1
|
Yao L, Wang X, Wang Z, Wang X, Guo X. Expression and functional analyses of TERF2 in esophageal carcinoma. Heliyon 2024; 10:e38040. [PMID: 39328506 PMCID: PMC11425175 DOI: 10.1016/j.heliyon.2024.e38040] [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: 07/15/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Background Esophageal cancer (ESCA) is a prevalent malignancy with a high incidence of morbidity and mortality, particularly in Asia. Telomeric Repeat-binding Factor 2 (TERF2) is a crucial component of the telomere-binding protein complex that maintains telomere stability. Aberrant TERF2 expression has been implicated in tumorigenesis, however, its specific role in ESCA remains largely unexplored. Methods The expression levels of TERF2 were assessed in esophageal squamous cell carcinoma (ESCC) samples using RT-PCR, IHC, and Western blotting (WB). Serum tumor marker concentrations were determined via electrochemiluminescence immunoassay (ECLIA) and chemiluminescent microparticle immunoassay (CMIA). Bioinformatics analyses were employed to elucidate TERF2's function in EC. The impact of TERF2 on ESCC cell proliferation was evaluated through cell counting kit-8 (CCK8) assays and flow cytometry. Results TERF2 protein and mRNA expression were elevated in ESCC tissues and correlated with age, sex, cancer stage, tumor grade, lymph node metastasis (LNM), and tumor histology. Univariate Cox regression analysis indicated TERF2 was an independent prognostic factor for overall survival (OS). TERF2 mRNA levels were associated with serum levels of carcinoembryonic antigen (CEA), cytokeratin 19 fragment (CYFRA21-1), and tissue polypeptide antigen (TPA) in patients with ESCC. Immune infiltration and chemokine profiles were linked to TERF2 expression in ESCA. TERF2 is involved in regulating ESCC cell proliferation may through the DDR/P53 signaling way. Conclusions TERF2 is overexpressed in ESCA and contributes to ESCC cell proliferation may via DDR/TP53 signaling pathway. These results suggest that TERF2 may serve as a potential target for developing treatments and diagnostic biomarker for ESCA.
Collapse
Affiliation(s)
- Lihua Yao
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, PR China
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Xinlu Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Zihao Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Xiaozhong Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Xiaolan Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, PR China
| |
Collapse
|
2
|
Abady MM, Jeong JS, Kwon HJ, Assiri AM, Cho J, Saadeldin IM. The reprotoxic adverse side effects of neurogenic and neuroprotective drugs: current use of human organoid modeling as a potential alternative to preclinical models. Front Pharmacol 2024; 15:1412188. [PMID: 38948466 PMCID: PMC11211546 DOI: 10.3389/fphar.2024.1412188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024] Open
Abstract
The management of neurological disorders heavily relies on neurotherapeutic drugs, but notable concerns exist regarding their possible negative effects on reproductive health. Traditional preclinical models often fail to accurately predict reprotoxicity, highlighting the need for more physiologically relevant systems. Organoid models represent a promising approach for concurrently studying neurotoxicity and reprotoxicity, providing insights into the complex interplay between neurotherapeutic drugs and reproductive systems. Herein, we have examined the molecular mechanisms underlying neurotherapeutic drug-induced reprotoxicity and discussed experimental findings from case studies. Additionally, we explore the utility of organoid models in elucidating the reproductive complications of neurodrug exposure. Have discussed the principles of organoid models, highlighting their ability to recapitulate neurodevelopmental processes and simulate drug-induced toxicity in a controlled environment. Challenges and future perspectives in the field have been addressed with a focus on advancing organoid technologies to improve reprotoxicity assessment and enhance drug safety screening. This review underscores the importance of organoid models in unraveling the complex relationship between neurotherapeutic drugs and reproductive health.
