1
|
Yang L, Cai M, Zhong L, Yin Y, Xie Y, Xie S, Hu Y, Zhang J. Yellow mealworm ( Tenebrio molitor) meal in diets of grass carp ( Ctenopharyngodon idellus): Effects on growth performance, antioxidant capacity, immunity, intestinal morphology, and intestinal microbiota. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 21:70-83. [PMID: 40416717 PMCID: PMC12099804 DOI: 10.1016/j.aninu.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/07/2024] [Accepted: 01/09/2025] [Indexed: 05/27/2025]
Abstract
The impacts of substituting dietary soybean meal (SBM) with yellow mealworm meal (YMM) were investigated during a 56-day feeding trial on growth, antioxidant capacity, immunity, intestinal morphology, and intestinal microbiota of grass carp. A total of 750 grass carp were divided into 5 groups (3 replications per group and 50 fish per replication) with different levels of YMM: SBM (control group), H25, H50, H75, and H100, for 8 weeks. The results showed that dietary YMM significantly increased final body weight (FW), weight gain (WG), and protein efficiency ratio (PER) in H25 group (P < 0.05); however, complete substitution showed the opposite trend (P < 0.05 for FW and WG). The liver antioxidant capacity was improved, manifested by enhanced superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) activities, glutathione (GSH) content and up-regulated antioxidant-related genes mediated by the Keap1-Nrf2 signaling pathway in the H25 group (P < 0.05). However, in the H100 group oxidative stress occurred in parallel with impairment of hepatic function. Intestinal inflammation was aggravated in the H100 group as evidenced by the up-regulated pro-inflammatory gene expression mediated by the NF-κB pathway (P < 0.05). Additionally, the activity of intestinal digestive enzymes for the grass carp was significantly reduced and accompanied with intestinal mucosal barrier dysfunction in the H100 group (P < 0.05). In summary, replacement of SBM with 25% YMM showed positive influences on growth, antioxidant capacity, immunity, and intestinal health. Conversely, complete replacement of SBM with YMM triggered oxidative stress, caused liver function disorder, and impaired intestinal health in grass carp.
Collapse
Affiliation(s)
- Linlin Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Minglang Cai
- College of Fisheries, Hunan Agricultural University, Changsha 410128, China
| | - Lei Zhong
- College of Fisheries, Hunan Agricultural University, Changsha 410128, China
| | - Yulong Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Yonghong Xie
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Shouqi Xie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Chinese Academy of Sciences, Wuhan 430072, China
| | - Yi Hu
- College of Fisheries, Hunan Agricultural University, Changsha 410128, China
| | - Junzhi Zhang
- College of Fisheries, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
2
|
Mansouri B, Moradi A, Saba F. Blood oxidative stress parameters in hospital workers occupationally exposed to low doses of ionizing radiation: A systematic review and meta-analysis. Heliyon 2024; 10:e39989. [PMID: 39748987 PMCID: PMC11693902 DOI: 10.1016/j.heliyon.2024.e39989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 01/04/2025] Open
Abstract
This study conducted a systematic review and meta-analysis to explore the relationship between blood oxidative stress biomarkers and exposure to low-dose ionizing radiation in medical radiation workers. The researchers searched PubMed, Scopus, Web of Science, and Google Scholar for relevant studies until February 2023. They assessed the quality of the studies using the Newcastle‒Ottawa Scale (NOS) and used a random-effects model to combine the results. The I-square test was employed to assess study heterogeneity. The effect sizes were represented by standardized mean differences (proxied by Hedges' g) with a 95 % confidence interval. Out of 295 initial articles, 38 studies met the inclusion criteria for the meta-analysis. The systematic review results revealed a significant difference in blood oxidative stress biomarkers with exposure to low-dose ionizing radiation in medical radiation workers. Furthermore, the overall effect size of MDA was notably higher than that of the control group (p < 0.05). However, the effect size did not show any significant difference between the two groups for other parameters (SMDs ranged from [-0.92, 2.10] for 8-OHdG, [-3.47, 4.48] for reduced glutathione, [-1.08, 3.61] for CAT, [-5.03, 18.35] for SOD, [-2.52, 2.56] for TAC (p > 0.05)).
Collapse
Affiliation(s)
- Borhan Mansouri
- Substance Abuse Prevention Research Center, Research Institute for Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Aida Moradi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fakhredin Saba
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Medical Laboratory Science, School of Paramedical, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
3
|
Xie Y, Liu X, Xie D, Zhang W, Zhao H, Guan H, Zhou PK. Voltage-dependent anion channel 1 mediates mitochondrial fission and glucose metabolic reprogramming in response to ionizing radiation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174246. [PMID: 38955266 DOI: 10.1016/j.scitotenv.2024.174246] [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: 02/04/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
The ionizing radiation (IR) represents a formidable challenge as an environmental factor to mitochondria, leading to disrupt cellular energy metabolism and posing health risks. Although the deleterious impacts of IR on mitochondrial function are recognized, the specific molecular targets remain incompletely elucidated. In this study, HeLa cells subjected to γ-rays exhibited concomitant oxidative stress, mitochondrial structural alterations, and diminished ATP production capacity. The γ-rays induced a dose-dependent induction of mitochondrial fission, simultaneously manifested by an elevated S616/S637 phosphorylation ratio of the dynamin-related protein 1 (DRP1) and a reduction in the expression of the mitochondrial fusion protein mitofusin 2 (MFN2). Knockdown of DRP1 effectively mitigated γ-rays-induced mitochondrial network damage, implying that DRP1 phosphorylation may act as an effector of radiation-induced mitochondrial damage. The mitochondrial outer membrane protein voltage-dependent anion channel 1 (VDAC1) was identified as a crucial player in IR-induced mitochondrial damage. The VDAC1 inhibitor 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS), counteracts the excessive mitochondrial fission induced by γ-rays, consequently rebalancing the glycolytic and oxidative phosphorylation equilibrium. This metabolic shift was uncovered to enhance glycolytic capacity, thus fortifying cellular resilience and elevating the radiosensitivity of cancer cells. These findings elucidate the intricate regulatory mechanisms governing mitochondrial morphology under radiation response. It is anticipated that the development of targeted drugs directed against VDAC1 may hold promise in augmenting the sensitivity of tumor cells to radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Ying Xie
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, PR China; Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, PR China
| | - Xiaochang Liu
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Dafei Xie
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Wen Zhang
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Hongling Zhao
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Hua Guan
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China.
