1
|
Hu P, Hao Y, Tang W, Diering GH, Zou F, Kafri T. Analysis of hepatic lentiviral vector transduction; implications for preclinical studies and clinical gene therapy protocols. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608805. [PMID: 39229157 PMCID: PMC11370356 DOI: 10.1101/2024.08.20.608805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Lentiviral vector-transduced T-cells were approved by the FDA as gene therapy anti-cancer medications. Little is known about the host genetic variation effects on the safety and efficacy of the lentiviral vector gene delivery system. To narrow this knowledge-gap, we characterized hepatic gene delivery by lentiviral vectors across the Collaborative Cross (CC) mouse genetic reference population. For 24 weeks, we periodically measured hepatic luciferase expression from lentiviral vectors in 41 CC mouse strains. Hepatic and splenic vector copy numbers were determined. We report that CC mouse strains showed highly diverse outcomes following lentiviral gene delivery. For the first time, moderate correlation between mouse strain-specific sleeping patterns and transduction efficiency was observed. We associated two quantitative trait loci (QTLs) with intra-strain variations in transduction phenotypes, which mechanistically relates to the phenomenon of metastable epialleles. An additional QTL was associated with the kinetics of hepatic transgene expression. Genes comprised in the above QTLs are potential targets to personalize gene therapy protocols. Importantly, we identified two mouse strains that open new directions in characterizing continuous viral vector silencing and HIV latency. Our findings suggest that wide-range patient-specific outcomes of viral vector-based gene therapy should be expected. Thus, novel escalating dose-based clinical protocols should be considered.
Collapse
Affiliation(s)
- Peirong Hu
- Gene Therapy Center, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- These authors contributed equally
| | - Yajing Hao
- Department of Biostatistics, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- These authors contributed equally
| | - Wei Tang
- Gene Therapy Center, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
| | - Graham H. Diering
- Department of Cell Biology and Physiology and UNC Neuroscience Center, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- Carolina Institute for developmental disabilities, 27510 Carrboro, North Carolina
| | - Fei Zou
- Department of Biostatistics, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
| | - Tal Kafri
- Gene Therapy Center, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, 27599 Chapel Hill, North Carolina
| |
Collapse
|
2
|
Costa DL. Grandchildren's Longevity and Their Grandfathers' POW Trauma in the U.S. Civil War. Demography 2024; 61:337-361. [PMID: 38393987 DOI: 10.1215/00703370-11191183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
I document the transmission of a grandfather's net nutritional deprivation and psychosocial stress in young adulthood across multiple generations using the grandfather's ex-prisoner of war (ex-POW) status in the U.S. Civil War (1861-1865). Using a newly created dataset, I uncover an association between a grandfather's ex-POW status and the longevity after age 45 of his sons and male-line grandsons but not of his daughters, granddaughters, female-line grandsons, children-in-law, or grandchildren-in-law. Male-line grandsons lost roughly a year of life at age 45 (4% of remaining life expectancy) if descended from ex-POWs who suffered severe captivity conditions than if descended from non-POWs. If their grandfathers faced a less harsh captivity, male-line grandsons lost less than a year of life compared with those descended from non-POWs. I find that the grandfather's age at exposure and the grandson's education, as well as the son's and the grandson's poor late gestational conditions (proxied by season of birth), mediate this relationship. I rule out socioeconomic status, marriage and mortality selection, and cultural or psychological transmission from grandfathers to grandsons as explanations. I cannot rule out an epigenetic explanation.
Collapse
Affiliation(s)
- Dora L Costa
- Department of Economics, University of California, Los Angeles, Los Angeles, CA, USA
- National Bureau of Economic Research, Cambridge, MA, USA
| |
Collapse
|
3
|
Costa DL. Overweight grandsons and grandfathers' starvation exposure. JOURNAL OF HEALTH ECONOMICS 2023; 91:102796. [PMID: 37541079 PMCID: PMC10593129 DOI: 10.1016/j.jhealeco.2023.102796] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 08/06/2023]
Abstract
Much of the increase in the prevalence of overweight and obesity has been in developing countries with a history of famines and malnutrition. This paper is the first to examine overweight among adult grandsons of grandfathers exposed to starvation during developmental ages. I study grandsons born to grandfathers who served in the Union Army during the US Civil War (1861-5) where some grandfathers experienced severe net malnutrition because they suffered a harsh POW experience. I find that male-line but not female-line grandsons of grandfathers who survived a severe captivity during their growing years faced a 21% increase in mean overweight and a 2% increase in mean BMI compared to grandsons of non-POWs. Male-line grandsons descended from grandfathers who experienced a harsh captivity faced a 22%-28% greater risk of dying every year after age 45 relative to grandsons descended from non-POWs, with overweight accounting for 9%-14% of the excess risk.
Collapse
Affiliation(s)
- Dora L Costa
- University of California, Los Angeles, United States of America; National Bureau of Economic Research, United States of America.
| |
Collapse
|
4
|
Brooks AL, Conca J, Glines WM, Waltar AE. How the Science of Radiation Biology Can Help Reduce the Crippling Fear of Low-level Radiation. HEALTH PHYSICS 2023; 124:407-424. [PMID: 36989223 DOI: 10.1097/hp.0000000000001677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
ABSTRACT The fear of radiation has been present almost since the discovery of radiation, but has intensified since the "dawn of the atomic age" over 75 y ago. This fear has often served as an impediment to the safe and beneficial uses of radiation and radioactive material. The underlying causes of such fear are varied, can be complex, and are often not associated with any scientific knowledge or understanding. The authors believe that a clear understanding of the current scientific knowledge and understanding of the effects of radiation exposure may be useful in helping to allay some of the fear of radiation. This manuscript attempts to (1) address several scientific questions that we believe have contributed to the fear of radiation, (2) review the data derived from research that can be used to address these questions, and (3) summarize how the results of such scientific research can be used to help address the fear of low-dose and low-dose-rate radiation. Several examples of how fear of radiation has affected public perception of radiological events are discussed, as well as a brief history of the etiology of radiation fear. Actions needed to reduce the public fear of radiation and help fulfill the full societal benefits of radiation and radioactive materials are suggested.
Collapse
Affiliation(s)
- Antone L Brooks
- Research Professor Emeritus, Washington State University, Chief Scientist, DOE Low Dose Program, 6802 W. 13th Avenue, Kennewick, WA 99338
| | - James Conca
- President UFA Ventures, Inc., Richland, WA, Science writer for Forbes
| | - Wayne M Glines
- Senior Technical Advisor (retired), Department of Energy, 2315 Camas Avenue, Richland, WA 99354
| | - Alan E Waltar
- Professor and Head (retired), Department of Nuclear Engineering, Texas A&M University, Past President, American Nuclear Society, 12449 Ingalls Creek Road, Peshastin, WA 98847
| |
Collapse
|
5
|
Usupzhanova DY, Astrelina TA, Kobzeva IV, Suchkova YB, Brunchukov VA, Rastorgueva AA, Nikitina VA, Samoilov AS. Evaluation of Changes in Some Functional Properties of Human Mesenchymal Stromal Cells Induced by Low Doses of Ionizing Radiation. Int J Mol Sci 2023; 24:6346. [PMID: 37047317 PMCID: PMC10094729 DOI: 10.3390/ijms24076346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Each person is inevitably exposed to low doses of ionizing radiation (LDIR) throughout their life. The research results of LDIR effects are ambiguous and an accurate assessment of the risks associated with the influence of LDIR is an important task. Mesenchymal stromal cells (MSCs) are the regenerative reserve of an adult organism; because of this, they are a promising model for studying the effects of LDIR. The qualitative and quantitative changes in their characteristics can also be considered promising criteria for assessing the risks of LDIR exposure. The MSCs from human connective gingiva tissue (hG-MSCs) were irradiated at doses of 50, 100, 250, and 1000 mGy by the X-ray unit RUST-M1 (Russia). The cells were cultured continuously for 64 days after irradiation. During the study, we evaluated the secretory profile of hG-MSCs (IL-10, IDO, IL-6, IL-8, VEGF-A) using an ELISA test, the immunophenotype (CD45, CD34, CD90, CD105, CD73, HLA-DR, CD44) using flow cytometry, and the proliferative activity using the xCelligence RTCA cell analyzer at the chosen time points. The results of study have indicated the development of stimulating effects in the early stages of cultivation after irradiation using low doses of X-ray radiation. On the contrary, the effects of the low doses were comparable with the effects of medium doses of X-ray radiation in the long-term periods of cultivation after irradiation and have indicated the inhibition of the functional activity of MSCs.
Collapse
Affiliation(s)
| | - Tatiana A. Astrelina
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123098 Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
6
|
Colwell ML, Townsel C, Petroff RL, Goodrich JM, Dolinoy DC. Epigenetics and the Exposome: DNA Methylation as a Proxy for Health Impacts of Prenatal Environmental Exposures. EXPOSOME 2023; 3:osad001. [PMID: 37333730 PMCID: PMC10275510 DOI: 10.1093/exposome/osad001] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The accumulation of every day exposures can impact health across the life course, but our understanding of such exposures is impeded by our ability to delineate the relationship between an individual's early life exposome and later life health effects. Measuring the exposome is challenging. Exposure assessed at a given time point captures a snapshot of the exposome but does not represent the full spectrum of exposures across the life course. In addition, the assessment of early life exposures and their effects is often further challenged by lack of relevant samples and the time gap between exposures and related health outcomes in later life. Epigenetics, specifically DNA methylation, has the potential to overcome these barriers as environmental epigenetic perturbances can be retained through time. In this review, we describe how DNA methylation can be framed in the world of the exposome. We offer three compelling examples of common environmental exposures, including cigarette smoke, the endocrine active compound bisphenol A (BPA), and the metal lead (Pb), to illustrate the application of DNA methylation as a proxy to measure the exposome. We discuss areas for future explorations and current limitations of this approach. Epigenetic profiling is a promising and rapidly developing tool and field of study, offering us a unique and powerful way to assess the early life exposome and its effects across different life stages.
