be treated symptomatically with no disease-modifying therapies that normalize the pathological tissue destruction driven by inflammation and remodelling. In search for novel therapeutic strategies for these diseases, several lines of evidence revealed the WNT pathway as an emerging target for regenerative strategies in the lung. WNT proteins, their receptors, and signalling effectors have central regulatory roles under (patho)physiological conditions underpinning lung function and (chronic) lung diseases and we summarize these roles and discuss how pharmacological targeting of the WNT pathway may be utilized for the treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yan Hu
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Chiara Ciminieri
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA.,Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Qianjiang Hu
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Mareike Lehmann
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Melanie Königshoff
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany. .,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
39
|
Yuan X, Dai M, Xu D. Telomere-related Markers for Cancer. Curr Top Med Chem 2020; 20:410-432. [PMID: 31903880 PMCID: PMC7475940 DOI: 10.2174/1568026620666200106145340] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/03/2019] [Accepted: 12/14/2019] [Indexed: 02/06/2023]
Abstract
Telomeres are structurally nucleoprotein complexes at termini of linear chromosomes and essential to chromosome stability/integrity. In normal human cells, telomere length erodes progressively with each round of cell divisions, which serves as an important barrier to uncontrolled proliferation and malignant transformation. In sharp contrast, telomere maintenance is a key feature of human malignant cells and required for their infinite proliferation and maintenance of other cancer hallmarks as well. Thus, a telomere-based anti-cancer strategy has long been suggested. However, clinically efficient and specific drugs targeting cancer telomere-maintenance have still been in their infancy thus far. To achieve this goal, it is highly necessary to elucidate how exactly cancer cells maintain functional telomeres. In the last two decades, numerous studies have provided profound mechanistic insights, and the identified mechanisms include the aberrant activation of telomerase or the alternative lengthening of telomere pathway responsible for telomere elongation, dysregulation and mutation of telomere-associated factors, and other telomere homeostasis-related signaling nodes. In the present review, these various strategies employed by malignant cells to regulate their telomere length, structure and function have been summarized, and potential implications of these findings in the rational development of telomere-based cancer therapy and other clinical applications for precision oncology have been discussed.
Collapse
Affiliation(s)
- Xiaotian Yuan
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
| | - Mingkai Dai
- Central Research Laboratory, Shandong University Second Hospital, Jinan, 250033, China.,Karolinska Institute Collaborative Laboratory for Cancer and Stem Cell Research, Shandong University Second Hospital, Jinan, 250033, China
| | - Dawei Xu
- Karolinska Institute Collaborative Laboratory for Cancer and Stem Cell Research, Shandong University Second Hospital, Jinan, 250033, China.,Department of Medicine, Division of Hematology, Center for Molecular Medicine (CMM) and Bioclinicum, Karolinska Institute and Karolinska University Hospital Solna, Solna 171 64, Sweden
| |
Collapse
|
40
|
Li W, Yan Y, Zheng Z, Zhu Q, Long Q, Sui S, Luo M, Chen M, Li Y, Hua Y, Deng W, Lai R, Li L. Targeting the NCOA3-SP1-TERT axis for tumor growth in hepatocellular carcinoma. Cell Death Dis 2020; 11:1011. [PMID: 33239622 PMCID: PMC7689448 DOI: 10.1038/s41419-020-03218-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) has a high mortality rate and lacks an effective therapeutic target. Elevated expression of human telomerase reverse transcriptase (TERT) is an important hallmark in cancers, but the mechanism by which TERT is activated differentially in cancers is poorly understood. Here, we have identified nuclear receptor coactivator-3 (NCOA3) as a new modulator of TERT expression and tumor growth in HCC. NACO3 specifically binds to the TERT promoter at the -234 to -144 region and transcriptionally activates TERT expression. NCOA3 promotes HCC cell growth and tumor progression in vitro and in vivo through upregulating the TERT signaling. Knockdown of NACO3 suppresses HCC cell viability and colony formation, whereas TERT overexpression rescues this suppression. NCOA3 interacts with and recruits SP1 binding on the TERT promoter. Knockdown of NCOA3 also inhibits the expression of the Wnt signaling-related genes but has no effect on the Notch signaling-targeting genes. Moreover, NCOA3 is positively correlated with TERT expression in HCC tumor tissues, and high expression of both NCOA3 and TERT predicts a poor prognosis in HCC patients. Our findings indicate that targeting the NCOA3-SP1-TERT signaling axis may benefit HCC patients.