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China.
| |
Collapse
|
4
|
Kumar S, Suman S, Angdisen J, Moon BH, Kallakury BVS, Datta K, Fornace AJ. Effects of High-Linear-Energy-Transfer Heavy Ion Radiation on Intestinal Stem Cells: Implications for Gut Health and Tumorigenesis. Cancers (Basel) 2024; 16:3392. [PMID: 39410012 PMCID: PMC11475725 DOI: 10.3390/cancers16193392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Heavy ion radiation, prevalent in outer space and relevant for radiotherapy, is densely ionizing and poses a risk to intestinal stem cells (ISCs), which are vital for maintaining intestinal homeostasis. Earlier studies have shown that heavy-ion radiation can cause chronic oxidative stress, persistent DNA damage, cellular senescence, and the development of a senescence-associated secretory phenotype (SASP) in mouse intestinal mucosa. However, the specific impact on different cell types, particularly Lgr5+ intestinal stem cells (ISCs), which are crucial for maintaining cellular homeostasis, GI function, and tumor initiation under genomic stress, remains understudied. Using an ISCs-relevant mouse model (Lgr5+ mice) and its GI tumor surrogate (Lgr5+Apc1638N/+ mice), we investigated ISCs-specific molecular alterations after high-LET radiation exposure. Tissue sections were assessed for senescence and SASP signaling at 2, 5 and 12 months post-exposure. Lgr5+ cells exhibited significantly greater oxidative stress following 28Si irradiation compared to γ-ray or controls. Both Lgr5+ cells and Paneth cells showed signs of senescence and developed a senescence-associated secretory phenotype (SASP) after 28Si exposure. Moreover, gene expression of pro-inflammatory and pro-growth SASP factors remained persistently elevated for up to a year post-28Si irradiation. Additionally, p38 MAPK and NF-κB signaling pathways, which are critical for stress responses and inflammation, were also upregulated after 28Si radiation. Transcripts involved in nutrient absorption and barrier function were also altered following irradiation. In Lgr5+Apc1638N/+ mice, tumor incidence was significantly higher in those exposed to 28Si radiation compared to the spontaneous tumorigenesis observed in control mice. Our results indicate that high-LET 28Si exposure induces persistent DNA damage, oxidative stress, senescence, and SASP in Lgr5+ ISCs, potentially predisposing astronauts to altered nutrient absorption, barrier function, and GI carcinogenesis during and after a long-duration outer space mission.
Collapse
Affiliation(s)
- Santosh Kumar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shubhankar Suman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jerry Angdisen
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Bo-Hyun Moon
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | - Kamal Datta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Albert J. Fornace
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
5
|
Nagata K, Nishimura M, Daino K, Nishimura Y, Hattori Y, Watanabe R, Iizuka D, Yokoya A, Suzuki K, Kakinuma S, Imaoka T. Luminal progenitor and mature cells are more susceptible than basal cells to radiation-induced DNA double-strand breaks in rat mammary tissue. JOURNAL OF RADIATION RESEARCH 2024; 65:640-650. [PMID: 39238338 PMCID: PMC11420845 DOI: 10.1093/jrr/rrae067] [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: 02/05/2024] [Revised: 06/07/2024] [Indexed: 09/07/2024]
Abstract
Ionizing radiation promotes mammary carcinogenesis. Induction of DNA double-strand breaks (DSBs) is the initial event after radiation exposure, which can potentially lead to carcinogenesis, but the dynamics of DSB induction and repair are not well understood at the tissue level. In this study, we used female rats, which have been recognized as a useful experimental model for studying radiation effects on the mammary gland. We focused on differences in DSB kinetics among basal cells, luminal progenitor and mature cells in different parts of the mammary duct. 53BP1 foci were used as surrogate markers of DSBs, and 53BP1 foci in each mammary epithelial cell in immunostained tissue sections were counted 1-24 h after irradiation and fitted to an exponential function of time. Basal cells were identified as cytokeratin (CK) 14+ cells, luminal progenitor cells as CK8 + 18low cells and luminal mature cells as CK8 + 18high cells. The number of DSBs per nucleus tended to be higher in luminal cells than basal cells at 1 h post-irradiation. A model analysis indicated that basal cells in terminal end buds (TEBs), which constitute the leading edge of the mammary duct, had significantly fewer initial DSBs than the two types of luminal cells, and there was no significant difference in initial amount among the cell types in the subtending duct. The repair rate did not differ among mammary epithelial cell types or their locations. Thus, luminal progenitor and mature cells are more susceptible to radiation-induced DSBs than are basal cells in TEBs.
