1
|
Suzuki M, Isobe R, Sato T, Ishikawa R, Suzuki K, Kino Y, Miura T, Inaba Y, Chida K, Fukumoto M. Establishment of acquired radioresistant cells to fractionated radiation from hTERT-immortalized normal human epithelial cell. RADIATION PROTECTION DOSIMETRY 2024; 200:1636-1640. [PMID: 39540470 DOI: 10.1093/rpd/ncae118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 11/16/2024]
Abstract
Senescence-like growth arrest (SLGA), which is a radiation-induced cell death pathway, is induced in immortalized normal human epithelial cell (hTERT-RPE1) by the daily fractionated X-irradiation with 1.5 Gy within 30 times. We here demonstrate that pre-treatment induces acquired radioresistance (ARR) that can survive from the lethal fractionated radiation. The parent cells were daily fractionated with 1.5 Gy for 5 d and then incubated for 7 d without fractionated radiation. After this, the daily fractionated radiation with 1.5 Gy was restarted. A small population of surviving cells appeared after 30 times of the daily fractionated radiation was completed and they were continuously growing up to 120 times of the daily fractionated radiation (RPE1-1.5Fr). We confirmed a higher basal expression level of p53, which functions in the activation of the SLGA pathway but fails to further accumulate after 1.5 Gy of single irradiation in RPE1-1.5Fr. It is the first report to induce ARR phenotype for fractionated radiation in normal human cells.
Collapse
Affiliation(s)
- Masatoshi Suzuki
- Laboratory of Radiological Disasters and Medical Science, International Research Institute of Disaster Science, Tohoku University, 519-1176 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-0845, Japan
- Radiological Technology, Health Science, Graduate School of Medicine, Tohoku University, 2-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | - Rio Isobe
- Laboratory of Radiological Disasters and Medical Science, International Research Institute of Disaster Science, Tohoku University, 519-1176 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Taku Sato
- Laboratory of Radiological Disasters and Medical Science, International Research Institute of Disaster Science, Tohoku University, 519-1176 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Ryoya Ishikawa
- Laboratory of Radiological Disasters and Medical Science, International Research Institute of Disaster Science, Tohoku University, 519-1176 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Keiji Suzuki
- Department of Radiation Medical Sciences, Nagasaki University Atomic Bomb Disease Institute, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yasushi Kino
- Radiochemistry, Graduate School of Science, Tohoku University, 6-3 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-8578, Japan
| | - Tomisato Miura
- Department of Risk Analysis and Biodosimetry, Institute of Radiation Emergency Medicine, Hirosaki University, 66-1 Hon-cho, Hirosaki, Aomori 036-8564, Japan
| | - Yohei Inaba
- Radiological Technology, Health Science, Graduate School of Medicine, Tohoku University, 2-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- Laboratory of Radiological Disasters and Medical Science, International Research Institute of Disaster Science, Tohoku University, 519-1176 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Koichi Chida
- Laboratory of Radiological Disasters and Medical Science, International Research Institute of Disaster Science, Tohoku University, 519-1176 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-0845, Japan
- Radiological Technology, Health Science, Graduate School of Medicine, Tohoku University, 2-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | - Manabu Fukumoto
- Pathology Informatics Team, RIKEN Center for Advance Intelligence Project, 1-4-1 Nihonbashi Chuo-ku, Tokyo 103-0027, Japan
- Laboratory of Radiological Disasters and Medical Science, International Research Institute of Disaster Science, Tohoku University, 519-1176 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-0845, Japan
| |
Collapse
|
2
|
Muthamil S, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Park JH. Biomarkers of Cellular Senescence and Aging: Current State-of-the-Art, Challenges and Future Perspectives. Adv Biol (Weinh) 2024; 8:e2400079. [PMID: 38935557 DOI: 10.1002/adbi.202400079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/29/2024] [Indexed: 06/29/2024]
Abstract
Population aging has increased the global prevalence of aging-related diseases, including cancer, sarcopenia, neurological disease, arthritis, and heart disease. Understanding aging, a fundamental biological process, has led to breakthroughs in several fields. Cellular senescence, evinced by flattened cell bodies, vacuole formation, and cytoplasmic granules, ubiquitously plays crucial roles in tissue remodeling, embryogenesis, and wound repair as well as in cancer therapy and aging. The lack of universal biomarkers for detecting and quantifying senescent cells, in vitro and in vivo, constitutes a major limitation. The applications and limitations of major senescence biomarkers, including senescence-associated β-galactosidase staining, telomere shortening, cell-cycle arrest, DNA methylation, and senescence-associated secreted phenotypes are discussed. Furthermore, explore senotherapeutic approaches for aging-associated diseases and cancer. In addition to the conventional biomarkers, this review highlighted the in vitro, in vivo, and disease models used for aging studies. Further, technologies from the current decade including multi-omics and computational methods used in the fields of senescence and aging are also discussed in this review. Understanding aging-associated biological processes by using cellular senescence biomarkers can enable therapeutic innovation and interventions to improve the quality of life of older adults.
Collapse
Affiliation(s)
- Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Hyun-Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu, 41062, Republic of Korea
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
- Korean Convergence Medicine Major, University of Science & Technology (UST), KIOM Campus, Daejeon, 34054, Republic of Korea
| |
Collapse
|
3
|
Maharajan N, Lee CM, Vijayakumar KA, Cho GW. Oxymatrine Improves Oxidative Stress-Induced Senescence in HT22 Cells and Mice via the Activation of AMP-Activated Protein Kinase. Antioxidants (Basel) 2023; 12:2078. [PMID: 38136198 PMCID: PMC10741246 DOI: 10.3390/antiox12122078] [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: 10/14/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
The accumulation of oxidative stress is one of the important factors causing cellular senescence. Oxymatrine (OM) is a natural quinolizidine alkaloid compound known for its antioxidant effects. This study aimed to investigate the anti-senescence potential of OM through oxidative stress-induced in vitro and in vivo models. By treating 600 μM of H2O2 to the HT22 mouse hippocampal neuronal cell line and by administering 150 mg/kg D-galactose to mice, we generated oxidative stress-induced senescence models. After providing 1, 2, and 4 μg/mL of OM to the HT22 mouse cell line and by administering 50 mg/kg OM to mice, we evaluated the enhancing effects. We evaluated different senescence markers, AMPK activity, and autophagy, along with DCFH-DA detection reaction and behavioral tests. In HT22 cells, OM showed a protective effect. OM, by reducing ROS and increasing p-AMPK expression, could potentially reduce oxidative stress-induced senescence. In the D-Gal-induced senescence mouse model, both the brain and heart tissues recovered AMPK activity, resulting in reduced levels of senescence. In neural tissue, to assess neurological recovery, including anxiety symptoms and exploration, we used a behavioral test. We also found that OM decreased the expression level of receptors for advanced glycation end products (RAGE). In heart tissue, we could observe the restoration of AMPK activity, which also increased the activity of autophagy. The results of our study suggest that OM ameliorates oxidative stress-induced senescence through its antioxidant action by restoring AMPK activity.
Collapse
Affiliation(s)
- Nagarajan Maharajan
- Department of Biological Science, College of Natural Sciences, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 501759, Republic of Korea; (N.M.); (C.-M.L.); (K.A.V.)
| | - Chang-Min Lee
- Department of Biological Science, College of Natural Sciences, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 501759, Republic of Korea; (N.M.); (C.-M.L.); (K.A.V.)
- BK21 FOUR Education Research Group for Age-Associated Disorder Control Technology, Department of Integrative Biological Science, Chosun University, Gwangju 61452, Republic of Korea
| | - Karthikeyan A. Vijayakumar
- Department of Biological Science, College of Natural Sciences, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 501759, Republic of Korea; (N.M.); (C.-M.L.); (K.A.V.)
| | - Gwang-Won Cho
- Department of Biological Science, College of Natural Sciences, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 501759, Republic of Korea; (N.M.); (C.-M.L.); (K.A.V.)
- BK21 FOUR Education Research Group for Age-Associated Disorder Control Technology, Department of Integrative Biological Science, Chosun University, Gwangju 61452, Republic of Korea
- The Basic Science Institute of Chosun University, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
4
|
Yang Y, Mihajlovic M, Masereeuw R. Protein-Bound Uremic Toxins in Senescence and Kidney Fibrosis. Biomedicines 2023; 11:2408. [PMID: 37760849 PMCID: PMC10525416 DOI: 10.3390/biomedicines11092408] [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: 08/07/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic kidney disease (CKD) is a progressive condition of kidney dysfunction due to diverse causes of injury. In healthy kidneys, protein-bound uremic toxins (PBUTs) are cleared from the systemic circulation by proximal tubule cells through the concerted action of plasma membrane transporters that facilitate their urinary excretion, but the endogenous metabolites are hardly removed with kidney dysfunction and may contribute to CKD progression. Accumulating evidence suggests that senescence of kidney tubule cells influences kidney fibrosis, the common endpoint for CKD with an excessive accumulation of extracellular matrix (ECM). Senescence is a special state of cells characterized by permanent cell cycle arrest and limitation of proliferation, which promotes fibrosis by releasing senescence-associated secretory phenotype (SASP) factors. The accumulation of PBUTs in CKD causes oxidative stress and increases the production of inflammatory (SASP) factors that could trigger fibrosis. Recent studies gave some clues that PBUTs may also promote senescence in kidney tubular cells. This review provides an overview on how senescence contributes to CKD, the involvement of PBUTs in this process, and how kidney senescence can be studied. Finally, some suggestions for future therapeutic options for CKD while targeting senescence are given.
Collapse
Affiliation(s)
- Yi Yang
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| | - Milos Mihajlovic
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium;
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| |
Collapse
|
5
|
Deng W, Jo JI, Morikuni H, Sasayama S, Hashimoto Y, Matsumoto N, Honda Y. Senescence-associated secretory phenotypes in rat-derived dedifferentiated fat cells with replicative senescence. Dent Mater J 2023. [PMID: 36775334 DOI: 10.4012/dmj.2022-242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Senescence-associated secretory phenotype (SASPs) secreted from senescent cells often cause the deleterious damages to the surrounding tissues. Although dedifferentiated fat (DFAT) cells prepared are considered a promising cell source for regenerative therapies, SASPs from DFAT cells undergoing long-term cell culture, which latently induce replicative senescence, have barely been explored. The present study was designed to investigate senescent behaviors in rat-derived DFAT cells at high passage numbers and to analyze the possible types of SASPs. Our data show that DFAT cells undergo senescence during replicative passaging, as determined by multiple senescent hallmarks including morphological changes in cell shape and nucleus. Moreover, RT2 PCR array analysis indicated that senescent DFAT cells expressed higher levels of 16 inflammatory cytokines (Ccl11, Ccl12, Ccl21, Ccl5, Csf2, Cxcl1, Cxcl12, Ifna2, IL11, IL12a, IL13, IL1a, IL1rn, IL6, Mif, and Tnf) associated with SASPs than non-senescent cells. This study implicates that rat DFAT cells undergo cellular senescence after long-term cell culture; cautious consideration should be paid to treat SASP secretion when senescent DFAT cells are used in regenerative medicine.
Collapse
Affiliation(s)
- Wenqi Deng
- Department of Orthodontics, Osaka Dental University
| | | | | | | | | | | | | |
Collapse
|
6
|
Mitochondrial Metabolism in X-Irradiated Cells Undergoing Irreversible Cell-Cycle Arrest. Int J Mol Sci 2023; 24:ijms24031833. [PMID: 36768155 PMCID: PMC9916319 DOI: 10.3390/ijms24031833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Irreversible cell-cycle-arrested cells not undergoing cell divisions have been thought to be metabolically less active because of the unnecessary consumption of energy for cell division. On the other hand, they might be actively involved in the tissue microenvironment through an inflammatory response. In this study, we examined the mitochondria-dependent metabolism in human cells irreversibly arrested in response to ionizing radiation to confirm this possibility. Human primary WI-38 fibroblast cells and the BJ-5ta fibroblast-like cell line were exposed to 20 Gy X-rays and cultured for up to 9 days after irradiation. The mitochondrial morphology and membrane potential were evaluated in the cells using the mitochondrial-specific fluorescent reagents MitoTracker Green (MTG) and 5,5',6,6'-tetraethyl-benzimidazolylcarbocyanine iodide (JC-1), respectively. The ratio of the mean MTG-stained total mitochondrial area per unit cell area decreased for up to 9 days after X-irradiation. The fraction of the high mitochondrial membrane potential area visualized by JC-1 staining reached its minimum 2 days after irradiation and then increased (particularly, WI-38 cells increased 1.8-fold the value of the control). Their chronological changes indicate that the mitochondrial volume in the irreversible cell-cycle-arrested cells showed significant increase concurrently with cellular volume expansion, indicating that the mitochondria-dependent energy metabolism was still active. These results indicate that the energy metabolism in X-ray-induced senescent-like cells is active compared to nonirradiated normal cells, even though they do not undergo cell divisions.