Collapse
Affiliation(s)
- Mariam M. Abady
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Republic of Korea
- Department of Nutrition and Food Science, National Research Centre, Cairo, Egypt
| | - Ji-Seon Jeong
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Ha-Jeong Kwon
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
| | - Abdullah M. Assiri
- Deperament of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Islam M. Saadeldin
- Deperament of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Engin AB, Engin A. Obesity-Senescence-Breast Cancer: Clinical Presentation of a Common Unfortunate Cycle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:821-850. [PMID: 39287873 DOI: 10.1007/978-3-031-63657-8_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
There are few convincing studies establishing the relationship between endogenous factors that cause obesity, cellular aging, and telomere shortening. Without a functional telomerase, a cell undergoing cell division has progressive telomere shortening. While obesity influences health and longevity as well as telomere dynamics, cellular senescence is one of the major drivers of the aging process and of age-related disorders. Oxidative stress induces telomere shortening, while decreasing telomerase activity. When progressive shortening of telomere length reaches a critical point, it triggers cell cycle arrest leading to senescence or apoptotic cell death. Telomerase activity cannot be detected in normal breast tissue. By contrast, maintenance of telomere length as a function of human telomerase is crucial for the survival of breast cancer cells and invasion. Approximately three-quarters of breast cancers in the general population are hormone-dependent and overexpression of estrogen receptors is crucial for their continued growth. In obesity, increasing leptin levels enhance aromatase messenger ribonucleic acid (mRNA) expression, aromatase content, and its enzymatic activity on breast cancer cells, simultaneously activating telomerase in a dose-dependent manner. Meanwhile, applied anti-estrogen therapy increases serum leptin levels and thus enhances leptin resistance in obese postmenopausal breast cancer patients. Many studies revealed that shorter telomeres of postmenopausal breast cancer have higher local recurrence rates and higher tumor grade. In this review, interlinked molecular mechanisms are looked over between the telomere length, lipotoxicity/glycolipotoxicity, and cellular senescence in the context of estrogen receptor alpha-positive (ERα+) postmenopausal breast cancers in obese women. Furthermore, the effect of the potential drugs, which are used for direct inhibition of telomerase and the inhibition of human telomerase reverse transcriptase (hTERT) or human telomerase RNA promoters as well as approved adjuvant endocrine therapies, the selective estrogen receptor modulator and selective estrogen receptor down-regulators are discussed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
4
|
Nguyen LNT, Nguyen LN, Zhao J, Schank M, Dang X, Cao D, Khanal S, Wu XY, Zhang Y, Zhang J, Ning S, Wang L, El Gazzar M, Moorman JP, Yao ZQ. TRF2 inhibition rather than telomerase disruption drives CD4T cell dysfunction during chronic viral infection. J Cell Sci 2022; 135:jcs259481. [PMID: 35660868 PMCID: PMC9377711 DOI: 10.1242/jcs.259481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/24/2022] [Indexed: 11/20/2022] Open
Abstract
We investigated the role of telomerase and telomere repeat-binding factor 2 (TRF2 or TERF2) in T-cell dysfunction in chronic viral infection. We found that the expression and activity of telomerase in CD4+ T (CD4T) cells from patients with hepatitis C virus (HCV) infections or people living with HIV (PLWH) were intact, but TRF2 expression was significantly inhibited at the post-transcriptional level, suggesting that TRF2 inhibition is responsible for the CD4T cell dysfunction observed during chronic viral infection. Silencing TRF2 expression in CD4T cells derived from healthy subjects induced telomeric DNA damage and CD4T cell dysfunction without affecting telomerase activity or translocation - similar to what we observed in CD4T cells from HCV patients and PLWH. These findings indicate that premature T-cell aging and dysfunction during chronic HCV or HIV infection are primarily caused by chronic immune stimulation and T-cell overactivation and/or proliferation that induce telomeric DNA damage due to TRF2 inhibition, rather than telomerase disruption. This study suggests that restoring TRF2 presents a novel approach to prevent telomeric DNA damage and premature T-cell aging, thus rejuvenating T-cell functions during chronic viral infection.
Collapse
Affiliation(s)
- Lam Ngoc Thao Nguyen
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Lam Nhat Nguyen
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Juan Zhao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Madison Schank
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Xindi Dang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Dechao Cao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Sushant Khanal
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Xiao Y. Wu
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Yi Zhang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Jinyu Zhang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Shunbin Ning
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Ling Wang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Mohamed El Gazzar
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Jonathan P. Moorman
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN 37684, USA
| | - Zhi Q. Yao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN 37684, USA
| |
Collapse
|
5
|
Vertecchi E, Rizzo A, Salvati E. Telomere Targeting Approaches in Cancer: Beyond Length Maintenance. Int J Mol Sci 2022; 23:ijms23073784. [PMID: 35409143 PMCID: PMC8998427 DOI: 10.3390/ijms23073784] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 12/19/2022] Open
Abstract
Telomeres are crucial structures that preserve genome stability. Their progressive erosion over numerous DNA duplications determines the senescence of cells and organisms. As telomere length homeostasis is critical for cancer development, nowadays, telomere maintenance mechanisms are established targets in cancer treatment. Besides telomere elongation, telomere dysfunction impinges on intracellular signaling pathways, in particular DNA damage signaling and repair, affecting cancer cell survival and proliferation. This review summarizes and discusses recent findings in anticancer drug development targeting different “telosome” components.