Collapse
Affiliation(s)
- Kento Nagata
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, 4–9–1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Mayumi Nishimura
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, 4–9–1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kazuhiro Daino
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, 4–9–1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Yukiko Nishimura
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, 4–9–1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Yuya Hattori
- Department of Electrical Engineering and Information Science, Faculty of Electrical Engineering and Information Science, National Institute of Technology Kure College, 2–2–11 Aga-minami, Kure, Hiroshima 737-8506, Japan
| | - Ritsuko Watanabe
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, 4–9–1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Daisuke Iizuka
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, 4–9–1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Akinari Yokoya
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, 4–9–1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Keiji Suzuki
- Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University, 1–12–4 Sakamoto, Nagasaki 852-8523, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, 4–9–1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Tatsuhiko Imaoka
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, 4–9–1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
6
|
Kumar K, Fornace AJ, Suman S. 8-OxodG: A Potential Biomarker for Chronic Oxidative Stress Induced by High-LET Radiation. DNA 2024; 4:221-238. [PMID: 39268222 PMCID: PMC11391509 DOI: 10.3390/dna4030015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Oxidative stress-mediated biomolecular damage is a characteristic feature of ionizing radiation (IR) injury, leading to genomic instability and chronic health implications. Specifically, a dose- and linear energy transfer (LET)-dependent persistent increase in oxidative DNA damage has been reported in many tissues and biofluids months after IR exposure. Contrary to low-LET photon radiation, high-LET IR exposure is known to cause significantly higher accumulations of DNA damage, even at sublethal doses, compared to low-LET IR. High-LET IR is prevalent in the deep space environment (i.e., beyond Earth's magnetosphere), and its exposure could potentially impair astronauts' health. Therefore, the development of biomarkers to assess and monitor the levels of oxidative DNA damage can aid in the early detection of health risks and would also allow timely intervention. Among the recognized biomarkers of oxidative DNA damage, 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-OxodG) has emerged as a promising candidate, indicative of chronic oxidative stress. It has been reported to exhibit differing levels following equivalent doses of low- and high-LET IR. This review discusses 8-OxodG as a potential biomarker of high-LET radiation-induced chronic stress, with special emphasis on its potential sources, formation, repair mechanisms, and detection methods. Furthermore, this review addresses the pathobiological implications of high-LET IR exposure and its association with 8-OxodG. Understanding the association between high-LET IR exposure-induced chronic oxidative stress, systemic levels of 8-OxodG, and their potential health risks can provide a framework for developing a comprehensive health monitoring biomarker system to safeguard the well-being of astronauts during space missions and optimize long-term health outcomes.
Collapse
Affiliation(s)
- Kamendra Kumar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Albert J Fornace
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shubhankar Suman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
7
|
Forenzo C, Larsen J. Bridging clinical radiotherapy and space radiation therapeutics through reactive oxygen species (ROS)-triggered delivery. Free Radic Biol Med 2024; 219:88-103. [PMID: 38631648 DOI: 10.1016/j.freeradbiomed.2024.04.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
This review explores the convergence of clinical radiotherapy and space radiation therapeutics, focusing on ionizing radiation (IR)-generated reactive oxygen species (ROS). IR, with high-energy particles, induces precise cellular damage, particularly in cancer treatments. The paper discusses parallels between clinical and space IR, highlighting unique characteristics of high-charge and energy particles in space and potential health risks for astronauts. Emphasizing the parallel occurrence of ROS generation in both clinical and space contexts, the review identifies ROS as a crucial factor with dual roles in cellular responses and potential disease initiation. The analysis covers ROS generation mechanisms, variations, and similarities in terrestrial and extraterrestrial environments leading to innovative ROS-responsive delivery systems adaptable for both clinical and space applications. The paper concludes by discussing applications of personalized ROS-triggered therapeutic approaches and discussing the challenges and prospects of implementing these strategies in clinical radiotherapy and extraterrestrial missions. Overall, it underscores the potential of ROS-targeted delivery for advancing therapeutic strategies in terrestrial clinical settings and space exploration, contributing to human health improvement on Earth and beyond.
Collapse
Affiliation(s)
- Chloe Forenzo
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, 29631, USA
| | - Jessica Larsen
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, 29631, USA; Department of Bioengineering, Clemson University, Clemson, SC, 29631, USA.
| |
Collapse
|
8
|
Ferraro S, Dave A, Cereda C, Verduci E, Marcovina S, Zuccotti G. Space research to explore novel biochemical insights on Earth. Clin Chim Acta 2024; 558:119673. [PMID: 38621588 DOI: 10.1016/j.cca.2024.119673] [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: 02/19/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/17/2024]
Abstract
Travel to space has overcome unprecedent technological challenges and this has resulted in transfer of these technological results on Earth to better our lives. Health technology, medical devices, and research advancements in human biology are the first beneficiaries of this transfer. The real breakthrough came with the International Space Station, which endorsed multidisciplinary international scientific collaborations and boosted the research on pathophysiological adaptation of astronauts to life on space. These studies evidenced that life in space appeared to have exposed the astronauts to an accelerated aging-related pathophysiological dysregulation across multiple systems. In this review we emphasize the interaction between several biomarkers and their alteration in concentrations/expression/function by space stress factors. These altered interactions, suggest that different biochemical and hormonal factors, and cell signals, contribute to a complex network of pathophysiological mechanisms, orchestrating the homeostatic dysregulation of various organs/metabolic pathways. The main effects of space travel on altering cell organelles biology, ultrastructure, and cross-talk, have been observed in cell aging as well as in the disruption of metabolic pathways, which are also the causal factor of rare inherited metabolic disorders, one of the major pediatric health issue. The pathophysiologic breakthrough from space research could allow the development of precision health both on Earth and Space by promoting the validation of improved biomarker-based risk scores and the exploration of new pathophysiologic hypotheses and therapeutic targets. Nonstandard abbreviations: International Space Station (ISS), Artificial Intelligence (AI), European Space Agency (ESA), National Aeronautics and Space Agency (NASA), Low Earth Orbit (LEO), high sensitive troponin (hs-cTn), high sensitive troponin I (hs-cTn I), high sensitive troponin T, Brain Natriuretic Peptide (BNP), N terminal Brain Natriuretic Peptide (NT-BNP), cardiovascular disease (CVD), parathyroid hormone (PTH), urinary hydroxyproline (uHP), urinary C- and N-terminal telopeptides (uCTX and uNTX), pyridinoline (PYD), deoxypyridinoline (DPD), half-time (HF), serum Bone Alkaline Phosphatase (sBSAP), serum Alkaline Phosphatase (sAP), Carboxy-terminal Propeptide of Type 1 Procollagen (P1CP), serum Osteocalcin (sOC)), advanced glycation end products (AGEs), glycated hemoglobin A1c (HbA1c), Insulin-like growth factor 1 (IGF1), Growth Hormone (GH), amino acid (AA), β-hydroxy-β methyl butyrate (HMB), maple syrup urine disease (MSUD), non-communicable diseases (NCDs).