Collapse
Affiliation(s)
- Mathia L. Colwell
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Courtney Townsel
- Department of Obstetrics and Gynecology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Rebekah L. Petroff
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| |
Collapse
|
7
|
Terracina S, Petrella C, Francati S, Lucarelli M, Barbato C, Minni A, Ralli M, Greco A, Tarani L, Fiore M, Ferraguti G. Antioxidant Intervention to Improve Cognition in the Aging Brain: The Example of Hydroxytyrosol and Resveratrol. Int J Mol Sci 2022; 23:15674. [PMID: 36555317 PMCID: PMC9778814 DOI: 10.3390/ijms232415674] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Both physiological and pathological aging processes induce brain alterations especially affecting the speed of processing, working memory, conceptual reasoning and executive functions. Many therapeutic approaches to reduce the impact of brain aging on cognitive functioning have been tested; unfortunately, there are no satisfactory results as a single therapy. As aging is partly contributed by free radical reactions, it has been proposed that exogenous antioxidants could have a positive impact on both aging and its associated manifestations. The aim of this report is to provide a summary and a subsequent review of the literature evidence on the role of antioxidants in preventing and improving cognition in the aging brain. Manipulation of endogenous cellular defense mechanisms through nutritional antioxidants or pharmacological compounds represents an innovative approach to therapeutic intervention in diseases causing brain tissue damage, such as neurodegeneration. Coherently with this notion, antioxidants, especially those derived from the Mediterranean diet such as hydroxytyrosol and resveratrol, seem to be able to delay and modulate the cognitive brain aging processes and decrease the occurrence of its effects on the brain. The potential preventive activity of antioxidants should be evaluated in long-term exposure clinical trials, using preparations with high bioavailability, able to bypass the blood-brain barrier limitation, and that are well standardized.
Collapse
Affiliation(s)
- Sergio Terracina
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC—CNR, 00185 Rome, Italy
| | - Silvia Francati
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Christian Barbato
- Institute of Biochemistry and Cell Biology, IBBC—CNR, 00185 Rome, Italy
| | - Antonio Minni
- Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology, IBBC—CNR, 00185 Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
8
|
A hypothesis: Retrotransposons as a relay of epigenetic marks in intergenerational epigenetic inheritance. Gene 2022; 817:146229. [PMID: 35063571 DOI: 10.1016/j.gene.2022.146229] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/13/2021] [Accepted: 01/13/2022] [Indexed: 12/19/2022]
Abstract
Epigenetic marks in gametes, which both respond to the parental environmental factors and shape offspring phenotypes, are usually positioned to mediate intergenerational or transgenerational epigenetic inheritance. Nonetheless, the mechanisms through which gametic epigenetic signatures encode parental acquired phenotypes, and further initiate a cascade of molecular events to affect offspring phenotypes during early embryonic development, remain unclear. Retrotransposons are mobile DNA elements that could resist to genomic epigenetic reprogramming at specific loci and rewire the core regulatory networks of embryogenesis. Increasing evidences show that retrotransposons in the embryonic genome could interact with gametic epigenetic marks, which provides a tentative possibility that retrotransposons may serve as a relay of gametic epigenetic marks to transmit parental acquired traits. Here, we summarize the recent progress in exploring the crosstalk between gametic epigenetic marks and retrotransposons, and the regulation of gene expression and early embryonic development by retrotransposons. Accordingly, deciphering the mystery of interactions between gametic epigenetic marks and retrotransposons during early embryonic development will provide valuable insights into the intergenerational or transgenerational transmission of acquired traits.
Collapse
|
9
|
Abstract
In this interview, Professor Randy L Jirtle speaks with Storm Johnson, Commissioning Editor for Epigenomics, on his work on genomic imprinting, environmental epigenomics and the fetal origins of disease susceptibility. Professor Randy Jirtle joined the Duke University Department of Radiology in 1977 and headed the Epigenetics and Imprinting Laboratory until 2012. He is now Professor of Epigenetics in the Department of Biological Sciences at North Carolina State University, Raleigh, NC, USA. Jirtle's research interests are in epigenetics, genomic imprinting and the fetal origins of disease susceptibility. He is known for his groundbreaking studies linking environmental exposures early in life to the development of adult diseases through changes in the epigenome and for determining the evolutionary origin of genomic imprinting in mammals. He has published over 200 peer-reviewed articles as well as the books Liver Regeneration and Carcinogenesis: Molecular and Cellular Mechanisms, Environmental Epigenomics in Health and Disease: Epigenetics and Disease Origins and Environmental Epigenomics in Health and Disease: Epigenetics and Complex Diseases. He was honored in 2006 with the Distinguished Achievement Award from the College of Engineering at the University of Wisconsin-Madison. In 2007, he was a featured scientist on the NOVA television program on epigenetics titled 'Ghost in Your Genes' and was nominated for Time Magazine's 'Person of the Year'. He was the inaugural recipient of the Epigenetic Medicine Award in 2008 and received the STARS Lecture Award in Nutrition and Cancer from the National Cancer Institute in 2009. Jirtle was presented the Linus Pauling Award from the Institute of Functional Medicine in 2014. In 2017, ShortCutsTV produced the English documentary 'Are You What Your Mother Ate? The Agouti Mouse Study' based on his pioneering epigenetic research. He received the 2018 Northern Communities Health Foundation Visiting Professorship Award at the University of Adelaide, Australia. The Personalized Lifestyle Medicine Institute presented Jirtle with the Research and Innovation Leadership Award in 2019. Dr Jirtle was also given the Alexander Hollaender Award in 2019 at the 50th annual meeting of the Environmental Mutagenesis and Genomics Society.
Collapse
Affiliation(s)
- Randy L Jirtle
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
10
|
López-Nieva P, González-Vasconcellos I, González-Sánchez L, Cobos-Fernández MA, Ruiz-García S, Sánchez Pérez R, Aroca Á, Fernández-Piqueras J, Santos J. Differential molecular response in mice and human thymocytes exposed to a combined-dose radiation regime. Sci Rep 2022; 12:3144. [PMID: 35210498 PMCID: PMC8873405 DOI: 10.1038/s41598-022-07166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/11/2022] [Indexed: 11/17/2022] Open
Abstract
In the quest for more effective radiation treatment options that can improve both cell killing and healthy tissue recovery, combined radiation therapies are lately in the spotlight. The molecular response to a combined radiation regime where exposure to an initial low dose (priming dose) of ionizing radiation is administered prior to a subsequent higher radiation dose (challenging dose) after a given latency period have not been thoroughly explored. In this study we report on the differential response to either a combined radiation regime or a single challenging dose both in mouse in vivo and in human ex vivo thymocytes. A differential cell cycle response including an increase in the subG1 fraction on cells exposed to the combined regime was found. Together with this, a differential protein expression profiling in several pathways including cell cycle control (ATM, TP53, p21CDKN1A), damage response (γH2AX) and cell death pathways such as apoptosis (Cleaved Caspase-3, PARP1, PKCδ and H3T45ph) and ferroptosis (xCT/GPX4) was demonstrated. This study also shows the epigenetic regulation following a combined regime that alters the expression of chromatin modifiers such as DNMTs (DNMT1, DNMT2, DNMT3A, DNMT3B, DNMT3L) and glycosylases (MBD4 and TDG). Furthermore, a study of the underlying cellular status six hours after the priming dose alone showed evidence of retained modifications on the molecular and epigenetic pathways suggesting that the priming dose infers a “radiation awareness phenotype” to the thymocytes, a sensitization key to the differential response seen after the second hit with the challenging dose. These data suggest that combined-dose radiation regimes could be more efficient at making cells respond to radiation and it would be interesting to further investigate how can these schemes be of use to potential new radiation therapies.
Collapse
Affiliation(s)
- Pilar López-Nieva
- Genome Dynamics and Function Program, Genome Decoding Unit, Severo Ochoa Molecular Biology Center, Madrid, Spain. .,Department of Biology, Madrid Autonomous University, 28049, Madrid, Spain. .,Institute of Health Research, Jiménez Díaz Foundation, 28040, Madrid, Spain.
| | - Iria González-Vasconcellos
- Genome Dynamics and Function Program, Genome Decoding Unit, Severo Ochoa Molecular Biology Center, Madrid, Spain. .,Department of Biology, Madrid Autonomous University, 28049, Madrid, Spain.
| | - Laura González-Sánchez
- Genome Dynamics and Function Program, Genome Decoding Unit, Severo Ochoa Molecular Biology Center, Madrid, Spain.,Institute of Health Research, Jiménez Díaz Foundation, 28040, Madrid, Spain.,Consorcio de Investigación Biomédica de Enfermedades Raras (CIBERER), Madrid, Spain
| | - María A Cobos-Fernández
- Genome Dynamics and Function Program, Genome Decoding Unit, Severo Ochoa Molecular Biology Center, Madrid, Spain.,Department of Biology, Madrid Autonomous University, 28049, Madrid, Spain
| | - Sara Ruiz-García
- Genome Dynamics and Function Program, Genome Decoding Unit, Severo Ochoa Molecular Biology Center, Madrid, Spain.,Department of Biology, Madrid Autonomous University, 28049, Madrid, Spain
| | - Raúl Sánchez Pérez
- Department of Congenital Cardiac Surgery, Hospital Universitario La Paz, 28046, Madrid, Spain
| | - Ángel Aroca
- Department of Congenital Cardiac Surgery, Hospital Universitario La Paz, 28046, Madrid, Spain
| | - José Fernández-Piqueras
- Genome Dynamics and Function Program, Genome Decoding Unit, Severo Ochoa Molecular Biology Center, Madrid, Spain.,Department of Biology, Madrid Autonomous University, 28049, Madrid, Spain.,Institute of Health Research, Jiménez Díaz Foundation, 28040, Madrid, Spain
| | - Javier Santos
- Genome Dynamics and Function Program, Genome Decoding Unit, Severo Ochoa Molecular Biology Center, Madrid, Spain.,Department of Biology, Madrid Autonomous University, 28049, Madrid, Spain.,Institute of Health Research, Jiménez Díaz Foundation, 28040, Madrid, Spain
| |
Collapse
|
11
|
Yushkova E. Radiobiological features in offspring of natural populations of Drosophila melanogaster after Chernobyl accident. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2022; 63:84-97. [PMID: 35275441 DOI: 10.1002/em.22476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/24/2022] [Accepted: 03/08/2022] [Indexed: 06/14/2023]
Abstract
In their natural habitats, populations of organisms are faced with different levels of chronic low-intensity radiation, causing a wide range of radiobiological effects (from radiosensitivity to radioadaptive response and hormesis). In this study, specimens of Drosophila melanogaster were selected from territories of the Chernobyl nuclear power plant with different levels of radioactive contamination. The isogenic stocks derived from these specimens represent the genetic systems of current populations and make it possible to study radioresistance and its mechanisms in future generations under controlled laboratory conditions. Previous studies have shown that transgenerational radiation effects at the level of lethal mutations and survival rate are unstable and depend not only on the level of chronic low-intensity irradiation, but also on other factors. A single acute irradiation exposure of offspring whose parents inhabited a site with a higher level of chronic irradiation made it possible to reveal pronounced radioresistant features in the offspring. And the offspring whose parents were exposed to radiation levels close to the natural radiation background, on the contrary, acquired radiosensitive features. Their response to acute exposure includes a high-frequency of lethal mutations and a short lifespan. The differential response to different levels of chronic parental exposure is caused by differences in the activities of certain transposons that destabilize the genome. Our data contribute to the understanding of genetic and epigenetic mechanisms (via transposon activity) of the effect of parental radiation exposure on the health and adaptive potential of populations affected by the technogenically increased radiation background.
