1
|
Abe JI, Allen BG, Beyer AM, Lewandowski D, Mapuskar KA, Subramanian V, Tamplin MR, Grumbach IM. Radiation-Induced Macrovessel/Microvessel Disease. Arterioscler Thromb Vasc Biol 2024. [PMID: 39445428 DOI: 10.1161/atvbaha.124.319866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Radiation therapy (RT) is a cornerstone in cancer treatment (used in 50% of cases), yet challenges persist because damage to normal tissue through direct impact of radiation or bystander effects is inevitable. Injury of macrovessels by RT manifests as obstructive disease, which is akin to atherosclerotic disease. Historically observed in coronary arteries of patients treated for breast cancer and lymphoma, it also affects patients receiving contemporary therapy for lung and chest cancers. Moreover, radiation at various sites can lead to peripheral vascular disease. An aspect of radiation-induced injury that has received little attention is microvascular injury, which typically results from damage to the endothelium and is considered the primary driver of RT-induced toxicity in the skin, kidney, and brain. This review delves into the clinical manifestations of RT-induced vascular disease, signaling pathways, cellular targets affected by radiation injury, and preclinical models of RT-induced vascular injury. The goal is to inspire the development of innovative strategies to prevent RT-related cardiovascular disease.
Collapse
Affiliation(s)
- Jun-Ichi Abe
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (J.-I.A.)
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine, University of Iowa. (B.G.A., K.A.M., I.M.G.)
| | - Andreas M Beyer
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee. (A.M.B.)
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee. (A.M.B.)
| | - David Lewandowski
- Department of Medicine, Medical College of Wisconsin, Milwaukee. (D.L.)
| | - Kranti A Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine, University of Iowa. (B.G.A., K.A.M., I.M.G.)
| | - Vikram Subramanian
- Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa. (V.S., M.R.T., I.M.G.)
| | - Michelle R Tamplin
- Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa. (V.S., M.R.T., I.M.G.)
| | - Isabella M Grumbach
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine, University of Iowa. (B.G.A., K.A.M., I.M.G.)
- Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa. (V.S., M.R.T., I.M.G.)
- Iowa City VA Healthcare System, Medical College of Wisconsin, Milwaukee. (I.M.G.)
| |
Collapse
|
2
|
Kozbenko T, Adam N, Grybas VS, Smith BJ, Alomar D, Hocking R, Abdelaziz J, Pace A, Boerma M, Azimzadeh O, Blattnig S, Hamada N, Yauk C, Wilkins R, Chauhan V. AOP report: Development of an adverse outcome pathway for deposition of energy leading to abnormal vascular remodeling. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65 Suppl 3:4-30. [PMID: 39440813 DOI: 10.1002/em.22636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Cardiovascular diseases (CVDs) are complex, encompassing many types of heart pathophysiologies and associated etiologies. Radiotherapy studies have shown that fractionated radiation exposure at high doses (3-17 Gy) to the heart increases the incidence of CVD. However, the effects of low doses of radiation on the cardiovascular system or the effects from space travel, where radiation and microgravity are important contributors to damage, are not clearly understood. Herein, the adverse outcome pathway (AOP) framework was applied to develop an AOP to abnormal vascular remodeling from the deposition of energy. Following the creation of a preliminary pathway with the guidance of field experts and authoritative reviews, a scoping review was conducted that informed final key event (KE) selection and evaluation of the Bradford Hill criteria for the KE relationships (KERs). The AOP begins with a molecular initiating event of deposition of energy; ionization events increase oxidative stress, which when persistent concurrently causes the release of pro-inflammatory mediators, suppresses anti-inflammatory mechanisms and alters stress response signaling pathways. These KEs alter nitric oxide levels leading to endothelial dysfunction, and subsequent abnormal vascular remodeling (the adverse outcome). The work identifies evidence needed to strengthen understanding of the causal associations for the KERs, emphasizing where there are knowledge gaps and uncertainties in both qualitative and quantitative understanding. The AOP is anticipated to direct future research to better understand the effects of space on the human body and potentially develop countermeasures to better protect future space travelers.
Collapse
Affiliation(s)
- Tatiana Kozbenko
- Health Canada, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | - Amanda Pace
- Carleton University, Ottawa, Ontario, Canada
| | - Marjan Boerma
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Omid Azimzadeh
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, Neuherberg, Germany
| | | | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| | - Carole Yauk
- University of Ottawa, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
3
|
Rahman MA, Islam MM, Ripon MAR, Islam MM, Hossain MS. Regulatory Roles of MicroRNAs in the Pathogenesis of Metabolic Syndrome. Mol Biotechnol 2024; 66:1599-1620. [PMID: 37393414 DOI: 10.1007/s12033-023-00805-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/17/2023] [Indexed: 07/03/2023]
Abstract
Metabolic syndrome refers to a group of several disease conditions together with high glucose triglyceride levels, high blood pressure, lower high-density lipoprotein level, and large waist circumference. About 400 million people worldwide, one-third of the Euro-American population and 27% Chinese population over age 50 have it. microRNAs, an abundant novel class of endogenous small, non-coding RNAs in eukaryotic cells, act as negative controllers of gene expression by promoting either degradation/translational repression of target messenger RNA. More than 2000 microRNAs in the human genome have been identified and they are implicated in various biological & pathophysiological processes, including glucose homeostasis, inflammatory response, and angiogenesis. Destruction of microRNAs has a crucial role in the pathogenesis of obesity, cardiovascular disease, and diabetes. Recently the discovery of circulating microRNAs in human serum may help to promote metabolic crosstalk between organs and serves as a novel approach for the identification of various diseases, like Type 2 diabetes & atherosclerosis. In this review, we will discuss the most recent and up-to-date research on the pathophysiology and histopathology of metabolic syndrome besides their historical background and epidemiological highlight. As well as search the methodologies employed in this field of research and the potential role of microRNAs as novel biomarkers and therapeutic targets for metabolic syndrome in the human body. Furthermore, the significance of microRNAs in promising strategies, like stem cell therapy, which holds enormous promise for regenerative medicine in the treatment of metabolic disorders will also be discussed.
Collapse
Affiliation(s)
- Md Abdur Rahman
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Md Mahmodul Islam
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Md Abdur Rahman Ripon
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Md Monirul Islam
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Mohammad Salim Hossain
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh.
- Bangladesh Obesity Research Network (BORN), Noakhali, 3814, Bangladesh.
| |
Collapse
|
4
|
Zablotska LB, Little MP, Hamada N. Revisiting an Inverse Dose-Fractionation Effect of Ionizing Radiation Exposure for Ischemic Heart Disease: Insights from Recent Studies. Radiat Res 2024; 202:80-86. [PMID: 38772552 PMCID: PMC11260496 DOI: 10.1667/rade-00230.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/26/2024] [Indexed: 05/23/2024]
Abstract
Over the last two decades, there has been emerging evidence suggesting that ionizing radiation exposures could be associated with elevated risks of cardiovascular disease (CVD), particularly ischemic heart disease (IHD). Excess CVD risks have been observed in a number of exposed groups, with generally similar risk estimates both at low and high radiation doses and dose rates. In 2014, we reported for the first time significantly higher risks of IHD mortality when radiation doses were delivered over a protracted period of time (an inverse dose-fractionation effect) in the Canadian Fluoroscopy Cohort Study. Here we review the current evidence on the dose-fractionation effect of radiation exposure, discuss potential implication for radiation protection policies and suggest further directions for research in this area.
Collapse
Affiliation(s)
- Lydia B Zablotska
- Department of Epidemiology & Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, California
| | - Mark P Little
- Radiation Epidemiology Branch, National Cancer Institute, Bethesda, Maryland; Faculty of Health and Life Sciences, Oxford Brookes University, Headington Campus, Oxford, United Kingdom
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| |
Collapse
|
5
|
Picano E, Vano E. Updated Estimates of Radiation Risk for Cancer and Cardiovascular Disease: Implications for Cardiology Practice. J Clin Med 2024; 13:2066. [PMID: 38610831 PMCID: PMC11012972 DOI: 10.3390/jcm13072066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
This review aims to furnish an updated assessment of the societal healthcare load, including cancer and cardiovascular disease resulting from diagnostic radiologic operations. The previously projected additional cancer risk of 0.9% in a United States 2004 study referred to radiological conditions in 1996 with an X-ray exposure of 0.50 millisievert (mSv) per capita annually. Radiological exposure (radiology + nuclear medicine) has escalated to 2.29 mSv (2016) per capita per year. Low-dose exposures were previously assumed to have a lower biological impact, since they allow the DNA repair system to mitigate molecular damage. However, epidemiological data matured and disproved this assumption, as shown by updated cancer risk assessments derived from the World Health Organization 2013 and the German Institute of Radioprotection 2014 data. The risk of cardiovascular disease aligns within the same order of magnitude as cancer risk and compounds it, as shown by a comprehensive meta-analysis of 93 studies. The collective societal burden arising from the augmented risks of cancer and cardiovascular disease attributable to diagnostic radiology and nuclear medicine is higher than previously thought.
Collapse
Affiliation(s)
- Eugenio Picano
- Cardiology Clinic, University Center Serbia, Medical School, University of Belgrade, 11000 Belgrade, Serbia
| | - Eliseo Vano
- Cardiology Department, Medical Faculty, Complutense University, 28040 Madrid, Spain;
| |
Collapse
|
6
|
Ibragimova M, Kussainova A, Aripova A, Bersimbaev R, Bulgakova O. The Molecular Mechanisms in Senescent Cells Induced by Natural Aging and Ionizing Radiation. Cells 2024; 13:550. [PMID: 38534394 DOI: 10.3390/cells13060550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
This review discusses the relationship between cellular senescence and radiation exposure. Given the wide range of ionizing radiation sources encountered by people in professional and medical spheres, as well as the influence of natural background radiation, the question of the effect of radiation on biological processes, particularly on aging processes, remains highly relevant. The parallel relationship between natural and radiation-induced cellular senescence reveals the common aspects underlying these processes. Based on recent scientific data, the key points of the effects of ionizing radiation on cellular processes associated with aging, such as genome instability, mitochondrial dysfunction, altered expression of miRNAs, epigenetic profile, and manifestation of the senescence-associated secretory phenotype (SASP), are discussed. Unraveling the molecular mechanisms of cellular senescence can make a valuable contribution to the understanding of the molecular genetic basis of age-associated diseases in the context of environmental exposure.
Collapse
Affiliation(s)
- Milana Ibragimova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Assiya Kussainova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
- Department of Health Sciences, University of Genova, Via Pastore 1, 16132 Genoa, Italy
| | - Akmaral Aripova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Rakhmetkazhi Bersimbaev
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Olga Bulgakova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| |
Collapse
|
7
|
Tavakol DN, Nash TR, Kim Y, He S, Fleischer S, Graney PL, Brown JA, Liberman M, Tamargo M, Harken A, Ferrando AA, Amundson S, Garty G, Azizi E, Leong KW, Brenner DJ, Vunjak-Novakovic G. Modeling and countering the effects of cosmic radiation using bioengineered human tissues. Biomaterials 2023; 301:122267. [PMID: 37633022 PMCID: PMC10528250 DOI: 10.1016/j.biomaterials.2023.122267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/28/2023]
Abstract
Cosmic radiation is the most serious risk that will be encountered during the planned missions to the Moon and Mars. There is a compelling need to understand the effects, safety thresholds, and mechanisms of radiation damage in human tissues, in order to develop measures for radiation protection during extended space travel. As animal models fail to recapitulate the molecular changes in astronauts, engineered human tissues and "organs-on-chips" are valuable tools for studying effects of radiation in vitro. We have developed a bioengineered tissue platform for studying radiation damage in individualized settings. To demonstrate its utility, we determined the effects of radiation using engineered models of two human tissues known to be radiosensitive: engineered cardiac tissues (eCT, a target of chronic radiation damage) and engineered bone marrow (eBM, a target of acute radiation damage). We report the effects of high-dose neutrons, a proxy for simulated galactic cosmic rays, on the expression of key genes implicated in tissue responses to ionizing radiation, phenotypic and functional changes in both tissues, and proof-of-principle application of radioprotective agents. We further determined the extent of inflammatory, oxidative stress, and matrix remodeling gene expression changes, and found that these changes were associated with an early hypertrophic phenotype in eCT and myeloid skewing in eBM. We propose that individualized models of human tissues have potential to provide insights into the effects and mechanisms of radiation during deep-space missions and allow testing of radioprotective measures.