Collapse
Affiliation(s)
- Eleonora Vertecchi
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy;
| | - Angela Rizzo
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy;
| | - Erica Salvati
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy;
- Correspondence:
| |
Collapse
|
6
|
Wu W, Du Z, Wu L. Dexmedetomidine attenuates hypoxia-induced cardiomyocyte injury by promoting telomere/telomerase activity: Possible involvement of ERK1/2-Nrf2 signaling pathway. Cell Biol Int 2022; 46:1036-1046. [PMID: 35312207 DOI: 10.1002/cbin.11799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/26/2021] [Accepted: 01/22/2022] [Indexed: 11/06/2022]
Abstract
Dexmedetomidine (Dex), an α2-adrenergic receptor (α2-AR) agonist, possesses cardioprotection against ischaemic/hypoxic injury, but the exact mechanism is not fully elucidated. Since telomere/telomerase dysfunction is involved in myocardial ischemic damage, the present study aimed to investigate whether Dex ameliorates cobalt chloride (CoCl2; a hypoxia mimic agent in vitro)-induced the damage of H9c2 cardiomyocytes by improving telomere/telomerase dysfunction and further explored the underlying mechanism focusing on ERK1/2-Nrf2 signaling pathway. Result showed that Dex increased cell viability, decreased apoptosis, and reduced cardiomyocyte hypertrophy as illustrated by the decreases in cell surface area and the biomarker levels for cardiac hypertrophy including atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and myosin heavy chain β (β-MHC) mRNA and protein in CoCl2 -exposed H9c2 cells. Intriguingly, Dex increased the telomere length and telomerase activity as well as telomere reverse transcriptase (TERT) protein and mRNA levels in H9c2 cells exposed to CoCl2 , indicating that Dex promotes telomere/telomerase function under hypoxia. In addition, Dex remarkably diminished the ROS generation, reduced MDA content, and increased antioxidative signaling as evidenced by the increases in SOD and GSH-Px activities. Furthermore, Dex increased the ratio of P-ERK1/2/T-ERK1/2 and P-Nrf2/T-Nrf2 and enhanced Nrf2 nuclear translocation in CoCl2 -subjected H9c2 cells, suggesting that Dex promotes the activation of the ERK1/2-Nrf2 signaling pathway. These novel findings indicated that Dex attenuates myocardial ischemic damage and reduces myocardial hypertrophy by promoting telomere/telomerase function, which may be associated with the activation of the ERK1/2-Nrf2 signaling pathway in vitro. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Wei Wu
- Department of Anesthesiology, Hunan Children's Hospital, Changsha, China 410007, People's Republic of China
| | - Zhen Du
- Department of Anesthesiology, Hunan Children's Hospital, Changsha, China 410007, People's Republic of China
| | - Lei Wu
- Department of Anesthesiology, Hunan Children's Hospital, Changsha, China 410007, People's Republic of China
| |
Collapse
|
7
|
Telomeres and Cancer. Life (Basel) 2021; 11:life11121405. [PMID: 34947936 PMCID: PMC8704776 DOI: 10.3390/life11121405] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/18/2022] Open
Abstract
Telomeres cap the ends of eukaryotic chromosomes and are indispensable chromatin structures for genome protection and replication. Telomere length maintenance has been attributed to several functional modulators, including telomerase, the shelterin complex, and the CST complex, synergizing with DNA replication, repair, and the RNA metabolism pathway components. As dysfunctional telomere maintenance and telomerase activation are associated with several human diseases, including cancer, the molecular mechanisms behind telomere length regulation and protection need particular emphasis. Cancer cells exhibit telomerase activation, enabling replicative immortality. Telomerase reverse transcriptase (TERT) activation is involved in cancer development through diverse activities other than mediating telomere elongation. This review describes the telomere functions, the role of functional modulators, the implications in cancer development, and the future therapeutic opportunities.
Collapse
|
8
|
Sołek P, Mytych J, Tabęcka-Łonczyńska A, Koziorowski M. Molecular Consequences of Depression Treatment: A Potential In Vitro Mechanism for Antidepressants-Induced Reprotoxic Side Effects. Int J Mol Sci 2021; 22:11855. [PMID: 34769286 PMCID: PMC8584852 DOI: 10.3390/ijms222111855] [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: 10/03/2021] [Revised: 10/24/2021] [Accepted: 10/30/2021] [Indexed: 11/16/2022] Open
Abstract
The incidence of depression among humans is growing worldwide, and so is the use of antidepressants. However, our fundamental understanding regarding the mechanisms by which these drugs function and their off-target effects against human sexuality remains poorly defined. The present study aimed to determine their differential toxicity on mouse spermatogenic cells and provide mechanistic data of cell-specific response to antidepressant and neuroleptic drug treatment. To directly test reprotoxicity, the spermatogenic cells (GC-1 spg and GC-2 spd cells) were incubated for 48 and 96 h with amitriptyline (hydrochloride) (AMI), escitalopram (ESC), fluoxetine (hydrochloride) (FLU), imipramine (hydrochloride) (IMI), mirtazapine (MIR), olanzapine (OLZ), reboxetine (mesylate) (REB), and venlafaxine (hydrochloride) (VEN), and several cellular and biochemical features were assessed. Obtained results reveal that all investigated substances showed considerable reprotoxic potency leading to micronuclei formation, which, in turn, resulted in upregulation of telomeric binding factor (TRF1/TRF2) protein expression. The TRF-based response was strictly dependent on p53/p21 signaling and was followed by irreversible G2/M cell cycle arrest and finally initiation of apoptotic cell death. In conclusion, our findings suggest that antidepressants promote a telomere-focused DNA damage response in germ cell lines, which broadens the established view of antidepressants' and neuroleptic drugs' toxicity and points to the need for further research in this topic with the use of in vivo models and human samples.