Collapse
Affiliation(s)
- Simona Ferraro
- Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy.
| | - Anilkumar Dave
- Space Economy and Open Innovation, Darwix srl, Venice, Italy
| | - Cristina Cereda
- Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy; Center of Functional Genomics and Rare Diseases
| | - Elvira Verduci
- Department of Health Sciences, University of Milan, Milan, Italy; Metabolic Diseases Unit, Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy
| | | | - Gianvincenzo Zuccotti
- Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy; Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| |
Collapse
|
9
|
Шеремета МС, Трухин АА, Ярцев ВД, Юдаков ДВ, Корчагина МО, Годжаева СА. [The lacrimal apparatus as an organ at risk during radionuclide therapy]. PROBLEMY ENDOKRINOLOGII 2024; 70:13-17. [PMID: 38433537 PMCID: PMC10926245 DOI: 10.14341/probl13163] [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: 08/22/2022] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 03/05/2024]
Abstract
Within the framework of the article, the authors analyzed the available information about the damage to the lacrimal apparatus during radionuclide therapy. In focus of article lesions of the lacrimal production system, the main and accessory lacrimal glands, as well as lacrimal drainage are considered. It was found that damage to the lacrimal apparatus is characteristic of 131I therapy for thyroid cancer, as well as for radioligand therapy using anti-PSMA antibodies labeled with 177Lu and 225Ac. 177Lu-PSMA and 225Ac-PSMA may damage the lacrimal gland with the formation of a clinically pronounced "dry eye syndrome". The pathogenesis of such lesions is associated with the accumulation of a radioisotope in the tissues of the lacrimal apparatus, while during therapy with 131I, accumulation is realized due to the expression of the sodium-iodine symporter in the nasolacrimal duct, and during therapy with 177Lu-PSMA and 225Ac-PSMA, the radiobiological effect is realized in connection with the expression PSMA by lacrimal tissue. An analysis of the available sources showed that to date there are no results of systematic studies on the problem, there is a lack of knowledge regarding the individual risks of developing these complications, methods for their prevention that have proven effectiveness have not been developed, and the treatment methods used, having relatively low efficiency, are not specialized. The authors concluded that the strengthening of interdisciplinary interaction, as well as the organization verification methodology and correct studies, can contribute to solving problems related to the study of the complications under consideration.
Collapse
Affiliation(s)
- М. С. Шеремета
- Национальный медицинский исследовательский центр эндокринологии
| | - А. А. Трухин
- Национальный медицинский исследовательский центр эндокринологии
| | - В. Д. Ярцев
- ФГБНУ «НИИ глазных болезней им. М.М. Краснова»
| | - Д. В. Юдаков
- Национальный медицинский исследовательский центр эндокринологии
| | - М. О. Корчагина
- Национальный медицинский исследовательский центр эндокринологии
| | - С. А. Годжаева
- ФГБНУ «НИИ глазных болезней им. М.М. Краснова»; Первый московский государственный медицинский университет имени И.М. Сеченова
| |
Collapse
|
10
|
Shakyawal S, Namdev N, Ahmad Z, Mahobiya P. Effects of Ultraviolet B Radiation on the Function of the Testicles, Expression of Caspase-3 and NOS-2, and the Protective Role of Naringin in Mice. Reprod Sci 2024; 31:452-468. [PMID: 37814202 DOI: 10.1007/s43032-023-01366-x] [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: 03/27/2023] [Accepted: 09/17/2023] [Indexed: 10/11/2023]
Abstract
In today's evolving global environment, reproductive dysfunctions brought on by various environmental toxins are of greatest concern. Radiation is a constant threat to living things, causing both genetic and cellular changes that result in mutations and cell death. It is thought that ultraviolet B (UVB) radiation we are exposed to daily has biological effects on rats and humans that are both short and long term. Due to the damaging effects of UVB radiation on the living system, this study explores the automatic mechanism by which a certain level of radiation induces oxidative stress, which is further controlled by the antioxidant activity of naringin (NG). In our study, male Swiss albino mice were exposed to UVB irradiation, which altered mice's body and testes weight, hormonal imbalance, biochemical parameters, and histo-morphometric parameter. In addition, we chose naringin's UVB irradiation deterrent effect. Twenty-four healthy adult male Swiss albino mice weighing 25-35 g were chosen at random. For 15 days of exposure, they were divided into four groups at random: group I-control, group II-UVB exposure (2 h per day), group III-UVB exposure with naringin (NG) (80 mg/kg, bw), and group IV-naringin (NG) (80 mg/kg, bw). Compared to the control group, UVB irradiation causes alterations in the animal body weight, testes weight, hormones, enzymatic and non-enzymatic assays, and histological parameters. It was seen that NG retrieved the alterations in parameters caused by UVB irradiation. The UVB radiation exposure on mice caused the testicular dysfunction drastically, while the naringin recapitulates testis functioning.