Collapse
|
7
|
The Molecular and Cellular Strategies of Glioblastoma and Non-Small-Cell Lung Cancer Cells Conferring Radioresistance. Int J Mol Sci 2022; 23:ijms232113577. [PMID: 36362359 PMCID: PMC9656305 DOI: 10.3390/ijms232113577] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Ionizing radiation (IR) has been shown to play a crucial role in the treatment of glioblastoma (GBM; grade IV) and non-small-cell lung cancer (NSCLC). Nevertheless, recent studies have indicated that radiotherapy can offer only palliation owing to the radioresistance of GBM and NSCLC. Therefore, delineating the major radioresistance mechanisms may provide novel therapeutic approaches to sensitize these diseases to IR and improve patient outcomes. This review provides insights into the molecular and cellular mechanisms underlying GBM and NSCLC radioresistance, where it sheds light on the role played by cancer stem cells (CSCs), as well as discusses comprehensively how the cellular dormancy/non-proliferating state and polyploidy impact on their survival and relapse post-IR exposure.
Collapse
|
8
|
Suzuki K, Kawamura K, Ujiie R, Nakayama T, Mitsutake N. Characterization of radiation-induced micronuclei associated with premature senescence, and their selective removal by senolytic drug, ABT-263. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 876-877:503448. [PMID: 35483779 DOI: 10.1016/j.mrgentox.2022.503448] [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: 09/26/2021] [Revised: 12/26/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
Radiotherapy is well-recognized as an efficient non-invasive remedy for cancer treatment. Since 10 Gy, a weekly total dose for conventional radiotherapy, was proven to create unreparable and residual DNA double-strand breaks (DSBs), they were found to give rise to mitotic failure, such as mitotic catastrophe, which resulted in multiple micronuclei associated with premature senescence. We demonstrated that pulverization of micronuclear DNA was caspase-dependent and triggered not ATM-dependent but DNA-PK-dependent DNA damage response, including phosphorylation of histone H2AX. Pulverization of micronuclear DNA and senescence-associated secretory phenotype (SASP) worsen tumor microenvironment after radiotherapy, so that senolytic drug was applied to eliminate senescent cancer cells. Prematurely senescent cancer cells with micronuclei caused by 10 Gy of γ-irradiation were subjected to 5 μM of ABT-263, a Bcl-2 family inhibitor, and selective cancer cell death by apoptosis was observed, while ABT-263 had little effect on growing cancer cells. Western blot analysis showed augmented expression of both apoptotic and anti-apoptotic proteins in senescent cells, indicating that increased apoptotic factors are essential for selective apoptotic cell death in combination with ABT-263. Our results suggested that selective elimination of senescent cells alleviates SASP and micronuclei-mediated the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) activation, both of which lead to unfavorable adverse effects caused by radiotherapy.
Collapse
Affiliation(s)
- Keiji Suzuki
- Department of Radiation Medical Sciences, Nagasaki University Atomic Bomb Disease Institute. 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan; Life Sciences and Radiation Research, Graduate School of Biomedical Sciences Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| | - Kasumi Kawamura
- Department of Radiation Medical Sciences, Nagasaki University Atomic Bomb Disease Institute. 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Risa Ujiie
- Life Sciences and Radiation Research, Graduate School of Biomedical Sciences Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Takahumi Nakayama
- Department of Molecular Medicine, Nagasaki University Atomic Bomb Disease Institute. 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Norisato Mitsutake
- Department of Radiation Medical Sciences, Nagasaki University Atomic Bomb Disease Institute. 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan; Life Sciences and Radiation Research, Graduate School of Biomedical Sciences Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| |
Collapse
|
9
|
Al-Jumayli M, Brown SL, Chetty IJ, Extermann M, Movsas B. The Biological Process of Aging and the Impact of Ionizing Radiation. Semin Radiat Oncol 2022; 32:172-178. [DOI: 10.1016/j.semradonc.2021.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
10
|
Vainshelbaum NM, Salmina K, Gerashchenko BI, Lazovska M, Zayakin P, Cragg MS, Pjanova D, Erenpreisa J. Role of the Circadian Clock "Death-Loop" in the DNA Damage Response Underpinning Cancer Treatment Resistance. Cells 2022; 11:880. [PMID: 35269502 PMCID: PMC8909334 DOI: 10.3390/cells11050880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/14/2022] [Accepted: 03/01/2022] [Indexed: 12/11/2022] Open
Abstract
Here, we review the role of the circadian clock (CC) in the resistance of cancer cells to genotoxic treatments in relation to whole-genome duplication (WGD) and telomere-length regulation. The CC drives the normal cell cycle, tissue differentiation, and reciprocally regulates telomere elongation. However, it is deregulated in embryonic stem cells (ESCs), the early embryo, and cancer. Here, we review the DNA damage response of cancer cells and a similar impact on the cell cycle to that found in ESCs—overcoming G1/S, adapting DNA damage checkpoints, tolerating DNA damage, coupling telomere erosion to accelerated cell senescence, and favouring transition by mitotic slippage into the ploidy cycle (reversible polyploidy). Polyploidy decelerates the CC. We report an intriguing positive correlation between cancer WGD and the deregulation of the CC assessed by bioinformatics on 11 primary cancer datasets (rho = 0.83; p < 0.01). As previously shown, the cancer cells undergoing mitotic slippage cast off telomere fragments with TERT, restore the telomeres by ALT-recombination, and return their depolyploidised offspring to telomerase-dependent regulation. By reversing this polyploidy and the CC “death loop”, the mitotic cycle and Hayflick limit count are thus again renewed. Our review and proposed mechanism support a life-cycle concept of cancer and highlight the perspective of cancer treatment by differentiation.
Collapse
Affiliation(s)
- Ninel Miriam Vainshelbaum
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
- Faculty of Biology, University of Latvia, LV-1050 Riga, Latvia
| | - Kristine Salmina
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Bogdan I. Gerashchenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, 03022 Kyiv, Ukraine;
| | - Marija Lazovska
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Pawel Zayakin
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Mark Steven Cragg
- Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK;
| | - Dace Pjanova
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Jekaterina Erenpreisa
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| |
Collapse
|
11
|
Leu JD, Wang CY, Lo CC, Lin MY, Chang CY, Hung WC, Lin ST, Wang BS, Lee YJ. Involvement of c-Myc in low dose radiation-induced senescence enhanced migration and invasion of unirradiated cancer cells. Aging (Albany NY) 2021; 13:22208-22231. [PMID: 34552037 PMCID: PMC8507273 DOI: 10.18632/aging.203527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/11/2021] [Indexed: 12/27/2022]
Abstract
Ionizing radiation is known to cause cell apoptosis at high dose range, but little is known about the cellular response to low dose radiation. In this study, we found that conditioned medium harvested from WI-38 lung fibroblasts and H1299 lung adenocarcinoma cells exposed to 0.1Gy to 1Gy could enhance the migration and invasion of unirradiated H1299 cells in both 2D and 3D culturing circumstances. Low dose radiation did not induce apoptosis, but induced senescence in irradiated cells. We next examined the expression of immediately early genes including c-Myc and K-Ras. Although both genes could be up-regulated by low dose radiation, induction of c-Myc was more specific to low dose range (0.5Gy) at transcriptional and translational levels. Knockdown of c-Myc by shRNA could repress the senescence induced by low dose radiation. The conditioned medium of irradiated cells induced migration of unirradiated cells was also repressed by knockdown of c-Myc. The c-Myc inhibitor 10058-F4 could suppress low dose radiation induced cell senescence, and the conditioned medium harvested from irradiated cells pretreated with 10058-F4 also lost the ability to enhance the migration of unirradiated cells. The cytokine array analysis revealed that immunosuppressive monocyte chemoattractant protein-1 increased by low dose radiation could be repressed by 10058-F4. We also showed that 10058-F4 could suppress low dose radiation induced tumor progression in a xenograft tumor model. Taken together, current data suggest that -Myc is involved in low dose radiation induced cell senescence and potent bystander effect to increase the motility of unirradiated cells.
Collapse
Affiliation(s)
- Jyh-Der Leu
- Department of Radiation Oncology, Taipei City Hospital, Taipei 110, Taiwan.,Institute of Neuroscience, National Cheng Chi University, Taipei 116, Taiwan
| | - Chung-Yih Wang
- Radiotherapy, Department of Medical Imaging, Cheng Hsin General Hospital, Taipei 112, Taiwan
| | - Chia-Chien Lo
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Min-Ying Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chun-Yuan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.,Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08903-2681, USA
| | - Wen-Chin Hung
- Department of Radiation Oncology, Taipei City Hospital, Taipei 110, Taiwan
| | - Shi-Ting Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Bo-Shen Wang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yi-Jang Lee
- Department of Radiation Oncology, Taipei City Hospital, Taipei 110, Taiwan.,Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
12
|
Nagane M, Yasui H, Kuppusamy P, Yamashita T, Inanami O. DNA damage response in vascular endothelial senescence: Implication for radiation-induced cardiovascular diseases. JOURNAL OF RADIATION RESEARCH 2021; 62:564-573. [PMID: 33912932 PMCID: PMC8273807 DOI: 10.1093/jrr/rrab032] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/24/2021] [Indexed: 05/27/2023]
Abstract
A post-exposure cohort study in Hiroshima and Nagasaki reported that low-dose exposure to radiation heightened the risk of cardiovascular diseases (CVD), such as stroke and myocardial infarction, by 14-18% per Gy. Moreover, the risk of atherosclerosis in the coronary arteries reportedly increases with radiation therapy of the chest, including breast and lung cancer treatment. Cellular senescence of vascular endothelial cells (ECs) is believed to play an important role in radiation-induced CVDs. The molecular mechanism of age-related cellular senescence is believed to involve genomic instability and DNA damage response (DDR); the chronic inflammation associated with senescence causes cardiovascular damage. Therefore, vascular endothelial cell senescence is believed to induce the pathogenesis of CVDs after radiation exposure. The findings of several prior studies have revealed that ionizing radiation (IR) induces cellular senescence as well as cell death in ECs. We have previously reported that DDR activates endothelial nitric oxide (NO) synthase, and NO production promotes endothelial senescence. Endothelial NO synthase (eNOS) is a major isoform expressed in ECs that maintains cardiovascular homeostasis. Therefore, radiation-induced NO production, a component of the DDR in ECs, may be involved in CVDs after radiation exposure. In this article, we describe the pathology of radiation-induced CVD and the unique radio-response to radiation exposure in ECs.
Collapse
Affiliation(s)
- Masaki Nagane
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa 252-5201, Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Periannan Kuppusamy
- Department of Radiology, The Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03756, US
| | - Tadashi Yamashita
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa 252-5201, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| |
Collapse
|
13
|
Choi HS, Kim JH, Jang SJ, Yun JW, Kang KM, Jeong H, Ha IB, Jeong BK. Synergistic Tumoricidal Effects of Alpha-Lipoic Acid and Radiotherapy on Human Breast Cancer Cells via HMGB1. Cancer Res Treat 2021; 53:685-694. [PMID: 33321563 PMCID: PMC8291200 DOI: 10.4143/crt.2020.1015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/13/2020] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Radiotherapy (RT) is one of main strategies of cancer treatment. However, some cancer cells are resistant to radiation-induced cell death, including apoptosis. Therefore, alternative approaches targeting different anti-tumor mechanisms such as cell senescence are required. This study aimed to investigate the synergistic effect of alpha-lipoic acid (ALA) on radiation-induced cell death and senescence in MDA-MB-231 human breast cancer cells. MATERIALS AND METHODS The cells were divided into four groups depending on the cell treatment (control, ALA, RT, and ALA+RT). Cells were analyzed for morphology, apoptotic cell death, mitochondrial reactive oxygen species, membrane potential, cellular senescence, and cell cycle. RESULTS Our data showed that ALA significantly promoted apoptotic cell death when combined with RT, as reflected by Annexin V staining, expression of apoptosis-related factors, mitochondrial damages as well as cell morphological changes and reduction of cell numbers. In addition, ALA significantly enhanced radiation-induced cellular senescence, which was shown by increased HMGB1 expression in the cytosol fraction compared to the control, increased p53 expression compared to the control, activation of p38 as well as nuclear factor кB, and G2/M cell cycle arrest. CONCLUSION The current study is the first report showing a new mode of action (senescence induction) of ALA beyond apoptotic cell death in MDA-MB-231 cancer cells known to be resistant to RT.