Collapse
Affiliation(s)
- Elena Yushkova
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the Russian Academy of Science, Syktyvkar, Russia
| |
Collapse
|
12
|
Kuzmina NS. Radiation-Induced DNA Methylation Disorders: In Vitro and In Vivo Studies. BIOL BULL+ 2022. [DOI: 10.1134/s1062359021110066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Abdel-Aziz N, Elkady AA, Elgazzar EM. Effect of Low-Dose Gamma Radiation and Lipoic Acid on High- Radiation-Dose Induced Rat Brain Injuries. Dose Response 2021; 19:15593258211044845. [PMID: 34759786 PMCID: PMC8573698 DOI: 10.1177/15593258211044845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/17/2021] [Indexed: 12/04/2022] Open
Abstract
Aim This work aims to investigate the possible radio-adaptive mechanisms induced by low-dose (LD) whole-body γ-irradiation alone or combined with alpha-lipoic acid (ALA) administration in modulating high-dose (HD) head irradiation–induced brain injury in rats. Materials and Methods Rats were irradiated with LD (.25 Gy) 24 hours prior HD (20 Gy), and subjected to ALA (100 mg/kg/day) 5 minutes after HD and continued for 10 days. At the end of the experiment, animals were sacrificed and brain samples were dissected for biochemical and histopathological examinations. Results HD irradiation-induced brain injury as manifested by elevation of oxidative stress, DNA damage, apoptotic, and inflammatory markers in brain tissue. Histological examination of brain sections showed marked alterations. However, LD alone or combined with ALA ameliorated the changes induced by HD. Conclusion Under the present experimental conditions, LD whole-body irradiation exhibited neuroprotective activity against detrimental effects of a subsequent HD head irradiation. This effect might be due to the adaptive response induced by LD that activated the anti-oxidative, anti-apoptotic, and anti-inflammatory mechanisms in the affected animals making them able to cope with the subsequent high-dose exposure. However, the combined LD exposure and ALA supplementation produced a further modulating effect in the HD-irradiated rats.
Collapse
Affiliation(s)
- Nahed Abdel-Aziz
- Radiation Biology Research Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Ahmed A Elkady
- Ahmed A. Elkady: Health Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Eman M Elgazzar
- Ahmed A. Elkady: Health Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| |
Collapse
|
14
|
Averbeck D, Rodriguez-Lafrasse C. Role of Mitochondria in Radiation Responses: Epigenetic, Metabolic, and Signaling Impacts. Int J Mol Sci 2021; 22:ijms222011047. [PMID: 34681703 PMCID: PMC8541263 DOI: 10.3390/ijms222011047] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Until recently, radiation effects have been considered to be mainly due to nuclear DNA damage and their management by repair mechanisms. However, molecular biology studies reveal that the outcomes of exposures to ionizing radiation (IR) highly depend on activation and regulation through other molecular components of organelles that determine cell survival and proliferation capacities. As typical epigenetic-regulated organelles and central power stations of cells, mitochondria play an important pivotal role in those responses. They direct cellular metabolism, energy supply and homeostasis as well as radiation-induced signaling, cell death, and immunological responses. This review is focused on how energy, dose and quality of IR affect mitochondria-dependent epigenetic and functional control at the cellular and tissue level. Low-dose radiation effects on mitochondria appear to be associated with epigenetic and non-targeted effects involved in genomic instability and adaptive responses, whereas high-dose radiation effects (>1 Gy) concern therapeutic effects of radiation and long-term outcomes involving mitochondria-mediated innate and adaptive immune responses. Both effects depend on radiation quality. For example, the increased efficacy of high linear energy transfer particle radiotherapy, e.g., C-ion radiotherapy, relies on the reduction of anastasis, enhanced mitochondria-mediated apoptosis and immunogenic (antitumor) responses.
Collapse
Affiliation(s)
- Dietrich Averbeck
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Correspondence:
| | - Claire Rodriguez-Lafrasse
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Department of Biochemistry and Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| |
Collapse
|
15
|
Bertozzi TM, Becker JL, Blake GET, Bansal A, Nguyen DK, Fernandez-Twinn DS, Ozanne SE, Bartolomei MS, Simmons RA, Watson ED, Ferguson-Smith AC. Variably methylated retrotransposons are refractory to a range of environmental perturbations. Nat Genet 2021; 53:1233-1242. [PMID: 34326545 PMCID: PMC7611517 DOI: 10.1038/s41588-021-00898-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/18/2021] [Indexed: 12/27/2022]
Abstract
The agouti viable yellow (Avy) allele is an insertional mutation in the mouse genome caused by a variably methylated intracisternal A particle (VM-IAP) retrotransposon. Avy expressivity is sensitive to a range of early-life chemical exposures and nutritional interventions, suggesting that environmental perturbations can have long-lasting effects on the methylome. However, the extent to which VM-IAP elements are environmentally labile with phenotypic implications is unknown. Using a recently identified repertoire of VM-IAPs, we assessed the epigenetic effects of different environmental contexts. A longitudinal aging analysis indicated that VM-IAPs are stable across the murine lifespan, with only small increases in DNA methylation detected for a subset of loci. No significant effects were observed after maternal exposure to the endocrine disruptor bisphenol A, an obesogenic diet or methyl donor supplementation. A genetic mouse model of abnormal folate metabolism exhibited shifted VM-IAP methylation levels and altered VM-IAP-associated gene expression, yet these effects are likely largely driven by differential targeting by polymorphic KRAB zinc finger proteins. We conclude that epigenetic variability at retrotransposons is not predictive of environmental susceptibility.
Collapse
Affiliation(s)
| | | | - Georgina E T Blake
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Amita Bansal
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Australian National University Medical School, John Curtin School of Medical Research, College of Health and Medicine, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Duy K Nguyen
- Department of Cell and Developmental Biology, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust Medical Research Council Institute of Metabolic Science, Cambridge, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust Medical Research Council Institute of Metabolic Science, Cambridge, UK
| | - Marisa S Bartolomei
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Erica D Watson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
16
|
Khan AUH, Blimkie M, Yang DS, Serran M, Pack T, Wu J, Kang JY, Laakso H, Lee SH, Le Y. Effects of Chronic Low-Dose Internal Radiation on Immune-Stimulatory Responses in Mice. Int J Mol Sci 2021; 22:7303. [PMID: 34298925 PMCID: PMC8306076 DOI: 10.3390/ijms22147303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 12/18/2022] Open
Abstract
The Linear-No-Threshold (LNT) model predicts a dose-dependent linear increase in cancer risk. This has been supported by biological and epidemiological studies at high-dose exposures. However, at low-doses (LDR ≤ 0.1 Gy), the effects are more elusive and demonstrate a deviation from linearity. In this study, the effects of LDR on the development and progression of mammary cancer in FVB/N-Tg(MMTVneu)202Mul/J mice were investigated. Animals were chronically exposed to total doses of 10, 100, and 2000 mGy via tritiated drinking water, and were assessed at 3.5, 6, and 8 months of age. Results indicated an increased proportion of NK cells in various organs of LDR exposed mice. LDR significantly influenced NK and T cell function and activation, despite diminishing cell proliferation. Notably, the expression of NKG2D receptor on NK cells was dramatically reduced at 3.5 months but was upregulated at later time-points, while the expression of NKG2D ligand followed the opposite trend, with an increase at 3.5 months and a decrease thereafter. No noticeable impact was observed on mammary cancer development, as measured by tumor load. Our results demonstrated that LDR significantly influenced the proportion, proliferation, activation, and function of immune cells. Importantly, to the best of our knowledge, this is the first report demonstrating that LDR modulates the cross-talk between the NKG2D receptor and its ligands.
Collapse
Affiliation(s)
- Abrar Ul Haq Khan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
| | - Melinda Blimkie
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Doo Seok Yang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
| | - Mandy Serran
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Tyler Pack
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Jin Wu
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Ji-Young Kang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
| | - Holly Laakso
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
- Centre for Infection, The University of Ottawa, Immunity and Inflammation, Ottawa, ON K1H 8M5, Canada
| | - Yevgeniya Le
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
- CANDU Owners Group Inc., Toronto, ON M5G 2K4, Canada
| |
Collapse
|
17
|
Hitchler MJ, Domann FE. The epigenetic and morphogenetic effects of molecular oxygen and its derived reactive species in development. Free Radic Biol Med 2021; 170:70-84. [PMID: 33450377 PMCID: PMC8217084 DOI: 10.1016/j.freeradbiomed.2021.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/11/2022]
Abstract
The development of multicellular organisms involves the unpacking of a complex genetic program. Extensive characterization of discrete developmental steps has revealed the genetic program is controlled by an epigenetic state. Shifting the epigenome is a group of epigenetic enzymes that modify DNA and proteins to regulate cell type specific gene expression. While the role of these modifications in development has been established, the input(s) responsible for electing changes in the epigenetic state remains unknown. Development is also associated with dynamic changes in cellular metabolism, redox, free radical production, and oxygen availability. It has previously been postulated that these changes are causal in development by affecting gene expression. This suggests that oxygen is a morphogenic compound that impacts the removal of epigenetic marks. Likewise, metabolism and reactive oxygen species influence redox signaling through iron and glutathione to limit the availability of key epigenetic cofactors such as α-ketoglutarate, ascorbate, NAD+ and S-adenosylmethionine. Given the close relationship between these cofactors and epigenetic marks it seems likely that the two are linked. Here we describe how changing these inputs might affect the epigenetic state during development to drive gene expression. Combined, these cofactors and reactive oxygen species constitute the epigenetic landscape guiding cells along differing developmental paths.