Collapse
Affiliation(s)
- Shashank Shakyawal
- Endocrinology Laboratory, Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh, India
| | - Narendra Namdev
- Endocrinology Laboratory, Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh, India
| | - Zaved Ahmad
- Cancer Biology Laboratory, Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh, India
| | - Payal Mahobiya
- Endocrinology Laboratory, Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh, India.
| |
Collapse
|
11
|
Veluthakal R, Esparza D, Hoolachan JM, Balakrishnan R, Ahn M, Oh E, Jayasena CS, Thurmond DC. Mitochondrial Dysfunction, Oxidative Stress, and Inter-Organ Miscommunications in T2D Progression. Int J Mol Sci 2024; 25:1504. [PMID: 38338783 PMCID: PMC10855860 DOI: 10.3390/ijms25031504] [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: 12/22/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Type 2 diabetes (T2D) is a heterogenous disease, and conventionally, peripheral insulin resistance (IR) was thought to precede islet β-cell dysfunction, promoting progression from prediabetes to T2D. New evidence suggests that T2D-lean individuals experience early β-cell dysfunction without significant IR. Regardless of the primary event (i.e., IR vs. β-cell dysfunction) that contributes to dysglycemia, significant early-onset oxidative damage and mitochondrial dysfunction in multiple metabolic tissues may be a driver of T2D onset and progression. Oxidative stress, defined as the generation of reactive oxygen species (ROS), is mediated by hyperglycemia alone or in combination with lipids. Physiological oxidative stress promotes inter-tissue communication, while pathological oxidative stress promotes inter-tissue mis-communication, and new evidence suggests that this is mediated via extracellular vesicles (EVs), including mitochondria containing EVs. Under metabolic-related stress conditions, EV-mediated cross-talk between β-cells and skeletal muscle likely trigger mitochondrial anomalies leading to prediabetes and T2D. This article reviews the underlying molecular mechanisms in ROS-related pathogenesis of prediabetes, including mitophagy and mitochondrial dynamics due to oxidative stress. Further, this review will describe the potential of various therapeutic avenues for attenuating oxidative damage, reversing prediabetes and preventing progression to T2D.
Collapse
Affiliation(s)
- Rajakrishnan Veluthakal
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope Beckman Research Institute, 1500 E. Duarte Rd, Duarte, CA 91010, USA; (D.E.); (J.M.H.); (R.B.); (M.A.); (E.O.); (C.S.J.)
| | | | | | | | | | | | | | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope Beckman Research Institute, 1500 E. Duarte Rd, Duarte, CA 91010, USA; (D.E.); (J.M.H.); (R.B.); (M.A.); (E.O.); (C.S.J.)
| |
Collapse
|
12
|
Chasara RS, Ajayi TO, Leshilo DM, Poka MS, Witika BA. Exploring novel strategies to improve anti-tumour efficiency: The potential for targeting reactive oxygen species. Heliyon 2023; 9:e19896. [PMID: 37809420 PMCID: PMC10559285 DOI: 10.1016/j.heliyon.2023.e19896] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
The cellular milieu in which malignant growths or cancer stem cells reside is known as the tumour microenvironment (TME). It is the consequence of the interactivity amongst malignant and non-malignant cells and directly affects cancer development and progression. Reactive oxygen species (ROS) are chemically reactive molecules that contain oxygen, they are generated because of numerous endogenous and external factors. Endogenous ROS produced from mitochondria is known to significantly increase intracellular oxidative stress. In addition to playing a key role in several biological processes both in healthy and malignant cells, ROS function as secondary messengers in cell signalling. At low to moderate concentrations, ROS serves as signalling transducers to promote cancer cell motility, invasion, angiogenesis, and treatment resistance. At high concentrations, ROS can induce oxidative stress, leading to DNA damage, lipid peroxidation and protein oxidation. These effects can result in cell death or trigger signalling pathways that lead to apoptosis. The creation of innovative therapies and cancer management techniques has been aided by a thorough understanding of the TME. At present, surgery, chemotherapy, and radiotherapy, occasionally in combination, are the most often used methods for tumour treatment. The current challenge that these therapies face is the lack of spatiotemporal application specifically at the lesion which results in toxic effects on healthy cells associated with off-target drug delivery and undesirably high doses. Nanotechnology can be used to specifically deliver various chemicals via nanocarriers to target tumour cells, thereby increasing the accumulation of ROS-inducing agents at the site of the tumour. Nanoparticles can be engineered to release ROS-inducing agents in a controlled manner to the TME that will in turn react with the ROS to either increase or decrease it, thereby improving antitumour efficiency. Nano-delivery systems such as liposomes, nanocapsules, solid lipid nanoparticles and nanostructured lipid carriers were explored for the up/down-regulation of ROS. This review will discuss the use of nanotechnology in targeting and altering the ROS in the TME.