Collapse
Affiliation(s)
- Hoon Sik Choi
- Department of Radiation Oncology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
- Institute of Health Science, Gyeongsang National University, Jinju, Korea
| | - Jin Hyun Kim
- Institute of Health Science, Gyeongsang National University, Jinju, Korea
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Korea
| | - Si Jung Jang
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Korea
| | - Jeong Won Yun
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Korea
| | - Ki Mun Kang
- Department of Radiation Oncology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
- Institute of Health Science, Gyeongsang National University, Jinju, Korea
| | - Hojin Jeong
- Institute of Health Science, Gyeongsang National University, Jinju, Korea
- Department of Radiation Oncology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| | - In Bong Ha
- Institute of Health Science, Gyeongsang National University, Jinju, Korea
- Department of Radiation Oncology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Bae Kwon Jeong
- Institute of Health Science, Gyeongsang National University, Jinju, Korea
- Department of Radiation Oncology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| |
Collapse
|
14
|
Giri S, Takada A, Paudel D, Yoshida K, Furukawa M, Kuramitsu Y, Matsushita K, Abiko Y, Furuichi Y. An in vitro senescence model of gingival epithelial cell induced by hydrogen peroxide treatment. Odontology 2021; 110:44-53. [PMID: 34143349 DOI: 10.1007/s10266-021-00630-3] [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: 04/26/2021] [Accepted: 06/14/2021] [Indexed: 11/30/2022]
Abstract
Gingival tissue shows progressive changes with aging and an in vitro model of gingival tissue could be useful in understanding age-associated oral diseases. The present study aims to establish a hydrogen peroxide (H2O2) treatment model to induce aging in human gingival epithelial cells. In addition, fisetin, a flavonoid component studied for the anti-aging property is used to examine if it could reverse the induced senescence. Primary human gingival epithelial progenitor (HGEPp) cells were cultured and treated with different concentrations of H2O2. A cell vitality and morphology, senescence-associated beta-galactosidase (SA-β-gal) staining, mRNA and protein expression analysis of known senescence markers p16, p21, and p53, and cell cycle assay were performed. The cells showed dose-dependent changes in vitality and morphology, SA-β-gal staining, relative mRNA and protein expression, and cell cycle assay after H2O2 treatment. Based on these results, 400 μM H2O2 was considered as an optimal concentration to induce senescence. Treatment of senescence-induced cells with fisetin downregulated all the senescence markers used in this study. In conclusion, a senescence model of gingival epithelial cells induced by hydrogen peroxide treatment was established which could be employed to study age-related periodontal diseases.
Collapse
Affiliation(s)
- Sarita Giri
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, 061-0293, Japan
| | - Ayuko Takada
- Division of Biochemistry, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Durga Paudel
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Koki Yoshida
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Masae Furukawa
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yasuhiro Kuramitsu
- Research Institute of Cancer Prevention, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Kenji Matsushita
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yoshihiro Abiko
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Yasushi Furuichi
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, 061-0293, Japan.
| |
Collapse
|
15
|
Dabrowska M, Uram L, Dabrowski M, Sikora E. Antigen presentation capability and AP-1 activation accompany methotrexate-induced colon cancer cell senescence in the context of aberrant β-catenin signaling. Mech Ageing Dev 2021; 197:111517. [PMID: 34139213 DOI: 10.1016/j.mad.2021.111517] [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: 12/05/2020] [Revised: 05/19/2021] [Accepted: 06/09/2021] [Indexed: 01/10/2023]
Abstract
Reversible cellular senescence was demonstrated previously to constitute colon cancer cell response to methotrexate. The current study presents a comparison of two senescent states of colon cancer cells, arrested and reversing, resulting from respectively, 120 h exposure to the drug, and 48 h exposure followed by 96 h regrowth in drug-free media. The upregulation of immunoproteasome subunit-coding genes and the increase in human leukocyte antigen HLA-A/B/C membrane level indicated MHC-I-restricted antigen presentation as common to both senescent states. Nuclear factor NF-κB p65 level decreased and activating protein AP-1: c-Jun, Fra2 and JunB nuclear levels increased in both senescent cell populations. Notably, the increase in AP-1- dependent transcription occurred after 48 h exposure to methotrexate. β-catenin nuclear level increased after 48 h exposure to the drug and remained as such only in senescence-arrested cells. β-catenin level was found uncoupled from the protein phosphorylation status indicating the deregulation of β-catenin signaling in colon cancer cells employed in the study. These findings carry implications for both, a general mechanism of senescence establishment and putative advantages for colon cancer treatment.
Collapse
Affiliation(s)
- Magdalena Dabrowska
- Laboratory of Molecular Bases of Ageing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093, Warszawa, Poland.
| | - Lukasz Uram
- Faculty of Chemistry, Rzeszow University of Technology, 6 Powstancow Warszawy Ave., 35-959, Rzeszow, Poland.
| | - Michal Dabrowski
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093, Warszawa, Poland.
| | - Ewa Sikora
- Laboratory of Molecular Bases of Ageing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093, Warszawa, Poland.
| |
Collapse
|
16
|
Suzuki K, Amrenova A, Mitsutake N. Recent advances in radiobiology with respect to pleiotropic aspects of tissue reaction. JOURNAL OF RADIATION RESEARCH 2021; 62:i30-i35. [PMID: 33978178 PMCID: PMC8114206 DOI: 10.1093/jrr/rraa086] [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] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/09/2020] [Indexed: 06/12/2023]
Abstract
DNA double-strand breaks (DSBs) induced by ionizing radiation are the major cause of cell death, leading to tissue/organ injuries, which is a fundamental mechanism underlying the development of tissue reaction. Since unscheduled senescence, predominantly induced among epithelial tissues/organs, is one of the major modes of cell death in response to radiation exposure, its role in tissue reaction has been extensively studied, and it has become clear that senescence-mediated secretion of soluble factors is an indispensable component of the manifestation of tissue reaction. Recently, an unexpected link between cytoplasmic DSBs and innate immunity was discovered. The activation of cyclic GMP-AMP (cGAMP) synthase (cGAS) results in the stimulation of the cGAS-stimulator of interferon genes (STING) pathway, which has been shown to regulate the transactivation of a variety of secretory factors that are the same as those secreted from senescent cells. Furthermore, it has been proven that cGAS-STING pathway also mediates execution of the senescence process by itself. Hence, an autocrine/paracrine feedback loop has been discussed in previous literature in relation to its effect on the tissue microenvironment. As the tissue microenvironment plays a crucial role in cancer development, tissue reaction could be involved in the late health effects caused by radiation exposure. In this paper, the novel findings in radiation biology, which should provide a better understanding of the mechanisms underlying radiation-induced carcinogenesis, are overviewed.
Collapse
Affiliation(s)
- Keiji Suzuki
- Department of Radiation Medical Sciences, Nagasaki University Atomic Bomb Disease Institute. 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
- Life Sciences and Radiation Research, Graduate School of Biomedical Sciences, Nagasaki University. 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Aidana Amrenova
- Department of Radiation Medical Sciences, Nagasaki University Atomic Bomb Disease Institute. 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
- Life Sciences and Radiation Research, Graduate School of Biomedical Sciences, Nagasaki University. 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Norisato Mitsutake
- Department of Radiation Medical Sciences, Nagasaki University Atomic Bomb Disease Institute. 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
- Life Sciences and Radiation Research, Graduate School of Biomedical Sciences, Nagasaki University. 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| |
Collapse
|
17
|
Zhang W, Yang J, Chen Y, Xue R, Mao Z, Lu W, Jiang Y. Lycorine hydrochloride suppresses stress-induced premature cellular senescence by stabilizing the genome of human cells. Aging Cell 2021; 20:e13307. [PMID: 33455051 PMCID: PMC7884038 DOI: 10.1111/acel.13307] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/09/2020] [Accepted: 12/25/2020] [Indexed: 12/11/2022] Open
Abstract
Lycorine, a natural compound isolated from the traditional Chinese medicinal herb Lycoris radiata, exhibits multiple pharmacological effects, such as anti-inflammatory, antiviral, and anticancer effects. Accumulating evidence also indicates that lycorine might hold the potential to treat age-associated Alzheimer's disease. However, whether lycorine is involved in delaying the onset of cellular senescence and its underlying mechanisms has not been determined. Here, we demonstrate that the salt of lycorine, lycorine hydrochloride, significantly suppressed stress-induced premature cellular senescence (SIPS) by ~2-fold, as determined by senescence-associated beta-galactosidase (SA-β-gal) staining and the expression of p16 and p21. In addition, pretreating cells with lycorine hydrochloride significantly inhibited the expression of CXCL1 and IL1α, two factors of the senescence-associated secreted phenotype (SASP) in SIPS cells. Further experiments revealed that lycorine hydrochloride promoted both the homologous recombination (HR) and nonhomologous end joining (NHEJ) pathways of DNA double-strand break (DSB) repair. Mechanistic studies suggested that lycorine hydrochloride treatment promoted the transcription of SIRT1 and SIRT6, critical longevity genes positively regulating both HR and NHEJ repair pathways, thereby stimulating DSB repair and stabilizing genomes. Inhibiting SIRT1 enzymatic activity abrogated the protective effect of lycorine hydrochloride on delaying the onset of SIPS, repairing DSBs, and restoring genome integrity. In summary, our work indicates that lycorine hydrochloride might hold therapeutic potential for treating age-associated diseases or promoting healthy aging by stabilizing genomes.
Collapse
Affiliation(s)
- Weina Zhang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Jiaqing Yang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Yu Chen
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Renhao Xue
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Zhiyong Mao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| | - Wen Lu
- Department of Gynecology of Shanghai First Maternity & Infant Hospital Tongji University School of Medicine Shanghai China
| | - Ying Jiang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research Frontier Science Center for Stem Cell Research School of Life Sciences and Technology Tongji University Shanghai China
| |
Collapse
|
18
|
Dutta B, Asami T, Imatomi T, Igarashi K, Nagata K, Watanabe-Asaka T, Yasuda T, Oda S, Shartl M, Mitani H. Strain difference in transgene-induced tumorigenesis and suppressive effect of ionizing radiation. JOURNAL OF RADIATION RESEARCH 2021; 62:12-24. [PMID: 33231252 PMCID: PMC7779347 DOI: 10.1093/jrr/rraa103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/30/2020] [Indexed: 06/11/2023]
Abstract
Transgenic expression in medaka of the Xiphophorus oncogene xmrk, under a pigment cell specific mitf promoter, induces hyperpigmentation and pigment cell tumors. In this study, we crossed the Hd-rR and HNI inbred strains because complete genome information is readily available for molecular and genetic analysis. We prepared an Hd-rR (p53+/-, p53-/-) and Hd-rR HNI hybrid (p53+/-) fish-based xmrk model system to study the progression of pigment cells from hyperpigmentation to malignant tumors on different genetic backgrounds. In all strains examined, most of the initial hyperpigmentation occurred in the posterior region. On the Hd-rR background, mitf:xmrk-induced tumorigenesis was less frequent in p53+/- fish than in p53-/- fish. The incidence of hyperpigmentation was more frequent in Hd-rR/HNI hybrids than in Hd-rR homozygotes; however, the frequency of malignant tumors was low, which suggested the presence of a tumor suppressor in HNI genetic background fish. The effects on tumorigenesis in xmrk-transgenic immature medaka of a single 1.3 Gy irradiation was assessed by quantifying tumor progression over 4 consecutive months. The results demonstrate that irradiation has a different level of suppressive effect on the frequency of hyperpigmentation in purebred Hd-rR compared with hybrids.