Collapse
Affiliation(s)
- Michael J Hitchler
- Department of Radiation Oncology, Kaiser Permanente Los Angeles Medical Center, 4950 Sunset Blvd, Los Angeles, CA, 90027, USA.
| | - Frederick E Domann
- Department of Radiation Oncology, Free Radical and Radiation Biology Program, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
18
|
Lozoya OA, Xu F, Grenet D, Wang T, Grimm SA, Godfrey V, Waidyanatha S, Woychik RP, Santos JH. Single Nucleotide Resolution Analysis Reveals Pervasive, Long-Lasting DNA Methylation Changes by Developmental Exposure to a Mitochondrial Toxicant. Cell Rep 2021; 32:108131. [PMID: 32937126 PMCID: PMC7553240 DOI: 10.1016/j.celrep.2020.108131] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/16/2020] [Accepted: 08/20/2020] [Indexed: 02/08/2023] Open
Abstract
Mitochondrial-driven alterations of the epigenome have been reported, but whether they are relevant at the organismal level remains unknown. The viable yellow agouti mouse (Avy) is a powerful epigenetic biosensor model that reports on the DNA methylation status of the Avy locus, which is established prior to the three-germ-layer separation, through the coat color of the animals. Here we show that maternal exposure to rotenone, a potent mitochondrial complex I inhibitor, not only changes the DNA methylation status of the Avy locus in the skin but broadly affects the liver DNA methylome of the offspring. These effects are accompanied by altered gene expression programs that persist throughout life, and which associate with impairment of antioxidant activity and mitochondrial function in aged animals. These pervasive and lasting genomic effects suggest a putative role for mitochondria in regulating life-long gene expression programs through developmental nuclear epigenetic remodeling. Lozoya et al. provide in vivo evidence of the epigenetic effects of mitochondrial dysfunction. Developmental-only exposure to rotenone through the mother’s diet inhibits mitochondrial complex I in the dams and results in lifelong nuclear DNA methylation and gene expression changes in the offspring. Aged offspring also show functional outcomes.
Collapse
Affiliation(s)
- Oswaldo A Lozoya
- Genome Integrity and Structural Biology Laboratory, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Fuhua Xu
- Genome Integrity and Structural Biology Laboratory, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Dagoberto Grenet
- Genome Integrity and Structural Biology Laboratory, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Tianyuan Wang
- Integrative Bioinformatics Support Group, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Sara A Grimm
- Integrative Bioinformatics Support Group, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Veronica Godfrey
- National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Suramya Waidyanatha
- National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Richard P Woychik
- Genome Integrity and Structural Biology Laboratory, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| | - Janine H Santos
- Genome Integrity and Structural Biology Laboratory, National Institutes of Health, Research Triangle Park, NC 27709, USA; National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
19
|
Stephen Inbaraj B, Chen BH. An overview on recent in vivo biological application of cerium oxide nanoparticles. Asian J Pharm Sci 2020; 15:558-575. [PMID: 33193860 PMCID: PMC7610205 DOI: 10.1016/j.ajps.2019.10.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/25/2019] [Accepted: 10/05/2019] [Indexed: 12/13/2022] Open
Abstract
Cerium oxide nanoparticles (CNPs) possess a great potential as therapeutic agents due to their ability to self-regenerate by reversibly switching between two valences +3 and +4. This article reviews recent articles dealing with in vivo studies of CNPs towards Alzheimer's disease, obesity, liver inflammation, cancer, sepsis, amyotrophic lateral sclerosis, acute kidney injury, radiation-induced tissue damage, hepatic ischemia reperfusion injury, retinal diseases and constipation. In vivo anti-cancer studies revealed the effectiveness of CNPs to reduce tumor growth and angiogenesis in melanoma, ovarian, breast and retinoblastoma cancer cell-induced mice, with their conjugation with folic acid, doxorubicin, CPM, or CXC receptor-4 antagonist ligand eliciting higher efficiency. After conjugation with triphenylphosphonium or magnetite nanoparticles, CNPs were shown to combat Alzheimer's disease by reducing amyloid-β, glial fibrillary acidic protein, inflammatory and oxidative stress markers in mice. By improving muscle function and longevity, the citrate/EDTA-stabilized CNPs could ameliorate amyotrophic lateral sclerosis. Also, they could effectively reduce obesity in mice by scavenging ROS and reducing adipogenesis, triglyceride synthesis, GAPDH enzyme activity, leptin and insulin levels. In CCl4-induced rats, stress signaling pathways due to inflammatory cytokines, liver enzymes, oxidative and endoplasmic reticulum messengers could be attenuated by CNPs. Commercial CNPs showed protective effects on rats with hepatic ischemia reperfusion and peritonitis-induced hepatic/cardiac injuries by decreasing oxidative stress and hepatic/cardiac inflammation. The same CNPs could improve kidney function by diminishing renal superoxide, hyperglycemia and tubular damage in peritonitis-induced acute kidney injury in rats. Radiation-induced lung and testicular tissue damage could be alleviated in mice, with the former showing improvement in pulmonary distress and bronchoconstriction and the latter exhibiting restoration in spermatogenesis rate and spermatid/spermatocyte number. Through enhancement of gastrointestinal motility, the CNPs could alleviate constipation in both young and old rats. They could also protect rat from light-induced retinal damage by slowing down neurodegenerative process and microglial activation.
Collapse
Affiliation(s)
| | - Bing-Huei Chen
- Department of Food Science, Fu Jen Catholic University, Taipei 242
| |
Collapse
|
20
|
Ionizing Radiation-Induced Epigenetic Modifications and Their Relevance to Radiation Protection. Int J Mol Sci 2020; 21:ijms21175993. [PMID: 32825382 PMCID: PMC7503247 DOI: 10.3390/ijms21175993] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
The present system of radiation protection assumes that exposure at low doses and/or low dose-rates leads to health risks linearly related to the dose. They are evaluated by a combination of epidemiological data and radiobiological models. The latter imply that radiation induces deleterious effects via genetic mutation caused by DNA damage with a linear dose-dependence. This picture is challenged by the observation of radiation-induced epigenetic effects (changes in gene expression without altering the DNA sequence) and of non-linear responses, such as non-targeted and adaptive responses, that in turn can be controlled by gene expression networks. Here, we review important aspects of the biological response to ionizing radiation in which epigenetic mechanisms are, or could be, involved, focusing on the possible implications to the low dose issue in radiation protection. We examine in particular radiation-induced cancer, non-cancer diseases and transgenerational (hereditary) effects. We conclude that more realistic models of radiation-induced cancer should include epigenetic contribution, particularly in the initiation and progression phases, while the impact on hereditary risk evaluation is expected to be low. Epigenetic effects are also relevant in the dispute about possible "beneficial" effects at low dose and/or low dose-rate exposures, including those given by the natural background radiation.
Collapse
|
21
|
Bertucci EM, Mason MW, Camus AC, Rhodes OE, Parrott BB. Chronic low dose irradiation alters hepatic transcriptional profiles, but not global DNA methylation in medaka (Oryzias latipes). THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 729:138680. [PMID: 32361431 DOI: 10.1016/j.scitotenv.2020.138680] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/26/2020] [Accepted: 04/11/2020] [Indexed: 06/11/2023]
Abstract
Ionizing radiation (IR) resulting from both natural and anthropogenic sources is ubiquitous throughout the environment. Historically, studies on the biological impacts of radiation primarily focused on responses to acute doses of radiation, with little advancement in our understanding of environmentally relevant exposures. Epigenetic mechanisms are capable of mediating organismal responses to environmental stressors and DNA methylation plays important roles in gene regulation and promoting chromosomal stability. Here, we assess broad-scale transcriptional and epigenetic variation resulting from chronic exposure to low doses of ionizing radiation (LDIR; 5.78, 53.76, or 520.23 mGy/day) using Japanese medaka fish (Oryzias latipes) in a replicated mesocosm design. We observed significant changes to the hepatic transcriptome induced by a 3-month chronic exposure to IR, whereas global DNA methylation appeared largely unaffected. Our findings reveal a set of genes, including those involved in immune function, responding to environmentally relevant IR exposures, which do not appear to be mediated by a systemic global shift in DNA methylation.
Collapse
Affiliation(s)
- Emily M Bertucci
- Odum School of Ecology, University of Georgia, 140 E. Green St., Athens 30602, GA, USA; Savannah River Ecology Laboratory, University of Georgia, P.O. Drawer E, Aiken 29802, SC, USA.
| | - Marilyn W Mason
- Savannah River Ecology Laboratory, University of Georgia, P.O. Drawer E, Aiken 29802, SC, USA.
| | - Alvin C Camus
- College of Veterinary Medicine Department of Pathology, University of Georgia, 501 D.W. Brooks Drive, Athens 30602, GA, USA.
| | - Olin E Rhodes
- Odum School of Ecology, University of Georgia, 140 E. Green St., Athens 30602, GA, USA; Savannah River Ecology Laboratory, University of Georgia, P.O. Drawer E, Aiken 29802, SC, USA.
| | - Benjamin B Parrott
- Odum School of Ecology, University of Georgia, 140 E. Green St., Athens 30602, GA, USA; Savannah River Ecology Laboratory, University of Georgia, P.O. Drawer E, Aiken 29802, SC, USA.
| |
Collapse
|
22
|
Murat El Houdigui S, Adam-Guillermin C, Armant O. Ionising Radiation Induces Promoter DNA Hypomethylation and Perturbs Transcriptional Activity of Genes Involved in Morphogenesis during Gastrulation in Zebrafish. Int J Mol Sci 2020; 21:ijms21114014. [PMID: 32512748 PMCID: PMC7312202 DOI: 10.3390/ijms21114014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/22/2022] Open
Abstract
Embryonic development is particularly vulnerable to stress and DNA damage, as mutations can accumulate through cell proliferation in a wide number of cells and organs. However, the biological effects of chronic exposure to ionising radiation (IR) at low and moderate dose rates (< 6 mGy/h) remain largely controversial, raising concerns for environmental protection. The present study focuses on the molecular effects of IR (0.005 to 50 mGy/h) on zebrafish embryos at the gastrula stage (6 hpf), at both the transcriptomics and epigenetics levels. Our results show that exposure to IR modifies the expression of genes involved in mitochondrial activity from 0.5 to 50 mGy/h. In addition, important developmental pathways, namely, the Notch, retinoic acid, BMP and Wnt signalling pathways, were altered at 5 and 50 mGy/h. Transcriptional changes of genes involved in the morphogenesis of the ectoderm and mesoderm were detected at all dose rates, but were prominent from 0.5 to 50 mGy/h. At the epigenetic level, exposure to IR induced a hypomethylation of DNA in the promoter of genes that colocalised with both H3K27me3 and H3Kme4 histone marks and correlated with changes in transcriptional activity. Finally, pathway enrichment analysis demonstrated that the DNA methylation changes occurred in the promoter of important developmental genes, including morphogenesis of the ectoderm and mesoderm. Together, these results show that the transcriptional program regulating morphogenesis in gastrulating embryos was modified at dose rates greater than or equal to 0.5 mGy/h, which might predict potential neurogenesis and somitogenesis defects observed at similar dose rates later in development.