Collapse
Affiliation(s)
| | - Trevor R Nash
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Youngbin Kim
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Siyu He
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Pamela L Graney
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Jessie A Brown
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Martin Liberman
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Manuel Tamargo
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Andrew Harken
- Center for Radiological Research, Columbia University, New York, NY 10032, USA
| | - Adolfo A Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Sally Amundson
- Center for Radiological Research, Columbia University, New York, NY 10032, USA
| | - Guy Garty
- Center for Radiological Research, Columbia University, New York, NY 10032, USA
| | - Elham Azizi
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - David J Brenner
- Center for Radiological Research, Columbia University, New York, NY 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA; Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA; Department of Medicine, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
8
|
Blutt SE, Coarfa C, Neu J, Pammi M. Multiomic Investigations into Lung Health and Disease. Microorganisms 2023; 11:2116. [PMID: 37630676 PMCID: PMC10459661 DOI: 10.3390/microorganisms11082116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Diseases of the lung account for more than 5 million deaths worldwide and are a healthcare burden. Improving clinical outcomes, including mortality and quality of life, involves a holistic understanding of the disease, which can be provided by the integration of lung multi-omics data. An enhanced understanding of comprehensive multiomic datasets provides opportunities to leverage those datasets to inform the treatment and prevention of lung diseases by classifying severity, prognostication, and discovery of biomarkers. The main objective of this review is to summarize the use of multiomics investigations in lung disease, including multiomics integration and the use of machine learning computational methods. This review also discusses lung disease models, including animal models, organoids, and single-cell lines, to study multiomics in lung health and disease. We provide examples of lung diseases where multi-omics investigations have provided deeper insight into etiopathogenesis and have resulted in improved preventative and therapeutic interventions.
Collapse
Affiliation(s)
- Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Josef Neu
- Department of Pediatrics, Section of Neonatology, University of Florida, Gainesville, FL 32611, USA;
| | - Mohan Pammi
- Department of Pediatrics, Section of Neonatology, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX 77030, USA
| |
Collapse
|
9
|
Azimzadeh O, Merl-Pham J, Subramanian V, Oleksenko K, Krumm F, Mancuso M, Pasquali E, Tanaka IB, Tanaka S, Atkinson MJ, Tapio S, Moertl S. Late Effects of Chronic Low Dose Rate Total Body Irradiation on the Heart Proteome of ApoE -/- Mice Resemble Premature Cardiac Ageing. Cancers (Basel) 2023; 15:3417. [PMID: 37444528 DOI: 10.3390/cancers15133417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Recent epidemiologic studies support an association between chronic low-dose radiation exposure and the development of cardiovascular disease (CVD). The molecular mechanisms underlying the adverse effect of chronic low dose exposure are not fully understood. To address this issue, we have investigated changes in the heart proteome of ApoE deficient (ApoE-/-) C57Bl/6 female mice chronically irradiated for 300 days at a very low dose rate (1 mGy/day) or at a low dose rate (20 mGy/day), resulting in cumulative whole-body doses of 0.3 Gy or 6.0 Gy, respectively. The heart proteomes were compared to those of age-matched sham-irradiated ApoE-/- mice using label-free quantitative proteomics. Radiation-induced proteome changes were further validated using immunoblotting, enzyme activity assays, immunohistochemistry or targeted transcriptomics. The analyses showed persistent alterations in the cardiac proteome at both dose rates; however, the effect was more pronounced following higher dose rates. The altered proteins were involved in cardiac energy metabolism, ECM remodelling, oxidative stress, and ageing signalling pathways. The changes in PPARα, SIRT, AMPK, and mTOR signalling pathways were found at both dose rates and in a dose-dependent manner, whereas more changes in glycolysis and ECM remodelling were detected at the lower dose rate. These data provide strong evidence for the possible risk of cardiac injury following chronic low dose irradiation and show that several affected pathways following chronic irradiation overlap with those of ageing-associated heart pathology.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Section of Radiation Biology, Federal Office of Radiation Protection (BfS), 85764 Nauenberg, Germany
| | - Juliane Merl-Pham
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Munich, Germany
| | - Vikram Subramanian
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kateryna Oleksenko
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Franziska Krumm
- Section of Radiation Biology, Federal Office of Radiation Protection (BfS), 85764 Nauenberg, Germany
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00196 Rome, Italy
| | - Emanuela Pasquali
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00196 Rome, Italy
| | - Ignacia B Tanaka
- Institute for Environmental Sciences (IES), Rokkasho, Aomori 039-3212, Japan
| | - Satoshi Tanaka
- Institute for Environmental Sciences (IES), Rokkasho, Aomori 039-3212, Japan
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- Radiation Oncology, Klinikum rechts der Isar, Technical University, 80333 Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Simone Moertl
- Section of Radiation Biology, Federal Office of Radiation Protection (BfS), 85764 Nauenberg, Germany
| |
Collapse
|
10
|
Little MP, Azizova TV, Richardson DB, Tapio S, Bernier MO, Kreuzer M, Cucinotta FA, Bazyka D, Chumak V, Ivanov VK, Veiga LHS, Livinski A, Abalo K, Zablotska LB, Einstein AJ, Hamada N. Ionising radiation and cardiovascular disease: systematic review and meta-analysis. BMJ 2023; 380:e072924. [PMID: 36889791 PMCID: PMC10535030 DOI: 10.1136/bmj-2022-072924] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2023] [Indexed: 03/10/2023]
Abstract
OBJECTIVE To systematically review and perform a meta-analysis of radiation associated risks of cardiovascular disease in all groups exposed to radiation with individual radiation dose estimates. DESIGN Systematic review and meta-analysis. MAIN OUTCOME MEASURES Excess relative risk per unit dose (Gy), estimated by restricted maximum likelihood methods. DATA SOURCES PubMed and Medline, Embase, Scopus, Web of Science Core collection databases. ELIGIBILITY CRITERIA FOR SELECTING STUDIES Databases were searched on 6 October 2022, with no limits on date of publication or language. Animal studies and studies without an abstract were excluded. RESULTS The meta-analysis yielded 93 relevant studies. Relative risk per Gy increased for all cardiovascular disease (excess relative risk per Gy of 0.11 (95% confidence interval 0.08 to 0.14)) and for the four major subtypes of cardiovascular disease (ischaemic heart disease, other heart disease, cerebrovascular disease, all other cardiovascular disease). However, interstudy heterogeneity was noted (P<0.05 for all endpoints except for other heart disease), possibly resulting from interstudy variation in unmeasured confounders or effect modifiers, which is markedly reduced if attention is restricted to higher quality studies or those at moderate doses (<0.5 Gy) or low dose rates (<5 mGy/h). For ischaemic heart disease and all cardiovascular disease, risks were larger per unit dose for lower dose (inverse dose effect) and for fractionated exposures (inverse dose fractionation effect). Population based excess absolute risks are estimated for a number of national populations (Canada, England and Wales, France, Germany, Japan, USA) and range from 2.33% per Gy (95% confidence interval 1.69% to 2.98%) for England and Wales to 3.66% per Gy (2.65% to 4.68%) for Germany, largely reflecting the underlying rates of cardiovascular disease mortality in these populations. Estimated risk of mortality from cardiovascular disease are generally dominated by cerebrovascular disease (around 0.94-1.26% per Gy), with the next largest contribution from ischaemic heart disease (around 0.30-1.20% per Gy). CONCLUSIONS Results provide evidence supporting a causal association between radiation exposure and cardiovascular disease at high dose, and to a lesser extent at low dose, with some indications of differences in risk between acute and chronic exposures, which require further investigation. The observed heterogeneity complicates a causal interpretation of these findings, although this heterogeneity is much reduced if only higher quality studies or those at moderate doses or low dose rates are considered. Studies are needed to assess in more detail modifications of radiation effect by lifestyle and medical risk factors. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42020202036.
Collapse
Affiliation(s)
- Mark P Little
- Radiation Epidemiology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Tamara V Azizova
- Clinical Department, Southern Urals Biophysics Institute, Ozyorsk, Chelyabinsk Region, Russia
| | - David B Richardson
- Department of Environmental and Occupational Health, Irvine Program in Public Health, University of California Irvine, Irvine, CA, USA
| | - Soile Tapio
- Technische Universität München, Munich, Germany
| | - Marie-Odile Bernier
- Institut de Radioprotection et de Sureté Nucléaire, Fontenay aux Roses, France
| | | | - Francis A Cucinotta
- Department of Health Physics and Diagnostic Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Dimitry Bazyka
- National Research Center for Radiation Medicine, National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
| | - Vadim Chumak
- National Research Center for Radiation Medicine, National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
| | - Victor K Ivanov
- Medical Radiological Research Center of Russian Academy of Medical Sciences, Obninsk, Russia
| | - Lene H S Veiga
- Radiation Epidemiology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Alicia Livinski
- National Institutes of Health Library, National Institutes of Health, Bethesda, MD, USA
| | - Kossi Abalo
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Department of Immunology Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | - Lydia B Zablotska
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew J Einstein
- Seymour, Paul, and Gloria Milstein Division of Cardiology, Department of Medicine, and Department of Radiology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Komae, Tokyo, Japan
| |
Collapse
|
11
|
Seara FAC, Maciel L, Kasai-Brunswick TH, Nascimento JHM, Campos-de-Carvalho AC. Extracellular Vesicles and Cardiac Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:33-56. [PMID: 37603271 DOI: 10.1007/978-981-99-1443-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Global population aging is a major challenge to health and socioeconomic policies. The prevalence of diseases progressively increases with aging, with cardiovascular disease being the major cause of mortality among elderly people. The allostatic overload imposed by the accumulation of cardiac senescent cells has been suggested to play a pivotal role in the aging-related deterioration of cardiovascular function. Senescent cells exhibit intrinsic disorders and release a senescence-associated secretory phenotype (SASP). Most of these SASP compounds and damaged molecules are released from senescent cells by extracellular vesicles (EVs). Once secreted, these EVs can be readily incorporated by recipient neighboring cells and elicit cellular damage or otherwise can promote extracellular matrix remodeling. This has been associated with the development of cardiac dysfunction, fibrosis, and vascular calcification, among others. The molecular signature of these EVs is highly variable and might provide important information for the development of aging-related biomarkers. Conversely, EVs released by the stem and progenitor cells can exert a rejuvenating effect, raising the possibility of future anti-aging therapies.
Collapse
Affiliation(s)
- Fernando A C Seara
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Physiological Sciences, Institute of Health and Biological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | - Leonardo Maciel
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Federal University of Rio de Janeiro, Campus Professor Geraldo, Duque de Caxias, Brazil
| | - Tais Hanae Kasai-Brunswick
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose H M Nascimento
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Health Sciences Centre, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Antonio C Campos-de-Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Yin B, Zhang X, Ren J, Chen F, Liang J, Zhang H, Pei H, Hu Z, Wang Y, Xue W, Yu X, Zhang R, Ma Y. The protective effects of procyanidin supplementation on PM 2.5-induced acute cardiac injury in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:10890-10900. [PMID: 36088442 DOI: 10.1007/s11356-022-22938-5] [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: 03/18/2022] [Accepted: 09/04/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE Numerous epidemiological and experimental studies have indicated that ambient fine particulate matter (PM2.5) exposure can lead to myocardial injury by inhibiting oxidative stress and apoptosis. The effects of procyanidin (PC) on PM2.5-induced cardiovascular diseases (CVDs) are still unknown. The purpose of this study was to explore the protective effect of PC supplementation on PM2.5-induced oxidative stress and cardiomyocyte apoptosis in rats. METHOD Rats were treated by gavage with three different PC concentrations (50, 100 and 200 mg/kg) for 21 days prior to exposure to 10 mg/kg PM2.5 suspension liquid by intratracheal instillation every other day for three times. We determined myocardial reactive oxygen species (ROS) and malondialdehyde (MDA) levels. Superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities in the myocardium were measured. The expression levels of apoptosis-related proteins, including p-Akt/Akt, Bcl-2, caspase-3 and Bax, were determined. In addition, histopathological examination was used to evaluate cardiac injury. RESULTS PM2.5 exposure noticeably elevated the contents of MDA and ROS and decreased the activities of GSH-Px and SOD. PM2.5 exposure inhibited Bcl-2 expression and up-regulated caspase-3 and Bax expression in the myocardium of rats. The anti-apoptosis-related index p-Akt/Akt was reduced. Moreover, pretreatment with PC could attenuate these PM2.5-induced changes. However, remarkable differences in the protective effect of different PC doses did not exist. CONCLUSIONS The results indicated that PC supplementation could effectively attenuate the oxidative stress and apoptosis induced by PM2.5 in rat myocardial tissue.