Collapse
Affiliation(s)
- Przemysław Sołek
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, Werynia 2, 36-100 Kolbuszowa, Poland; (A.T.-Ł.); (M.K.)
| | - Jennifer Mytych
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, Werynia 2, 36-100 Kolbuszowa, Poland; (A.T.-Ł.); (M.K.)
| | | | | |
Collapse
|
9
|
Panner Selvam MK, Baskaran S, Sikka SC. Telomere Signaling and Maintenance Pathways in Spermatozoa of Infertile Men Treated With Antioxidants: An in silico Approach Using Bioinformatic Analysis. Front Cell Dev Biol 2021; 9:768510. [PMID: 34708049 PMCID: PMC8542908 DOI: 10.3389/fcell.2021.768510] [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: 08/31/2021] [Accepted: 09/23/2021] [Indexed: 12/03/2022] Open
Abstract
Telomere shortening is considered as a marker of cellular senescence and it is regulated by various signaling pathways. Sperm telomere appears to play important role in its longevity and function. Antioxidant intake has been known to prevent the shortening of telomere. In the management of male infertility, antioxidants are commonly used to counterbalance the seminal oxidative stress. It is important to understand how antioxidants treatment may modulate telomere signaling in sperm. In the current study, we have identified 377 sperm proteins regulated by antioxidants based on data mining of published literature. Bioinformatic analysis revealed involvement of 399 upstream regulators and 806 master regulators associated with differentially expressed sperm proteins. Furthermore, upstream regulator analysis indicated activation of kinases (EGFR and MAPK3) and transcription factors (CCNE1, H2AX, MYC, RB1, and TP53). Hence, it is evident that antioxidant supplementation activates molecules associated with telomere function in sperm. The outcome of this in silico study suggests that antioxidant therapy has beneficial effects on certain transcription factors and kinases associated with sperm telomere maintenance and associated signaling pathways that may play an important role in the management of male factor infertility.
Collapse
Affiliation(s)
| | - Saradha Baskaran
- Department of Urology, Tulane University Health Sciences Center, New Orleans, LA, United States
| | - Suresh C Sikka
- Department of Urology, Tulane University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
10
|
Imran SAM, Yazid MD, Cui W, Lokanathan Y. The Intra- and Extra-Telomeric Role of TRF2 in the DNA Damage Response. Int J Mol Sci 2021; 22:ijms22189900. [PMID: 34576063 PMCID: PMC8470803 DOI: 10.3390/ijms22189900] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
Telomere repeat binding factor 2 (TRF2) has a well-known function at the telomeres, which acts to protect the telomere end from being recognized as a DNA break or from unwanted recombination. This protection mechanism prevents DNA instability from mutation and subsequent severe diseases caused by the changes in DNA, such as cancer. Since TRF2 actively inhibits the DNA damage response factors from recognizing the telomere end as a DNA break, many more studies have also shown its interactions outside of the telomeres. However, very little has been discovered on the mechanisms involved in these interactions. This review aims to discuss the known function of TRF2 and its interaction with the DNA damage response (DDR) factors at both telomeric and non-telomeric regions. In this review, we will summarize recent progress and findings on the interactions between TRF2 and DDR factors at telomeres and outside of telomeres.
Collapse
Affiliation(s)
- Siti A. M. Imran
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.)
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.)
| | - Wei Cui
- Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK;
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.)
- Correspondence: ; Tel.: +603-9145-7704
| |
Collapse
|
11
|
Wang Z, Wu X. Abnormal function of telomere protein TRF2 induces cell mutation and the effects of environmental tumor‑promoting factors (Review). Oncol Rep 2021; 46:184. [PMID: 34278498 PMCID: PMC8273685 DOI: 10.3892/or.2021.8135] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/14/2021] [Indexed: 01/30/2023] Open
Abstract
Recent studies have found that somatic gene mutations and environmental tumor-promoting factors are both indispensable for tumor formation. Telomeric repeat-binding factor (TRF)2 is the core component of the telomere shelterin complex, which plays an important role in chromosome stability and the maintenance of normal cell physiological states. In recent years, TRF2 and its role in tumor formation have gradually become a research hot topic, which has promoted in-depth discussions into tumorigenesis and treatment strategies, and has achieved promising results. Some cells bypass elimination, due to either aging, apoptosis via mutations or abnormal prolongation of the mitotic cycle, and enter the telomere crisis period, where large-scale DNA reorganization occurs repeatedly, which manifests as the precancerous cell cycle. Finally, at the end of the crisis cycle, the mutation activates either the expression level of telomerase or activates the alternative lengthening of telomere mechanism to extend the local telomeres. Under the protection of TRF2, chromosomes are gradually stabilized, immortal cells are formed and the stagewise mutation-driven transformation of normal cells to cancer cells is completed. In addition, TRF2 also shares the characteristics of environmental tumor-promoting factors. It acts on multiple signal transduction pathway-related proteins associated with cell proliferation, and affects peripheral angiogenesis, inhibits the immune recognition and killing ability of the microenvironment, and maintains the stemness characteristics of tumor cells. TRF2 levels are abnormally elevated by a variety of tumor control proteins, which are more conducive to the protection of telomeres and the survival of tumor cells. In brief, the various regulatory mechanisms which tumor cells rely on to survive are organically integrated around TRF2, forming a regulatory network, which is conducive to the optimization of the survival direction of heterogeneous tumor cells, and promotes their survival and adaptability. In terms of clinical application, TRF2 is expected to become a new type of cancer prognostic marker and a new tumor treatment target. Inhibition of TRF2 overexpression could effectively cut off the core network regulating tumor cell survival, reduce drug resistance, or bypass the mutation under the pressure of tumor treatment selection, which may represent a promising therapeutic strategy for the complete eradication of tumors in the clinical setting. Based on recent research, the aim of the present review was to systematically elaborate on the basic structure and functional characteristics of TRF2 and its role in tumor formation, and to analyze the findings indicating that TRF2 deficiency or overexpression could cause severe damage to telomere function and telomere shortening, and induce DNA damage response and chromosomal instability.