Collapse
Affiliation(s)
- Sophia Murat El Houdigui
- PSE-ENV/SRTE/LECO, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Cadarache, 13115 Saint-Paul-Lez-Durance, France;
| | - Christelle Adam-Guillermin
- PSE-SANTE/SDOS/LMDN, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Cadarache, 13115 Saint-Paul-Lez-Durance, France;
| | - Olivier Armant
- PSE-ENV/SRTE/LECO, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Cadarache, 13115 Saint-Paul-Lez-Durance, France;
- Correspondence:
| |
Collapse
|
23
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020. [PMID: 32399610 DOI: 10.1007/s00204-020-02752-z)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
24
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020; 94:1511-1549. [PMID: 32399610 PMCID: PMC7261741 DOI: 10.1007/s00204-020-02752-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
25
|
Jin S, Jiang H, Cai L. New understanding of the low-dose radiation-induced hormesis. RADIATION MEDICINE AND PROTECTION 2020. [DOI: 10.1016/j.radmp.2020.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
26
|
Bertozzi TM, Ferguson-Smith AC. Metastable epialleles and their contribution to epigenetic inheritance in mammals. Semin Cell Dev Biol 2020; 97:93-105. [PMID: 31551132 DOI: 10.1016/j.semcdb.2019.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 02/02/2023]
Abstract
Many epigenetic differences between individuals are driven by genetic variation. Mammalian metastable epialleles are unusual in that they show variable DNA methylation states between genetically identical individuals. The occurrence of such states across generations has resulted in their consideration by many as strong evidence for epigenetic inheritance in mammals, with the classic Avy and AxinFu mouse models - each products of repeat element insertions - being the most widely accepted examples. Equally, there has been interest in exploring their use as epigenetic biosensors given their susceptibility to environmental compromise. Here we review the classic murine metastable epialleles as well as more recently identified candidates, with the aim of providing a more holistic understanding of their biology. We consider the extent to which epigenetic inheritance occurs at metastable epialleles and explore the limited mechanistic insights into the establishment of their variable epigenetic states. We discuss their environmental modulation and their potential relevance in genome regulation. In light of recent whole-genome screens for novel metastable epialleles, we point out the need to reassess their biological relevance in multi-generational studies and we highlight their value as a model to study repeat element silencing as well as the mechanisms and consequences of mammalian epigenetic stochasticity.
Collapse
Affiliation(s)
- Tessa M Bertozzi
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | |
Collapse
|
27
|
Perera BP, Faulk C, Svoboda LK, Goodrich JM, Dolinoy DC. The role of environmental exposures and the epigenome in health and disease. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:176-192. [PMID: 31177562 PMCID: PMC7252203 DOI: 10.1002/em.22311] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 05/02/2023]
Abstract
The genetic material of every organism exists within the context of regulatory networks that govern gene expression, collectively called the epigenome. Epigenetics has taken center stage in the study of diseases such as cancer and diabetes, but its integration into the field of environmental health is still emerging. As the Environmental Mutagenesis and Genomics Society (EMGS) celebrates its 50th Anniversary this year, we have come together to review and summarize the seminal advances in the field of environmental epigenomics. Specifically, we focus on the role epigenetics may play in multigenerational and transgenerational transmission of environmentally induced health effects. We also summarize state of the art techniques for evaluating the epigenome, environmental epigenetic analysis, and the emerging field of epigenome editing. Finally, we evaluate transposon epigenetics as they relate to environmental exposures and explore the role of noncoding RNA as biomarkers of environmental exposures. Although the field has advanced over the past several decades, including being recognized by EMGS with its own Special Interest Group, recently renamed Epigenomics, we are excited about the opportunities for environmental epigenetic science in the next 50 years. Environ. Mol. Mutagen. 61:176-192, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bambarendage P.U. Perera
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Christopher Faulk
- Department of Animal Sciences, University of Minnesota, St. Paul, Minnesota
| | - Laurie K. Svoboda
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Correspondence to: Dana C. Dolinoy, Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan.
| |
Collapse
|
28
|
Kuzmina NS, Lapteva NS, Rusinova GG, Azizova TV, Vyazovskaya NS, Rubanovich AV. Dose Dependence of Hypermethylation of Gene Promoters in Blood Leukocytes in Humans Occupationally Exposed to External γ-Radiation. BIOL BULL+ 2019. [DOI: 10.1134/s1062359019110062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
29
|
Birkett N, Al-Zoughool M, Bird M, Baan RA, Zielinski J, Krewski D. Overview of biological mechanisms of human carcinogens. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2019; 22:288-359. [PMID: 31631808 DOI: 10.1080/10937404.2019.1643539] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
This review summarizes the carcinogenic mechanisms for 109 Group 1 human carcinogens identified as causes of human cancer through Volume 106 of the IARC Monographs. The International Agency for Research on Cancer (IARC) evaluates human, experimental and mechanistic evidence on agents suspected of inducing cancer in humans, using a well-established weight of evidence approach. The monographs provide detailed mechanistic information about all carcinogens. Carcinogens with closely similar mechanisms of action (e.g. agents emitting alpha particles) were combined into groups for the review. A narrative synopsis of the mechanistic profiles for the 86 carcinogens or carcinogen groups is presented, based primarily on information in the IARC monographs, supplemented with a non-systematic review. Most carcinogens included a genotoxic mechanism.
Collapse
Affiliation(s)
- Nicholas Birkett
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- McLaughlin Centre for Population Health Risk Assessment, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Mustafa Al-Zoughool
- Department of Community and Environmental Health, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Michael Bird
- McLaughlin Centre for Population Health Risk Assessment, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Robert A Baan
- International Agency for Research on Cancer, Lyon, France
| | - Jan Zielinski
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- McLaughlin Centre for Population Health Risk Assessment, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Daniel Krewski
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- McLaughlin Centre for Population Health Risk Assessment, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Risk Sciences International, Ottawa, Canada
| |
Collapse
|
30
|
Sokolenko VL, Sokolenko SV. Manifestations of allostatic load in residents of radiation contaminated areas aged 18–24 years. REGULATORY MECHANISMS IN BIOSYSTEMS 2019. [DOI: 10.15421/021963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
We studied the features of allostatic load (AL) in 100 students aged 18–24 years old who, from birth to adulthood, lived in the territories assigned to the IV radiation zone after the Chornobyl accident (density of soil contamination by isotopes 137Cs 3.7–18.5∙104 Bq/m2) and underwent prolonged exposure to small doses of ionizing radiation. The examined students did not have any clinical signs of the immune-neuroendocrine system dysfunction. 50 people had signs of vegetative-vascular dystonia syndrome (VVD), 48 had signs of moderate hyperthyroidism and 21 had signs of moderate hypothyroidism. During the examination session, as a factor of additional psycho-emotional load, in 66 of the examined the immunoregulatory index CD4+/CD8+ went below the lower limit of the homeostatic norm, in 62 of the examined low density lipoprotein cholesterol (LDL-C) exceeded the upper level. The relative risk (RR) and attributable risk (AR) of the participation of potential secondary factors of allostatic load formation in CD4+/CD8+ immunoregulatory index going below the lower limit were calculated. The presence of statistically significant relative risk of participation in the formation of suppression of the index CD4+/CD8+: the state of hyperthyroidism, state of hypothyroidism, vegetative-vascular dystonia syndrome, higher than normal LDL-C. When the examined students combined the signs of hyperthyroidism, vegetative-vascular dystonia syndrome and higher level of LDL-C; with combination of signs of hypothyroidism, vegetative-vascular dystonia syndrome and higher level of LDL-C. The attributable risk in all cases exceeded 0.10, which confirmed the importance of some of these factors and their complexes in the formation of the effect of reduced immunoregulatory index. The CD4+/CD8+ index can be considered an important biomarker of AL and premature age-related changes in the immune system in residents of radiation-contaminated areas. The risk of AL formation in the case of occurrence of a complex of mediated secondary biomarkers (vegetative-vascular dystonia syndrome, thyroid dysfunction, hypercholesterolemia) is higher compared to their individual significance.
Collapse
|
31
|
Skinner M, Lumey L, Fleming TP, Sapienza C, Hoyo C, Aronica L, Thompson J, Nichol PF. RW-2018-Research Workshop: The Effect of Nutrition on Epigenetic Status, Growth, and Health. JPEN J Parenter Enteral Nutr 2019; 43:627-637. [PMID: 30997688 PMCID: PMC6625918 DOI: 10.1002/jpen.1536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/26/2019] [Indexed: 12/22/2022]
Abstract
The goal of the 2018 American Society for Parenteral and Enteral Nutrition (ASPEN) Research Workshop was to explore the influence of nutrition and dietary exposure to xenobiotics on the epigenome during critical periods in development and how these exposures influence both disease incidence and severity transgenerationally. A growing compendium of research indicates that the incidence and severity of common and costly human diseases may be influenced by dietary exposures and deficiencies that modify the epigenome. The greatest periods of vulnerability to these exposures are the periconception period and early childhood. Xenobiotics in the food chain, protein malnutrition, and methyl donor deficiencies could have a profound bearing on the risk of developing heart disease, diabetes, obesity, hypertension, and mental illness over multiple generations. The financial impact and the life burden of these diseases are enormous. These and other aspects of nutrition, epigenetics, and health are explored in this research workshop.
Collapse
Affiliation(s)
- Michael Skinner
- Center for Reproductive Biology School of Biological Sciences, Washington State University Pullman WA
| | - L.H. Lumey
- Department of Epidemiology Mailman School of Public Health Columbia University Medical Center, New York, NY
| | - Tom P. Fleming
- Biological Sciences, University of Southampton, Southampton, UK
| | - Carmen Sapienza
- Fels Institute for Cancer Research and Molecular Biology Lewis Katz School of Medicine Temple University Philadelphia, PA
| | - Cathrine Hoyo
- Department of Biological Sciences, Center for Human Health and the Environment, Director, Epidemiology and Environmental Epigenomics Laboratory, North Carolina State University, Raleigh, NC
| | - Lucia Aronica
- Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, California, USA
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | | | - Peter F. Nichol
- Department of Surgery, Division of Pediatric Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
32
|
Effect of low dose gamma rays on certain essential metals and oxidative stress in different rat organs. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2019. [DOI: 10.1016/j.jrras.2013.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
33
|
Kozumbo WJ, Calabrese EJ. Two decades (1998-2018) of research Progress on Hormesis: advancing biological understanding and enabling novel applications. J Cell Commun Signal 2019; 13:273-275. [PMID: 30997652 DOI: 10.1007/s12079-019-00517-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/11/2019] [Indexed: 01/10/2023] Open
Abstract
This commentary briefly summarizes the extraordinary resurgence of hormesis within the biological, biomedical, toxicological and risk assessment domains over the past two decades. It places this resurgence within the context of challenging the scientific validity of the threshold and linear dose responses. It argues that conducting research on mechanisms that actuate and regulate the stimulatory response features of hormesis will provide the knowledge needed to develop potentially transformational applications aimed at protecting and enhancing biological resiliency as well as treating/curing a multitude of diverse medical conditions.