Collapse
Affiliation(s)
- Bowen Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiao Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, China
| | - Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Fengge Chen
- Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang, 050017, China
| | - Jufeng Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hanning Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Huanting Pei
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhaohui Hu
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yan Wang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Wei Xue
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xinyue Yu
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Rui Zhang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
13
|
Azimzadeh O, Moertl S, Ramadan R, Baselet B, Laiakis EC, Sebastian S, Beaton D, Hartikainen JM, Kaiser JC, Beheshti A, Salomaa S, Chauhan V, Hamada N. Application of radiation omics in the development of adverse outcome pathway networks: an example of radiation-induced cardiovascular disease. Int J Radiat Biol 2022; 98:1722-1751. [PMID: 35976069 DOI: 10.1080/09553002.2022.2110325] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Epidemiological studies have indicated that exposure of the heart to doses of ionizing radiation as low as 0.5 Gy increases the risk of cardiac morbidity and mortality with a latency period of decades. The damaging effects of radiation to myocardial and endothelial structures and functions have been confirmed radiobiologically at high dose, but much less is known at low dose. Integration of radiation biology and epidemiology data is a recommended approach to improve the radiation risk assessment process. The adverse outcome pathway (AOP) framework offers a comprehensive tool to compile and translate mechanistic information into pathological endpoints which may be relevant for risk assessment at the different levels of a biological system. Omics technologies enable the generation of large volumes of biological data at various levels of complexity, from molecular pathways to functional organisms. Given the quality and quantity of available data across levels of biology, omics data can be attractive sources of information for use within the AOP framework. It is anticipated that radiation omics studies could improve our understanding of the molecular mechanisms behind the adverse effects of radiation on the cardiovascular system. In this review, we explored the available omics studies on radiation-induced cardiovascular disease (CVD) and their applicability to the proposed AOP for CVD. RESULTS The results of 80 omics studies published on radiation-induced CVD over the past 20 years have been discussed in the context of the AOP of CVD proposed by Chauhan et al. Most of the available omics data on radiation-induced CVD are from proteomics, transcriptomics, and metabolomics, whereas few datasets were available from epigenomics and multi-omics. The omics data presented here show great promise in providing information for several key events of the proposed AOP of CVD, particularly oxidative stress, alterations of energy metabolism, extracellular matrix and vascular remodeling. CONCLUSIONS The omics data presented here shows promise to inform the various levels of the proposed AOP of CVD. However, the data highlight the urgent need of designing omics studies to address the knowledge gap concerning different radiation scenarios, time after exposure and experimental models. This review presents the evidence to build a qualitative omics-informed AOP and provides views on the potential benefits and challenges in using omics data to assess risk-related outcomes.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Simone Moertl
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Raghda Ramadan
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Bjorn Baselet
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Evagelia C Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | | | | | - Jaana M Hartikainen
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
| | - Jan Christian Kaiser
- Helmholtz Zentrum München, Institute of Radiation Medicine (HMGU-IRM), 85764 Neuherberg, Germany
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Sisko Salomaa
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vinita Chauhan
- Environmental Health Science Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Komae, Tokyo 201-8511, Japan
| |
Collapse
|
14
|
Walls GM, O'Kane R, Ghita M, Kuburas R, McGarry CK, Cole AJ, Jain S, Butterworth KT. Murine models of radiation cardiotoxicity: A systematic review and recommendations for future studies. Radiother Oncol 2022; 173:19-31. [PMID: 35533784 DOI: 10.1016/j.radonc.2022.04.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/13/2022] [Accepted: 04/29/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE The effects of radiation on the heart are dependent on dose, fractionation, overall treatment time, and pre-existing cardiovascular pathology. Murine models have played a central role in improving our understanding of the radiation response of the heart yet a wide range of exposure parameters have been used. We evaluated the study design of published murine cardiac irradiation experiments to assess gaps in the literature and to suggest guidance for the harmonisation of future study reporting. METHODS AND MATERIALS A systematic review of mouse/rat studies published 1981-2021 that examined the effect of radiation on the heart was performed. The protocol was published on PROSPERO (CRD42021238921) and the findings were reported in accordance with the PRISMA guidance. Risk of bias was assessed using the SYRCLE checklist. RESULTS 159 relevant full-text original articles were reviewed. The heart only was the target volume in 67% of the studies and simulation details were unavailable for 44% studies. Dosimetry methods were reported in 31% studies. The pulmonary effects of whole and partial heart irradiation were reported in 13% studies. Seventy-eight unique dose-fractionation schedules were evaluated. Large heterogeneity was observed in the endpoints measured, and the reporting standards were highly variable. CONCLUSIONS Current murine models of radiation cardiotoxicity cover a wide range of irradiation configurations and latency periods. There is a lack of evidence describing clinically relevant dose-fractionations, circulating biomarkers and radioprotectants. Recommendations for the consistent reporting of methods and results of in vivo cardiac irradiation studies are made to increase their suitability for informing the design of clinical studies.
Collapse
Affiliation(s)
- Gerard M Walls
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland; Cancer Centre Belfast City Hospital, Belfast Health & Social Care Trust, Lisburn Road, Belfast, Northern Ireland.
| | - Reagan O'Kane
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland
| | - Mihaela Ghita
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland
| | - Refik Kuburas
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland
| | - Conor K McGarry
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland; Cancer Centre Belfast City Hospital, Belfast Health & Social Care Trust, Lisburn Road, Belfast, Northern Ireland
| | - Aidan J Cole
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland; Cancer Centre Belfast City Hospital, Belfast Health & Social Care Trust, Lisburn Road, Belfast, Northern Ireland
| | - Suneil Jain
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland; Cancer Centre Belfast City Hospital, Belfast Health & Social Care Trust, Lisburn Road, Belfast, Northern Ireland
| | - Karl T Butterworth
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, Northern Ireland
| |
Collapse
|
15
|
Akh LA, Ishak MO, Harris JF, Glaros TG, Sasiene ZJ, Mach PM, Lilley LM, McBride EM. -Omics potential of in vitro skin models for radiation exposure. Cell Mol Life Sci 2022; 79:390. [PMID: 35776214 PMCID: PMC11073334 DOI: 10.1007/s00018-022-04394-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/12/2022] [Accepted: 05/24/2022] [Indexed: 11/12/2022]
Abstract
There is a growing need to uncover biomarkers of ionizing radiation exposure that leads to a better understanding of how exposures take place, including dose type, rate, and time since exposure. As one of the first organs to be exposed to external sources of ionizing radiation, skin is uniquely positioned in terms of model systems for radiation exposure study. The simultaneous evolution of both MS-based -omics studies, as well as in vitro 3D skin models, has created the ability to develop a far more holistic understanding of how ionizing radiation affects the many interconnected biomolecular processes that occur in human skin. However, there are a limited number of studies describing the biomolecular consequences of low-dose ionizing radiation to the skin. This review will seek to explore the current state-of-the-art technology in terms of in vitro 3D skin models, as well as track the trajectory of MS-based -omics techniques and their application to ionizing radiation research, specifically, the search for biomarkers within the low-dose range.
Collapse
Affiliation(s)
- Leyla A Akh
- Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Mohammad O Ishak
- Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Jennifer F Harris
- Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Trevor G Glaros
- Bioenergy and Biome Sciences Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Zachary J Sasiene
- Bioenergy and Biome Sciences Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Phillip M Mach
- Bioenergy and Biome Sciences Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Laura M Lilley
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA.
| | - Ethan M McBride
- Bioenergy and Biome Sciences Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA.
| |
Collapse
|
16
|
Iacobazzi D, Alvino VV, Caputo M, Madeddu P. Accelerated Cardiac Aging in Patients With Congenital Heart Disease. Front Cardiovasc Med 2022; 9:892861. [PMID: 35694664 PMCID: PMC9177956 DOI: 10.3389/fcvm.2022.892861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/11/2022] [Indexed: 01/03/2023] Open
Abstract
An increasing number of patients with congenital heart disease (CHD) survive into adulthood but develop long-term complications including heart failure (HF). Cellular senescence, classically defined as stable cell cycle arrest, is implicated in biological processes such as embryogenesis, wound healing, and aging. Senescent cells have a complex senescence-associated secretory phenotype (SASP), involving a range of pro-inflammatory factors with important paracrine and autocrine effects on cell and tissue biology. While senescence has been mainly considered as a cause of diseases in the adulthood, it may be also implicated in some of the poor outcomes seen in patients with complex CHD. We propose that patients with CHD suffer from multiple repeated stress from an early stage of the life, which wear out homeostatic mechanisms and cause premature cardiac aging, with this term referring to the time-related irreversible deterioration of the organ physiological functions and integrity. In this review article, we gathered evidence from the literature indicating that growing up with CHD leads to abnormal inflammatory response, loss of proteostasis, and precocious age in cardiac cells. Novel research on this topic may inspire new therapies preventing HF in adult CHD patients.
Collapse
Affiliation(s)
| | | | | | - Paolo Madeddu
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
17
|
Babini G, Baiocco G, Barbieri S, Morini J, Sangsuwan T, Haghdoost S, Yentrapalli R, Azimzadeh O, Rombouts C, Aerts A, Quintens R, Ebrahimian T, Benotmane MA, Ramadan R, Baatout S, Tapio S, Harms-Ringdahl M, Ottolenghi A. A systems radiation biology approach to unravel the role of chronic low-dose-rate gamma-irradiation in inducing premature senescence in endothelial cells. PLoS One 2022; 17:e0265281. [PMID: 35286349 PMCID: PMC8920222 DOI: 10.1371/journal.pone.0265281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/24/2022] [Indexed: 12/13/2022] Open
Abstract
Purpose The aim of this study was to explore the effects of chronic low-dose-rate gamma-radiation at a multi-scale level. The specific objective was to obtain an overall view of the endothelial cell response, by integrating previously published data on different cellular endpoints and highlighting possible different mechanisms underpinning radiation-induced senescence. Materials and methods Different datasets were collected regarding experiments on human umbilical vein endothelial cells (HUVECs) which were chronically exposed to low dose rates (0, 1.4, 2.1 and 4.1 mGy/h) of gamma-rays until cell replication was arrested. Such exposed cells were analyzed for different complementary endpoints at distinct time points (up to several weeks), investigating cellular functions such as proliferation, senescence and angiogenic properties, as well as using transcriptomics and proteomics profiling. A mathematical model was proposed to describe proliferation and senescence. Results Simultaneous ceasing of cell proliferation and senescence onset as a function of time were well reproduced by the logistic growth curve, conveying shared equilibria between the two endpoints. The combination of all the different endpoints investigated highlighted a dose-dependence for prematurely induced senescence. However, the underpinning molecular mechanisms appeared to be dissimilar for the different dose rates, thus suggesting a more complex scenario. Conclusions This study was conducted integrating different datasets, focusing on their temporal dynamics, and using a systems biology approach. Results of our analysis highlight that different dose rates have different effects in inducing premature senescence, and that the total cumulative absorbed dose also plays an important role in accelerating endothelial cell senescence.
Collapse
Affiliation(s)
| | | | - Sofia Barbieri
- Physics Department, University of Pavia, Pavia, Italy
- Faculty of Medicine, Department of Cellular Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Jacopo Morini
- Physics Department, University of Pavia, Pavia, Italy
| | - Traimate Sangsuwan
- Department of Molecular Bioscience, Centre for Radiation Protection Research, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Siamak Haghdoost
- Department of Molecular Bioscience, Centre for Radiation Protection Research, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- ARIA Laboratory, University of Caen Normandy, CIMAP-GANIL, 14076, Caen, France
| | - Ramesh Yentrapalli
- Institute of Radiation Biology, Helmholtz Zentrum Muenchen—German Research Centre for Environmental Health, Neuherberg, Germany
| | - Omid Azimzadeh
- Institute of Radiation Biology, Helmholtz Zentrum Muenchen—German Research Centre for Environmental Health, Neuherberg, Germany
- Section Radiation Biology, Federal Office for Radiation Protection, Munich, Germany
| | - Charlotte Rombouts
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK CEN, Boeretang, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - An Aerts
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK CEN, Boeretang, Belgium
| | - Roel Quintens
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK CEN, Boeretang, Belgium
| | - Teni Ebrahimian
- Laboratoire de Radiobiologie et RadioToxicologie expérimentale, Service de recherche des effets biologiques et sanitaires des rayonnements ionisants, Pôle santé, F-92262, Fontenay-aux-Roses, France
| | | | - Raghda Ramadan
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK CEN, Boeretang, Belgium
- * E-mail:
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK CEN, Boeretang, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum Muenchen—German Research Centre for Environmental Health, Neuherberg, Germany
| | - Mats Harms-Ringdahl
- Department of Molecular Bioscience, Centre for Radiation Protection Research, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | |
Collapse
|
18
|
Langston JC, Rossi MT, Yang Q, Ohley W, Perez E, Kilpatrick LE, Prabhakarpandian B, Kiani MF. Omics of endothelial cell dysfunction in sepsis. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2022; 4:R15-R34. [PMID: 35515704 PMCID: PMC9066943 DOI: 10.1530/vb-22-0003] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/07/2022] [Indexed: 12/19/2022]
Abstract
During sepsis, defined as life-threatening organ dysfunction due to dysregulated host response to infection, systemic inflammation activates endothelial cells and initiates a multifaceted cascade of pro-inflammatory signaling events, resulting in increased permeability and excessive recruitment of leukocytes. Vascular endothelial cells share many common properties but have organ-specific phenotypes with unique structure and function. Thus, therapies directed against endothelial cell phenotypes are needed to address organ-specific endothelial cell dysfunction. Omics allow for the study of expressed genes, proteins and/or metabolites in biological systems and provide insight on temporal and spatial evolution of signals during normal and diseased conditions. Proteomics quantifies protein expression, identifies protein-protein interactions and can reveal mechanistic changes in endothelial cells that would not be possible to study via reductionist methods alone. In this review, we provide an overview of how sepsis pathophysiology impacts omics with a focus on proteomic analysis of mouse endothelial cells during sepsis/inflammation and its relationship with the more clinically relevant omics of human endothelial cells. We discuss how omics has been used to define septic endotype signatures in different populations with a focus on proteomic analysis in organ-specific microvascular endothelial cells during sepsis or septic-like inflammation. We believe that studies defining septic endotypes based on proteomic expression in endothelial cell phenotypes are urgently needed to complement omic profiling of whole blood and better define sepsis subphenotypes. Lastly, we provide a discussion of how in silico modeling can be used to leverage the large volume of omics data to map response pathways in sepsis.