Collapse
Affiliation(s)
- Wenbin Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.,Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Yue Yan
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Zongheng Zheng
- The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiaohua Zhu
- Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
| | - Qian Long
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Silei Sui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Meihua Luo
- Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
| | - Miao Chen
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yizhuo Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yijun Hua
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| | - Renchun Lai
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| | - Liren Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| |
Collapse
|
41
|
Fundamental insights into the interaction between telomerase/TERT and intracellular signaling pathways. Biochimie 2020; 181:12-24. [PMID: 33232793 DOI: 10.1016/j.biochi.2020.11.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 11/07/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Telomerase activity is critical for cancer cells to provide unrestricted proliferation and cellular immortality through maintaining telomeres. Telomerase enzymatic activity is regulatable at the level of DNA, mRNA, post translational modifications, cellular transport and enzyme assembly. More recent studies confirm the interaction of the telomerase with various intracellular signaling pathways including PI3K/AKT/mTOR, NF-κB and Wnt/β-catenin which mainly participating in inflammation, epithelial to mesenchymal transition (EMT) and tumor cell invasion and metastasis. Furthermore, hTERT protein has been detected in non-nuclear sites such as the mitochondria and cytoplasm in cells. Mitochondrial TERT indicates various non-telomere-related functions such as decreasing reactive oxygen species (ROS) generation, boosting the respiration rate, protecting mtDNA by direct binding, interacting with mitochondrial tRNAs and increasing mitochondrial membrane potential which can lead to higher chemoresistance rate in cancer cells during therapies. Understanding the molecular mechanisms of the TERT function and depended interactions in tumor cells can suggest novel therapeutic approaches. Hence, in this review we will explain the telomerase activity regulation in translational and post translational levels besides the established correlations with various cell signaling pathways with possible pathways for therapeutic targeting.
Collapse
|
42
|
Kubo C, Ogawa M, Uehara N, Katakura Y. Fisetin Promotes Hair Growth by Augmenting TERT Expression. Front Cell Dev Biol 2020; 8:566617. [PMID: 33178686 PMCID: PMC7593534 DOI: 10.3389/fcell.2020.566617] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/24/2020] [Indexed: 12/16/2022] Open
Abstract
Although thinning hair and alopecia are not recognized as severe diseases, hair loss has implications for mental health and quality of life; therefore, a large number of studies have been carried out to develop novel hair growth agents. In the present study, we aimed to examine the potential of telomerase reverse transcriptase (TERT), because TERT overexpression in skin activates resting hair follicle bulge stem cells, which triggers initiation of a new hair follicle growth phase and promotes hair synthesis. To this end, we screened polyphenols that activate TERT expression in keratinocytes, and identified resveratrol and fisetin as strong hTERT-augmenting compounds. These polyphenols also regulated the gene expression of cytokines such as IGF-1 and KGF, which activate the β-catenin pathway, and TGF-β1, which plays an important role in maintaining the niche of hair follicle stem cells, thus are thought to play roles in promoting hair growth. We additionally showed that these polyphenols, especially fisetin, promoted hair growth from the shaved dorsal skin of mice, which suggests that these polyphenols activate the transition from telogen to anagen phase. Histological studies indicated that the dorsal skin of mice treated with these polyphenols contained numerous hair follicles and was thickened compared with that in control mice. Furthermore, on the dorsal skin of mice treated with resveratrol and fisetin, a number of proliferating cells (Ki67+ cells) were observed around the hair papilla. These results suggest that resveratrol and fisetin induce a shift from telogen to anagen in the hair follicle by inducing proliferation of hair follicle bulge stem cells, thus promoting hair growth.