Collapse
Affiliation(s)
| | - Edward J Calabrese
- Department of Environmental Health Sciences, University of Massachusetts, Morrill I, N344, Amherst, MA, 01003, USA.
| |
Collapse
|
34
|
The LNT model for cancer induction is not supported by radiobiological data. Chem Biol Interact 2019; 301:34-53. [PMID: 30763552 DOI: 10.1016/j.cbi.2019.01.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/18/2022]
Abstract
The hallmarks of cancer have been the focus of much research and have influenced the development of risk models for radiation-induced cancer. However, natural defenses against cancer, which constitute the hallmarks of cancer prevention, have largely been neglected in developing cancer risk models. These natural defenses are enhanced by low doses and dose rates of ionizing radiation, which has aided in the continuation of human life over many generations. Our natural defenses operate at the molecular, cellular, tissue, and whole-body levels and include epigenetically regulated (epiregulated) DNA damage repair and antioxidant production, selective p53-independent apoptosis of aberrant cells (e.g. neoplastically transformed and tumor cells), suppression of cancer-promoting inflammation, and anticancer immunity (both innate and adaptive components). This publication reviews the scientific bases for the indicated cancer-preventing natural defenses and evaluates their implication for assessing cancer risk after exposure to low radiation doses and dose rates. Based on the extensive radiobiological evidence reviewed, it is concluded that the linear-no-threshold (LNT) model (which ignores natural defenses against cancer), as it relates to cancer risk from ionizing radiation, is highly implausible. Plausible models include dose-threshold and hormetic models. More research is needed to establish when a given model (threshold, hormetic, or other) applies to a given low-dose-radiation exposure scenario.
Collapse
|
35
|
Brooks AL. The impact of dose rate on the linear no threshold hypothesis. Chem Biol Interact 2019; 301:68-80. [PMID: 30763551 DOI: 10.1016/j.cbi.2018.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/17/2018] [Accepted: 12/11/2018] [Indexed: 12/13/2022]
Abstract
The goal of this manuscript is to define the role of dose rate and dose protraction on the induction of biological changes at all levels of biological organization. Both total dose and the time frame over which it is delivered are important as the body has great capacity to repair all types of biological damage. The importance of dose rate has been recognized almost from the time that radiation was discovered and has been included in radiation standards as a Dose, Dose Rate, Effectiveness Factor (DDREF) and a Dose Rate Effectiveness Factor (DREF). This manuscript will evaluate the role of dose rate at the molecular, cellular, tissue, experimental animals and humans to demonstrate that dose rate is an important variable in estimating radiation cancer risk and other biological effects. The impact of low-dose rates on the Linear-No-Threshold Hypothesis (LNTH) will be reviewed since if the LNTH is not valid it is not possible to calculate a single value for a DDREF or DREF. Finally, extensive human experience is briefly reviewed to show that the radiation risks are not underestimated and that radiation at environmental levels has limited impact on total human cancer risk.
Collapse
Affiliation(s)
- Antone L Brooks
- Environmental Science, Washington State University, Richland, WA, USA.
| |
Collapse
|
36
|
Tharmalingam S, Sreetharan S, Brooks AL, Boreham DR. Re-evaluation of the linear no-threshold (LNT) model using new paradigms and modern molecular studies. Chem Biol Interact 2019; 301:54-67. [PMID: 30763548 DOI: 10.1016/j.cbi.2018.11.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/13/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
The linear no-threshold (LNT) model is currently used to estimate low dose radiation (LDR) induced health risks. This model lacks safety thresholds and postulates that health risks caused by ionizing radiation is directly proportional to dose. Therefore even the smallest radiation dose has the potential to cause an increase in cancer risk. Advances in LDR biology and cell molecular techniques demonstrate that the LNT model does not appropriately reflect the biology or the health effects at the low dose range. The main pitfall of the LNT model is due to the extrapolation of mutation and DNA damage studies that were conducted at high radiation doses delivered at a high dose-rate. These studies formed the basis of several outdated paradigms that are either incorrect or do not hold for LDR doses. Thus, the goal of this review is to summarize the modern cellular and molecular literature in LDR biology and provide new paradigms that better represent the biological effects in the low dose range. We demonstrate that LDR activates a variety of cellular defense mechanisms including DNA repair systems, programmed cell death (apoptosis), cell cycle arrest, senescence, adaptive memory, bystander effects, epigenetics, immune stimulation, and tumor suppression. The evidence presented in this review reveals that there are minimal health risks (cancer) with LDR exposure, and that a dose higher than some threshold value is necessary to achieve the harmful effects classically observed with high doses of radiation. Knowledge gained from this review can help the radiation protection community in making informed decisions regarding radiation policy and limits.
Collapse
Affiliation(s)
- Sujeenthar Tharmalingam
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Rd, Sudbury, ON, P3E 2C6, Canada.
| | - Shayenthiran Sreetharan
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, 1280 Main Street W, Hamilton ON, L8S 4K1, Canada
| | - Antone L Brooks
- Environmental Science, Washington State University, Richland, WA, USA
| | - Douglas R Boreham
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Rd, Sudbury, ON, P3E 2C6, Canada; Bruce Power, Tiverton, ON(3), UK.
| |
Collapse
|
37
|
Lin S, Ren A, Wang L, Santos C, Huang Y, Jin L, Li Z, Greene NDE. Aberrant methylation of Pax3 gene and neural tube defects in association with exposure to polycyclic aromatic hydrocarbons. Clin Epigenetics 2019; 11:13. [PMID: 30665459 PMCID: PMC6341549 DOI: 10.1186/s13148-019-0611-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/08/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Neural tube defects (NTDs) are common and severe congenital malformations. Pax3 is an essential gene for neural tube closure in mice but it is unknown whether altered expression or methylation of PAX3 contributes to human NTDs. We examined the potential role of hypermethylation of Pax3 in the development of NTDs by analyzing human NTD cases and a mouse model in which NTDs were induced by benzo[a]pyrene (BaP), a widely studied polycyclic aromatic hydrocarbon (PAH). METHODS We extracted methylation information of PAX3 in neural tissues from array data of ten NTD cases and eight non-malformed controls. A validation study was then performed in a larger independent population comprising 73 NTD cases and 29 controls. Finally, we examined methylation patterns and expression of Pax3 in neural tissues from mouse embryos of dams exposed to BaP or BaP and vitamin E. RESULTS Seven CpG sites in PAX3 were hypermethylated in NTD fetuses as compared to controls in the array data. In the validation phase, significantly higher methylation levels in the body region of PAX3 were observed in NTD cases than in controls (P = 0.003). And mean methylation intensity in the body region of PAX3 in fetal neural tissues was positively correlated with median concentrations of PAH in maternal serum. In the mouse model, BaP-induced NTDs were associated with hypermethylation of specific CpG sites within both the promoter and body region of Pax3. Supplementation with vitamin E via chow decreased the rate of NTDs, partly recovered the repressed total antioxidant capacity in mouse embryos exposed to BaP, and this was accompanied by the normalization of Pax3 methylation level and gene expression. CONCLUSION Hypermethylation of Pax3 may play a role in the development of NTDs; DNA methylation aberration may be caused by exposure to BaP, with possible involvement of oxidative stress.
Collapse
Affiliation(s)
- Shanshan Lin
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Centre, Peking University, Beijing, 100191, China.,Division of Birth Cohort Study, and Department of Neonatal Surgery, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Aiguo Ren
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Centre, Peking University, Beijing, 100191, China.
| | - Linlin Wang
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Centre, Peking University, Beijing, 100191, China.
| | - Chloe Santos
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Yun Huang
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Centre, Peking University, Beijing, 100191, China
| | - Lei Jin
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Centre, Peking University, Beijing, 100191, China
| | - Zhiwen Li
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Centre, Peking University, Beijing, 100191, China
| | - Nicholas D E Greene
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, WC1N 1EH, UK
| |
Collapse
|
38
|
Abstract
Abstract
Three complementary points to Jaswal & Akhtar are raised: (1) As a person with autism, I desire sociality despite vulnerability to others’ antisocial behaviour; (2) Asperger's conflation of autism with psychopathy (Czech 2018) likely caused clinicians to disregard social motivation among those with autism; and (3) adverse experiences cause social-engagement diversity to develop in all people, not just those on the spectrum.
Collapse
|
39
|
Das S, Neal CJ, Ortiz J, Seal S. Engineered nanoceria cytoprotection in vivo: mitigation of reactive oxygen species and double-stranded DNA breakage due to radiation exposure. NANOSCALE 2018; 10:21069-21075. [PMID: 30226515 DOI: 10.1039/c8nr04640a] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Cerium oxide nanomaterials are known to absorb ionizing radiation energy, as well as to neutralize free radicals in solution, by undergoing redox changes. We, therefore, proposed that ceria nanoparticles could be used in biomedical applications as an injectable, radio-protectant material. In this study, we examine the effectiveness of engineered nanoparticles in protecting germ cells from the damaging effects of irradiation-induced cell death, in vivo. C57BL/6J male mice were used as a model and irradiation was localized to the scrotal region at 2.5, 5, and/or 10 Gy intensities. Ceria nanoparticles were introduced as 100 μL injections at 100 nM and 100 μM via tail vein injections, weekly, for one month. Following this, the animals were sacrificed and their organs (heart, brain, kidneys) were harvested. Tissues were fixed, sectioned, and stained for instances of cell death, DNA damage (TUNEL assay), and ROS (nitro-tyrosine evolution). Tissues from mice treated with ceria nanoparticles showed significantly less (∼13% decrease; *P < 0.05) tissue damage (per immunohistochemistry) over controls at up to 5 Gy radiation. DNA damage and ROS also decrease substantially with ceria treatment, confirming ceria's capacity as an injectable, radio-protectant material. The study also highlights the ability of ceria nanoparticles to protect cells/tissues from both direct and indirect effects of ionizing radiation.