Collapse
Affiliation(s)
- Jordan C Langston
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania, USA
| | | | - Qingliang Yang
- Department of Mechanical Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | - William Ohley
- Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Edwin Perez
- Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Laurie E Kilpatrick
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology and Inflammation, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Balabhaskar Prabhakarpandian
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology and Inflammation, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Mohammad F Kiani
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania, USA
- Department of Mechanical Engineering, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
19
|
Morilla I, Chan P, Caffin F, Svilar L, Selbonne S, Ladaigue S, Buard V, Tarlet G, Micheau B, Paget V, François A, Souidi M, Martin JC, Vaudry D, Benadjaoud MA, Milliat F, Guipaud O. Deep models of integrated multiscale molecular data decipher the endothelial cell response to ionizing radiation. iScience 2022; 25:103685. [PMID: 35106469 PMCID: PMC8786676 DOI: 10.1016/j.isci.2021.103685] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/04/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
The vascular endothelium is a hot spot in the response to radiation therapy for both tumors and normal tissues. To improve patient outcomes, interpretable systemic hypotheses are needed to help radiobiologists and radiation oncologists propose endothelial targets that could protect normal tissues from the adverse effects of radiation therapy and/or enhance its antitumor potential. To this end, we captured the kinetics of multi-omics layers-i.e. miRNome, targeted transcriptome, proteome, and metabolome-in irradiated primary human endothelial cells cultured in vitro. We then designed a strategy of deep learning as in convolutional graph networks that facilitates unsupervised high-level feature extraction of important omics data to learn how ionizing radiation-induced endothelial dysfunction may evolve over time. Last, we present experimental data showing that some of the features identified using our approach are involved in the alteration of angiogenesis by ionizing radiation.
Collapse
Affiliation(s)
- Ian Morilla
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
- Corresponding author
| | - Philippe Chan
- Normandie Univ, UNIROUEN, PISSARO Proteomic Platform, 76821 Mont Saint-Aignan, France
| | - Fanny Caffin
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
| | - Ljubica Svilar
- Aix Marseille Univ, INSERM, INRA, C2VN, 13007 Marseille, France
- CriBioM, Criblage Biologique Marseille, Faculté de Médecine de la Timone, 13205 Marseille Cedex 01, France
| | - Sonia Selbonne
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
| | - Ségolène Ladaigue
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
- Sorbonne University, Doctoral College, 75005 Paris, France
| | - Valérie Buard
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
| | - Georges Tarlet
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
| | - Béatrice Micheau
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
| | - Vincent Paget
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
| | - Agnès François
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
| | - Maâmar Souidi
- IRSN, Radiobiology of Accidental Exposure Laboratory (LRAcc), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
| | - Jean-Charles Martin
- Aix Marseille Univ, INSERM, INRA, C2VN, 13007 Marseille, France
- CriBioM, Criblage Biologique Marseille, Faculté de Médecine de la Timone, 13205 Marseille Cedex 01, France
| | - David Vaudry
- Normandie Univ, UNIROUEN, PISSARO Proteomic Platform, 76821 Mont Saint-Aignan, France
| | - Mohamed-Amine Benadjaoud
- IRSN, Radiobiology and Regenerative Medicine Research Service (SERAMED), 92260 Fontenay-Aux-Roses, France
| | - Fabien Milliat
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
| | - Olivier Guipaud
- IRSN, Radiobiology of Medical Exposure Laboratory (LRMed), Human Health Radiation Protection Unit, 92260 Fontenay-Aux-Roses, France
- Corresponding author
| |
Collapse
|
20
|
Allen BD, Limoli CL. Breaking barriers: Neurodegenerative repercussions of radiotherapy induced damage on the blood-brain and blood-tumor barrier. Free Radic Biol Med 2022; 178:189-201. [PMID: 34875340 PMCID: PMC8925982 DOI: 10.1016/j.freeradbiomed.2021.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/20/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023]
Abstract
Exposure to radiation during the treatment of CNS tumors leads to detrimental damage of the blood brain barrier (BBB) in normal tissue. Effects are characterized by leakage of the vasculature which exposes the brain to a host of neurotoxic agents potentially leading to white matter necrosis, parenchymal calcification, and an increased chance of stroke. Vasculature of the blood tumor barrier (BTB) is irregular leading to poorly perfused and hypoxic tissue throughout the tumor that becomes resistant to radiation. While current clinical applications of cranial radiotherapy use dose fractionation to reduce normal tissue damage, these treatments still cause significant alterations to the cells that make up the neurovascular unit of the BBB and BTB. Damage to the vasculature manifests as reduction in tight junction proteins, alterations to membrane transporters, impaired cell signaling, apoptosis, and cellular senescence. While radiotherapy treatments are detrimental to normal tissue, adapting combined strategies with radiation targeted to damage the BTB could aid in drug delivery. Understanding differences between the BBB and the BTB may provide valuable insight allowing clinicians to improve treatment outcomes. Leveraging this information should allow advances in the development of therapeutic modalities that will protect the normal tissue while simultaneously improving CNS tumor treatments.
Collapse
Affiliation(s)
- Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, CA, 92697, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
21
|
Ionizing Radiation-Induced Brain Cell Aging and the Potential Underlying Molecular Mechanisms. Cells 2021; 10:cells10123570. [PMID: 34944078 PMCID: PMC8700624 DOI: 10.3390/cells10123570] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/10/2023] Open
Abstract
Population aging is occurring rapidly worldwide, challenging the global economy and healthcare services. Brain aging is a significant contributor to various age-related neurological and neuropsychological disorders, including Alzheimer's disease and Parkinson's disease. Several extrinsic factors, such as exposure to ionizing radiation, can accelerate senescence. Multiple human and animal studies have reported that exposure to ionizing radiation can have varied effects on organ aging and lead to the prolongation or shortening of life span depending on the radiation dose or dose rate. This paper reviews the effects of radiation on the aging of different types of brain cells, including neurons, microglia, astrocytes, and cerebral endothelial cells. Further, the relevant molecular mechanisms are discussed. Overall, this review highlights how radiation-induced senescence in different cell types may lead to brain aging, which could result in the development of various neurological and neuropsychological disorders. Therefore, treatment targeting radiation-induced oxidative stress and neuroinflammation may prevent radiation-induced brain aging and the neurological and neuropsychological disorders it may cause.
Collapse
|
22
|
Azimzadeh O, Subramanian V, Sievert W, Merl-Pham J, Oleksenko K, Rosemann M, Multhoff G, Atkinson MJ, Tapio S. Activation of PPARα by Fenofibrate Attenuates the Effect of Local Heart High Dose Irradiation on the Mouse Cardiac Proteome. Biomedicines 2021; 9:biomedicines9121845. [PMID: 34944662 PMCID: PMC8698387 DOI: 10.3390/biomedicines9121845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/18/2021] [Accepted: 12/02/2021] [Indexed: 01/13/2023] Open
Abstract
Radiation-induced cardiovascular disease is associated with metabolic remodeling in the heart, mainly due to the inactivation of the transcription factor peroxisome proliferator-activated receptor alpha (PPARα), thereby inhibiting lipid metabolic enzymes. The objective of the present study was to investigate the potential protective effect of fenofibrate, a known agonist of PPARα on radiation-induced cardiac toxicity. To this end, we compared, for the first time, the cardiac proteome of fenofibrate- and placebo-treated mice 20 weeks after local heart irradiation (16 Gy) using label-free proteomics. The observations were further validated using immunoblotting, enzyme activity assays, and ELISA. The analysis showed that fenofibrate restored signalling pathways that were negatively affected by irradiation, including lipid metabolism, mitochondrial respiratory chain, redox response, tissue homeostasis, endothelial NO signalling and the inflammatory status. The results presented here indicate that PPARα activation by fenofibrate attenuates the cardiac proteome alterations induced by irradiation. These findings suggest a potential benefit of fenofibrate administration in the prevention of cardiovascular diseases, following radiation exposure.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Section Radiation Biology, Federal Office for Radiation Protection, 85764 Neuherberg, Germany
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Correspondence: ; Tel.: +49-030/18333-2242
| | - Vikram Subramanian
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa, IA 52242, USA
| | - Wolfgang Sievert
- Department of Radiation Oncology, Campus Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany; (W.S.); (G.M.)
- Central Institute for Translational Cancer Research-TranslaTUM, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Juliane Merl-Pham
- Research Unit Protein Science, Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, 80939 Munich, Germany;
| | - Kateryna Oleksenko
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
| | - Michael Rosemann
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
| | - Gabriele Multhoff
- Department of Radiation Oncology, Campus Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany; (W.S.); (G.M.)
- Central Institute for Translational Cancer Research-TranslaTUM, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Michael J. Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Chair of Radiation Biology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| |
Collapse
|
23
|
Xu F, Wang Y, Gao H, Zhang X, Hu Y, Han T, Shen B, Zhang L, Wu Q. X-Ray Causes mRNA Transcripts Change to Enhance Orai2-Mediated Ca 2+ Influx in Rat Brain Microvascular Endothelial Cells. Front Mol Biosci 2021; 8:646730. [PMID: 34595206 PMCID: PMC8477418 DOI: 10.3389/fmolb.2021.646730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/05/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Radiation-induced brain injury is a serious and treatment-limiting complication of brain radiation therapy. Although endothelial cell dysfunction plays a critical role in the development of this pathogenesis, the underlying molecular mechanisms remain elusive. Methods: Primary cultured rat brain microvascular endothelial cells (BMECs) were divided into five groups without or with exposure of x-rays delivered at 5 Gy or 20 Gy. For the irradiated groups, cells were continued to cultivate for 12 or 24 h after being irradiated. Then the mRNA libraries of each group were established and applied for next-generation sequencing. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were conducted to analyze the sequencing results. Quantitative polymerase chain reaction, western blotting, cck8 assay and intracellular calcium concentration assays were conducted to analyze the role of Orai2-associated SOCE in x-ray induced cellular injury. Results: In total, 3,005 transcripts in all the four x-ray-exposed groups of BMECs showed expression level changes compared with controls. With the dose of x-ray augment and the following cultured time extension, the numbers of differentially expressed genes (DEGs) increased significantly in BMECs. Venn diagrams identified 40 DEGs common to all four exposure groups. Functional pathway enrichment analyses indicated that those 40 DEGs were enriched in the calcium signaling pathway. Among those 40 DEGs, mRNA and protein expression levels of Orai2 were significantly upregulated for 24 h. Similarly, calcium influx via store-operated calcium entry, which is modulated by Orai2, was also significantly increased for 24 h in x-ray-exposed BMECs. Moreover, the change in SOCE was suppressed by btp-2, which is a non-selective inhibitor of Orai. Additionally, x-ray exposure induced a significant decrease of proliferation in BMECs in the dose- and time-dependent manner. Conclusion: These findings provide evidence for molecular mechanisms underlying BMECs dysfunction in development of radiation-induced brain injury and suggest new approaches for therapeutic targets.
Collapse
Affiliation(s)
- Fangfang Xu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yang Wang
- Department of Otolaryngology-Head and Neck Surgery, Lu'an People's Hospital, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, China.,School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Huiwen Gao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xinchen Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yu Hu
- Department of Otolaryngology-Head and Neck Surgery, Lu'an People's Hospital, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, China
| | - Tingting Han
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lesha Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qibing Wu
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
24
|
Chauhan V, Hamada N, Monceau V, Ebrahimian T, Adam N, Wilkins RC, Sebastian S, Patel ZS, Huff JL, Simonetto C, Iwasaki T, Kaiser JC, Salomaa S, Moertl S, Azimzadeh O. Expert consultation is vital for adverse outcome pathway development: a case example of cardiovascular effects of ionizing radiation. Int J Radiat Biol 2021; 97:1516-1525. [PMID: 34402738 DOI: 10.1080/09553002.2021.1969466] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND The circulatory system distributes nutrients, signaling molecules, and immune cells to vital organs and soft tissues. Epidemiological, animal, and in vitro cellular mechanistic studies have highlighted that exposure to ionizing radiation (IR) can induce molecular changes in cellular and subcellular milieus leading to long-term health impacts, particularly on the circulatory system. Although the mechanisms for the pathologies are not fully elucidated, endothelial dysfunction is proven to be a critical event via radiation-induced oxidative stress mediators. To delineate connectivities of events specifically to cardiovascular disease (CVD) initiation and progression, the adverse outcome pathway (AOP) approach was used with consultation from field experts. AOPs are a means to organize information around a disease of interest to a regulatory question. An AOP begins with a molecular initiating event and ends in an adverse outcome via sequential linkages of key event relationships that are supported by evidence in the form of the modified Bradford-Hill criteria. Detailed guidelines on building AOPs are provided by the Organisation for Economic Cooperation and Development (OECD) AOP program. Here, we report on the questions and discussions needed to develop an AOP for CVD resulting from IR exposure. A recent workshop jointly organized by the MELODI (Multidisciplinary European Low Dose Initiative) and the ALLIANCE (European Radioecology Alliance) associations brought together experts from the OECD to present the AOP approach and tools with examples from the toxicology field. As part of this workshop, four working groups were formed to discuss the identification of adverse outcomes relevant to radiation exposures and development of potential AOPs, one of which was focused on IR-induced cardiovascular effects. Each working group comprised subject matter experts and radiation researchers interested in the specific disease area and included an AOP coach. CONCLUSION The CVD working group identified the critical questions of interest for AOP development, including the exposure scenario that would inform the evidence, the mechanisms of toxicity, the initiating event, intermediate key events/relationships, and the type of data currently available. This commentary describes the four-day discussion of the CVD working group, its outcomes, and demonstrates how collaboration and expert consultation is vital to informing AOP construction.