Collapse
Affiliation(s)
- Zhengyi Wang
- Good Clinical Practice Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610071, P.R. China
| | - Xiaoying Wu
- Ministry of Education and Training, Chengdu Second People's Hospital, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
12
|
Different epigenetic signatures of newborn telomere length and telomere attrition rate in early life. Aging (Albany NY) 2021; 13:14630-14650. [PMID: 34086604 PMCID: PMC8221291 DOI: 10.18632/aging.203117] [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: 01/26/2021] [Accepted: 05/17/2021] [Indexed: 12/22/2022]
Abstract
Telomere length (TL) and telomere shortening are biological indicators of aging, and epigenetic associates have been found for TL in adults. However, the role of epigenetic signatures in setting newborn TL and early life telomere dynamics is unknown. In the present study, based on 247 participating newborns from the ENVIRONAGE birth cohort, whole-genome DNA methylation, profiled on the Illumina MethylationEPIC BeadChip microarray, and TL were measured in cord blood. In a follow-up visit at a mean age of 4.58 years, leukocyte TL was evaluated. We combined an epigenome-wide association study and a statistical learning method with re-sampling to select CpGs and their two-way interactions to model baseline (cord blood) TL and early-life telomere attrition rate, where distinct epigenetic signatures were identified for the two outcomes. In addition, a stronger epigenetic regulation was suggested in setting newborn TL than that of telomere dynamics in early life: 47 CpGs and 7 between-CpG interactions explained 76% of the variance in baseline TLs, while 72% of the total variance in telomere attrition rate was explained by 31 CpGs and 5 interactions. Functional enrichment analysis based on the selected CpGs in the two models revealed GLUT4 translocation and immune cell signaling pathways, respectively. These CpGs and interactions, as well as the cellular pathways, are potential novel targets of further investigation of telomere biology and aging.
Collapse
|
13
|
Khodadadi E, Mir SM, Memar MY, Sadeghi H, Kashiri M, Faeghiniya M, Jamalpoor Z, Sheikh Arabi M. Shelterin complex at telomeres: Roles in cancers. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
14
|
Thiruthaneeswaran N, Bibby BAS, Yang L, Hoskin PJ, Bristow RG, Choudhury A, West C. Lost in application: Measuring hypoxia for radiotherapy optimisation. Eur J Cancer 2021; 148:260-276. [PMID: 33756422 DOI: 10.1016/j.ejca.2021.01.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022]
Abstract
The history of radiotherapy is intertwined with research on hypoxia. There is level 1a evidence that giving hypoxia-targeting treatments with radiotherapy improves locoregional control and survival without compromising late side-effects. Despite coming in and out of vogue over decades, there is now an established role for hypoxia in driving molecular alterations promoting tumour progression and metastases. While tumour genomic complexity and immune profiling offer promise, there is a stronger evidence base for personalising radiotherapy based on hypoxia status. Despite this, there is only one phase III trial targeting hypoxia modification with full transcriptomic data available. There are no biomarkers in routine use for patients undergoing radiotherapy to aid management decisions, and a roadmap is needed to ensure consistency and provide a benchmark for progression to application. Gene expression signatures address past limitations of hypoxia biomarkers and could progress biologically optimised radiotherapy. Here, we review recent developments in generating hypoxia gene expression signatures and highlight progress addressing the challenges that must be overcome to pave the way for their clinical application.
Collapse
Affiliation(s)
- Niluja Thiruthaneeswaran
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.
| | - Becky A S Bibby
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Lingjang Yang
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Peter J Hoskin
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Mount Vernon Cancer Centre, Northwood, UK
| | - Robert G Bristow
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; CRUK Manchester Institute and Manchester Cancer Research Centre, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| |
Collapse
|
15
|
On K, Crevel G, Cotterill S, Itoh M, Kato Y. Drosophila telomere capping protein HOAP interacts with DSB sensor proteins Mre11 and Nbs. Genes Cells 2021; 26:219-229. [PMID: 33556205 DOI: 10.1111/gtc.12836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/15/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022]
Abstract
In eukaryotes, specific DNA-protein structures called telomeres exist at linear chromosome ends. Telomere stability is maintained by a specific capping protein complex. This capping complex is essential for the inhibition of the DNA damage response (DDR) at telomeres and contributes to genome integrity. In Drosophila, the central factors of telomere capping complex are HOAP and HipHop. Furthermore, a DDR protein complex Mre11-Rad50-Nbs (MRN) is known to be important for the telomere association of HOAP and HipHop. However, whether MRN interacts with HOAP and HipHop, and the telomere recognition mechanisms of HOAP and HipHop are poorly understood. Here, we show that Nbs interacts with Mre11 and transports the Mre11-Rad50 complex from the cytoplasm to the nucleus. In addition, we report that HOAP interacts with both Mre11 and Nbs. The N-terminal region of HOAP is essential for its co-localization with HipHop. Finally, we reveal that Nbs interacts with the N-terminal region of HOAP.