Collapse
Affiliation(s)
- Chisato Kubo
- Laboratory of Cellular Regulation Technology, Department of Bioscience and Biotechnology, Graduate School of Bioresources and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Mizuki Ogawa
- Laboratory of Cellular Regulation Technology, Department of Bioscience and Biotechnology, Graduate School of Bioresources and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Norihisa Uehara
- Department of Molecular Cell Biology and Oral Anatomy, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yoshinori Katakura
- Laboratory of Cellular Regulation Technology, Department of Bioscience and Biotechnology, Graduate School of Bioresources and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan.,Laboratory of Cellular Regulation Technology, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| |
Collapse
|
43
|
Shen L, Zeng J, Ma L, Li S, Chen C, Jia J, Liang X. Helicobacter pylori Induces a Novel NF-kB/LIN28A/let-7a/hTERT Axis to Promote Gastric Carcinogenesis. Mol Cancer Res 2020; 19:74-85. [PMID: 33004623 DOI: 10.1158/1541-7786.mcr-19-0678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 11/18/2019] [Accepted: 09/28/2020] [Indexed: 11/16/2022]
Abstract
Reactivated telomerase is a crucial event in the development and progression of a variety of tumors. However, how telomerase is activated in gastric carcinogenesis has not been fully uncovered yet. Here, we identified a key role of the NF-κB/LIN28A/let-7a axis to promote human telomerase reverse transcriptase (hTERT) expression for gastric cancer initiation. Mechanistically, LIN28A expression was upregulated by H. pylori-induced NF-κB activation. And LIN28A, in turn, suppressed let-7a expression, forming the NF-κB/LIN28A/let-7a axis to regulate gene expression upon H. pylori infection. Of note, we first discovered hTERT as a direct target of let-7a, which inhibited hTERT expression by binding to its 3'UTR of mRNA. Therefore, H. pylori-triggered let-7a downregulation enhanced hTERT protein translation, resulting in telomerase reactivation. Furthermore, hTERT enhanced LIN28A expression, forming the positive feedback regulation between hTERT and NF-κB/LIN28A/let-7a axis to maintain the sustained overexpression of hTERT in gastric cancer. IMPLICATIONS: The NF-κB/LIN28A/Let-7a axis was crucial for the overexpression of hTERT upon H. pylori infection during gastric cancer development and may serve as a potential target to suppress hTERT expression for gastric cancer prevention and treatment.
Collapse
Affiliation(s)
- Li Shen
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China
| | - Jiping Zeng
- Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China
| | - Lin Ma
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China
| | - Shuyan Li
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China
| | - Chunyan Chen
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Jihui Jia
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China.,Cancer Research Laboratory, Shandong University-Karolinska Institutet collaborative Laboratory, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China
| | - Xiuming Liang
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China. .,Cancer Research Laboratory, Shandong University-Karolinska Institutet collaborative Laboratory, School of Basic Medical Science, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
44
|
Yuan X, Liu T, Xu D. Telomerase reverse transcriptase promoter mutations in thyroid carcinomas: implications in precision oncology-a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1244. [PMID: 33178776 PMCID: PMC7607115 DOI: 10.21037/atm-20-5024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Telomerase is a ribonucleoprotein enzyme with telomerase reverse transcriptase (TERT) as a catalytic component. In normal human follicular thyroid cells or thyrocytes, telomerase is silent due to the TERT gene being tightly repressed. However, during the formation of thyroid carcinoma (TC), telomerase becomes activated via TERT induction. The TERT promoter’s gain-of-function mutation has recently been identified in TCs and many other malignancies. The mutation creates a de novo ETS-binding motif through which TERT transcription is de-repressed and telomerase is activated; through this, the mutant TERT promoter promotes the development of TC, contributes to disease aggressiveness and treatment resistance, and thereby leads to poor patient outcomes. From a clinical point of view, the strong association between the TERT promoter mutation and disease malignancy and aggressiveness holds great promise for its value in TC diagnostics, risk stratification, prognostication, treatment decision, and follow-up design. In the present review article, we summarize the recent findings of studies of TERT promoter mutations in TC and underscore the implications of TERT hyperactivity driven by genetic events in the pathogenesis and management of TC. Finally, the targeting of TERT promoter mutations and the disruption of telomere maintenance are considered as potential therapeutic strategies against TC.