Collapse
Affiliation(s)
- Rumbidzai Sharon Chasara
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0204, South Africa
| | - Taiwo Oreoluwa Ajayi
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0204, South Africa
| | - Dineo Motjoadi Leshilo
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0204, South Africa
| | - Madan Sai Poka
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0204, South Africa
| | - Bwalya Angel Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0204, South Africa
| |
Collapse
|
13
|
Golomb BA, Han JH. Adverse effect propensity: A new feature of Gulf War illness predicted by environmental exposures. iScience 2023; 26:107363. [PMID: 37554469 PMCID: PMC10405325 DOI: 10.1016/j.isci.2023.107363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 05/26/2023] [Accepted: 07/10/2023] [Indexed: 08/10/2023] Open
Abstract
A third of 1990-1 Gulf-deployed personnel developed drug/chemical-induced multisymptom illness, "Gulf War illness" (GWI). Veterans with GWI (VGWI) report increased drug/exposure adverse effects (AEs). Using previously collected data from a case-control study, we evaluated whether the fraction of exposures that engendered AEs ("AE Propensity") is increased in VGWI (it was); whether AE Propensity is related to self-rated "chemical sensitivity" (it did); and whether specific exposures "predicted" AE Propensity (they did). Pesticides and radiation exposure were significant predictors, with copper significantly "protective"-in the total sample (adjusted for GWI-status) and separately in VGWI and controls, on multivariable regression. Mitochondrial impairment and oxidative stress (OS) underlie AEs from many exposures irrespective of nominal specific mechanism. We hypothesize that mitochondrial toxicity and interrelated OS from pesticides and radiation position people on the steep part of the curve of mitochondrial impairment and OS versus symptom/biological disruption, amplifying impact of new exposures. Copper, meanwhile, is involved in critical OS detoxification processes.
Collapse
Affiliation(s)
- Beatrice A. Golomb
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jun Hee Han
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
14
|
Shichijo K, Takatsuji T, Uzbekov D, Chaizhunusova N, Shabdarbaeva D, Kurisu M, Takahashi Y, Stepanenko V, Azhimkhanov A, Hoshi M. Radiation makes cells select the form of death dependent on external or internal exposure: apoptosis or pyroptosis. Sci Rep 2023; 13:12002. [PMID: 37491560 PMCID: PMC10368746 DOI: 10.1038/s41598-023-38789-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 07/14/2023] [Indexed: 07/27/2023] Open
Abstract
Internal radiation exposure from neutron-induced radioisotopes environmentally activated following atomic bombing or nuclear accidents should be considered for a complete picture of pathologic effects on survivors. Acute and localized high dose radiation exposure from hot particles taken into the body must induce cell death and severe damage to tissues, whether they are proliferating or not. However, very little the cellular and molecular mechanisms underlying this internal radiation pathology has been investigated. Male Wistar rats were internally exposed to 56MnO2 powder by inhalation. Small intestine samples were investigated by histological staining at acute phase (6 h, 3 days and 14 days) and late phase (2, 6 and 8 months) after the exposure. Histological location and chemical properties of the hot particles embedded in small intestinal tissues were analyzed by synchrotron radiation-X-ray fluorescence-X-ray absorption near-edge structure (SR-XRF-XANES). Hot particles located in the intestinal cavity were identified as accumulations of Mn and iron. Pathological changes showed evidence of crypt shortening, massive cell death at the position of stem cell zone, including apoptosis and pyroptosis from 6 h through 8 months in the internal exposed rats.
Collapse
Affiliation(s)
- Kazuko Shichijo
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| | - Toshihiro Takatsuji
- Nagasaki University, 1-14 Bunkyo, Nagasaki, 852-8521, Japan
- School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi, 321-0293, Japan
| | - Darkhan Uzbekov
- Department of Pathological Anatomy and Forensic Medicine, Semey State Medical University, Abay Str., 103, Semey, 071400, Kazakhstan
| | - Nailya Chaizhunusova
- Department of Pathological Anatomy and Forensic Medicine, Semey State Medical University, Abay Str., 103, Semey, 071400, Kazakhstan
| | - Dariya Shabdarbaeva
- Department of Pathological Anatomy and Forensic Medicine, Semey State Medical University, Abay Str., 103, Semey, 071400, Kazakhstan
| | - Minako Kurisu
- Department of Earth and Planetary Science, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Research Institute for Marine Resources Utilization, Japan Agency for Marine-Earth Science and Technology, 2-15 Natsusima-cho, Yokosuka-shi, Kanagawa, 237-0061, Japan
| | - Yoshio Takahashi
- Department of Earth and Planetary Science, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Valeriy Stepanenko
- A.Tsyb Medical Radiological Research Center-National Medical Research Center of Radiology, Ministry of Health of Russian Federation, 249036, Obninsk, Russia
| | - Almas Azhimkhanov
- National Nuclear Center of the Republic of Kazakhstan, Beibyt atom st., 2B, Kurchatov, 071100, Kazakhstan
| | - Masaharu Hoshi
- The Center for Peace, Hiroshima University, Higashi-senda-machi, Naka-ku, Hiroshima, 730-0053, Japan
| |
Collapse
|
15
|
Kumar K, Kumar S, Datta K, Fornace AJ, Suman S. High-LET-Radiation-Induced Persistent DNA Damage Response Signaling and Gastrointestinal Cancer Development. Curr Oncol 2023; 30:5497-5514. [PMID: 37366899 DOI: 10.3390/curroncol30060416] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/28/2023] Open
Abstract
Ionizing radiation (IR) dose, dose rate, and linear energy transfer (LET) determine cellular DNA damage quality and quantity. High-LET heavy ions are prevalent in the deep space environment and can deposit a much greater fraction of total energy in a shorter distance within a cell, causing extensive DNA damage relative to the same dose of low-LET photon radiation. Based on the DNA damage tolerance of a cell, cellular responses are initiated for recovery, cell death, senescence, or proliferation, which are determined through a concerted action of signaling networks classified as DNA damage response (DDR) signaling. The IR-induced DDR initiates cell cycle arrest to repair damaged DNA. When DNA damage is beyond the cellular repair capacity, the DDR for cell death is initiated. An alternative DDR-associated anti-proliferative pathway is the onset of cellular senescence with persistent cell cycle arrest, which is primarily a defense mechanism against oncogenesis. Ongoing DNA damage accumulation below the cell death threshold but above the senescence threshold, along with persistent SASP signaling after chronic exposure to space radiation, pose an increased risk of tumorigenesis in the proliferative gastrointestinal (GI) epithelium, where a subset of IR-induced senescent cells can acquire a senescence-associated secretory phenotype (SASP) and potentially drive oncogenic signaling in nearby bystander cells. Moreover, DDR alterations could result in both somatic gene mutations as well as activation of the pro-inflammatory, pro-oncogenic SASP signaling known to accelerate adenoma-to-carcinoma progression during radiation-induced GI cancer development. In this review, we describe the complex interplay between persistent DNA damage, DDR, cellular senescence, and SASP-associated pro-inflammatory oncogenic signaling in the context of GI carcinogenesis.