Collapse
Affiliation(s)
- Kinyo On
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
| | - Gilles Crevel
- Department of Basic Medical Sciences, St Georges, University of London, London, UK
| | - Sue Cotterill
- Department of Basic Medical Sciences, St Georges, University of London, London, UK
| | - Masanobu Itoh
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan.,Advanced Insect Research Promotion Center, Kyoto Institute of Technology, Kyoto, Japan
| | - Yasuko Kato
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan.,Advanced Insect Research Promotion Center, Kyoto Institute of Technology, Kyoto, Japan
| |
Collapse
|
16
|
Engin AB, Engin A. The Connection Between Cell Fate and Telomere. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:71-100. [PMID: 33539012 DOI: 10.1007/978-3-030-49844-3_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abolition of telomerase activity results in telomere shortening, a process that eventually destabilizes the ends of chromosomes, leading to genomic instability and cell growth arrest or death. Telomere shortening leads to the attainment of the "Hayflick limit", and the transition of cells to state of senescence. If senescence is bypassed, cells undergo crisis through loss of checkpoints. This process causes massive cell death concomitant with further telomere shortening and spontaneous telomere fusions. In functional telomere of mammalian cells, DNA contains double-stranded tandem repeats of TTAGGG. The Shelterin complex, which is composed of six different proteins, is required for the regulation of telomere length and stability in cells. Telomere protection by telomeric repeat binding protein 2 (TRF2) is dependent on DNA damage response (DDR) inhibition via formation of T-loop structures. Many protein kinases contribute to the DDR activated cell cycle checkpoint pathways, and prevent DNA replication until damaged DNA is repaired. Thereby, the connection between cell fate and telomere length-associated telomerase activity is regulated by multiple protein kinase activities. Contrarily, inactivation of DNA damage checkpoint protein kinases in senescent cells can restore cell-cycle progression into S phase. Therefore, telomere-initiated senescence is a DNA damage checkpoint response that is activated with a direct contribution from dysfunctional telomeres. In this review, in addition to the above mentioned, the choice of main repair pathways, which comprise non-homologous end joining and homologous recombination in telomere uncapping telomere dysfunctions, are discussed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
17
|
[Are telomeres and telomerase still relevant targets in oncology?]. Bull Cancer 2020; 108:30-38. [PMID: 33256968 DOI: 10.1016/j.bulcan.2020.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
|
18
|
Fathi E, Farahzadi R, Javanmardi S, Vietor I. L-carnitine Extends the Telomere Length of the Cardiac Differentiated CD117 +- Expressing Stem Cells. Tissue Cell 2020; 67:101429. [PMID: 32861877 DOI: 10.1016/j.tice.2020.101429] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 01/06/2023]
Abstract
Stem cell-based therapy has emerged as an attractive method for regenerating and repairing the lost heart organ. On other hand, poor survival and maintenance of the cells transferred into the damaged heart tissue are broadly accepted as serious barriers to enhance the efficacy of the regenerative therapy. For this reason, external factors, such as antioxidants are used as a favorite strategy by the investigators to improve the cell survival and retention properties. Therefore, the present study was conducted to investigate the In -vitro effect of L-carnitine (LC) on the telomere length and human telomerase reverse transcriptase (hTERT) gene expression in the cardiac differentiated bone marrow resident CD117+ stem cells through Wnt3/β-catenin and ERK1/2 pathways. To do this, bone marrow resident CD117+ stem cells were enriched by the magnetic-activated cell sorting (MACS) method, and were differentiated to the cardiac cells in the absence (-LC) and presence of the LC (+LC). Also, characterization of the enriched c-kit+ cells was performed using the flow cytometry and immunocytochemistry. At the end of the treatment period, the cells were subjected to the real-time PCR technique along with western blotting assay for measurement of the telomere length and assessment of mRNA and protein, respectively. The results showed that 0.2 mM LC caused the elongation of the telomere length and increased the hTERT gene expression in the cardiac differentiated CD117+ stem cells. In addition, a significant increase was observed in the mRNA and protein expression of Wnt3, β-catenin and ERK1/2 as key components of these pathways. It can be concluded that the LC can increase the telomere length as an effective factor in increasing the cell survival and maintenance of the cardiac differentiated bone marrow resident CD117+ stem cells via Wnt3/β-catenin and ERK1/2 signaling pathway components.