Collapse
Affiliation(s)
- Bibek Dutta
- Laboratory of Genome Stability, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562, Kashiwa, Japan
| | - Taichi Asami
- Laboratory of Genome Stability, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562, Kashiwa, Japan
| | - Tohru Imatomi
- Laboratory of Genome Stability, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562, Kashiwa, Japan
| | - Kento Igarashi
- Laboratory of Genome Stability, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562, Kashiwa, Japan
| | - Kento Nagata
- Laboratory of Genome Stability, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562, Kashiwa, Japan
| | - Tomomi Watanabe-Asaka
- Laboratory of Genome Stability, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562, Kashiwa, Japan
| | - Takako Yasuda
- Laboratory of Genome Stability, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562, Kashiwa, Japan
| | - Shoji Oda
- Laboratory of Genome Stability, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562, Kashiwa, Japan
| | - Manfred Shartl
- University of Wuerzburg, Physiological Chemistry, Biocenter, 97074 Wuerzburg, Germany and the Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas, 78666, USA
| | - Hiroshi Mitani
- Corresponding author. Laboratory of Genome Stability, Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha 277-8562, Kashiwa, Japan. Tel: +81(4) 7136-3670; Fax:+81(4)7136-3669;
| |
Collapse
|
19
|
The conformation-specific Hsp90 inhibition interferes with the oncogenic RAF kinase adaptation and triggers premature cellular senescence, hence, acts as a tumor suppressor mechanism. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118943. [PMID: 33359710 DOI: 10.1016/j.bbamcr.2020.118943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/23/2020] [Accepted: 12/18/2020] [Indexed: 12/28/2022]
Abstract
Cancer emergence is associated with cellular adaptations to altered signal transduction mechanisms arbitrated by mutated kinases. Since conventional kinase inhibitors can exhibit certain limitations to such kinase adaptations, overcoming kinase adaptation for cancer treatment gains importance. The cancer chaperone, Hsp90, is implicated in the conformational maturation and functional stabilization of mutated gene products. However, its role in kinase adaptations is not explored in detail. Therefore, the present study aims to understand the mechanisms of Hsp90-dependent kinase adaptation and develop a novel antitumor strategy. We chose malignant human lung cancer cells to demonstrate Hsp90-dependent RAF oncogene adaptation. We show that RAF oncogene adaptations were predominant over wild type RAF and are facilitated by conformation-specific Hsp90. Consequently, the conformation-specific Hsp90 inhibitor, 17AAG, interfered with oncogenic RAF stability and function and inhibited cell proliferation. The enforced cytostasis further triggered premature cellular senescence and acted as an efficient and irreversible tumor suppressor mechanism. Our results also display that oncogenic RAF interactions with Hsp90 require the middle-charged region of the chaperone. Our mice xenografts revealed that 17AAG pretreated tumor cells lost their ability to proliferate and metastasize in vivo. In summary, we demonstrated Hsp90-dependent kinase adaptation in tumor cells and the effect of Hsp90 inhibition in triggering premature senescence to interfere with the tumor progression. Our findings are of both biological relevance and clinical importance.
Collapse
|
20
|
Kaur A, Macip S, Stover CM. An Appraisal on the Value of Using Nutraceutical Based Senolytics and Senostatics in Aging. Front Cell Dev Biol 2020; 8:218. [PMID: 32309282 PMCID: PMC7145958 DOI: 10.3389/fcell.2020.00218] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/13/2020] [Indexed: 01/10/2023] Open
Abstract
The average human life expectancy has increased globally, and continues to rise, owing to the substantive progress made in healthcare, medicine, sanitation, housing and education. This ultimately enriches society with a greater proportion of elderly people. Sustaining a healthy aged population is key to diminish the societal and economic impact of age-related infirmities. This is especially challenging because tissue function, and thus wellbeing, naturally progressively decline as humans age. With age increasing the risk of developing diseases, one of the therapeutic options is to interfere with the molecular and cellular pathways involved in age-related tissue dysfunction, which is in part caused by the accumulation of senescent cells. One strategy to prevent this could be using drugs that selectively kill these cells (senolytics). In parallel, some compounds have been identified that prevent or slow down the progression of senescence or some of its features (senostatics). Senolytic and senostatic therapies have been shown to be efficient in vivo, but they also have unwanted dose-dependent side effects, including toxicity. Important advances might be made using bioactive compounds from plants and foods (nutraceuticals) if, as is proposed, they offer similar effectiveness with fewer side effects. The focus of this review is on the use of nutraceuticals in interfering with cellular senescence.
Collapse
Affiliation(s)
- Amanpreet Kaur
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Salvador Macip
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom.,Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Cordula M Stover
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
21
|
Gupta S, Silveira DA, Mombach JCM. ATM/miR‐34a‐5p axis regulates a p21‐dependent senescence‐apoptosis switch in non‐small cell lung cancer: a Boolean model of G1/S checkpoint regulation. FEBS Lett 2019; 594:227-239. [DOI: 10.1002/1873-3468.13615] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/16/2019] [Accepted: 09/19/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Shantanu Gupta
- Department of Physics Universidade Federal de Santa Maria Brazil
| | | | | |
Collapse
|
22
|
The DNA-damage response and nuclear events as regulators of nonapoptotic forms of cell death. Oncogene 2019; 39:1-16. [PMID: 31462710 DOI: 10.1038/s41388-019-0980-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/05/2019] [Accepted: 08/09/2019] [Indexed: 12/20/2022]
Abstract
The maintenance of genome stability is essential for the cell as the integrity of genomic information guaranties reproduction of a whole organism. DNA damage occurring in response to different natural and nonnatural stimuli (errors in DNA replication, UV radiation, chemical agents, etc.) is normally detected by special cellular machinery that induces DNA repair. However, further accumulation of genetic lesions drives the activation of cell death to eliminate cells with defective genome. This particular feature is used for targeting fast-proliferating tumor cells during chemo-, radio-, and immunotherapy. Among different cell death modalities induced by DNA damage, apoptosis is the best studied. Nevertheless, nonapoptotic cell death and adaptive stress responses are also activated following genotoxic stress and play a crucial role in the outcome of anticancer therapy. Here, we provide an overview of nonapoptotic cell death pathways induced by DNA damage and discuss their interplay with cellular senescence, mitotic catastrophe, and autophagy.
Collapse
|
23
|
Kornienko JS, Smirnova IS, Pugovkina NA, Ivanova JS, Shilina MA, Grinchuk TM, Shatrova AN, Aksenov ND, Zenin VV, Nikolsky NN, Lyublinskaya OG. High doses of synthetic antioxidants induce premature senescence in cultivated mesenchymal stem cells. Sci Rep 2019; 9:1296. [PMID: 30718685 PMCID: PMC6361906 DOI: 10.1038/s41598-018-37972-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/13/2018] [Indexed: 12/19/2022] Open
Abstract
Stress-induced premature senescence program is known to be activated in cells by various genotoxic stressors, and oxidative stress is considered to be the main of those. To this end, many studies discover antioxidants as protective anti-aging agents. In the current study, we examined the effects of different antioxidants (Tempol, resveratrol, NAC, DPI) on the mesenchymal stem cells maintained in normal physiological conditions. We used high, but non-cytotoxic antioxidant doses which are widely used in laboratory practice to protect cells from oxidative damage. We show that these substances induce reversible block of cell proliferation and do not cause any genotoxic effects when applied to the quiescent cells. However, the same doses of the same substances, when applied to the proliferating cells, can induce irreversible cell cycle arrest, DNA strand breaks accumulation and DNA damage response activation. As a consequence, antioxidant-induced DNA damage results in the stress-induced premature senescence program activation. We conclude that high doses of antioxidants, when applied to the proliferating cells that maintain physiological levels of reactive oxygen species, can cause DNA damage and induce premature senescence which suggests to re-estimate believed unconditional anti-aging antioxidant properties.
Collapse
Affiliation(s)
- Ju S Kornienko
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - I S Smirnova
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - N A Pugovkina
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - Ju S Ivanova
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - M A Shilina
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - T M Grinchuk
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - A N Shatrova
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - N D Aksenov
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - V V Zenin
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - N N Nikolsky
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - O G Lyublinskaya
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia.
| |
Collapse
|
24
|
Prevost V, Sichel F, Pottier I, Leduc A, Lagadu S, Laurent C. Production of early and late nuclear DNA damage and extracellular 8-oxodG in normal human skin fibroblasts after carbon ion irradiation compared to X-rays. Toxicol In Vitro 2018; 52:116-121. [PMID: 29879454 DOI: 10.1016/j.tiv.2018.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Virginie Prevost
- Normandie Univ, UNICAEN, INSERM U 1086, ANTICIPE, 14000 Caen, France; CLCC François Baclesse, 3 avenue du Général Harris, 14076 Caen, France
| | - François Sichel
- CLCC François Baclesse, 3 avenue du Général Harris, 14076 Caen, France; Normandie Univ, UNICAEN, UNIROUEN, ABTE, 14000 Caen, France
| | - Ivannah Pottier
- CLCC François Baclesse, 3 avenue du Général Harris, 14076 Caen, France; Normandie Univ, UNICAEN, UNIROUEN, ABTE, 14000 Caen, France
| | - Alexandre Leduc
- SAPHYN (ARCHADE Program), 3 avenue du Général Harris, 14076 Caen, France
| | - Stéphanie Lagadu
- CLCC François Baclesse, 3 avenue du Général Harris, 14076 Caen, France; Normandie Univ, UNICAEN, UNIROUEN, ABTE, 14000 Caen, France
| | - Carine Laurent
- CLCC François Baclesse, 3 avenue du Général Harris, 14076 Caen, France; Normandie Univ, UNICAEN, UNIROUEN, ABTE, 14000 Caen, France; SAPHYN (ARCHADE Program), 3 avenue du Général Harris, 14076 Caen, France.
| |
Collapse
|
25
|
Cortese F, Klokov D, Osipov A, Stefaniak J, Moskalev A, Schastnaya J, Cantor C, Aliper A, Mamoshina P, Ushakov I, Sapetsky A, Vanhaelen Q, Alchinova I, Karganov M, Kovalchuk O, Wilkins R, Shtemberg A, Moreels M, Baatout S, Izumchenko E, de Magalhães JP, Artemov AV, Costes SV, Beheshti A, Mao XW, Pecaut MJ, Kaminskiy D, Ozerov IV, Scheibye-Knudsen M, Zhavoronkov A. Vive la radiorésistance!: converging research in radiobiology and biogerontology to enhance human radioresistance for deep space exploration and colonization. Oncotarget 2018; 9:14692-14722. [PMID: 29581875 PMCID: PMC5865701 DOI: 10.18632/oncotarget.24461] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022] Open
Abstract
While many efforts have been made to pave the way toward human space colonization, little consideration has been given to the methods of protecting spacefarers against harsh cosmic and local radioactive environments and the high costs associated with protection from the deleterious physiological effects of exposure to high-Linear energy transfer (high-LET) radiation. Herein, we lay the foundations of a roadmap toward enhancing human radioresistance for the purposes of deep space colonization and exploration. We outline future research directions toward the goal of enhancing human radioresistance, including upregulation of endogenous repair and radioprotective mechanisms, possible leeways into gene therapy in order to enhance radioresistance via the translation of exogenous and engineered DNA repair and radioprotective mechanisms, the substitution of organic molecules with fortified isoforms, and methods of slowing metabolic activity while preserving cognitive function. We conclude by presenting the known associations between radioresistance and longevity, and articulating the position that enhancing human radioresistance is likely to extend the healthspan of human spacefarers as well.
Collapse
Affiliation(s)
- Franco Cortese
- Biogerontology Research Foundation, London, UK
- Department of Biomedical and Molecular Sciences, Queen's University School of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Dmitry Klokov
- Canadian Nuclear Laboratories, Chalk River, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Andreyan Osipov
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Jakub Stefaniak
- Biogerontology Research Foundation, London, UK
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Alexey Moskalev
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, Russia
| | - Jane Schastnaya
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
| | - Charles Cantor
- Boston University, Department of Biomedical Engineering, Boston, MA, USA
| | - Alexander Aliper
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- Laboratory of Bioinformatics, D. Rogachev Federal Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Polina Mamoshina
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- Computer Science Department, University of Oxford, Oxford, UK
| | - Igor Ushakov
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow, Russia
| | - Alex Sapetsky
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow, Russia
| | - Quentin Vanhaelen
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
| | - Irina Alchinova
- Laboratory of Physicochemical and Ecological Pathophysiology, Institute of General Pathology and Pathophysiology, Moscow, Russia
- Research Institute for Space Medicine, Federal Medical Biological Agency, Moscow, Russia
| | - Mikhail Karganov
- Laboratory of Physicochemical and Ecological Pathophysiology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Olga Kovalchuk
- Canada Cancer and Aging Research Laboratories, Ltd., Lethbridge, Alberta, Canada
- University of Lethbridge, Lethbridge, Alberta, Canada
| | - Ruth Wilkins
- Environmental and Radiation and Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Andrey Shtemberg
- Laboratory of Extreme Physiology, Institute of Medical and Biological Problems RAS, Moscow, Russia
| | - Marjan Moreels
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre, (SCK·CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre, (SCK·CEN), Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Evgeny Izumchenko
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- The Johns Hopkins University, School of Medicine, Department of Otolaryngology, Head and Neck Cancer Research, Baltimore, MD, USA
| | - João Pedro de Magalhães
- Biogerontology Research Foundation, London, UK
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Artem V. Artemov
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
| | | | - Afshin Beheshti
- Wyle Laboratories, Space Biosciences Division, NASA Ames Research Center, Mountain View, CA, USA
- Division of Hematology/Oncology, Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, USA
| | - Michael J. Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, USA
| | - Dmitry Kaminskiy
- Biogerontology Research Foundation, London, UK
- Deep Knowledge Life Sciences, London, UK
| | - Ivan V. Ozerov
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow, Russia
| | | | - Alex Zhavoronkov
- Biogerontology Research Foundation, London, UK
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
26
|
Begolly S, Olschowka JA, Love T, Williams JP, O'Banion MK. Fractionation enhances acute oligodendrocyte progenitor cell radiation sensitivity and leads to long term depletion. Glia 2017; 66:846-861. [PMID: 29288597 DOI: 10.1002/glia.23288] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 12/18/2022]
Abstract
Ionizing radiation (IR) is commonly used to treat central nervous system (CNS) cancers and metastases. While IR promotes remission, frequent side effects including impaired cognition and white matter loss occur following treatment. Fractionation is used to minimize these CNS late side effects, as it reduces IR effects in differentiated normal tissue, but not rapidly proliferating normal or tumor tissue. However, side effects occur even with the use of fractionated paradigms. Oligodendrocyte progenitor cells (OPCs) are a proliferative population within the CNS affected by radiation. We hypothesized that fractionated radiation would lead to OPC loss, which could contribute to the delayed white matter loss seen after radiation exposure. We found that fractionated IR induced a greater early loss of OPCs than an equivalent single dose exposure. Furthermore, OPC recovery was impaired following fractionated IR. Finally, reduced OPC differentiation and mature oligodendrocyte numbers occurred in single dose and fractionated IR paradigms. This work demonstrates that fractionation does not spare normal brain tissue and, importantly, highlights the sensitivity of OPCs to fractionated IR, suggesting that fractionated schedules may promote white matter dysfunction, a point that should be considered in radiotherapy.