Collapse
Affiliation(s)
- Vinita Chauhan
- Consumer and Clinical Radiation Bureau, Health Canada, Ottawa, Canada
| | - Nobuyuki Hamada
- Radiation Safety Unit, Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Tokyo, Japan
| | - Virginie Monceau
- Institute of Radiation and Nuclear Safety (IRSN), Radiotoxicology and Radiobiology Research Laboratory (LRTOX), Fontenay-Aux-Roses, France
| | - Teni Ebrahimian
- Institute of Radiation and Nuclear Safety (IRSN), Radiotoxicology and Radiobiology Research Laboratory (LRTOX), Fontenay-Aux-Roses, France
| | - Nadine Adam
- Consumer and Clinical Radiation Bureau, Health Canada, Ottawa, Canada
| | - Ruth C Wilkins
- Consumer and Clinical Radiation Bureau, Health Canada, Ottawa, Canada
| | - Soji Sebastian
- Radiobiology, Canadian Nuclear Laboratories, Chalk River, Canada
| | - Zarana S Patel
- KBR Inc, Houston, TX, USA.,NASA Johnson Space Center, Houston, TX, USA
| | | | - Cristoforo Simonetto
- Helmholtz Zentrum München, Institute of Radiation Medicine (HMGU-IRM), Neuherberg, Germany
| | - Toshiyasu Iwasaki
- Radiation Safety Unit, Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Tokyo, Japan
| | - Jan Christian Kaiser
- Helmholtz Zentrum München, Institute of Radiation Medicine (HMGU-IRM), Neuherberg, Germany
| | - Sisko Salomaa
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Simone Moertl
- Section Radiation Biology, Federal Office for Radiation Protection (BfS), Neuherberg, Germany
| | - Omid Azimzadeh
- Section Radiation Biology, Federal Office for Radiation Protection (BfS), Neuherberg, Germany
| |
Collapse
|
25
|
Ahmadi M, Barnard S, Ainsbury E, Kadhim M. Early Responses to Low-Dose Ionizing Radiation in Cellular Lens Epithelial Models. Radiat Res 2021; 197:78-91. [PMID: 34324666 DOI: 10.1667/rade-20-00284.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 07/06/2021] [Indexed: 11/03/2022]
Abstract
Cataract is the leading cause of visual impairment which can result in blindness. Cataract formation has been associated with radiation exposure; however, the mechanistic understanding of this phenomenon is still lacking. The goal of this study was to investigate mechanisms of cataract induction in isolated lens epithelial cells (LEC) exposed to ionizing radiation. Human LECs from different genetic backgrounds (SV40 immortalized HLE-B3 and primary HLEC cells) were exposed to varying doses of 137Cs gamma rays (0, 0.1, 0.25 and 0.5 Gy), at low (0.065 Gy/min) and higher (0.3 Gy/min) dose rates. Different assays were used to measure LEC response for, e.g., viability, oxidative stress, DNA damage studies, senescence and changes to telomere length/telomerase activity at two time points (1 h and 24 h, or 24 h and 15 days, depending on the type of assay and expected response time). The viability of cells decreased in a dose-dependent manner within 24 h of irradiation. Measurement of reactive oxygen species showed an increase at 1 h postirradiation, which was alleviated within 24 h. This was consistent with DNA damage results showing high DNA damage after 1 h postirradiation which reduced significantly (but not completely) within 24 h. Induction of senescence was also observed 15 days postirradiation, but this was not attributed to telomere erosion or telomerase activity reduction. Overall, these findings provide a mechanistic understanding of low-dose radiation-induced cataractogenesis which will ultimately help to inform judgements on the magnitude of risk and improve existing radiation protection procedures.
Collapse
Affiliation(s)
- Maryam Ahmadi
- Genomic Instability and Cell Communication Research Group, Department of Biological and Medical Science, Oxford Brookes University, Oxford, United Kingdom.,Kidney Genetics Group, Academic Unit of Nephrology, The Medical School, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Stephen Barnard
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Oxford, United Kingdom
| | - Elizabeth Ainsbury
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Oxford, United Kingdom
| | - Munira Kadhim
- Genomic Instability and Cell Communication Research Group, Department of Biological and Medical Science, Oxford Brookes University, Oxford, United Kingdom
| |
Collapse
|
26
|
Azimzadeh O, von Toerne C, Subramanian V, Sievert W, Multhoff G, Atkinson MJ, Tapio S. Data-Independent Acquisition Proteomics Reveals Long-Term Biomarkers in the Serum of C57BL/6J Mice Following Local High-Dose Heart Irradiation. Front Public Health 2021; 9:678856. [PMID: 34277544 PMCID: PMC8283568 DOI: 10.3389/fpubh.2021.678856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/18/2021] [Indexed: 12/23/2022] Open
Abstract
Background and Purpose: Cardiotoxicity is a well-known adverse effect of radiation therapy. Measurable abnormalities in the heart function indicate advanced and often irreversible heart damage. Therefore, early detection of cardiac toxicity is necessary to delay and alleviate the development of the disease. The present study investigated long-term serum proteome alterations following local heart irradiation using a mouse model with the aim to detect biomarkers of radiation-induced cardiac toxicity. Materials and Methods: Serum samples from C57BL/6J mice were collected 20 weeks after local heart irradiation with 8 or 16 Gy X-ray; the controls were sham-irradiated. The samples were analyzed by quantitative proteomics based on data-independent acquisition mass spectrometry. The proteomics data were further investigated using bioinformatics and ELISA. Results: The analysis showed radiation-induced changes in the level of several serum proteins involved in the acute phase response, inflammation, and cholesterol metabolism. We found significantly enhanced expression of proinflammatory cytokines (TNF-α, TGF-β, IL-1, and IL-6) in the serum of the irradiated mice. The level of free fatty acids, total cholesterol, low-density lipoprotein (LDL), and oxidized LDL was increased, whereas that of high-density lipoprotein was decreased by irradiation. Conclusions: This study provides information on systemic effects of heart irradiation. It elucidates a radiation fingerprint in the serum that may be used to elucidate adverse cardiac effects after radiation therapy.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Section Radiation Biology, Federal Office for Radiation Protection, Oberschleissheim, Germany
| | - Christine von Toerne
- Research Unit Protein Science, Helmholtz Zentrum München - German Research Center for Environmental Health, Munich, Germany
| | - Vikram Subramanian
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfgang Sievert
- Department of Radiation Oncology, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Radiation Biology, Technical University of Munich, Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Institute for Biological and Medical Imaging, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
27
|
Abstract
PURPOSE The aim of this article is to describe the technical development in proteomics during the last two decades with the focus on its use in radiation biology. It is written from a subjective point of view and aims not to be a scientific review of the subject. CONCLUSION Proteomics is a fast developing technique and it has already contributed greatly to our understanding of biological mechanisms following radiation exposure. Novel proteomics approaches can be used in adequately designed cellular and animal experiments and above all in big clinical trials to investigate effects of ionizing radiation in the future.
Collapse
Affiliation(s)
- Soile Tapio
- Institute of Radiation Biology and Institute for Biological and Medical Imaging, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
28
|
Sun X, Feinberg MW. Vascular Endothelial Senescence: Pathobiological Insights, Emerging Long Noncoding RNA Targets, Challenges and Therapeutic Opportunities. Front Physiol 2021; 12:693067. [PMID: 34220553 PMCID: PMC8242592 DOI: 10.3389/fphys.2021.693067] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/07/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a stable form of cell cycle arrest in response to various stressors. While it serves as an endogenous pro-resolving mechanism, detrimental effects ensue when it is dysregulated. In this review, we introduce recent advances for cellular senescence and inflammaging, the underlying mechanisms for the reduction of nicotinamide adenine dinucleotide in tissues during aging, new knowledge learned from p16 reporter mice, and the development of machine learning algorithms in cellular senescence. We focus on pathobiological insights underlying cellular senescence of the vascular endothelium, a critical interface between blood and all tissues. Common causes and hallmarks of endothelial senescence are highlighted as well as recent advances in endothelial senescence. The regulation of cellular senescence involves multiple mechanistic layers involving chromatin, DNA, RNA, and protein levels. New targets are discussed including the roles of long noncoding RNAs in regulating endothelial cellular senescence. Emerging small molecules are highlighted that have anti-aging or anti-senescence effects in age-related diseases and impact homeostatic control of the vascular endothelium. Lastly, challenges and future directions are discussed including heterogeneity of endothelial cells and endothelial senescence, senescent markers and detection of senescent endothelial cells, evolutionary differences for immune surveillance in mice and humans, and long noncoding RNAs as therapeutic targets in attenuating cellular senescence. Accumulating studies indicate that cellular senescence is reversible. A better understanding of endothelial cellular senescence through lifestyle and pharmacological interventions holds promise to foster a new frontier in the management of cardiovascular disease risk.
Collapse
Affiliation(s)
- Xinghui Sun
- Department of Biochemistry, University of Nebraska–Lincoln, Lincoln, NE, United States
- Nebraska Center for the Prevention of Obesity Diseases Through Dietary Molecules, University of Nebraska–Lincoln, Lincoln, NE, United States
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska–Lincoln, Lincoln, NE, United States
| | - Mark W. Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
29
|
Abdelgawad IY, Sadak KT, Lone DW, Dabour MS, Niedernhofer LJ, Zordoky BN. Molecular mechanisms and cardiovascular implications of cancer therapy-induced senescence. Pharmacol Ther 2021; 221:107751. [PMID: 33275998 PMCID: PMC8084867 DOI: 10.1016/j.pharmthera.2020.107751] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022]
Abstract
Cancer treatment has been associated with accelerated aging that can lead to early-onset health complications typically experienced by older populations. In particular, cancer survivors have an increased risk of developing premature cardiovascular complications. In the last two decades, cellular senescence has been proposed as an important mechanism of premature cardiovascular diseases. Cancer treatments, specifically anthracyclines and radiation, have been shown to induce senescence in different types of cardiovascular cells. Additionally, clinical studies identified increased systemic markers of senescence in cancer survivors. Preclinical research has demonstrated the potential of several approaches to mitigate cancer therapy-induced senescence. However, strategies to prevent and/or treat therapy-induced cardiovascular senescence have not yet been translated to the clinic. In this review, we will discuss how therapy-induced senescence can contribute to cardiovascular complications. Thereafter, we will summarize the current in vitro, in vivo, and clinical evidence regarding cancer therapy-induced cardiovascular senescence. Then, we will discuss interventional strategies that have the potential to protect against therapy-induced cardiovascular senescence. To conclude, we will highlight challenges and future research directions to mitigate therapy-induced cardiovascular senescence in cancer survivors.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Karim T Sadak
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA; University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Diana W Lone
- University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA
| | - Mohamed S Dabour
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.
| |
Collapse
|
30
|
Coates JTT, Pirovano G, El Naqa I. Radiomic and radiogenomic modeling for radiotherapy: strategies, pitfalls, and challenges. J Med Imaging (Bellingham) 2021; 8:031902. [PMID: 33768134 PMCID: PMC7985651 DOI: 10.1117/1.jmi.8.3.031902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
The power of predictive modeling for radiotherapy outcomes has historically been limited by an inability to adequately capture patient-specific variabilities; however, next-generation platforms together with imaging technologies and powerful bioinformatic tools have facilitated strategies and provided optimism. Integrating clinical, biological, imaging, and treatment-specific data for more accurate prediction of tumor control probabilities or risk of radiation-induced side effects are high-dimensional problems whose solutions could have widespread benefits to a diverse patient population-we discuss technical approaches toward this objective. Increasing interest in the above is specifically reflected by the emergence of two nascent fields, which are distinct but complementary: radiogenomics, which broadly seeks to integrate biological risk factors together with treatment and diagnostic information to generate individualized patient risk profiles, and radiomics, which further leverages large-scale imaging correlates and extracted features for the same purpose. We review classical analytical and data-driven approaches for outcomes prediction that serve as antecedents to both radiomic and radiogenomic strategies. Discussion then focuses on uses of conventional and deep machine learning in radiomics. We further consider promising strategies for the harmonization of high-dimensional, heterogeneous multiomics datasets (panomics) and techniques for nonparametric validation of best-fit models. Strategies to overcome common pitfalls that are unique to data-intensive radiomics are also discussed.