Collapse
Affiliation(s)
- Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Javanmardi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ilja Vietor
- Institute of Cell Biology, Medical University of Innsbruck, Biocenter, Innsbruck, Austria
| |
Collapse
|
19
|
CDK phosphorylation of TRF2 controls t-loop dynamics during the cell cycle. Nature 2019; 575:523-527. [PMID: 31723267 PMCID: PMC6874499 DOI: 10.1038/s41586-019-1744-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/30/2019] [Indexed: 01/05/2023]
Abstract
Telomere end-protection by the shelterin complex prevents DNA damage signalling and promiscuous repair at chromosome ends. Evidence suggests that the 3’ single-stranded telomere end can assemble into a lasso-like t-loop configuration1,2, which has been proposed to safeguard chromosome ends from being recognized as DNA double strand breaks2. Mechanisms must also exist to transiently disassemble t-loops to allow faithful telomere replication and to permit telomerase access to the 3’-end to solve the end replication problem. However, the regulation and physiological importance of t-loops in end-protection remains uncertain. Here, we identify a CDK phosphorylation site in the shelterin subunit, TRF2 (Ser365), whose dephosphorylation in S-phase by the PP6C/R3 phosphatase provides a narrow window during which the helicase RTEL1 is able to transiently access and unwind t-loops to facilitate telomere replication. Re-phosphorylation of TRF2 on Ser365 outside of S-phase is required to release RTEL1 from telomeres, which not only protects t-loops from promiscuous unwinding and inappropriate ATM activation, but also counteracts replication conflicts at DNA secondary structures arising within telomeres and across the genome. Hence, a phospho-switch in TRF2 coordinates assembly and disassembly of t-loops during the cell cycle, which protects telomeres from replication stress and an unscheduled DNA damage response.
Collapse
|
20
|
Bejarano L, Bosso G, Louzame J, Serrano R, Gómez‐Casero E, Martínez‐Torrecuadrada J, Martínez S, Blanco‐Aparicio C, Pastor J, Blasco MA. Multiple cancer pathways regulate telomere protection. EMBO Mol Med 2019; 11:e10292. [PMID: 31273934 PMCID: PMC6609915 DOI: 10.15252/emmm.201910292] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 11/09/2022] Open
Abstract
Telomeres are considered as universal anti-cancer targets, as telomere maintenance is essential to sustain indefinite cancer growth. Mutations in telomerase, the enzyme that maintains telomeres, are among the most frequently found in cancer. In addition, mutations in components of the telomere protective complex, or shelterin, are also found in familial and sporadic cancers. Most efforts to target telomeres have focused in telomerase inhibition; however, recent studies suggest that direct targeting of the shelterin complex could represent a more effective strategy. In particular, we recently showed that genetic deletion of the TRF1 essential shelterin protein impairs tumor growth in aggressive lung cancer and glioblastoma (GBM) mouse models by direct induction of telomere damage independently of telomere length. Here, we screen for TRF1 inhibitory drugs using a collection of FDA-approved drugs and drugs in clinical trials, which cover the majority of pathways included in the Reactome database. Among other targets, we find that inhibition of several kinases of the Ras pathway, including ERK and MEK, recapitulates the effects of Trf1 genetic deletion, including induction of telomeric DNA damage, telomere fragility, and inhibition of cancer stemness. We further show that both bRAF and ERK2 kinases phosphorylate TRF1 in vitro and that these modifications are essential for TRF1 location to telomeres in vivo. Finally, we use these new TRF1 regulatory pathways as the basis to discover novel drug combinations based on TRF1 inhibition, with the goal of effectively blocking potential resistance to individual drugs in patient-derived glioblastoma xenograft models.
Collapse
Affiliation(s)
- Leire Bejarano
- Telomeres and Telomerase GroupMolecular Oncology ProgramSpanish National Cancer Centre (CNIO)MadridSpain
| | - Giuseppe Bosso
- Telomeres and Telomerase GroupMolecular Oncology ProgramSpanish National Cancer Centre (CNIO)MadridSpain
| | - Jessica Louzame
- Telomeres and Telomerase GroupMolecular Oncology ProgramSpanish National Cancer Centre (CNIO)MadridSpain
| | - Rosa Serrano
- Telomeres and Telomerase GroupMolecular Oncology ProgramSpanish National Cancer Centre (CNIO)MadridSpain
| | - Elena Gómez‐Casero
- Experimental Therapeutics ProgramSpanish National Cancer Centre (CNIO)MadridSpain
| | | | - Sonia Martínez
- Experimental Therapeutics ProgramSpanish National Cancer Centre (CNIO)MadridSpain
| | | | - Joaquín Pastor
- Experimental Therapeutics ProgramSpanish National Cancer Centre (CNIO)MadridSpain
| | - Maria A Blasco
- Telomeres and Telomerase GroupMolecular Oncology ProgramSpanish National Cancer Centre (CNIO)MadridSpain
| |
Collapse
|
21
|
Cherfils-Vicini J, Gilson E. Inhibiting TRF1 upstream signaling pathways to target telomeres in cancer cells. EMBO Mol Med 2019; 11:e10845. [PMID: 31273935 PMCID: PMC6609909 DOI: 10.15252/emmm.201910845] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In the context of tumorigenesis, telomere shortening is associated with apparent antagonistic outcomes: On one side, it favors cancer initiation through mechanisms involving genome instability, while on the other side, it prevents cancer progression, due to the activation of the DNA damage response (DDR) checkpoint behaving as a cell-intrinsic proliferation barrier. Consequently, telomerase, which can compensate for replicative erosion by adding telomeric DNA repeats at the chromosomal DNA extremities, is crucial for cancer progression and is upregulated in nearly 90% of human cancers. Therefore, telomeres are considered potential anti-cancer targets and, to date, most of the studies have focused on telomerase inhibition. However, the development of clinically efficient telomerase targeting therapies is still in its infancy. In this context, the findings reported in this issue of EMBO Molecular Medicine by Bejarano et al (2019) open new avenues for alternative telomere therapies.