Collapse
Affiliation(s)
- Xiaotian Yuan
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Medicine, Division of Hematology, Bioclinicum and Center for Molecular Medicine (CMM), Karolinska Institutet and Karolinska University Hospital Solna, Solna, Sweden
| | - Tiantian Liu
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dawei Xu
- Department of Medicine, Division of Hematology, Bioclinicum and Center for Molecular Medicine (CMM), Karolinska Institutet and Karolinska University Hospital Solna, Solna, Sweden.,Karolinska Institute-Shandong University Collaborative Laboratory for Cancer and Stem Cell Research, Jinan, China
| |
Collapse
|
45
|
Chattopadhyay S, Law S. Morphogen signaling by Wnt/β-catenin pathway and microenvironmental alteration in the bone marrow of agricultural pesticide exposure-induced experimental aplastic anemia. J Biochem Mol Toxicol 2020; 34:e22523. [PMID: 32410290 DOI: 10.1002/jbt.22523] [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: 10/12/2019] [Revised: 03/14/2020] [Accepted: 04/27/2020] [Indexed: 11/08/2022]
Abstract
The etiologic link between pesticide toxicity and aplastic anemia in agricultural and agro-industrial setting has been frequently reported in epidemiological studies conducted worldwide. Chronic pesticide toxicity causes long-term bone marrow injury and perturbs the normal hematopoietic physiology, including survival of hematopoietic progenitor cells and bone marrow's blood cell forming ability. The purpose of this study is to understand the mechanism of pesticide toxicity-mediated bone marrow aplasia by studying Wnt/β-catenin signaling pathway and microenvironmental stromal components. An agricultural pesticide formulation comprising of cypermethrin, chlorpyriphos, and hexaconazole was used to induce bone marrow aplasia in inbred Swiss albino mice. Marrow failure followed by the onset of aplastic condition was confirmed by pancytopenic peripheral blood and hypocellular bone marrow filled with adipocytes. Significant downregulation of canonical Wnt/β-catenin signaling was identified by expression analysis of Wnt3a, β-catenin, and telomerase reverse transcriptase in the aplastic bone marrow hematopoietic stem/progenitor compartment. Along with signaling deregulation, disruption in both the osteoblastic and vascular stromal components was observed in the pesticide-exposed bone marrow microenvironment when compared to control. In this study, we tried to establish the correlation among disease pathophysiology, signaling deregulation in the hematopoietic cells, and bone marrow microenvironmental alteration during environmental exposure-mediated aplastic hematopoietic catastrophe, which may shed light on the unexplored mechanistic perspective of this fatal blood disease.
Collapse
Affiliation(s)
- Sukalpa Chattopadhyay
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, Kolkata, West Bengal, India
| | - Sujata Law
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, Kolkata, West Bengal, India
| |
Collapse
|
46
|
Jeong YJ, Park SH, Jeon CH. Detection of circulating tumor cells in patients with breast cancer using the conditionally reprogrammed cell culture method and reverse transcription-PCR of hTERT and MAGE A1-6. Oncol Lett 2020; 20:78. [PMID: 32863911 PMCID: PMC7436930 DOI: 10.3892/ol.2020.11939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/17/2020] [Indexed: 12/21/2022] Open
Abstract
The present study aimed to verify the efficacy of the conditionally reprogrammed cell (CRC) culture method for the detection of circulating tumor cells (CTCs) in breast cancer. CTCs were isolated from the peripheral blood of patients with breast cancer, and culture of the collected CTCs was performed according to the conditional reprogramming protocol. Total RNA was extracted from cultured CTCs, and the hTERT and MAGE A1-6 genes were amplified using reverse transcription-PCR (RT-PCR). In addition, RNA extraction from another blood sample was performed and the expression of the two genes was analyzed by RT-PCR only. Following CRC culture, grown CTCs were observed in 7 samples (23.3%). The CTC detection rates by RT-PCR for the hTERT and MAGE A1-6 genes in CTCs grown using the CRC culture method were 26.7 and 10.0%, respectively. The positive expression rates for the hTERT and MAGE genes in CTCs assessed by RT-PCR only were 44.1 and 23.5%, respectively. When combining the positive expression rates of RT-PCR only and CRC culture for the hTERT and MAGE A1-6 genes, CTC detection rates increased to 53.3 and 23.3%, respectively. Additionally, when combining the positive expression rates of the two genes by either method, the CTC detection rate was the highest value observed. In conclusion, the present study revealed the potential of CRC culture in the detection of CTCs in breast cancer. Furthermore, a combination of CRC culture and RT-PCR for the hTERT and MAGE A1-6 genes is useful in enhancing the detection rate of CTCs in the blood.