Collapse
Affiliation(s)
- James T. T. Coates
- Massachusetts General Hospital & Harvard Medical School, Center for Cancer Research, Boston, Massachusetts, United States
| | - Giacomo Pirovano
- Memorial Sloan Kettering Cancer Center, Department of Radiology, New York, New York, United States
| | - Issam El Naqa
- Moffitt Cancer Center and Research Institute, Department of Machine Learning, Tampa, Florida, United States
| |
Collapse
|
31
|
X‑irradiation induces acute and early term inflammatory responses in atherosclerosis‑prone ApoE‑/‑ mice and in endothelial cells. Mol Med Rep 2021; 23:399. [PMID: 33786610 PMCID: PMC8025474 DOI: 10.3892/mmr.2021.12038] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
Thoracic radiotherapy is an effective treatment for many types of cancer; however it is also associated with an increased risk of developing cardiovascular disease (CVD), appearing mainly ≥10 years after radiation exposure. The present study investigated acute and early term physiological and molecular changes in the cardiovascular system after ionizing radiation exposure. Female and male ApoE‑/‑ mice received a single exposure of low or high dose X‑ray thoracic irradiation (0.1 and 10 Gy). The level of cholesterol and triglycerides, as well as a large panel of inflammatory markers, were analyzed in serum samples obtained at 24 h and 1 month after irradiation. The secretion of inflammatory markers was further verified in vitro in coronary artery and microvascular endothelial cell lines after exposure to low and high dose of ionizing radiation (0.1 and 5 Gy). Local thoracic irradiation of ApoE‑/‑ mice increased serum growth differentiation factor‑15 (GDF‑15) and C‑X‑C motif chemokine ligand 10 (CXCL10) levels in both female and male mice 24 h after high dose irradiation, which were also secreted from coronary artery and microvascular endothelial cells in vitro. Sex‑specific responses were observed for triglyceride and cholesterol levels, and some of the assessed inflammatory markers as detailed below. Male ApoE‑/‑ mice demonstrated elevated intercellular adhesion molecule‑1 and P‑selectin at 24 h, and adiponectin and plasminogen activator inhibitor‑1 at 1 month after irradiation, while female ApoE‑/‑ mice exhibited decreased monocyte chemoattractant protein‑1 and urokinase‑type plasminogen activator receptor at 24 h, and basic fibroblast growth factor 1 month after irradiation. The inflammatory responses were mainly significant following high dose irradiation, but certain markers showed significant changes after low dose exposure. The present study revealed that acute/early inflammatory responses occurred after low and high dose thoracic irradiation. However, further research is required to elucidate early asymptomatic changes in the cardiovascular system post thoracic X‑irradiation and to investigate whether GDF‑15 and CXCL10 could be considered as potential biomarkers for the early detection of CVD risk in thoracic radiotherapy‑treated patients.
Collapse
|
32
|
Late Health Effects of Partial Body Irradiation Injury in a Minipig Model Are Associated with Changes in Systemic and Cardiac IGF-1 Signaling. Int J Mol Sci 2021; 22:ijms22063286. [PMID: 33807089 PMCID: PMC8005067 DOI: 10.3390/ijms22063286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Clinical, epidemiological, and experimental evidence demonstrate non-cancer, cardiovascular, and endocrine effects of ionizing radiation exposure including growth hormone deficiency, obesity, metabolic syndrome, diabetes, and hyperinsulinemia. Insulin-like growth factor-1 (IGF-1) signaling perturbations are implicated in development of cardiovascular disease and metabolic syndrome. The minipig is an emerging model for studying radiation effects given its high analogy to human anatomy and physiology. Here we use a minipig model to study late health effects of radiation by exposing male Göttingen minipigs to 1.9–2.0 Gy X-rays (lower limb tibias spared). Animals were monitored for 120 days following irradiation and blood counts, body weight, heart rate, clinical chemistry parameters, and circulating biomarkers were assessed longitudinally. Collagen deposition, histolopathology, IGF-1 signaling, and mRNA sequencing were evaluated in tissues. Our findings indicate a single exposure induced histopathological changes, attenuated circulating IGF-1, and disrupted cardiac IGF-1 signaling. Electrolytes, lipid profiles, liver and kidney markers, and heart rate and rhythm were also affected. In the heart, collagen deposition was significantly increased and transforming growth factor beta-1 (TGF-beta-1) was induced following irradiation; collagen deposition and fibrosis were also observed in the kidney of irradiated animals. Our findings show Göttingen minipigs are a suitable large animal model to study long-term effects of radiation exposure and radiation-induced inhibition of IGF-1 signaling may play a role in development of late organ injuries.
Collapse
|
33
|
Mechanisms of radiation-induced endothelium damage: Emerging models and technologies. Radiother Oncol 2021; 158:21-32. [PMID: 33581220 DOI: 10.1016/j.radonc.2021.02.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/24/2022]
Abstract
Radiation-induced endothelial/vascular injury is a major complicating factor in radiotherapy and a leading cause of morbidity and mortality in nuclear or radiological catastrophes. Exposure of tissue to ionizing radiation (IR) leads to the release of oxygen radicals and proteases that result in loss of endothelial barrier function and leukocyte dysfunction leading to tissue injury and organ damage. Microvascular endothelial cells are particularly sensitive to IR and radiation-induced alterations in endothelial cell function are thought to be a critical factor in organ damage through endothelial cell activation, enhanced leukocyte-endothelial cell interactions, increased barrier permeability and initiation of apoptotic pathways. These radiation-induced inflammatory responses are important in early and late radiation pathologies in various organs. A better understanding of mechanisms of radiation-induced endothelium dysfunction is therefore vital, as radiobiological response of endothelium is of major importance for medical management and therapeutic development for radiation injuries. In this review, we summarize the current knowledge of cellular and molecular mechanisms of radiation-induced endothelium damage and their impact on early and late radiation injury. Furthermore, we review established and emerging in vivo and in vitro models that have been developed to study the mechanisms of radiation-induced endothelium damage and to design, develop and rapidly screen therapeutics for treatment of radiation-induced vascular damage. Currently there are no specific therapeutics available to protect against radiation-induced loss of endothelial barrier function, leukocyte dysfunction and resulting organ damage. Developing therapeutics to prevent endothelium dysfunction and normal tissue damage during radiotherapy can serve as the urgently needed medical countermeasures.
Collapse
|
34
|
Tapio S, Little MP, Kaiser JC, Impens N, Hamada N, Georgakilas AG, Simar D, Salomaa S. Ionizing radiation-induced circulatory and metabolic diseases. ENVIRONMENT INTERNATIONAL 2021; 146:106235. [PMID: 33157375 PMCID: PMC10686049 DOI: 10.1016/j.envint.2020.106235] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/09/2020] [Accepted: 10/08/2020] [Indexed: 05/23/2023]
Abstract
Risks to health are the prime consideration in all human situations of ionizing radiation exposure and therefore of relevance to radiation protection in all occupational, medical, and public exposure situations. Over the past few decades, advances in therapeutic strategies have led to significant improvements in cancer survival rates. However, a wide range of long-term complications have been reported in cancer survivors, in particular circulatory diseases and their major risk factors, metabolic diseases. However, at lower levels of exposure, the evidence is less clear. Under real-life exposure scenarios, including radiotherapy, radiation effects in the whole organism will be determined mainly by the response of normal tissues receiving relatively low doses, and will be mediated and moderated by systemic effects. Therefore, there is an urgent need for further research on the impact of low-dose radiation. In this article, we review radiation-associated risks of circulatory and metabolic diseases in clinical, occupational or environmental exposure situations, addressing epidemiological, biological, risk modelling, and systems biology aspects, highlight the gaps in knowledge and discuss future directions to address these gaps.
Collapse
Affiliation(s)
- Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health GmbH, Neuherberg, Germany.
| | - Mark P Little
- Radiation Epidemiology Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), MD, USA
| | - Jan Christian Kaiser
- Institute of Radiation Medicine, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Nathalie Impens
- Institute of Environment, Health and Safety, Biosphere Impact Studies, SCK•CEN, Mol, Belgium
| | - Nobuyuki Hamada
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Tokyo, Japan
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece
| | - David Simar
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Sisko Salomaa
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
35
|
Sampath C, Okoro EU, Gipson MJ, Chukkapalli SS, Farmer-Dixon CM, Gangula PR. Porphyromonas gingivalis infection alters Nrf2-phase II enzymes and nitric oxide in primary human aortic endothelial cells. J Periodontol 2020; 92:54-65. [PMID: 33128253 DOI: 10.1002/jper.20-0444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Periodontal disease (PD) is known to be associated with endothelial dysfunction in patients with coronary artery and/or cardiovascular disease. In our study, we sought to explore the virulence of P. gingivalis (Pg) affecting glycogen synthase kinase 3 beta (GSK-3β)/nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/tetrahydrobiopterin (BH4 )/ nitric oxide synthase (NOS) expression in primary human aortic endothelial cells (pHAECs). METHODS pHAECs were infected for 48 hours with Pg in vitro using the Human oxygen-Bacteria anaerobic coculture technique. Cell viability was determined, and target gene expression changes were evaluated by quantitative real-time polymerase chain reaction at the end of each incubation period. RESULTS Pg impaired pHAEC viability 24 hours post-infection. Pg infection reduced mRNA expression levels of endothelial NOS (eNOS), Nrf2, and Phase II enzymes (heme oxygenase-1, catalase, superoxide dismutase-1) in a time-dependent manner. Significant (P <0.05) increase in the inflammatory markers (interleukin [IL]-1β, IL-6, and tumor necrosis factor-α) were observed in the medium as well as in the infected cells. Interestingly, inducible NOS mRNA levels showed a significant (P <0.05) increase at 12 hours and 24 hours and were reduced at later time points. BH4 (cofactor of eNOS) biosynthesis enzyme dihydrofolate reductase (DHFR, salvage pathway) mRNA levels showed a significant (P <0.05) decrease, while mRNA levels of GSK-3β were elevated. CONCLUSIONS These results suggest that periodontal bacterial infection may cause significant changes in the endothelial GSK-3β/BH4 /eNOS/Nrf2 pathways, which may lead to impaired vascular relaxation. Greater understanding of the factors that adversely affect endothelial cell function could contribute to the development of new therapeutic compounds to treat PD-induced vascular diseases.
Collapse
Affiliation(s)
- Chethan Sampath
- Department of ODS & Research, Meharry Medical College, Nashville, TN
| | - Emmanuel U Okoro
- Department of Microbiology, Immunology & Physiology, Meharry Medical College, Nashville, TN
| | - Michael J Gipson
- Department of ODS & Research, Meharry Medical College, Nashville, TN
| | | | | | - Pandu R Gangula
- Department of ODS & Research, Meharry Medical College, Nashville, TN
| |
Collapse
|
36
|
Philipp J, Le Gleut R, von Toerne C, Subedi P, Azimzadeh O, Atkinson MJ, Tapio S. Radiation Response of Human Cardiac Endothelial Cells Reveals a Central Role of the cGAS-STING Pathway in the Development of Inflammation. Proteomes 2020; 8:proteomes8040030. [PMID: 33114474 PMCID: PMC7709117 DOI: 10.3390/proteomes8040030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/06/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Radiation-induced inflammation leading to the permeability of the endothelial barrier may increase the risk of cardiovascular disease. The aim of this study was to investigate potential mechanisms in vitro at the level of the proteome in human coronary artery endothelial cells (HCECest2) that were exposed to radiation doses of 0, 0.25, 0.5, 2.0 and 10 Gy (60Co-γ). Proteomics analysis was performed using mass spectrometry in a label-free data-independent acquisition mode. The data were validated using bioinformatics and immunoblotting. The low- and moderate-dose-irradiated samples (0.25 Gy, 0.5 Gy) showed only scarce proteome changes. In contrast, an activation of DNA-damage repair, inflammation, and oxidative stress pathways was seen after the high-dose treatments (2 and 10 Gy). The level of the DNA damage response protein DDB2 was enhanced early at the 10 Gy dose. The expression of proteins belonging to the inflammatory response or cGAS-STING pathway (STING, STAT1, ICAM1, ISG15) increased in a dose-dependent manner, showing the strongest effects at 10 Gy after one week. This study suggests a connection between the radiation-induced DNA damage and the induction of inflammation which supports the inhibition of the cGAS-STING pathway in the prevention of radiation-induced cardiovascular disease.