Collapse
Affiliation(s)
- Kamendra Kumar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Santosh Kumar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Kamal Datta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology and Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Albert J Fornace
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology and Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shubhankar Suman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology and Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
16
|
Mousavikia SN, Bahreyni Toossi MT, Khademi S, Soukhtanloo M, Azimian H. Evaluation of micronuclei and antioxidant status in hospital radiation workers occupationally exposed to low-dose ionizing radiation. BMC Health Serv Res 2023; 23:540. [PMID: 37226157 DOI: 10.1186/s12913-023-09516-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/08/2023] [Indexed: 05/26/2023] Open
Abstract
PURPOSE There is scientific evidence that ionizing radiation (IR) can be responsible for various health hazards that are one of the concerns in occupational exposure. This study was performed to evaluate DNA damage and antioxidant status in hospital workers who are occupationally exposed to low doses of IR. MATERIALS AND METHODS In this study, twenty occupationally exposed to low doses of IR (CT and angiography) comprising with control groups which matched them. In order to investigate the effects of chronic irradiation of radiation workers, Micronuclei (MN) frequency and the antioxidant activity of Superoxide Dismutase (SOD), Catalase (CAT) and Total Antioxidant Capacity (TAC) were measured. Then, to check adaptation against high challenge dose, the samples (in all groups) were irradiated in vitro and MN frequency was compared. Finally, to investigated the effect of the high dose after the acute and chronic low dose of ionizing radiation, MN frequency was compared in two groups (the control group that was to in-vitro irradiated (acute low dose + high dose) and radiation workers (chronic low dose + high dose)). RESULTS MN frequency in the occupationally exposed group (n = 30) increased significantly when compared to the control group (p-value < 0.0001). However, chronic irradiation of radiation workers could not lead to an adaptive Sresponse, while acute low-doses could produce this effect (p-value ˂ 0.05). In addition, the activity levels of antioxidant enzymes SOD, CAT, and TAC were not statistically different between the radiation workers and the control group (p-value > 0.05). CONCLUSIONS We observed that exposure to low doses of IR leads to increased cytogenetic damage, could not cause an adaptive-response, and improve antioxidant capacity in radiation workers. Controlling healthcare workers' exposure is the first step to improving the health of hospital workers and the quality of patient care, thus decreasing human and economic costs.
Collapse
Affiliation(s)
- S N Mousavikia
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - M T Bahreyni Toossi
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - S Khademi
- Department of Radiology Technology, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - M Soukhtanloo
- Department of Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - H Azimian
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Mirieri CK, Abd-Alla AM, Ros VI, van Oers MM. Evaluating the Effect of Irradiation on the Densities of Two RNA Viruses in Glossina morsitans morsitans. INSECTS 2023; 14:397. [PMID: 37103212 PMCID: PMC10140815 DOI: 10.3390/insects14040397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 06/19/2023]
Abstract
Tsetse flies are cyclic vectors of Trypanosoma parasites, which cause debilitating diseases in humans and animals. To decrease the disease burden, the number of flies is reduced using the sterile insect technique (SIT), where male flies are sterilized through irradiation and released into the field. This procedure requires the mass rearing of high-quality male flies able to compete with wild male flies for mating with wild females. Recently, two RNA viruses, an iflavirus and a negevirus, were discovered in mass-reared Glossina morsitans morsitans and named GmmIV and GmmNegeV, respectively. The aim of this study was to evaluate whether the densities of these viruses in tsetse flies are affected by the irradiation treatment. Therefore, we exposed tsetse pupae to various doses (0-150 Gy) of ionizing radiation, either in air (normoxia) or without air (hypoxia), for which oxygen was displaced by nitrogen. Pupae and/or emerging flies were collected immediately afterwards, and at three days post irradiation, virus densities were quantified through RT-qPCR. Generally, the results show that irradiation exposure had no significant impact on the densities of GmmIV and GmmNegeV, suggesting that the viruses are relatively radiation-resistant, even at higher doses. However, sampling over a longer period after irradiation would be needed to verify that densities of these insect viruses are not changed by the sterilisation treatment.