Collapse
Affiliation(s)
- Julien Cherfils-Vicini
- Université Côte d'Azur (UCA), Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Eric Gilson
- Université Côte d'Azur (UCA), Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
- Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, Nice, France
| |
Collapse
|
22
|
Trajano LADSN, Sergio LPDS, Stumbo AC, Mencalha AL, Fonseca ADSD. Low power lasers on genomic stability. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2018; 180:186-197. [DOI: 10.1016/j.jphotobiol.2018.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/02/2018] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
|
23
|
Basken J, Stuart SA, Kavran AJ, Lee T, Ebmeier CC, Old WM, Ahn NG. Specificity of Phosphorylation Responses to Mitogen Activated Protein (MAP) Kinase Pathway Inhibitors in Melanoma Cells. Mol Cell Proteomics 2017; 17:550-564. [PMID: 29255136 DOI: 10.1074/mcp.ra117.000335] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 11/08/2017] [Indexed: 01/01/2023] Open
Abstract
The BRAF-MKK1/2-ERK1/2 pathway is constitutively activated in response to oncogenic mutations of BRAF in many cancer types, including melanoma. Although small molecules that inhibit oncogenic BRAF and MAP kinase kinase (MKK)1/2 have been successful in clinical settings, resistance invariably develops. High affinity inhibitors of ERK1/2 have been shown in preclinical studies to bypass the resistance of melanoma and colon cancer cells to BRAF and MKK1/2 inhibitors, and are thus promising additions to current treatment protocols. But still unknown is how molecular responses to ERK1/2 inhibitors compare with inhibitors currently in clinical use. Here, we employ quantitative phosphoproteomics to evaluate changes in phosphorylation in response to the ERK inhibitors, SCH772984 and GDC0994, and compare these to the clinically used MKK1/2 inhibitor, trametinib. Combined with previous studies measuring phosphoproteomic responses to the MKK1/2 inhibitor, selumetinib, and the BRAF inhibitor, vemurafenib, the outcomes reveal key insights into pathway organization, phosphorylation specificity and off-target effects of these inhibitors. The results demonstrate linearity in signaling from BRAF to MKK1/2 and from MKK1/2 to ERK1/2. They identify likely targets of direct phosphorylation by ERK1/2, as well as inhibitor off-targets, including an off-target regulation of the p38α mitogen activated protein kinase (MAPK) pathway by the MKK1/2 inhibitor, trametinib, at concentrations used in the literature but higher than in vivo drug concentrations. In addition, several known phosphorylation targets of ERK1/2 are insensitive to MKK or ERK inhibitors, revealing variability in canonical pathway responses between different cell systems. By comparing multiple inhibitors targeted to multiple tiers of protein kinases in the MAPK pathway, we gain insight into regulation and new targets of the oncogenic BRAF driver pathway in cancer cells, and a useful approach for evaluating the specificity of drugs and drug candidates.
Collapse
Affiliation(s)
- Joel Basken
- From the ‡Department of Chemistry and Biochemistry
| | | | - Andrew J Kavran
- From the ‡Department of Chemistry and Biochemistry.,§BioFrontiers Institute
| | - Thomas Lee
- From the ‡Department of Chemistry and Biochemistry
| | - Christopher C Ebmeier
- ¶Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO 80303
| | - William M Old
- ¶Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO 80303
| | - Natalie G Ahn
- From the ‡Department of Chemistry and Biochemistry, .,§BioFrontiers Institute
| |
Collapse
|
24
|
Sergio LPDS, de Paoli F, Mencalha AL, da Fonseca ADS. Chronic Obstructive Pulmonary Disease: From Injury to Genomic Stability. COPD 2017; 14:439-450. [DOI: 10.1080/15412555.2017.1332025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Luiz Philippe da Silva Sergio
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Rio de Janeiro, Brazil
| | - Flavia de Paoli
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, São Pedro, Juiz de Fora, Minas Gerais, Brazil
| | - Andre Luiz Mencalha
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Rio de Janeiro, Brazil
| | - Adenilson de Souza da Fonseca
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Rio de Janeiro, Brazil
- Departamento de Ciências Fisiológicas, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Centro de Ciências da Saúde, Centro Universitário Serra dos Órgãos, Teresópolis, Rio de Janeiro, Brazil
| |
Collapse
|