Collapse
Affiliation(s)
- Jos Philipp
- Institute of Radiation Biology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.P.); (P.S.); (O.A.); (M.J.A.)
| | - Ronan Le Gleut
- Institute of Computational Biology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany;
| | - Christine von Toerne
- Research Unit Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany;
| | - Prabal Subedi
- Institute of Radiation Biology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.P.); (P.S.); (O.A.); (M.J.A.)
- Federal Office for Radiation Protection, BfS, 85764 Neuherberg, Germany
| | - Omid Azimzadeh
- Institute of Radiation Biology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.P.); (P.S.); (O.A.); (M.J.A.)
| | - Michael J. Atkinson
- Institute of Radiation Biology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.P.); (P.S.); (O.A.); (M.J.A.)
- Chair of Radiation Biology, Technical University of Munich, 80333 Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.P.); (P.S.); (O.A.); (M.J.A.)
- Correspondence: ; Tel.: +49-89-3187-3445
| |
Collapse
|
37
|
Ping Z, Peng Y, Lang H, Xinyong C, Zhiyi Z, Xiaocheng W, Hong Z, Liang S. Oxidative Stress in Radiation-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3579143. [PMID: 32190171 PMCID: PMC7071808 DOI: 10.1155/2020/3579143] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/03/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023]
Abstract
There is a distinct increase in the risk of heart disease in people exposed to ionizing radiation (IR). Radiation-induced heart disease (RIHD) is one of the adverse side effects when people are exposed to ionizing radiation. IR may come from various forms, such as diagnostic imaging, radiotherapy for cancer treatment, nuclear disasters, and accidents. However, RIHD was mainly observed after radiotherapy for chest malignant tumors, especially left breast cancer. Radiation therapy (RT) has become one of the main ways to treat all kinds of cancer, which is used to reduce the recurrence of cancer and improve the survival rate of patients. The potential cause of radiation-induced cardiotoxicity is unclear, but it may be relevant to oxidative stress. Oxidative stress, an accumulation of reactive oxygen species (ROS), disrupts intracellular homeostasis through chemical modification and damages proteins, lipids, and DNA; therefore, it results in a series of related pathophysiological changes. The purpose of this review was to summarise the studies of oxidative stress in radiotherapy-induced cardiotoxicity and provide prevention and treatment methods to reduce cardiac damage.
Collapse
Affiliation(s)
- Zhang Ping
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Yang Peng
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Hong Lang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Cai Xinyong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Zeng Zhiyi
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Wu Xiaocheng
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Zeng Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Shao Liang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| |
Collapse
|
38
|
Livingston K, Schlaak RA, Puckett LL, Bergom C. The Role of Mitochondrial Dysfunction in Radiation-Induced Heart Disease: From Bench to Bedside. Front Cardiovasc Med 2020; 7:20. [PMID: 32154269 PMCID: PMC7047199 DOI: 10.3389/fcvm.2020.00020] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/05/2020] [Indexed: 12/25/2022] Open
Abstract
Radiation is a key modality in the treatment of many cancers; however, it can also affect normal tissues adjacent to the tumor, leading to toxic effects. Radiation to the thoracic region, such as that received as part of treatment for breast and lung cancer, can result in incidental dose to the heart, leading to cardiac dysfunction, such as pericarditis, coronary artery disease, ischemic heart disease, conduction defects, and valvular dysfunction. The underlying mechanisms for these morbidities are currently being studied but are not entirely understood. There has been increasing focus on the role of radiation-induced mitochondrial dysfunction and the ensuing impact on various cardiac functions in both preclinical models and in humans. Cardiomyocyte mitochondria are critical to cardiac function, and mitochondria make up a substantial part of a cardiomyocyte's volume. Mitochondrial dysfunction can also alter other cell types in the heart. This review summarizes several factors related to radiation-induced mitochondrial dysfunction in cardiomyocytes and endothelial cells. These factors include mitochondrial DNA mutations, oxidative stress, alterations in various mitochondrial function-related transcription factors, and apoptosis. Through improved understanding of mitochondria-dependent mechanisms of radiation-induced heart dysfunction, potential therapeutic targets can be developed to assist in prevention and treatment of radiation-induced heart damage.
Collapse
Affiliation(s)
- Katie Livingston
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Rachel A Schlaak
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lindsay L Puckett
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Carmen Bergom
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
39
|
Zeng ZM, Du HY, Xiong L, Zeng XL, Zhang P, Cai J, Huang L, Liu AW. BRCA1 protects cardiac microvascular endothelial cells against irradiation by regulating p21-mediated cell cycle arrest. Life Sci 2020; 244:117342. [PMID: 31978450 DOI: 10.1016/j.lfs.2020.117342] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/12/2020] [Accepted: 01/20/2020] [Indexed: 12/14/2022]
Abstract
AIMS Microvascular endothelial cell dysfunction is a leading cause of radiation-induced heart disease (RIHD). BRCA1 plays an important role in DNA damage repair. The study aims to explore the effect of BRCA1 in endothelial cells involved in RIHD. MATERIALS AND METHODS BRCA1 and p21 expression were detected in human umbilical vein endothelial cells (HUVECs) and in mouse heart tissue after irradiation exposure. The effects of BRCA1 on cell proliferation, cell cycle and radiosensitivity were determined in HUVECs with overexpression and knockdown of BRCA1. A mouse model of RIHD was established. Heart damage was detected in C57BL/6J mice and endothelial cell specific knockout BRCA1 mice (EC-BRCA1-/-). KEY FINDINGS BRCA1 and p21 expression was significantly increased both in vitro and vivo response to irradiation. BRCA1 overexpression in endothelial cells enhanced cell growth and G1/S phase arrest, and the opposite results were observed in BRCA1 knockdown endothelial cells. BRCA1 downregulated endothelial cell cycle-related genes cyclin A, cyclin D1, cyclin E and p-Rb through increasing p21 expression, and HUVECs with BRCA1 gene knockdown were more sensitive to radiation. In vivo, a decrease in cardiac microvascular density, as well as cardiomyocyte hypoxia and apoptosis were observed in a time-dependent manner. EC-BRCA1-/- mice were more prone to severe RIHD than EC-BRCA1+/- mice after 16Gy radiation exposure due to endothelial dysfunction caused by loss of BRCA1, and p21 was declined in EC-BRCA1-/- mice heart. SIGNIFICANCE These findings indicate that BRCA1 plays a protective role in RIHD by regulating endothelial cell cycle arrest mediated by p21 signal.
Collapse
Affiliation(s)
- Zhi-Min Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
| | - Hai-Yang Du
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
| | - Le Xiong
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
| | - Xiao-Li Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Peng Zhang
- Department of Radiation Oncology, Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, PR China
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Long Huang
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China.
| | - An-Wen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China.
| |
Collapse
|
40
|
Philipp J, Sievert W, Azimzadeh O, von Toerne C, Metzger F, Posch A, Hladik D, Subedi P, Multhoff G, Atkinson MJ, Tapio S. Data independent acquisition mass spectrometry of irradiated mouse lung endothelial cells reveals a STAT-associated inflammatory response. Int J Radiat Biol 2020; 96:642-650. [PMID: 31914348 DOI: 10.1080/09553002.2020.1712492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Purpose: Pulmonary inflammation is an adverse consequence of radiation therapy in breast cancer. The aim of this study was to elucidate biological pathways leading to this pathology.Materials and methods: Lung endothelial cells were isolated 24 h after thorax-irradiation (sham or 10 Gy X-ray) from female C57Bl/6 mice and cultivated for 6 days.Results: Quantitative proteomic analysis of lung endothelial cells was done using data independent acquisition (DIA) mass spectrometry. The data were analyzed using Ingenuity Pathway Analysis and STRINGdb. In total, 4220 proteins were identified using DIA of which 60 were dysregulated in the irradiated samples (fold change ≥2.00 or ≤0.50; q-value <0.05). Several (12/40) upregulated proteins formed a cluster of inflammatory proteins with STAT1 and IRF3 as predicted upstream regulators. The several-fold increased expression of STAT1 and STAT-associated ISG15 was confirmed by immunoblotting. The expression of antioxidant proteins SOD1 and PRXD5 was downregulated suggesting radiation-induced oxidative stress. Similarly, the phosphorylated (active) forms of STING and IRF3, both members of the cGAS/STING pathway, were downregulated.Conclusions: These data suggest the involvement of JAK/STAT and cGas/STING pathways in the genesis of radiation-induced lung inflammation. These pathways may be used as novel targets for the prevention of radiation-induced lung damage.
Collapse
Affiliation(s)
- Jos Philipp
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Wolfgang Sievert
- Radiation Immuno Oncology Group, Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Omid Azimzadeh
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Christine von Toerne
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Research Unit Protein Science, Munich, Germany
| | - Fabian Metzger
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Research Unit Protein Science, Munich, Germany
| | - Anton Posch
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Daniela Hladik
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Prabal Subedi
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Gabriele Multhoff
- Radiation Immuno Oncology Group, Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Michael J Atkinson
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Soile Tapio
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| |
Collapse
|
41
|
Effects and Mechanisms of Traditional Chinese Herbal Medicine in the Treatment of Ischemic Cardiomyopathy. Pharmacol Res 2020; 151:104488. [DOI: 10.1016/j.phrs.2019.104488] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/28/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022]
|
42
|
Unthank JL, Ortiz M, Trivedi H, Pelus LM, Sampson CH, Sellamuthu R, Fisher A, Chua HL, Plett A, Orschell CM, Cohen EP, Miller SJ. Cardiac and Renal Delayed Effects of Acute Radiation Exposure: Organ Differences in Vasculopathy, Inflammation, Senescence and Oxidative Balance. Radiat Res 2019; 191:383-397. [PMID: 30901530 DOI: 10.1667/rr15130.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We have previously shown significant pathology in the heart and kidney of murine hematopoietic-acute radiation syndrome (H-ARS) survivors of 8.7-9.0 Gy total-body irradiation (TBI). The goal of this study was to determine temporal relationships in the development of vasculopathy and the progression of renal and cardiovascular delayed effects of acute radiation exposure (DEARE) at TBI doses less than 9 Gy and to elucidate the potential roles of senescence, inflammation and oxidative stress. Our results show significant loss of endothelial cells in coronary arteries by 4 months post-TBI (8.53 or 8.72 Gy of gamma radiation). This loss precedes renal dysfunction and interstitial fibrosis and progresses to abnormalities in the arterial media and adventitia and loss of coronary arterioles. Major differences in radiation-induced pathobiology exist between the heart and kidney in terms of vasculopathy progression and also in indices of inflammation, senescence and oxidative imbalance. The results of this work suggest a need for different medical countermeasures for multiple targets in different organs and at various times after acute radiation injury to prevent the progression of DEARE.
Collapse
Affiliation(s)
- Joseph L Unthank
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Miguel Ortiz
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Hina Trivedi
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Louis M Pelus
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Carol H Sampson
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Rajendran Sellamuthu
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Alexa Fisher
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Hui Lin Chua
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Artur Plett
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christie M Orschell
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Eric P Cohen
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Steven J Miller
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
43
|
Tao X, Sun M, Chen M, Ying R, Su W, Zhang J, Xie X, Wei W, Meng X. HMGB1-modified mesenchymal stem cells attenuate radiation-induced vascular injury possibly via their high motility and facilitation of endothelial differentiation. Stem Cell Res Ther 2019; 10:92. [PMID: 30867070 PMCID: PMC6416980 DOI: 10.1186/s13287-019-1197-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/28/2022] Open
Abstract
Background Vascular injury is one of the most common detrimental effects of cancer radiotherapy on healthy tissues. Since the efficacy of current preventive and therapeutic strategies remains limited, the exploration of new approaches to treat radiation-induced vascular injury (RIV) is on high demands. The use of mesenchymal stem cells (MSCs) to treat RIV holds great promise thanks to their well-documented function of mediating tissue regeneration after injury. Recently, we genetically modified MSCs with high mobility group box 1 (HMGB1) and demonstrated the high efficacy of these cells in treating graft atherosclerosis. The current study was to investigate the protective effect of HMGB1-modified MSCs (MSC-H) on RIV by using a rat model. Methods Female F344 rats received an intravenous injection of male F344 MSC-H cells or vehicle control at four doses of 2 × 106 cells with a 15-day interval starting from 30 days after irradiation to the abdominal aorta. The aortas were procured for histological and biomedical analysis at 90 days after irradiation. Cell migration to irradiated aortas was traced by green fluorescent protein and sex determination region on the Y chromosome. In vitro cell migration and endothelial differentiation of MSC-H cells were analyzed by stromal-derived factor 1-induced transwell assay and RNA microarray, respectively. The contribution of extracellular HMGB1 to the bioactivity of MSC-H cells was investigated by inhibition experiments with HMGB1 antibody. Result MSC-H cell infusion alleviated neointimal formation, vascular inflammation, and fibrosis in irradiated aortas, which was associated with local migration and endothelial differentiation of MSC-H cells. The MSC-H cells showed high motility and potential of endothelial differentiation in vitro. Microarray analysis suggested multiple pathways like MAPK and p53 signaling were activated during endothelial differentiation. MSC-H cells highly expressed CXC chemokine receptor 4 and migrated progressively after stromal-derived factor 1 stimulation, which was blocked by the antagonist of CXC chemokine receptor 4. Finally, the migration and endothelial differentiation of MSC-H cells were inhibited by HMGB1 antibody. Conclusion MSC-H cell infusion significantly attenuated RIV, which was associated with their high motility and endothelial differentiation potential. Multiple pathways that possibly contributed to the efficacy of MSC-H cells were suggested and deserved further investigation. Electronic supplementary material The online version of this article (10.1186/s13287-019-1197-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuan Tao
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingyang Sun
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Chen
- Department of Gastroenterology, Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, China
| | - Rongchao Ying
- Department of Gastroenterological Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjie Su
- Department of Gastroenterological Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Zhang
- Department of Gastroenterological Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaodong Xie
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wei
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China. .,Department of Gastroenterological Surgery, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China.