Collapse
Affiliation(s)
- Young Ju Jeong
- Department of Surgery, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| | - Sung Hwan Park
- Department of Surgery, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| | - Chang-Ho Jeon
- Department of Laboratory Medicine, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| |
Collapse
|
47
|
Zhang X, Bai J, Yin H, Long L, Zheng Z, Wang Q, Chen F, Yu X, Zhou Y. Exosomal miR-1255b-5p targets human telomerase reverse transcriptase in colorectal cancer cells to suppress epithelial-to-mesenchymal transition. Mol Oncol 2020; 14:2589-2608. [PMID: 32679610 PMCID: PMC7530775 DOI: 10.1002/1878-0261.12765] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 06/04/2020] [Accepted: 06/14/2020] [Indexed: 12/27/2022] Open
Abstract
Cancer cells undergo epithelial‐to‐mesenchymal transition (EMT) in response to hypoxia. Exosomes produced in tumor microenvironments carry microRNAs (miRNAs) that affect proliferation, metastasis, and EMT. Hypoxic regulation of EMT is associated with telomerase content and stability, but the underlying mechanisms remain unclear. We identified a targeting relationship between tumor‐suppressing miR‐1255b‐5p and human telomerase reverse transcriptase (hTERT) via clinical screening of serum samples in colorectal cancer (CRC) patients. EMT suppression via exosomal miR‐1255b‐5p delivery was investigated by assessing hTERT expression, Wnt/β‐catenin signaling, and telomerase activity. We revealed that hypoxia directly affected exosomal miR‐1255b‐5p content, the delivery of which between CRC cells significantly impacted cell invasion, EMT‐related protein expression, and telomerase stability. Specifically, miR‐1255b‐5p suppressed EMT by inhibiting Wnt/β‐catenin activation via hTERT inhibition. Hypoxia reduced exosomal miR‐1255b‐5p secretion by CRC cells, thereby increasing hTERT expression to enhance EMT and telomerase activity. In a mouse CRC model, hypoxic exosomes containing overexpressed miR‐1255b‐5p attenuated EMT, tumor progression, and liver metastasis. Our results suggest the antitumor role of miR‐1255b‐5p and its involvement in the regulation of hTERT‐mediated EMT. We propose that miRNA‐targeted regulation of telomerase is a promising therapeutic strategy for future CRC treatment.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Jian Bai
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China.,Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hang Yin
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Long Long
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Zhewen Zheng
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Qingqing Wang
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Fengxia Chen
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Xiaoyan Yu
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yunfeng Zhou
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| |
Collapse
|
48
|
Baruah J, Chaudhuri S, Mastej V, Axen C, Hitzman R, Ribeiro IMB, Wary KK. Low-Level Nanog Expression in the Regulation of Quiescent Endothelium. Arterioscler Thromb Vasc Biol 2020; 40:2244-2264. [PMID: 32640900 PMCID: PMC7447188 DOI: 10.1161/atvbaha.120.314875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Supplemental Digital Content is available in the text. Nanog is expressed in adult endothelial cells (ECs) at a low-level, however, its functional significance is not known. The goal of our study was to elucidate the role of Nanog in adult ECs using a genetically engineered mouse model system.