Collapse
Affiliation(s)
- Sage Begolly
- Departments of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York
| | - John A Olschowka
- Department of Neuroscience and Del Monte Neuroscience Institute, University of Rochester School of Medicine & Dentistry, Rochester, New York
| | - Tanzy Love
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine & Dentistry, Rochester, New York
| | - Jacqueline P Williams
- Departments of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York.,Department of Radiation Oncology, University of Rochester School of Medicine & Dentistry, Rochester, New York
| | - M Kerry O'Banion
- Department of Neuroscience and Del Monte Neuroscience Institute, University of Rochester School of Medicine & Dentistry, Rochester, New York.,Department of Neurology, University of Rochester School of Medicine & Dentistry, Rochester, New York
| |
Collapse
|
27
|
McCart EA, Thangapazham RL, Lombardini ED, Mog SR, Panganiban RAM, Dickson KM, Mansur RA, Nagy V, Kim SY, Selwyn R, Landauer MR, Darling TN, Day RM. Accelerated senescence in skin in a murine model of radiation-induced multi-organ injury. JOURNAL OF RADIATION RESEARCH 2017; 58:636-646. [PMID: 28340212 PMCID: PMC5737212 DOI: 10.1093/jrr/rrx008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/10/2017] [Indexed: 05/24/2023]
Abstract
Accidental high-dose radiation exposures can lead to multi-organ injuries, including radiation dermatitis. The types of cellular damage leading to radiation dermatitis are not completely understood. To identify the cellular mechanisms that underlie radiation-induced skin injury in vivo, we evaluated the time-course of cellular effects of radiation (14, 16 or 17 Gy X-rays; 0.5 Gy/min) in the skin of C57BL/6 mice. Irradiation of 14 Gy induced mild inflammation, observed histologically, but no visible hair loss or erythema. However, 16 or 17 Gy radiation induced dry desquamation, erythema and mild ulceration, detectable within 14 days post-irradiation. Histological evaluation revealed inflammation with mast cell infiltration within 14 days. Fibrosis occurred 80 days following 17 Gy irradiation, with collagen deposition, admixed with neutrophilic dermatitis, and necrotic debris. We found that in cultures of normal human keratinocytes, exposure to 17.9 Gy irradiation caused the upregulation of p21/waf1, a marker of senescence. Using western blot analysis of 17.9 Gy-irradiated mice skin samples, we also detected a marker of accelerated senescence (p21/waf1) 7 days post-irradiation, and a marker of cellular apoptosis (activated caspase-3) at 30 days, both preceding histological evidence of inflammatory infiltrates. Immunohistochemistry revealed reduced epithelial stem cells from hair follicles 14-30 days post-irradiation. Furthermore, p21/waf1 expression was increased in the region of the hair follicle stem cells at 14 days post 17 Gy irradiation. These data indicate that radiation induces accelerated cellular senescence in the region of the stem cell population of the skin.
Collapse
Affiliation(s)
- Elizabeth A McCart
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Rajesh L Thangapazham
- Department of Dermatology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Eric D Lombardini
- Current address: Public Health Activity-Fort Carson, 1661 O'Connell Blvd, Fort Carson, CO 80913, USA
| | - Steven R Mog
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, 5001 Campus Drive, College Park, MD 20740, USA
| | - Ronald Allan M Panganiban
- Current address: Molecular and Integrative Physiological Sciences, Harvard TH Chan School of Public Health, 677 Huntington Ave., Boston, MA 02115, USA
| | - Kelley M Dickson
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Rihab A Mansur
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Vitaly Nagy
- Department of Radiation Dosimetry, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Sung-Yop Kim
- Current address: Department of Radiology, University of New Mexico, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Reed Selwyn
- Current address: Department of Radiology, University of New Mexico, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Michael R Landauer
- Radiation Countermeasures Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Thomas N Darling
- Department of Dermatology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Regina M Day
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
28
|
Mirzayans R, Andrais B, Murray D. Do Multiwell Plate High Throughput Assays Measure Loss of Cell Viability Following Exposure to Genotoxic Agents? Int J Mol Sci 2017; 18:ijms18081679. [PMID: 28767065 PMCID: PMC5578069 DOI: 10.3390/ijms18081679] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 12/31/2022] Open
Abstract
Cell-based assays in multiwell plates are widely used for radiosensitivity and chemosensitivity assessment with different mammalian cell types. Despite their relative ease of performance, such assays lack specificity as they do not distinguish between the cytostatic (reversible/sustained growth arrest) and cytotoxic (loss of viability) effects of genotoxic agents. We recently reported studies with solid tumor-derived cell lines demonstrating that radiosensitivity as measured by multiwell plate colorimetric (e.g., XTT) and fluorimetric (e.g., CellTiter-Blue) assays reflects growth arrest but not loss of viability. Herein we report similar observations with cancer cell lines expressing wild-type p53 (A549 lung carcinoma) or mutant p53 (MDA–MB-231 breast carcinoma) after treatment with the chemotherapeutic drug cisplatin. Importantly, we show that treatment of cancer cells with concentrations of cisplatin that result in 50% effect (i.e., IC50) in multiwell plate assays trigger the emergence of growth-arrested cells that exhibit highly enlarged morphology, remain viable and adherent to the culture dish, and metabolize the tetrazolium salt 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) to its formazan derivative. The emergence of markedly enlarged viable cells complicates the interpretation of chemosensitivity data obtained with multiwell plate high throughput assays. Relying solely on IC50 values could be misleading.
Collapse
Affiliation(s)
- Razmik Mirzayans
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada.
| | - Bonnie Andrais
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada.
| | - David Murray
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada.
| |
Collapse
|
29
|
Wu Q, Allouch A, Martins I, Brenner C, Modjtahedi N, Deutsch E, Perfettini JL. Modulating Both Tumor Cell Death and Innate Immunity Is Essential for Improving Radiation Therapy Effectiveness. Front Immunol 2017; 8:613. [PMID: 28603525 PMCID: PMC5445662 DOI: 10.3389/fimmu.2017.00613] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 05/09/2017] [Indexed: 12/17/2022] Open
Abstract
Radiation therapy is one of the cornerstones of cancer treatment. In tumor cells, exposure to ionizing radiation (IR) provokes DNA damages that trigger various forms of cell death such as apoptosis, necrosis, autophagic cell death, and mitotic catastrophe. IR can also induce cellular senescence that could serve as an additional antitumor barrier in a context-dependent manner. Moreover, accumulating evidence has demonstrated that IR interacts profoundly with tumor-infiltrating immune cells, which cooperatively drive treatment outcomes. Recent preclinical and clinical successes due to the combination of radiation therapy and immune checkpoint blockade have underscored the need for a better understanding of the interplay between radiation therapy and the immune system. In this review, we will present an overview of cell death modalities induced by IR, summarize the immunogenic properties of irradiated cancer cells, and discuss the biological consequences of IR on innate immune cell functions, with a particular attention on dendritic cells, macrophages, and NK cells. Finally, we will discuss their potential applications in cancer treatment.
Collapse
Affiliation(s)
- Qiuji Wu
- Cell Death and Aging Team, Gustave Roussy Cancer Campus, Villejuif, France.,Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France.,Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Awatef Allouch
- Cell Death and Aging Team, Gustave Roussy Cancer Campus, Villejuif, France.,Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| | - Isabelle Martins
- Cell Death and Aging Team, Gustave Roussy Cancer Campus, Villejuif, France.,Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| | - Catherine Brenner
- Laboratory of Signaling and Cardiovascular Pathophysiology, INSERM UMR-S 1180, Université Paris-Sud, Faculté de Pharmacie, Châtenay-Malabry, France
| | - Nazanine Modjtahedi
- Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| | - Eric Deutsch
- Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| | - Jean-Luc Perfettini
- Cell Death and Aging Team, Gustave Roussy Cancer Campus, Villejuif, France.,Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Villejuif, France
| |
Collapse
|
30
|
Acyl-CoA thioesterase 7 is involved in cell cycle progression via regulation of PKCζ-p53-p21 signaling pathway. Cell Death Dis 2017; 8:e2793. [PMID: 28518146 PMCID: PMC5584527 DOI: 10.1038/cddis.2017.202] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 03/14/2017] [Accepted: 04/06/2017] [Indexed: 11/08/2022]
Abstract
Acyl-CoA thioesterase 7 (ACOT7) is a major isoform of the ACOT family that catalyzes hydrolysis of fatty acyl-CoAs to free fatty acids and CoA-SH. However, canonical and non-canonical functions of ACOT7 remain to be discovered. In this study, for the first time, ACOT7 was shown to be responsive to genotoxic stresses such as ionizing radiation (IR) and the anti-cancer drug doxorubicin in time- and dose-dependent manners. ACOT7 knockdown induced cytostasis via activation of the p53-p21 signaling pathway without a DNA damage response. PKCζ was specifically involved in ACOT7 depletion-mediated cell cycle arrest as an upstream molecule of the p53-p21 signaling pathway in MCF7 human breast carcinoma and A549 human lung carcinoma cells. Of the other members of the ACOT family, including ACOT1, 4, 8, 9, 11, 12, and 13 that were expressed in human, ACOT4, 8, and 12 were responsive to genotoxic stresses. However, none of those had a role in cytostasis via activation of the PKCζ-p53-p21 signaling pathway. Analysis of the ACOT7 prognostic value revealed that low ACOT7 levels prolonged overall survival periods in breast and lung cancer patients. Furthermore, ACOT7 mRNA levels were higher in lung cancer patient tissues compared to normal tissues. We also observed a synergistic effect of ACOT7 depletion in combination with either IR or doxorubicin on cell proliferation in breast and lung cancer cells. Together, our data suggest that a low level of ACOT7 may be involved, at least in part, in the prevention of human breast and lung cancer development via regulation of cell cycle progression.
Collapse
|
31
|
Significance of Wild-Type p53 Signaling in Suppressing Apoptosis in Response to Chemical Genotoxic Agents: Impact on Chemotherapy Outcome. Int J Mol Sci 2017; 18:ijms18050928. [PMID: 28452953 PMCID: PMC5454841 DOI: 10.3390/ijms18050928] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/18/2017] [Accepted: 04/25/2017] [Indexed: 12/17/2022] Open
Abstract
Our genomes are subject to potentially deleterious alterations resulting from endogenous sources (e.g., cellular metabolism, routine errors in DNA replication and recombination), exogenous sources (e.g., radiation, chemical agents), and medical diagnostic and treatment applications. Genome integrity and cellular homeostasis are maintained through an intricate network of pathways that serve to recognize the DNA damage, activate cell cycle checkpoints and facilitate DNA repair, or eliminate highly injured cells from the proliferating population. The wild-type p53 tumor suppressor and its downstream effector p21WAF1 (p21) are key regulators of these responses. Although extensively studied for its ability to control cell cycle progression, p21 has emerged as a multifunctional protein capable of downregulating p53, suppressing apoptosis, and orchestrating prolonged growth arrest through stress-induced premature senescence. Studies with solid tumors and solid tumor-derived cell lines have revealed that such growth-arrested cancer cells remain viable, secrete growth-promoting factors, and can give rise to progeny with stem-cell-like properties. This article provides an overview of the mechanisms by which p53 signaling suppresses apoptosis following genotoxic stress, facilitating repair of genomic injury under physiological conditions but having the potential to promote tumor regrowth in response to cancer chemotherapy.