| | - Xiaohu Meng
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
44
|
Chung NC, Mirza B, Choi H, Wang J, Wang D, Ping P, Wang W. Unsupervised classification of multi-omics data during cardiac remodeling using deep learning. Methods 2019; 166:66-73. [PMID: 30853547 DOI: 10.1016/j.ymeth.2019.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
Integration of multi-omics in cardiovascular diseases (CVDs) presents high potentials for translational discoveries. By analyzing abundance levels of heterogeneous molecules over time, we may uncover biological interactions and networks that were previously unidentifiable. However, to effectively perform integrative analysis of temporal multi-omics, computational methods must account for the heterogeneity and complexity in the data. To this end, we performed unsupervised classification of proteins and metabolites in mice during cardiac remodeling using two innovative deep learning (DL) approaches. First, long short-term memory (LSTM)-based variational autoencoder (LSTM-VAE) was trained on time-series numeric data. The low-dimensional embeddings extracted from LSTM-VAE were then used for clustering. Second, deep convolutional embedded clustering (DCEC) was applied on images of temporal trends. Instead of a two-step procedure, DCEC performes a joint optimization for image reconstruction and cluster assignment. Additionally, we performed K-means clustering, partitioning around medoids (PAM), and hierarchical clustering. Pathway enrichment analysis using the Reactome knowledgebase demonstrated that DL methods yielded higher numbers of significant biological pathways than conventional clustering algorithms. In particular, DCEC resulted in the highest number of enriched pathways, suggesting the strength of its unified framework based on visual similarities. Overall, unsupervised DL is shown to be a promising analytical approach for integrative analysis of temporal multi-omics.
Collapse
Affiliation(s)
- Neo Christopher Chung
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA; Institute of Informatics, Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland.
| | - Bilal Mirza
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Howard Choi
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA; Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jie Wang
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ding Wang
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Peipei Ping
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA; Scalable Analytics Institute (ScAi), University of California Los Angeles, Los Angeles, CA 90095, USA; Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine (Cardiology), University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Wei Wang
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA; Scalable Analytics Institute (ScAi), University of California Los Angeles, Los Angeles, CA 90095, USA; Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
45
|
Baselet B, Sonveaux P, Baatout S, Aerts A. Pathological effects of ionizing radiation: endothelial activation and dysfunction. Cell Mol Life Sci 2019; 76:699-728. [PMID: 30377700 PMCID: PMC6514067 DOI: 10.1007/s00018-018-2956-z] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/19/2018] [Accepted: 10/23/2018] [Indexed: 01/13/2023]
Abstract
The endothelium, a tissue that forms a single layer of cells lining various organs and cavities of the body, especially the heart and blood as well as lymphatic vessels, plays a complex role in vascular biology. It contributes to key aspects of vascular homeostasis and is also involved in pathophysiological processes, such as thrombosis, inflammation, and hypertension. Epidemiological data show that high doses of ionizing radiation lead to cardiovascular disease over time. The aim of this review is to summarize the current knowledge on endothelial cell activation and dysfunction after ionizing radiation exposure as a central feature preceding the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Bjorn Baselet
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK•CEN), Mol, Belgium
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics, Université catholique de Louvain (UCL), Brussels, Belgium
| | - Pierre Sonveaux
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics, Université catholique de Louvain (UCL), Brussels, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK•CEN), Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - An Aerts
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK•CEN), Mol, Belgium.
| |
Collapse
|
46
|
Mirza B, Wang W, Wang J, Choi H, Chung NC, Ping P. Machine Learning and Integrative Analysis of Biomedical Big Data. Genes (Basel) 2019; 10:E87. [PMID: 30696086 PMCID: PMC6410075 DOI: 10.3390/genes10020087] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/08/2019] [Accepted: 01/21/2019] [Indexed: 12/11/2022] Open
Abstract
Recent developments in high-throughput technologies have accelerated the accumulation of massive amounts of omics data from multiple sources: genome, epigenome, transcriptome, proteome, metabolome, etc. Traditionally, data from each source (e.g., genome) is analyzed in isolation using statistical and machine learning (ML) methods. Integrative analysis of multi-omics and clinical data is key to new biomedical discoveries and advancements in precision medicine. However, data integration poses new computational challenges as well as exacerbates the ones associated with single-omics studies. Specialized computational approaches are required to effectively and efficiently perform integrative analysis of biomedical data acquired from diverse modalities. In this review, we discuss state-of-the-art ML-based approaches for tackling five specific computational challenges associated with integrative analysis: curse of dimensionality, data heterogeneity, missing data, class imbalance and scalability issues.
Collapse
Affiliation(s)
- Bilal Mirza
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Wei Wang
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Scalable Analytics Institute (ScAi), University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Bioinformatics, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Jie Wang
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Howard Choi
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Bioinformatics, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Neo Christopher Chung
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Institute of Informatics, Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland.
| | - Peipei Ping
- NIH BD2K Center of Excellence for Biomedical Computing, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Scalable Analytics Institute (ScAi), University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Bioinformatics, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Medicine (Cardiology), University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
47
|
Wu K, Chen Z, Peng Q, Chen G, Yan W, Chen X. Ku86 alleviates human umbilical vein endothelial cellular apoptosis and senescence induced by a low dose of ionizing radiation. J Int Med Res 2019; 47:893-904. [PMID: 30616437 PMCID: PMC6381507 DOI: 10.1177/0300060518805302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Objective The aim of this study was to observe the effect of Ku86 on cellular senescence and apoptosis induced by various doses of ionizing radiation in human umbilical vein endothelial cells (HUVECs). Methods Senescence-associated β-galactosidase activity was detected to evaluate cell senescence. Apoptosis was determined by flow cytometry and a caspase enzyme determination kit. p16Ink4a, Sirt1, superoxide dismutase 2 (SOD2), xanthine oxidase (XOD), and Bcl-2 protein expression levels were measured by western blotting. Results Low doses of ionizing radiation induced cellular senescence and apoptosis in a dose-dependent manner. The Ku86 protein was negatively correlated with ionization intensity. After transfection of Ku86 with a vector (pcDNA 3.1), or interference with siRNA (si-Ku86), apoptosis/senescence and related protein expression were observed. Western blot results revealed that this induction of senescence was associated with activated Sirt1 and SOD2, and downregulation of p16Ink4a and XOD in 0.2 Gy ionizing radiation. The expression levels of apoptosis-associated proteins, such as Bcl-2, cleaved caspase-3, caspase-8, and caspase-9, were significantly altered in both the presence and absence of Ku86 with ionizing radiation (0.2 Gy). Conclusions Our study revealed that Ku86 overexpression inhibits HUVEC apoptosis and senescence induced by low doses of ionizing radiation.
Collapse
Affiliation(s)
- Kai Wu
- 1 Department of Cardiovascular Medicine, 903 Hospital, and Center for Medical Radiation Biology, Institute of Materials, China Academy of Engineering Physics, Mianyang, China.,2 Department of Cardiovascular Medicine, West China Hospital, Mianyang, China.,3 Department of Cardiovascular Medicine, Guangyuan Central Hospital, Guangyuan, China
| | - Zejin Chen
- 1 Department of Cardiovascular Medicine, 903 Hospital, and Center for Medical Radiation Biology, Institute of Materials, China Academy of Engineering Physics, Mianyang, China
| | - Qing Peng
- 2 Department of Cardiovascular Medicine, West China Hospital, Mianyang, China
| | - Guojian Chen
- 1 Department of Cardiovascular Medicine, 903 Hospital, and Center for Medical Radiation Biology, Institute of Materials, China Academy of Engineering Physics, Mianyang, China
| | - Weihong Yan
- 1 Department of Cardiovascular Medicine, 903 Hospital, and Center for Medical Radiation Biology, Institute of Materials, China Academy of Engineering Physics, Mianyang, China
| | - Xiaoping Chen
- 2 Department of Cardiovascular Medicine, West China Hospital, Mianyang, China
| |
Collapse
|
48
|
Cataractogenic load – A concept to study the contribution of ionizing radiation to accelerated aging in the eye lens. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2019; 779:68-81. [DOI: 10.1016/j.mrrev.2019.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 12/11/2022]
|
49
|
Soloviev AI, Kizub IV. Mechanisms of vascular dysfunction evoked by ionizing radiation and possible targets for its pharmacological correction. Biochem Pharmacol 2018; 159:121-139. [PMID: 30508525 DOI: 10.1016/j.bcp.2018.11.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
Abstract
Ionizing radiation (IR) leads to a variety of the cardiovascular diseases, including the arterial hypertension. A number of studies have demonstrated that blood vessels represent important target for IR, and the endothelium is one of the most vulnerable components of the vascular wall. IR causes an inhibition of nitric oxide (NO)-mediated endothelium-dependent vasodilatation and generation of reactive oxygen (ROS) and nitrogen (RNS) species trigger this process. Inhibition of NO-mediated vasodilatation could be due to endothelial NO synthase (eNOS) down-regulation, inactivation of endothelium-derived NO, and abnormalities in diffusion of NO from the endothelial cells (ECs) leading to a decrease in NO bioavailability. Beside this, IR suppresses endothelial large conductance Ca2+-activated K+ channels (BKCa) activity, which control NO synthesis. IR also leads to inhibition of the BKCa current in vascular smooth muscle cells (SMCs) which is mediated by protein kinase C (PKC). On the other hand, IR-evoked enhanced vascular contractility may result from PKC-mediated increase in SMCs myofilament Ca2+ sensitivity. Also, IR evokes vascular wall inflammation and atherosclerosis development. Vascular function damaged by IR can be effectively restored by quercetin-filled phosphatidylcholine liposomes and mesenchymal stem cells injection. Using RNA-interference technique targeted to different PKC isoforms can also be a perspective approach for pharmacological treatment of IR-induced vascular dysfunction.
Collapse
Affiliation(s)
- Anatoly I Soloviev
- Department of Pharmacology of Cellular Signaling Systems and Experimental Therapy, Institute of Pharmacology and Toxicology, National Academy of Medical Sciences of Ukraine, 14 Eugene Pottier Street, Kiev 03068, Ukraine
| | - Igor V Kizub
- Department of Pharmacology, New York Medical College, 15 Dana Road, Valhalla 10595, NY, United States.
| |
Collapse
|
50
|
Aratani S, Tagawa M, Nagasaka S, Sakai Y, Shimizu A, Tsuruoka S. Radiation-induced premature cellular senescence involved in glomerular diseases in rats. Sci Rep 2018; 8:16812. [PMID: 30429495 PMCID: PMC6235850 DOI: 10.1038/s41598-018-34893-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/27/2018] [Indexed: 12/17/2022] Open
Abstract
Currently, cellular senescence has emerged as a fundamental contributor to chronic organ diseases. Radiation is one of the stress factors that induce cellular senescence. Although the kidney is known as a radiosensitive organ, whether and how radiation-induced cellular senescence is associated with kidney diseases remains unclear. In this study, we performed experiments on 7–8-week-old male rats that received a single dose of 18-Gy radiation in the unilateral kidney. The irradiated kidneys showed hallmarks of cellular senescence, including increased SA-β-gal activity, upregulation of cyclin-dependent kinase inhibitor (p53, p21, and p16), and absence of DNA proliferation marker (Ki-67). Furthermore, combined with in-vitro experiments, we demonstrated that radiation-induced senescent glomerular endothelial cells acquired altered gene expression, namely, senescence-associated secretory phenotype (particularly, IL-6), which might be triggered by NF-kB signaling pathway. Pathological analysis suggested severe glomerular endothelial cell injury, as evidenced by thrombotic microangiopathy, collapsing glomeruli, and reduced endothelial cell numbers. We suggested that glomerular endothelial cells were more susceptible to radiation-induced cellular senescence. In conclusion, the current study is the first to identify the important role of radiation-induced cellular senescence, mainly derived from glomerular endothelial cells, for the development of glomerular injury.
Collapse
Affiliation(s)
- Sae Aratani
- Department of Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan. .,Department of Analytic Human Pathology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | - Masako Tagawa
- Department of Analytic Human Pathology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Shinya Nagasaka
- Department of Analytic Human Pathology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yukinao Sakai
- Department of Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Shuichi Tsuruoka
- Department of Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|