Collapse
Affiliation(s)
- Soumen Das
- Materials Science and Engineering, Advanced Materials Processing Center, University of Central Florida, Orlando, FL 32816, USA.
| | | | | | | |
Collapse
|
40
|
Mordaunt CE, Shibata NM, Kieffer DA, Członkowska A, Litwin T, Weiss KH, Gotthardt DN, Olson K, Wei D, Cooper S, Wan YJY, Ali MR, LaSalle JM, Medici V. Epigenetic changes of the thioredoxin system in the tx-j mouse model and in patients with Wilson disease. Hum Mol Genet 2018; 27:3854-3869. [PMID: 30010856 PMCID: PMC6216211 DOI: 10.1093/hmg/ddy262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 06/02/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
Abstract
Wilson disease (WD) is caused by mutations in the copper transporter ATP7B, leading to copper accumulation in the liver and brain. Excess copper inhibits S-adenosyl-L-homocysteine hydrolase, leading to variable WD phenotypes from widespread alterations in DNA methylation and gene expression. Previously, we demonstrated that maternal choline supplementation in the Jackson toxic milk (tx-j) mouse model of WD corrected higher thioredoxin 1 (TNX1) transcript levels in fetal liver. Here, we investigated the effect of maternal choline supplementation on genome-wide DNA methylation patterns in tx-j fetal liver by whole-genome bisulfite sequencing (WGBS). Tx-j Atp7b genotype-dependent differences in DNA methylation were corrected by choline for genes including, but not exclusive to, oxidative stress pathways. To examine phenotypic effects of postnatal choline supplementation, tx-j mice were randomized to one of six treatment groups: with or without maternal and/or continued choline supplementation, and with or without copper chelation with penicillamine (PCA) treatment. Hepatic transcript levels of TXN1 and peroxiredoxin 1 (Prdx1) were significantly higher in mice receiving maternal and continued choline with or without PCA treatment compared to untreated mice. A WGBS comparison of human WD liver and tx-j mouse liver demonstrated a significant overlap of differentially methylated genes associated with ATP7B deficiency. Further, eight genes in the thioredoxin (TXN) pathway were differentially methylated in human WD liver samples. In summary, Atp7b deficiency and choline supplementation have a genome-wide impact, including on TXN system-related genes, in tx-j mice. These findings could explain the variability of WD phenotype and suggest new complementary treatment options for WD.
Collapse
Affiliation(s)
- Charles E Mordaunt
- Department of Medical Microbiology and Immunology, Genome Center, and MIND Institute, University of California, Davis, California, USA
| | - Noreene M Shibata
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California, Davis, California, USA
| | - Dorothy A Kieffer
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California, Davis, California, USA
| | - Anna Członkowska
- Second Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Karl Heinz Weiss
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel N Gotthardt
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Kristin Olson
- Department of Pathology, University of California, Davis, California, USA
| | - Dongguang Wei
- Department of Pathology, University of California, Davis, California, USA
| | - Stewart Cooper
- California Pacific Medical Center, San Francisco, California, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology, University of California, Davis, California, USA
| | - Mohamed R Ali
- Department of Surgery, University of California, Davis, California, USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, Genome Center, and MIND Institute, University of California, Davis, California, USA
| | - Valentina Medici
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California, Davis, California, USA
| |
Collapse
|
41
|
Abstract
We study whether paternal trauma is transmitted to the children of survivors of Confederate prisoner of war (POW) camps during the US Civil War (1861-1865) to affect their longevity at older ages, the mechanisms behind this transmission, and the reversibility of this transmission. We examine children born after the war who survived to age 45, comparing children whose fathers were non-POW veterans and ex-POWs imprisoned in very different camp conditions. We also compare children born before and after the war within the same family by paternal ex-POW status. The sons of ex-POWs imprisoned when camp conditions were at their worst were 1.11 times more likely to die than the sons of non-POWs and 1.09 times more likely to die than the sons of ex-POWs when camp conditions were better. Paternal ex-POW status had no impact on daughters. Among sons born in the fourth quarter, when maternal in utero nutrition was adequate, there was no impact of paternal ex-POW status. In contrast, among sons born in the second quarter, when maternal nutrition was inadequate, the sons of ex-POWs who experienced severe hardship were 1.2 times more likely to die than the sons of non-POWs and ex-POWs who fared better in captivity. Socioeconomic effects, family structure, father-specific survival traits, and maternal effects, including quality of paternal marriages, cannot explain our findings. While we cannot rule out fully psychological or cultural effects, our findings are most consistent with an epigenetic explanation.
Collapse
|
42
|
Leak RK, Calabrese EJ, Kozumbo WJ, Gidday JM, Johnson TE, Mitchell JR, Ozaki CK, Wetzker R, Bast A, Belz RG, Bøtker HE, Koch S, Mattson MP, Simon RP, Jirtle RL, Andersen ME. Enhancing and Extending Biological Performance and Resilience. Dose Response 2018; 16:1559325818784501. [PMID: 30140178 PMCID: PMC6096685 DOI: 10.1177/1559325818784501] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 05/15/2018] [Indexed: 12/17/2022] Open
Abstract
Human performance, endurance, and resilience have biological limits that are genetically and epigenetically predetermined but perhaps not yet optimized. There are few systematic, rigorous studies on how to raise these limits and reach the true maxima. Achieving this goal might accelerate translation of the theoretical concepts of conditioning, hormesis, and stress adaptation into technological advancements. In 2017, an Air Force-sponsored conference was held at the University of Massachusetts for discipline experts to display data showing that the amplitude and duration of biological performance might be magnified and to discuss whether there might be harmful consequences of exceeding typical maxima. The charge of the workshop was "to examine and discuss and, if possible, recommend approaches to control and exploit endogenous defense mechanisms to enhance the structure and function of biological tissues." The goal of this white paper is to fulfill and extend this workshop charge. First, a few of the established methods to exploit endogenous defense mechanisms are described, based on workshop presentations. Next, the white paper accomplishes the following goals to provide: (1) synthesis and critical analysis of concepts across some of the published work on endogenous defenses, (2) generation of new ideas on augmenting biological performance and resilience, and (3) specific recommendations for researchers to not only examine a wider range of stimulus doses but to also systematically modify the temporal dimension in stimulus inputs (timing, number, frequency, and duration of exposures) and in measurement outputs (interval until assay end point, and lifespan). Thus, a path forward is proposed for researchers hoping to optimize protocols that support human health and longevity, whether in civilians, soldiers, athletes, or the elderly patients. The long-term goal of these specific recommendations is to accelerate the discovery of practical methods to conquer what were once considered intractable constraints on performance maxima.
Collapse
Affiliation(s)
- Rehana K. Leak
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Edward J. Calabrese
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
| | | | - Jeffrey M. Gidday
- Departments of Ophthalmology, Neuroscience, and Physiology, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Thomas E. Johnson
- Department of Integrative Physiology, University of Colorado, Boulder, CO, USA
| | - James R. Mitchell
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - C. Keith Ozaki
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Reinhard Wetzker
- Institute for Molecular Cell Biology, University of Jena, Jena, Germany
| | - Aalt Bast
- Department of Pharmacology and Toxicology, Maastricht University, Maastricht, The Netherlands
| | - Regina G. Belz
- Hans-Ruthenberg-Institute, Agroecology Unit, University of Hohenheim, Stuttgart, Germany
| | - Hans E. Bøtker
- Department of Clinical Medicine, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Sebastian Koch
- Department of Neurology, University of Miami, Miller School of Medicine, FL, USA
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | - Roger P. Simon
- Departments of Medicine and Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Randy L. Jirtle
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | | |
Collapse
|
43
|
Vaiserman A, Koliada A, Zabuga O, Socol Y. Health Impacts of Low-Dose Ionizing Radiation: Current Scientific Debates and Regulatory Issues. Dose Response 2018; 16:1559325818796331. [PMID: 30263019 PMCID: PMC6149023 DOI: 10.1177/1559325818796331] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/31/2022] Open
Abstract
Health impacts of low-dose ionizing radiation are significant in important fields such as X-ray imaging, radiation therapy, nuclear power, and others. However, all existing and potential applications are currently challenged by public concerns and regulatory restrictions. We aimed to assess the validity of the linear no-threshold (LNT) model of radiation damage, which is the basis of current regulation, and to assess the justification for this regulation. We have conducted an extensive search in PubMed. Special attention has been given to papers cited in comprehensive reviews of the United States (2006) and French (2005) Academies of Sciences and in the United Nations Scientific Committee on Atomic Radiation 2016 report. Epidemiological data provide essentially no evidence for detrimental health effects below 100 mSv, and several studies suggest beneficial (hormetic) effects. Equally significant, many studies with in vitro and in animal models demonstrate that several mechanisms initiated by low-dose radiation have beneficial effects. Overall, although probably not yet proven to be untrue, LNT has certainly not been proven to be true. At this point, taking into account the high price tag (in both economic and human terms) borne by the LNT-inspired regulation, there is little doubt that the present regulatory burden should be reduced.
Collapse
|
44
|
Adam-Guillermin C, Hertal-Aas T, Oughton D, Blanchard L, Alonzo F, Armant O, Horemans N. Radiosensitivity and transgenerational effects in non-human species. Ann ICRP 2018; 47:327-341. [PMID: 29745724 DOI: 10.1177/0146645318756844] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The ALLIANCE working group on effects of ionising radiation on wildlife brings together European researchers to work on the topics of radiosensitivity and transgenerational effects in non-human biota. Differences in radiation sensitivity across species and phyla are poorly understood, but have important implications for understanding the overall effects of radiation and for radiation protection; for example, sensitive species may require special attention in monitoring and radiation protection, and differences in sensitivity between species also lead to overall effects at higher levels (community, ecosystem), since interactions between species can be altered. Hence, understanding the mechanisms of interspecies radiation sensitivity differences may help to clarify mechanisms underpinning intraspecies variation. Differences in sensitivity may only be revealed when organisms are exposed to ionising radiation over several generations. This issue of potential long-term or hereditary effects for both humans and wildlife exposed to low doses of ionising radiation is a major concern. Animal and plant studies suggest that gamma irradiation can lead to observable effects in the F1 generation that are not attributable to inheritance of a rare stable DNA mutation. Several studies have provided evidence of an increase in genomic instability detected in germ or somatic cells of F1 organisms from exposed F0 organisms. This can lead to induced radiosensitivity, and can result in phenotypic effects or lead to reproductive effects and teratogenesis. In particular, studies have been conducted to understand the possible role of epigenetic modifications, such as DNA methylation, histone modifications, or expression of non-coding RNAs in radiosensitivity, as well as in adaptation effects. As such, research using biological models in which the relative contribution of genetic and epigenetic processes can be elucidated is highly valuable.