Collapse
|
32
|
Gopas J, Stern E, Zurgil U, Ozer J, Ben-Ari A, Shubinsky G, Braiman A, Sinay R, Ezratty J, Dronov V, Balachandran S, Benharroch D, Livneh E. Reed-Sternberg cells in Hodgkin's lymphoma present features of cellular senescence. Cell Death Dis 2016; 7:e2457. [PMID: 27831553 PMCID: PMC5287295 DOI: 10.1038/cddis.2016.185] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 12/20/2022]
Abstract
Hodgkin's Lymphoma (HL) is one of the most prevailing malignancies in young adults. Reed–Sternberg (RS) cells in HL have distinctive large cell morphology, are characteristic of the disease and their presence is essential for diagnosis. Enlarged cells are one of the hallmarks of senescence, but whether RS cells are senescent has not been previously investigated. Here we show that RS cells have characteristics of senescent cells; RS cells in HL biopsies specifically express the senescence markers and cell cycle inhibitors p21Cip1 and p16INK4a and are negative for the proliferation marker Ki-67, suggesting that these cells have ceased to proliferate. Moreover, the RS-like cells in HL lines, stained specifically for senescence-associated β-galactosidase (SA-β-gal). Oxidative stress promoted senescence in these cells as demonstrated by their staining for p21Cip1, p16INK4a, p53 and γH2AX. Senescent cells produce copious amounts of inflammatory cytokines termed ‘senescence-associated secretory phenotype' (SASP), primarily regulated by Nuclear Factor κB (NF-κB). Indeed, we show that NF-κB activity and NF-κB-dependent cytokines production (e.g., IL-6, TNF-α, GM-CSF) were elevated in RS-like cells. Furthermore, NF-κB inhibitors, JSH-23 and curcumin reduced IL-6 secretion from RS-like cells. Thus, defining RS cells as senescent offers new insights on the origin of the proinflammatory microenvironment in HL.
Collapse
Affiliation(s)
- J Gopas
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel.,Department of Oncology, Soroka University Medical Center, Beer Sheva 84105, Israel
| | - E Stern
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel
| | - U Zurgil
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel
| | - J Ozer
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel
| | - A Ben-Ari
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel
| | - G Shubinsky
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel.,Flow Cytometry Unit, Hematology Laboratory and Institute of Hematology, Beer Sheva 84105, Israel
| | - A Braiman
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel
| | - R Sinay
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel
| | - J Ezratty
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel
| | - V Dronov
- Department of Pathology, Soroka University Medical Center, Beer Sheva 84105, Israel
| | - S Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - D Benharroch
- Department of Pathology, Soroka University Medical Center, Beer Sheva 84105, Israel
| | - E Livneh
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of The Negev, Beer Sheva 84105, Israel
| |
Collapse
|
33
|
Yamaguchi Y, Madhyastha H, Madhyastha R, Choijookhuu N, Hishikawa Y, Pengjam Y, Nakajima Y, Maruyama M. Arsenic acid inhibits proliferation of skin fibroblasts, and increases cellular senescence through ROS mediated MST1-FOXO signaling pathway. J Toxicol Sci 2016; 41:105-13. [PMID: 26763397 DOI: 10.2131/jts.41.105] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Arsenic exposure through drinking water is a major public health problem. It causes a number of toxic effects on skin. Arsenic has been reported to inhibit cell proliferation in in vitro conditions. However, reports about the molecular mechanisms are limited. Here, we investigated the mechanism involved in arsenic acid-mediated inhibition of cell proliferation using mouse skin fibroblast cell line. The present study found that 10 ppm arsenic acid inhibited cell proliferation, without any effect on cell death. Arsenic acid induced the generation of reactive oxygen species (ROS), resulting in oxidative stress to DNA. It also activated the mammalian Ste20-like protein kinase 1 (MST1); however the serine/threonine kinase Akt was downregulated. Forkhead box O (FOXO) transcription factors are activated through phosphorylation by MST1 under stress conditions. They are inhibited by phosphorylation by Akt through external and internal stimuli. Activation of FOXOs results in their nuclear localization, followed by an increase in transcriptional activity. Our results showed that arsenic induced the nuclear translocation of FOXO1 and FOXO3a, and altered the cell cycle, with cells accumulating at the G2/M phase. These effects caused cellular senescence. Taken together, our results indicate that arsenic acid inhibited cell proliferation through cellular senescence process regulated by MST1-FOXO signaling pathway.
Collapse
Affiliation(s)
- Yuya Yamaguchi
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Gewirtz DA, Alotaibi M, Yakovlev VA, Povirk LF. Tumor Cell Recovery from Senescence Induced by Radiation with PARP Inhibition. Radiat Res 2016; 186:327-332. [PMID: 27588595 DOI: 10.1667/rr14437.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inhibitors of poly(ADP-ribose) polymerase (PARP) are clinically used as single-agent therapy for tumors with BRCA1 or BRCA2 mutations. One approach to expanding the use of PARP inhibitors to a wider range of tumors is to combine them with cytotoxic chemotherapy or radiotherapy. Preclinical studies in experimental animals and tumor cells in culture indicate that PARP inhibition modestly sensitizes most tumor cells to ionizing radiation. Studies of cell behavior after these combined treatments show that radiosensitization is manifested predominantly in an increase in prolonged growth arrest and senescence, with little or no contribution from apoptosis. The secretory phenotype associated with senescence can target these tumor cells for immune surveillance, and therefore increased senescence can effectively contribute to tumor control. However, the possible recovery of senescent cells and re-entry into cell cycle after prolonged arrest also needs to be considered. Such recovery could lead to tumor recurrence, yet may not be reflected in short-term assays commonly used to assess radiosensitization.
Collapse
Affiliation(s)
| | - Moureq Alotaibi
- Department of a Pharmacology, Toxicology and Medicine and.,c College of Pharmacy, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Vasily A Yakovlev
- b Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298; and
| | | |
Collapse
|
35
|
Wang Y, Boerma M, Zhou D. Ionizing Radiation-Induced Endothelial Cell Senescence and Cardiovascular Diseases. Radiat Res 2016; 186:153-61. [PMID: 27387862 DOI: 10.1667/rr14445.1] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Exposure to ionizing radiation induces not only apoptosis but also senescence. While the role of endothelial cell apoptosis in mediating radiation-induced acute tissue injury has been extensively studied, little is known about the role of endothelial cell senescence in the pathogenesis of radiation-induced late effects. Senescent endothelial cells exhibit decreased production of nitric oxide and expression of thrombomodulin, increased expression of adhesion molecules, elevated production of reactive oxygen species and inflammatory cytokines and an inability to proliferate and form capillary-like structures in vitro. These findings suggest that endothelial cell senescence can lead to endothelial dysfunction by dysregulation of vasodilation and hemostasis, induction of oxidative stress and inflammation and inhibition of angiogenesis, which can potentially contribute to radiation-induced late effects such as cardiovascular diseases (CVDs). In this article, we discuss the mechanisms by which radiation induces endothelial cell senescence, the roles of endothelial cell senescence in radiation-induced CVDs and potential strategies to prevent, mitigate and treat radiation-induced CVDs by targeting senescent endothelial cells.
Collapse
Affiliation(s)
- Yingying Wang
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas Medical Sciences, Little Rock, Arkansas 72205
| | - Marjan Boerma
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas Medical Sciences, Little Rock, Arkansas 72205
| | - Daohong Zhou
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas Medical Sciences, Little Rock, Arkansas 72205
| |
Collapse
|
36
|
Stress-Induced Premature Senescence of Endothelial and Endothelial Progenitor Cells. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 77:281-306. [PMID: 27451101 DOI: 10.1016/bs.apha.2016.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This brief overview of premature senescence of dysfunctional endothelial and endothelial progenitor cells provides information on endothelial cell differentiation and specialization, their ontogeny, and controversies related to endothelial stem and progenitor cells. Stressors responsible for the dysfunction of endothelial and endothelial progenitor cells, as well as cellular mechanisms and consequences of endothelial cell dysfunction are presented. Metabolic signatures of dysfunctional endothelial cells and senescence pathways are described. Emerging strategies to rejuvenate endothelial and endothelial progenitor cells conclude the review.
Collapse
|
37
|
Lee J, Kim MR, Kim HJ, An YS, Yi JY. TGF-β1 accelerates the DNA damage response in epithelial cells via Smad signaling. Biochem Biophys Res Commun 2016; 476:420-425. [PMID: 27237972 DOI: 10.1016/j.bbrc.2016.05.136] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 11/30/2022]
Abstract
The evidence suggests that transforming growth factor-beta (TGF-β) regulates the DNA-damage response (DDR) upon irradiation, and we previously reported that TGF-β1 induced DNA ligase IV (Lig4) expression and enhanced the nonhomologous end-joining repair pathway in irradiated cells. In the present study, we investigated the effects of TGF-β1 on the irradiation-induced DDRs of A431 and HaCaT cells. Cells were pretreated with or without TGF-β1 and irradiated. At 30 min post-irradiation, DDRs were detected by immunoblotting of phospho-ATM, phospho-Chk2, and the presence of histone foci (γH2AX). The levels of all three factors were similar right after irradiation regardless of TGF-β1 pretreatment. However, they soon thereafter exhibited downregulation in TGF-β1-pretreated cells, indicating the acceleration of the DDR. Treatment with a TGF-β type I receptor inhibitor (SB431542) or transfections with siRNAs against Smad2/3 or DNA ligase IV (Lig4) reversed this acceleration of the DDR. Furthermore, the frequency of irradiation-induced apoptosis was decreased by TGF-β1 pretreatment in vivo, but this effect was abrogated by SB431542. These results collectively suggest that TGF-β1 could enhance cell survival by accelerating the DDR via Smad signaling and Lig4 expression.
Collapse
Affiliation(s)
- Jeeyong Lee
- Division of Basic Radiation Bioscience, Korea Institute of Radiation and Medical Sciences, Seoul, 01812, Republic of Korea
| | - Mi-Ra Kim
- Division of Basic Radiation Bioscience, Korea Institute of Radiation and Medical Sciences, Seoul, 01812, Republic of Korea
| | - Hyun-Ji Kim
- Division of Basic Radiation Bioscience, Korea Institute of Radiation and Medical Sciences, Seoul, 01812, Republic of Korea
| | - You Sun An
- Division of Basic Radiation Bioscience, Korea Institute of Radiation and Medical Sciences, Seoul, 01812, Republic of Korea
| | - Jae Youn Yi
- Division of Basic Radiation Bioscience, Korea Institute of Radiation and Medical Sciences, Seoul, 01812, Republic of Korea.
| |
Collapse
|
38
|
Therapeutic Implications for Overcoming Radiation Resistance in Cancer Therapy. Int J Mol Sci 2015; 16:26880-913. [PMID: 26569225 PMCID: PMC4661850 DOI: 10.3390/ijms161125991] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/29/2015] [Accepted: 10/29/2015] [Indexed: 12/17/2022] Open
Abstract
Ionizing radiation (IR), such as X-rays and gamma (γ)-rays, mediates various forms of cancer cell death such as apoptosis, necrosis, autophagy, mitotic catastrophe, and senescence. Among them, apoptosis and mitotic catastrophe are the main mechanisms of IR action. DNA damage and genomic instability contribute to IR-induced cancer cell death. Although IR therapy may be curative in a number of cancer types, the resistance of cancer cells to radiation remains a major therapeutic problem. In this review, we describe the morphological and molecular aspects of various IR-induced types of cell death. We also discuss cytogenetic variations representative of IR-induced DNA damage and genomic instability. Most importantly, we focus on several pathways and their associated marker proteins responsible for cancer resistance and its therapeutic implications in terms of cancer cell death of various types and characteristics. Finally, we propose radiation-sensitization strategies, such as the modification of fractionation, inflammation, and hypoxia and the combined treatment, that can counteract the resistance of tumors to IR.