Collapse
Affiliation(s)
- Jugajyoti Baruah
- From the Department of Psychiatry, Harvard Medical School, Boston, MA (J.B.).,Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital, Belmont, MA (J.B.)
| | - Suhnrita Chaudhuri
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Victoria Mastej
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Cassondra Axen
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Ryan Hitzman
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Isabella M B Ribeiro
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Kishore K Wary
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| |
Collapse
|
49
|
Trybek T, Kowalik A, Góźdź S, Kowalska A. Telomeres and telomerase in oncogenesis. Oncol Lett 2020; 20:1015-1027. [PMID: 32724340 PMCID: PMC7377093 DOI: 10.3892/ol.2020.11659] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/25/2020] [Indexed: 12/24/2022] Open
Abstract
Telomeres are located at the ends of chromosomes and protect them from degradation. Suppressing the activity of telomerase, a telomere-synthesizing enzyme, and maintaining short telomeres is a protective mechanism against cancer in humans. In most human somatic cells, the expression of telomerase reverse transcriptase (TERT) is repressed and telomerase activity is inhibited. This leads to the progressive shortening of telomeres and inhibition of cell growth in a process called replicative senescence. Most types of primary cancer exhibit telomerase activation, which allows uncontrolled cell proliferation. Previous research indicates that TERT activation also affects cancer development through activities other than the canonical function of mediating telomere elongation. Recent studies have improved the understanding of the structure and function of telomeres and telomerase as well as key mechanisms underlying the activation of TERT and its role in oncogenesis. These advances led to a search for drugs that inhibit telomerase as a target for cancer therapy. The present review article summarizes the organization and function of telomeres, their role in carcinogenesis, and advances in telomerase-targeted therapy.
Collapse
Affiliation(s)
- Tomasz Trybek
- Endocrinology Clinic, Holycross Cancer Center, 25-734 Kielce, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holycross Cancer Center, 25-734 Kielce, Poland
| | - Stanisław Góźdź
- The Faculty of Health Sciences, Jan Kochanowski University, 25-319 Kielce, Poland.,Oncology Clinic, Holycross Cancer Center, 25-734 Kielce, Poland
| | - Aldona Kowalska
- Endocrinology Clinic, Holycross Cancer Center, 25-734 Kielce, Poland.,The Faculty of Health Sciences, Jan Kochanowski University, 25-319 Kielce, Poland
| |
Collapse
|
50
|
Bajaj S, Kumar MS, Peters GJ, Mayur YC. Targeting telomerase for its advent in cancer therapeutics. Med Res Rev 2020; 40:1871-1919. [PMID: 32391613 DOI: 10.1002/med.21674] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 12/24/2022]
Abstract
Telomerase has emerged as an important primary target in anticancer therapy. It is a distinctive reverse transcriptase enzyme, which extends the length of telomere at the 3' chromosomal end, and uses telomerase reverse transcriptase (TERT) and telomerase RNA template-containing domains. Telomerase has a vital role and is a contributing factor in human health, mainly affecting cell aging and cell proliferation. Due to its unique feature, it ensures unrestricted cell proliferation in malignancy and plays a major role in cancer disease. The development of telomerase inhibitors with increased specificity and better pharmacokinetics is being considered to design and develop newer potent anticancer agents. Use of natural and synthetic compounds for the inhibition of telomerase activity can lead to an opening of new vistas in cancer treatment. This review details about the telomerase biochemistry, use of natural and synthetic compounds; vaccines and oncolytic virus in therapy that suppress the telomerase activity. We have discussed structure-activity relationships of various natural and synthetic telomerase inhibitors to help medicinal chemists and chemical biology researchers with a ready reference and updated status of their clinical trials. Suppression of human TERT (hTERT) activity through inhibition of hTERT promoter is an important approach for telomerase inhibition.
Collapse
Affiliation(s)
| | | | - G J Peters
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Y C Mayur
- SPPSPTM, SVKM's NMIMS, Mumbai, India
| |
Collapse
|