Collapse
Affiliation(s)
- C Adam-Guillermin
- a Institut de Radioprotection et de Sûreté Nucléaire, PSE-ENV/SRTE/LECO, Cadarache, Saint Paul Lez Durance, France
| | | | - D Oughton
- b Norwegian University of Life Sciences, Norway
| | - L Blanchard
- c Commissariat à l'énergie atomique et aux énergies alternatives, France.,d Centre national de la recherche scientifique, France.,e Aix-Marseille Université, France
| | - F Alonzo
- a Institut de Radioprotection et de Sûreté Nucléaire, PSE-ENV/SRTE/LECO, Cadarache, Saint Paul Lez Durance, France
| | - O Armant
- a Institut de Radioprotection et de Sûreté Nucléaire, PSE-ENV/SRTE/LECO, Cadarache, Saint Paul Lez Durance, France
| | - N Horemans
- f Belgian Nuclear Research Centre, Belgium
| |
Collapse
|
45
|
Fatehi D, Rostamzadeh A. LETTER TO THE EDITOR: POTENTIAL EFFECTS OF DIAGNOSTIC IRRADIATION FROM COMPUTED TOMOGRAPHY ON REPRODUCTIVE SYSTEM AND GENERAL PROTECTIVE STRATEGIES. RADIATION PROTECTION DOSIMETRY 2018; 179:196-197. [PMID: 29069495 DOI: 10.1093/rpd/ncx223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 09/27/2017] [Indexed: 06/07/2023]
Affiliation(s)
- Daryoush Fatehi
- Department of Medical Physics, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ayoob Rostamzadeh
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
46
|
Epigenetic-based hormesis and age-dependent altruism: Additions to the behavioural constellation of deprivation. Behav Brain Sci 2018; 40:e320. [PMID: 29342772 DOI: 10.1017/s0140525x17001194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We support Pepper and Nettle's (P&N's) hypothesised adaptive responses to deprivation. However, we argue that adaptive responses to stress shift with age. Specifically, present-oriented behaviours are adaptive for young people (e.g., in terms of mating and reproduction) but costly for older people in deprived communities who would benefit from investing in grandchildren. Epigenetic mechanisms may be responsible for age-related tactical shifts.
Collapse
|
47
|
Volkova PY, Geras'kin SA, Horemans N, Makarenko ES, Saenen E, Duarte GT, Nauts R, Bondarenko VS, Jacobs G, Voorspoels S, Kudin M. Chronic radiation exposure as an ecological factor: Hypermethylation and genetic differentiation in irradiated Scots pine populations. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 232:105-112. [PMID: 28931465 DOI: 10.1016/j.envpol.2017.08.123] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/14/2017] [Accepted: 08/24/2017] [Indexed: 05/18/2023]
Abstract
Genetic and epigenetic changes were investigated in chronically irradiated Scots pine (Pinus sylvestris L.) populations from territories that were heavily contaminated by radionuclides as result of the Chernobyl Nuclear Power Plant accident. In comparison to the reference site, the genetic diversity revealed by electrophoretic mobility of AFLPs was found to be significantly higher at the radioactively contaminated areas. In addition, the genome of pine trees was significantly hypermethylated at 4 of the 7 affected sites.
Collapse
Affiliation(s)
- P Yu Volkova
- Institute of Radiology and Agroecology, 249030, Obninsk, Russian Federation.
| | - S A Geras'kin
- Institute of Radiology and Agroecology, 249030, Obninsk, Russian Federation
| | - N Horemans
- Belgian Nuclear Research Centre SCK•CEN, Biosphere Impact Studies, Boeretang 200, 2400, Mol, Belgium
| | - E S Makarenko
- Institute of Radiology and Agroecology, 249030, Obninsk, Russian Federation
| | - E Saenen
- Belgian Nuclear Research Centre SCK•CEN, Biosphere Impact Studies, Boeretang 200, 2400, Mol, Belgium
| | - G T Duarte
- Institute of Radiology and Agroecology, 249030, Obninsk, Russian Federation
| | - R Nauts
- Belgian Nuclear Research Centre SCK•CEN, Biosphere Impact Studies, Boeretang 200, 2400, Mol, Belgium
| | - V S Bondarenko
- Institute of Radiology and Agroecology, 249030, Obninsk, Russian Federation
| | - G Jacobs
- Flemish Institute for Technological Research (VITO NV), Boeretang 200, 2400 Mol, Belgium
| | - S Voorspoels
- Flemish Institute for Technological Research (VITO NV), Boeretang 200, 2400 Mol, Belgium
| | - M Kudin
- Polessye State Radiation Ecological Reserve, 247618, Belarus
| |
Collapse
|
48
|
Schofield PN, Kondratowicz M. Evolving paradigms for the biological response to low dose ionizing radiation; the role of epigenetics. Int J Radiat Biol 2017; 94:769-781. [PMID: 29157078 DOI: 10.1080/09553002.2017.1388548] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE In the late 1990s, it had become clear that the long-standing paradigm for the action of radiation on living cells and organisms did not have sufficient power to explain the observed effects of low dose ionizing radiation. The purpose of this commentary is to examine the experiments that lead up to the modification of the classic paradigm consequent on these observations, their historical precedents, and the development of our understanding of the role of epigenetics in low dose radiation effects. RESULTS AND CONCLUSIONS We discuss how parallel advances in epigenetics from developmental biology and cancer studies, and the discovery of epigenetic modifications of chromatin, such as DNA methylation, impacted on the development of an epigenetic paradigm for low dose effects. We also assess the impact of technology development in supporting the paradigm shift. We then examine recent accumulated data on epigenetic modification in response to irradiation since that shift took place, and identify areas where bringing together data from developmental biology and cancer might answer some of the paradoxes and contradictions in this data. We predict that further paradigm shifts are imminent.
Collapse
Affiliation(s)
- Paul N Schofield
- a Department of Physiology, Development, and Neuroscience , University of Cambridge , Cambridge , UK
| | - Monika Kondratowicz
- a Department of Physiology, Development, and Neuroscience , University of Cambridge , Cambridge , UK
| |
Collapse
|
49
|
Paunesku T, Haley B, Brooks A, Woloschak GE. Biological basis of radiation protection needs rejuvenation. Int J Radiat Biol 2017; 93:1056-1063. [PMID: 28287035 PMCID: PMC7340141 DOI: 10.1080/09553002.2017.1294773] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/04/2017] [Accepted: 02/09/2017] [Indexed: 12/30/2022]
Abstract
PURPOSE Human beings encounter radiation in many different situations - from proximity to radioactive waste sites to participation in medical procedures using X-rays etc. Limits for radiation exposures are legally regulated; however, current radiation protection policy does not explicitly acknowledge that biological, cellular and molecular effects of low doses and low dose rates of radiation differ from effects induced by medium and high dose radiation exposures. Recent technical developments in biology and medicine, from single cell techniques to big data computational research, have enabled new approaches for study of biology of low doses of radiation. Results of the work done so far support the idea that low doses of radiation have effects that differ from those associated with high dose exposures; this work, however, is far from sufficient for the development of a new theoretical framework needed for the understanding of low dose radiation exposures. CONCLUSIONS Mechanistic understanding of radiation effects at low doses is necessary in order to develop better radiation protection policy.
Collapse
Affiliation(s)
- Tatjana Paunesku
- a Department of Radiation Oncology , Northwestern University , Chicago , IL , USA
| | - Benjamin Haley
- a Department of Radiation Oncology , Northwestern University , Chicago , IL , USA
| | - Antone Brooks
- a Department of Radiation Oncology , Northwestern University , Chicago , IL , USA
| | - Gayle E Woloschak
- a Department of Radiation Oncology , Northwestern University , Chicago , IL , USA
| |
Collapse
|
50
|
Kim M, Na H, Kasai H, Kawai K, Li YS, Yang M. Comparison of Blueberry ( Vaccinium spp.) and Vitamin C via Antioxidative and Epigenetic Effects in Human. J Cancer Prev 2017; 22:174-181. [PMID: 29018782 PMCID: PMC5624458 DOI: 10.15430/jcp.2017.22.3.174] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 09/11/2017] [Indexed: 01/06/2023] Open
Abstract
Background Chemopreventive effects and the underlying mechanisms of blueberry (Vaccinium spp.) are not clearly understood in human. We hypothesized blueberry would work via antioxidative and epigenetic modulation, which is similar to vitamin C. Methods We performed a pilot and non-inferiority study in healthy young women (n = 12), who consumed vitamin C (1 g/d) or 240 mL of blueberry juice (total polyphenols 300 mg and proanthocyanidin 76 mg/d) for 2 weeks. We analyzed 8-hydroxydeoxyguanosine (8-OHdG) and malondialdehyde (MDA) levels in their urine, and global and specific DNA methylation at the NAD(P)H quinone oxidoreductase 1 (NQO1), methylenetetrahydrofolate reductase (MTHFR), or DNA methyltransferase 1 (DNMT1) genes in their blood. Results Urinary 8-OHdG levels were reduced by blueberry consumption rather than by vitamin C. The methylation (%) of the MTHFR was significantly decreased in blueberry-consumers and the antioxidant-susceptible subgroup, whose urinary MDA levels were decreased by the intervention. We also found a positive correlation between changes of urinary 8-OHdG and of DNA methylation at the MTHFR or the DNMT1 (P < 0.05). However, the genetic polymorphism of the MTHFR (C677T in exon 4) did not affect any above markers. Conclusions Blueberry juice shows similar anti-oxidative or anti-premutagenic activity to vitamin C and the potential as a methylation inhibitor for the MTHFR and the DNMT1 in human.
Collapse
Affiliation(s)
- Minju Kim
- Research Center for Cell Fate Control and College of Pharmacy, Sookmyung Women's University, Seoul, Korea
| | - Hyunkyung Na
- Research Center for Cell Fate Control and College of Pharmacy, Sookmyung Women's University, Seoul, Korea
| | - Hiroshi Kasai
- Department of Environmental Oncology and Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazuaki Kawai
- Department of Environmental Oncology and Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yun-Shan Li
- Department of Environmental Oncology and Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Mihi Yang
- Research Center for Cell Fate Control and College of Pharmacy, Sookmyung Women's University, Seoul, Korea
| |
Collapse
|