Collapse
|
39
|
Babizhayev MA, Yegorov YE. Tissue formation and tissue engineering through host cell recruitment or a potential injectable cell-based biocomposite with replicative potential: Molecular mechanisms controlling cellular senescence and the involvement of controlled transient telomerase activation therapies. J Biomed Mater Res A 2015; 103:3993-4023. [PMID: 26034007 DOI: 10.1002/jbm.a.35515] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 05/18/2015] [Indexed: 01/04/2023]
Abstract
Accumulated data indicate that wound-care products should have a composition equivalent to that of the skin: a combination of particular growth factors and extracellular matrix (ECM) proteins endogenous to the skin, together with viable epithelial cells, fibroblasts, and mesenchymal stem cells (MSCs). Strategies consisting of bioengineered dressings and cell-based products have emerged for widespread clinical use; however, their performance is not optimal because chronic wounds persist as a serious unmet medical need. Telomerase, the ribonucleoprotein complex that adds telomeric repeats to the ends of chromosomes, is responsible for telomere maintenance, and its expression is associated with cell immortalization and, in certain cases, cancerogenesis. Telomerase contains a catalytic subunit, the telomerase reverse transcriptase (hTERT). Introduction of TERT into human cells extends both their lifespan and their telomeres to lengths typical of young cells. The regulation of TERT involves transcriptional and posttranscriptional molecular biology mechanisms. The manipulation, regulation of telomerase is multifactorial in mammalian cells, involving overall telomerase gene expression, post-translational protein-protein interactions, and protein phosphorylation. Reactive oxygen species (ROS) have been implicated in aging, apoptosis, and necrosis of cells in numerous diseases. Upon production of high levels of ROS from exogenous or endogenous generators, the redox balance is perturbed and cells are shifted into a state of oxidative stress, which subsequently leads to modifications of intracellular proteins and membrane lipid peroxidation and to direct DNA damage. When the oxidative stress is severe, survival of the cell is dependent on the repair or replacement of damaged molecules, which can result in induction of apoptosis in the injured with ROS cells. ROS-mediated oxidative stress induces the depletion of hTERT from the nucleus via export through the nuclear pores. Nuclear export is initiated by ROS-induced phosphorylation of tyrosine 707 within hTERT by the Src kinase family. It might be presumed that protection of mitochondria against oxidative stress is an important telomere length-independent function for telomerase in cell survival. Biotechnology companies are focused on development of therapeutic telomerase vaccines, telomerase inhibitors, and telomerase promoter-driven cell killing in oncology, have a telomerase antagonist in late preclinical studies. Anti-aging medicine-oriented groups have intervened on the market with products working on telomerase activation for a broad range of degenerative diseases in which replicative senescence or telomere dysfunction may play an important role. Since oxidative damage has been shown to shorten telomeres in tissue culture models, the adequate topical, transdermal, or systemic administration of antioxidants (such as, patented ocular administration of 1% N-acetylcarnosine lubricant eye drops in the treatment of cataracts) may be beneficial at preserving telomere lengths and delaying the onset or in treatment of disease in susceptible individuals. Therapeutic strategies toward controlled transient activation of telomerase are targeted to cells and replicative potential in cell-based therapies, tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Mark A Babizhayev
- Innovative Vision Products, Inc., 3511 Silverside Road, Suite 105, County of New Castle, Delaware, 19810
| | - Yegor E Yegorov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov Street, Moscow, 119991, Russian Federation
| |
Collapse
|
40
|
Liu S, Wang X, Zhao Q, Liu S, Zhang H, Shi J, Li N, Lei X, Zhao H, Deng Z, Cao Y, Ning L, Xia G, Duan E. Senescence of human skin-derived precursors regulated by Akt-FOXO3-p27(KIP¹)/p15(INK⁴b) signaling. Cell Mol Life Sci 2015; 72:2949-60. [PMID: 25753771 PMCID: PMC11113525 DOI: 10.1007/s00018-015-1877-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 12/17/2022]
Abstract
Multipotent skin-derived precursors (SKPs) are dermal stem cells with the capacity to reconstitute the dermis and other tissues, such as muscles and the nervous system. Thus, the easily available human SKPs (hSKPs) hold great promises in regenerative medicine. However, long-term expansion is difficult for hSKPs in vitro. We previously demonstrated that hSKPs senesced quickly under routine culture conditions. To identify the underlying mechanisms so as to find an effective way to expand hSKPs, time-dependent microarray analysis of gene expression in hSKPs during in vitro culture was performed. We found that the senescence of hSKPs had a unique gene expression pattern that differs from reported typical senescence. Subsequent investigation ruled out the role of DNA damage and classical p53 and p16(INK4a) signaling in hSKP senescence. Examination of cyclin-dependent kinase inhibitors revealed the involvement of p15(INK4b) and p27(KIP1). Further exploration about upstream signals indicated the contribution of Akt hypo-activity and FOXO3 to hSKP senescence. Forced activation of Akt and knockdown of FOXO3, p15(INK4b) and p27(KIP1) effectively inhibited hSKP senescence and promoted hSKP proliferation. The unique senescent phenotype of human dermal stem cells and the role of Akt-FOXO3-p27(KIP1)/p15(INK4b) signaling in regulating hSKP senescence provide novel insights into the senescence and self-renewal regulation of adult stem cells. The present study also points out a way to propagate hSKPs in vitro so as to fulfill their promises in regenerative medicine.
Collapse
Affiliation(s)
- Shuang Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
| | - Xinyue Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
| | - Qian Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, No. 2 Yuanmingyuan Xilu, Haidian District, Beijing, 100193 China
| | - Shu Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
| | - Huishan Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
| | - Junchao Shi
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Na Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Xiaohua Lei
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
| | - Huashan Zhao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zhili Deng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yujing Cao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
| | - Lina Ning
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, No. 2 Yuanmingyuan Xilu, Haidian District, Beijing, 100193 China
| | - Enkui Duan
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101 China
| |
Collapse
|
41
|
Jung YR, Kim EJ, Choi HJ, Park JJ, Kim HS, Lee YJ, Park MJ, Lee M. Aspirin Targets SIRT1 and AMPK to Induce Senescence of Colorectal Carcinoma Cells. Mol Pharmacol 2015. [PMID: 26219912 DOI: 10.1124/mol.115.098616] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer therapies attempt to destroy the entire tumor, but this tends to require toxic compounds and high doses of radiation. Recently, considerable attention has focused on therapy-induced senescence (TIS), which can be induced in cancer cells by low doses of therapeutic drugs or radiation and provides a barrier to tumor development. However, the molecular mechanisms governing TIS remain elusive. Special attention has been paid to the potential chemopreventive effect of aspirin against human colorectal cancer. In this study, we investigated the effects of aspirin on TIS of human colorectal carcinoma (CRC) cells and show that it occurs via sirtuin 1 (SIRT1) and AMP-activated protein kinase (AMPK), two key regulators of cellular metabolism. Aspirin increased the senescence of CRC cells, increased the protein levels of SIRT1, phospho-AMPK (T172), and phospho-acetyl CoA carboxylase (S79), and reduced the cellular level of ATP. Small-interfering RNA-mediated downregulation or pharmacological inhibition of SIRT1 or AMPK significantly attenuated the aspirin-induced cellular senescence in CRC cells. In contrast, treatment with a SIRT1 agonist or an AMP analog induced cellular senescence. Remarkably, SIRT1 knockdown abrogated the aspirin-induced activation of AMPK, and vice versa. During the progression of aspirin-induced cellular senescence in CRC cells, SIRT1 showed increased deacetylase activity at a relatively early time point but was characterized by decreased activity with increased cytoplasmic localization at a later time point. Collectively, these novel findings suggest that aspirin could provide anticancer effects by inducing senescence in human CRC cells through the reciprocal regulation of SIRT1-AMPK pathways.
Collapse
Affiliation(s)
- Yu Ri Jung
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Eun Ju Kim
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hyeong Jwa Choi
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Jung-Jin Park
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hak-Su Kim
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Yoon-Jin Lee
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Myung-Jin Park
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Minyoung Lee
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| |
Collapse
|
42
|
Widel M, Lalik A, Krzywon A, Poleszczuk J, Fujarewicz K, Rzeszowska-Wolny J. The different radiation response and radiation-induced bystander effects in colorectal carcinoma cells differing in p53 status. Mutat Res 2015; 778:61-70. [PMID: 26099456 DOI: 10.1016/j.mrfmmm.2015.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/15/2015] [Accepted: 06/07/2015] [Indexed: 01/18/2023]
Abstract
Radiation-induced bystander effect, appearing as different biological changes in cells that are not directly exposed to ionizing radiation but are under the influence of molecular signals secreted by irradiated neighbors, have recently attracted considerable interest due to their possible implication for radiotherapy. However, various cells present diverse radiosensitivity and bystander responses that depend, inter alia, on genetic status including TP53, the gene controlling the cell cycle, DNA repair and apoptosis. Here we compared the ionizing radiation and bystander responses of human colorectal carcinoma HCT116 cells with wild type or knockout TP53 using a transwell co-culture system. The viability of exposed to X-rays (0-8 Gy) and bystander cells of both lines showed a roughly comparable decline with increasing dose. The frequency of micronuclei was also comparable at lower doses but at higher increased considerably, especially in bystander TP53-/- cells. Moreover, the TP53-/- cells showed a significantly elevated frequency of apoptosis, while TP53+/+ counterparts expressed high level of senescence. The cross-matched experiments where irradiated cells of one line were co-cultured with non-irradiated cells of opposite line show that both cell lines were also able to induce bystander effects in their counterparts, however different endpoints revealed with different strength. Potential mediators of bystander effects, IL-6 and IL-8, were also generated differently in both lines. The knockout cells secreted IL-6 at lower doses whereas wild type cells only at higher doses. Secretion of IL-8 by TP53-/- control cells was many times lower than that by TP53+/+ but increased significantly after irradiation. Transcription of the NFκBIA was induced in irradiated TP53+/+ mainly, but in bystanders a higher level was observed in TP53-/- cells, suggesting that TP53 is required for induction of NFκB pathway after irradiation but another mechanism of activation must operate in bystander cells.
Collapse
Affiliation(s)
- Maria Widel
- Biosystems Group, Institute of Automatic Control, Silesian University of Technology, 16 Akademicka Street, 44-100 Gliwice, Poland.
| | - Anna Lalik
- Biosystems Group, Institute of Automatic Control, Silesian University of Technology, 16 Akademicka Street, 44-100 Gliwice, Poland
| | - Aleksandra Krzywon
- Biosystems Group, Institute of Automatic Control, Silesian University of Technology, 16 Akademicka Street, 44-100 Gliwice, Poland
| | - Jan Poleszczuk
- College of Inter-faculty Individual Studies in Mathematics and Natural Sciences, University of Warsaw, 93 Zwirki i Wigury Street, 02-089 Warsaw, Poland; Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Krzysztof Fujarewicz
- Biosystems Group, Institute of Automatic Control, Silesian University of Technology, 16 Akademicka Street, 44-100 Gliwice, Poland
| | - Joanna Rzeszowska-Wolny
- Biosystems Group, Institute of Automatic Control, Silesian University of Technology, 16 Akademicka Street, 44-100 Gliwice, Poland
| |
Collapse
|
43
|
Liu S, Wang X, Zhao Q, Liu S, Zhang H, Shi J, Li N, Lei X, Zhao H, Deng Z, Cao Y, Ning L, Xia G, Duan E. Senescence of human skin-derived precursors regulated by Akt-FOXO3-p27KIP1/p15INK4b signaling. Cell Mol Life Sci 2015. [DOI: doi 10.1007/s00018-015-1877-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
44
|
Kim EJ, Lee J, Jung YR, Park JJ, Park MJ, Lee JS, Kim CH, Lee YJ, Lee M. Involvement of corin downregulation in ionizing radiation-induced senescence of myocardial cells. Int J Mol Med 2014; 35:731-8. [PMID: 25543718 DOI: 10.3892/ijmm.2014.2048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 12/16/2014] [Indexed: 11/06/2022] Open
Abstract
Radiation-induced heart disease (RIHD) is becoming an increasing concern for patients and clinicians alike due to the use of radiotherapy for the treatment of breast cancer, Hodgkin's lymphoma, pediatric cancer and tumors of the thorax. However, the mechanisms underlying this phenomenon remain largely unknown. As the senescent cell fraction following irradiation is known to increase, in the present study, we investigated whether ionizing radiation (IR) causes the onset of heart disease by inducing cellular senescence in cardiomyocytes. In the present study, we evaluated the effects of IR on HL-1 and H9C2 cells, cells predominantly used in in vitro myocardial cell models. We found that the exposure of the HL-1 and H9C2 cells to IR induced reactive oxygen species (ROS)-mediated cellular senescence, as shown by staining of senescence-associated β-galactosidase (SA-β-gal). The levels of ROS in irradiated cells were determined using the fluorescent dye, 2', 7'-dichlorodihydrofluorescein diacetate (DCF-DA). Notably, the expression of corin, a cardiac protease that is essential for the proteolytic cleavage of natriuretic peptides, was significantly decreased following the exposure of the cells to IR. Importantly, the knockdown of corin by RNA interference enhanced IR-induced senescence. On the contrary, the overexpression of natriuretic peptides reversed the IR-induced senescence. Taken together, our data suggest that defects in corin function and the inhibition of natriuretic peptides following exposure to IR may contribute to the development of RIHD through the acceleration of cellular senescence.
Collapse
Affiliation(s)
- Eun Ju Kim
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Jeok Lee
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Yu Ri Jung
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Jung-Jin Park
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Myung-Jin Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Jae-Seon Lee
- Division of Biomedical Sciences, College of Medicine, Inha University, Incheon 400-712, Republic of Korea
| | - Chun-Ho Kim
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Yoon-Jin Lee
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Minyoung Lee
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| |
Collapse
|
45
|
Zurgil U, Ben-Ari A, Atias K, Isakov N, Apte R, Livneh E. PKCη promotes senescence induced by oxidative stress and chemotherapy. Cell Death Dis 2014; 5:e1531. [PMID: 25412309 PMCID: PMC4260739 DOI: 10.1038/cddis.2014.481] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 10/06/2014] [Accepted: 10/09/2014] [Indexed: 12/21/2022]
Abstract
Senescence is characterized by permanent cell-cycle arrest despite continued viability and metabolic activity, in conjunction with the secretion of a complex mixture of extracellular proteins and soluble factors known as the senescence-associated secretory phenotype (SASP). Cellular senescence has been shown to prevent the proliferation of potentially tumorigenic cells, and is thus generally considered a tumor suppressive process. However, some SASP components may act as pro-tumorigenic mediators on premalignant cells in the microenvironment. A limited number of studies indicated that protein kinase C (PKC) has a role in senescence, with different isoforms having opposing effects. It is therefore important to elucidate the functional role of specific PKCs in senescence. Here we show that PKCη, an epithelial specific and anti-apoptotic kinase, promotes senescence induced by oxidative stress and DNA damage. We further demonstrate that PKCη promotes senescence through its ability to upregulate the expression of the cell cycle inhibitors p21Cip1 and p27Kip1 and enhance transcription and secretion of interleukin-6 (IL-6). Moreover, we demonstrate that PKCη creates a positive loop for reinforcing senescence by increasing the transcription of both IL-6 and IL-6 receptor, whereas the expression of IL-8 is specifically suppressed by PKCη. Thus, the presence/absence of PKCη modulates major components of SASP. Furthermore, we show that the human polymorphic variant of PKCη, 374I, that exhibits higher kinase activity in comparison to WT-374V, is also more effective in IL-6 secretion, p21Cip1 expression and the promotion of senescence, further supporting a role for PKCη in senescence. As there is now considerable interest in senescence activation/elimination to control tumor progression, it is first crucial to reveal the molecular regulators of senescence. This will improve our ability to develop new strategies to harness senescence as a potential cancer therapy in the future.
Collapse
Affiliation(s)
- U Zurgil
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - A Ben-Ari
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - K Atias
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - N Isakov
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - R Apte
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - E Livneh
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
46
|
Reste J, Zvigule G, Zvagule T, Kurjane N, Eglite M, Gabruseva N, Berzina D, Plonis J, Miklasevics E. Telomere length in Chernobyl accident recovery workers in the late period after the disaster. JOURNAL OF RADIATION RESEARCH 2014; 55:1089-100. [PMID: 25015931 PMCID: PMC4229925 DOI: 10.1093/jrr/rru060] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 06/08/2014] [Accepted: 06/09/2014] [Indexed: 05/02/2023]
Abstract
The outcome of the Chernobyl nuclear power plant (CNPP) accident was that a huge number of people were exposed to ionizing radiation. Previous studies of CNPP clean-up workers from Latvia revealed a high occurrence of age-associated degenerative diseases and cancer in young adults, as well as a high mortality as a result of cardiovascular disorders at age 45-54 years. DNA tandem repeats that cap chromosome ends, known as telomeres, are sensitive to oxidative damage and exposure to ionizing radiation. Telomeres are important in aging processes and carcinogenesis. The aim of this study was to investigate the long-term effect of protracted ionizing radiation exposure on telomere length in CNPP clean-up workers. Relative telomere length (RTL) was measured in peripheral blood leukocytes of 595 CNPP clean-up workers and 236 gender- and age-matched controls using real-time quantitative polymerase chain reaction (q-PCR). Close attention was paid to participation year and tasks performed during the worker's stay in Chernobyl, health status, and RTL differences between subgroups. Telomere shortening was not found in CNPP clean-up workers; on the contrary, their RTL was slightly greater than in controls (P = 0.001). Longer telomeres were found in people who worked during 1986, in those undertaking 'dirty' tasks (digging and deactivation), and in people with cancer. Shorter telomeres appeared frequently in those with cataract, osteoporosis, atherosclerosis, or coronary heart disease. We conclude that the longer telomeres revealed in people more heavily exposed to ionizing radiation probably indicate activation of telomerase as a chromosome healing mechanism following damage, and reflect defects in telomerase regulation that could potentiate carcinogenesis.
Collapse
Affiliation(s)
- Jelena Reste
- Institute of Occupational Safety and Environmental Health, Riga Stradins University, Dzirciema Street 16, Riga, LV-1007, Latvia Centre of Occupational and Radiological Medicine, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, Riga, LV-1002, Latvia
| | - Gunda Zvigule
- Institute of Oncology, Riga Stradins University, Dzirciema Street 16, Riga, LV-1007, Latvia
| | - Tija Zvagule
- Institute of Occupational Safety and Environmental Health, Riga Stradins University, Dzirciema Street 16, Riga, LV-1007, Latvia Centre of Occupational and Radiological Medicine, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, Riga, LV-1002, Latvia
| | - Natalja Kurjane
- Institute of Occupational Safety and Environmental Health, Riga Stradins University, Dzirciema Street 16, Riga, LV-1007, Latvia Centre of Occupational and Radiological Medicine, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, Riga, LV-1002, Latvia
| | - Maija Eglite
- Institute of Occupational Safety and Environmental Health, Riga Stradins University, Dzirciema Street 16, Riga, LV-1007, Latvia Centre of Occupational and Radiological Medicine, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, Riga, LV-1002, Latvia
| | - Natalija Gabruseva
- Centre of Occupational and Radiological Medicine, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, Riga, LV-1002, Latvia
| | - Dace Berzina
- Institute of Oncology, Riga Stradins University, Dzirciema Street 16, Riga, LV-1007, Latvia
| | - Juris Plonis
- Institute of Oncology, Riga Stradins University, Dzirciema Street 16, Riga, LV-1007, Latvia
| | - Edvins Miklasevics
- Institute of Oncology, Riga Stradins University, Dzirciema Street 16, Riga, LV-1007, Latvia
| |
Collapse
|
47
|
Abstract
Cellular senescence is a physiological process of irreversible cell-cycle arrest that contributes to various physiological and pathological processes of aging. Whereas replicative senescence is associated with telomere attrition after repeated cell division, stress-induced premature senescence occurs in response to aberrant oncogenic signaling, oxidative stress, and DNA damage which is independent of telomere dysfunction. Recent evidence indicates that cellular senescence provides a barrier to tumorigenesis and is a determinant of the outcome of cancer treatment. However, the senescence-associated secretory phenotype, which contributes to multiple facets of senescent cancer cells, may influence both cancer-inhibitory and cancer-promoting mechanisms of neighboring cells. Conventional treatments, such as chemo- and radiotherapies, preferentially induce premature senescence instead of apoptosis in the appropriate cellular context. In addition, treatment-induced premature senescence could compensate for resistance to apoptosis via alternative signaling pathways. Therefore, we believe that an intensive effort to understand cancer cell senescence could facilitate the development of novel therapeutic strategies for improving the efficacy of anticancer therapies. This review summarizes the current understanding of molecular mechanisms, functions, and clinical applications of cellular senescence for anticancer therapy.
Collapse
Affiliation(s)
- Minyoung Lee
- Research Center for Radio-senescence, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Korea
| | - Jae-Seon Lee
- Research Center for Radio-senescence, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Korea; Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 400-712, Korea
| |
Collapse
|
48
|
Murray D, McBride WH, Schwartz JL. Radiation biology in the context of changing patterns of radiotherapy. Radiat Res 2014; 182:259-72. [PMID: 25029108 DOI: 10.1667/rr13740.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The last decade has witnessed a revolution in the clinical application of high-dose "ablative" radiation therapy. Initially this approach was limited to the treatment of brain tumors, but more recently we have seen its successful extension to tumors outside the brain, e.g., for small lung nodules. These advances have been driven largely by improvements in image-guided inverse treatment planning that allow the dose per fraction to the tumor to be increased over the conventional 2 Gy dose while keeping the late normal tissue complications at an acceptable level by dose limitation. Despite initial concerns about excessive late complications, as might be expected based on dose extrapolations using the linear-quadratic equation, these approaches have shown considerable clinical promise. Our knowledge of the biological consequences of high-doses of ionizing radiation in normal and cancerous tissues has lagged behind these clinical advances. Our intent here is to survey recent experimental findings from the perspective of better understanding the biological effects of high-dose therapy and whether they are truly different from conventional doses. We will also consider the implications of this knowledge for further refining and improving these approaches on the basis of underlying mechanisms.
Collapse
Affiliation(s)
- David Murray
- a Department of Oncology, Division of Experimental Oncology, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
49
|
Loseva O, Shubbar E, Haghdoost S, Evers B, Helleday T, Harms-Ringdahl M. Chronic Low Dose Rate Ionizing Radiation Exposure Induces Premature Senescence in Human Fibroblasts that Correlates with Up Regulation of Proteins Involved in Protection against Oxidative Stress. Proteomes 2014; 2:341-362. [PMID: 28250385 PMCID: PMC5302754 DOI: 10.3390/proteomes2030341] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 06/16/2014] [Accepted: 06/25/2014] [Indexed: 12/16/2022] Open
Abstract
The risks of non-cancerous diseases associated with exposure to low doses of radiation are at present not validated by epidemiological data, and pose a great challenge to the scientific community of radiation protection research. Here, we show that premature senescence is induced in human fibroblasts when exposed to chronic low dose rate (LDR) exposure (5 or 15 mGy/h) of gamma rays from a 137Cs source. Using a proteomic approach we determined differentially expressed proteins in cells after chronic LDR radiation compared to control cells. We identified numerous proteins involved in protection against oxidative stress, suggesting that these pathways protect against premature senescence. In order to further study the role of oxidative stress for radiation induced premature senescence, we also used human fibroblasts, isolated from a patient with a congenital deficiency in glutathione synthetase (GS). We found that these GS deficient cells entered premature senescence after a significantly shorter time of chronic LDR exposure as compared to the GS proficient cells. In conclusion, we show that chronic LDR exposure induces premature senescence in human fibroblasts, and propose that a stress induced increase in reactive oxygen species (ROS) is mechanistically involved.
Collapse
Affiliation(s)
- Olga Loseva
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm S-171 21, Sweden.
| | - Emman Shubbar
- Sahlgrenska Cancer Center, Department of Clinical Genetics, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg SE-41345, Sweden.
| | - Siamak Haghdoost
- Center for Radiation Protections Research, Department of Molecular Biosciences, The Wenner Gren Institute, Stockholm University, Stockholm S-106 91, Sweden.
| | - Bastiaan Evers
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands.
| | - Thomas Helleday
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm S-171 21, Sweden.
| | - Mats Harms-Ringdahl
- Center for Radiation Protections Research, Department of Molecular Biosciences, The Wenner Gren Institute, Stockholm University, Stockholm S-106 91, Sweden.
| |
Collapse
|
50
|
Serra MP, Marongiu F, Sini M, Marongiu M, Contini A, Wolff H, Rave-Frank M, Krause P, Laconi E, Koenig S. Hepatocyte senescence induced by radiation and partial hepatectomy in rat liver. Int J Radiat Biol 2014; 90:876-83. [PMID: 24827852 DOI: 10.3109/09553002.2014.922714] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Exposure to radiation primes the liver for extensive replacement of the resident parenchymal cells by transplanted hepatocytes. The mechanisms underlying this repopulation remain to be clarified. In these studies, we examined the possible occurrence of cell senescence in vivo following radiation-associated preconditioning of the host liver. MATERIALS AND METHODS Fischer 344 rats underwent external-beam, computed-tomography-based partial liver irradiation. A single dose of 25 Gy was delivered to the right liver lobes (40% of liver mass). An additional group of animals received a 1/3 partial hepatectomy (removal of the left anterior lobe) four days after irradiation. Non-irradiated groups served as controls. All rats were sacrificed four weeks after the initial treatment. RESULTS The irradiated livers displayed several markers of cell senescence, including expression of senescence-associated-β-galactosidase (SA-β-gal), increase in cell size, and up-regulation of cyclin-dependent kinase inhibitors (CDK-I) p16 and p21. Furthermore, quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) analysis revealed activation of the senescence-associated secretory phenotype (SASP), including the cytokines interleukin 6 (IL6) and 1α (IL1α). The senescence-related changes were more prominent in rats undergoing partial hepatectomy (PH) following irradiation (IR). CONCLUSIONS We conclude that priming with radiation for liver repopulation results in the induction of cell senescence and the up-regulation of a senescence-associated secretory phenotype. The latter can contribute to the extensive growth of transplanted cells in this system.
Collapse
|