Collapse
Affiliation(s)
- Caroline K. Mirieri
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400 Vienna, Austria;
| | - Adly M.M. Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400 Vienna, Austria;
| | - Vera I.D. Ros
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Monique M. van Oers
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
18
|
Li Y, Bansal S, Sridharan V, Bansal S, Jayatilake MM, Fernández JA, Griffin JH, Boerma M, Cheema AK. Urinary Metabolomics for the Prediction of Radiation-Induced Cardiac Dysfunction. Metabolites 2023; 13:metabo13040525. [PMID: 37110184 PMCID: PMC10146652 DOI: 10.3390/metabo13040525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/02/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Survivors of acute radiation exposures are likely to experience delayed effects that manifest as injury in late-responding organs such as the heart. Noninvasive indicators of radiation-induced cardiac dysfunction are important in the prediction and diagnosis of this disease. In this study, we aimed to identify urinary metabolites indicative of radiation-induced cardiac damage by analyzing previously collected urine samples from a published study. The samples were collected from male and female wild-type (C57BL/6N) and transgenic mice constitutively expressing Activated Protein C (APCHi), a circulating protein with potential cardiac protective properties, that were exposed to 9.5 Gy of γ-rays. We utilized LC-MS-based metabolomics and lipidomics for the analysis of urine samples collected at 24 h, 1 week, 1 month, 3 months, and 6 months post-irradiation. Radiation caused perturbations in the TCA cycle, glycosphingolipid metabolism, fatty acid oxidation, purine catabolism, and amino acid metabolites, which were more prominent in wild-type (WT) mice compared to APCHi mice, suggesting a differential response between the two genotypes. After combining genotypes and sexes, we identified a multi-analyte urinary panel at early post-irradiation time points that predicted heart dysfunction using a logistic regression model with a discovery validation study design. These studies demonstrate the utility of a molecular phenotyping approach to develop a urinary biomarker panel predictive of delayed effects of ionizing radiation. It is important to note that no live mice were used or assessed in this study; instead, we focused solely on analyzing previously collected urine samples.
Collapse
Affiliation(s)
- Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA
- Departments of Biochemistry, Molecular, and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shivani Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, Department of Pharmaceutical Sciences, 4301 West Markham #522-10, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sunil Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Meth M. Jayatilake
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jose A. Fernández
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - John H. Griffin
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, 4301 West Markham #522-10, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Amrita K. Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA
- Departments of Biochemistry, Molecular, and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
19
|
Liu Y, Liu Z, Hu L, He L, Yang L, Qin Z, Xie Y, Peng X, Dai L. Function of stem cells in radiation-induced damage. Int J Radiat Biol 2023; 99:1483-1494. [PMID: 36912588 DOI: 10.1080/09553002.2023.2188935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/27/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE The aim of this review is to discuss previous studies on the function of stem cells in radiation-induced damage, and the factors affecting these processes, in the hope of improving our understanding of the different stem cells and the communication networks surrounding them. This is essential for the development of effective stem cell-based therapies to regenerate or replace normal tissues damaged by radiation. CONCLUSION In salivary glands, senescence-associated cytokines and inflammation-associated cells have a greater effect on stem cells. In the intestinal glands, Paneth cells strongly affect stem cell-mediated tissue regeneration after radiation treatment. In the pancreas, β-cells as well as protein C receptor positive (Procr) cells are expected to be key cells in the treatment of diabetes. In the bone marrow, a variety of cytokines such as CXC-chemokine ligand 12 (CXCL12) and stem cell factor (SCF), contribute to the functional recovery of hematopoietic stem cells after irradiation.
Collapse
Affiliation(s)
- Yingtong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Liqiang Hu
- West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ling He
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lianlian Yang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zijian Qin
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuping Xie
- Department of Oncology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Kumar S, Suman S, Moon BH, Fornace AJ, Datta K. Low dose radiation upregulates Ras/p38 and NADPH oxidase in mouse colon two months after exposure. Mol Biol Rep 2023; 50:2067-2076. [PMID: 36542238 PMCID: PMC10119992 DOI: 10.1007/s11033-022-08186-3] [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: 10/26/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Exposure to ionizing is known to cause persistent cellular oxidative stress and NADPH oxidase (Nox) is a major source of cellular oxidant production. Chronic oxidative stress is associated with a myriad of human diseases including gastrointestinal cancer. However, the roles of NADPH oxidase in relation of long-term oxidative stress in colonic epithelial cells after radiation exposure are yet to be clearly established. METHODS AND RESULTS Mice were exposed either to sham or to 0.5 Gy γ radiation, and NADPH oxidase, oxidative stress, and related signaling pathways were assessed in colon samples 60 days after exposure. Radiation exposure led to increased expression of colon-specific NADPH oxidase isoform, Nox1, as well as upregulation of its modifiers such as Noxa1 and Noxo1 at the mRNA and protein level. Co-immunoprecipitation experiments showed enhanced binding of Rac1, an activator of NADPH oxidase, to Nox1. Increased 4-hydroxynonenal, 8-oxo-dG, and γH2AX along with higher protein carbonylation levels suggest increased oxidative stress after radiation exposure. Immunoblot analysis demonstrates upregulation of Ras/p38 pathway, and Gata6 and Hif1α after irradiation. Increased staining of β-catenin, cyclinD1, and Ki67 after radiation was also observed. CONCLUSIONS In summary, data show that exposure to a low dose of radiation was associated with upregulation of NADPH oxidase and its modifiers along with increased Ras/p38/Gata6 signaling in colon. When considered along with oxidative damage and proliferative markers, our observations suggest that the NADPH oxidase pathway could be playing a critical role in propagating long-term oxidative stress after radiation with implications for colon carcinogenesis.
Collapse
Affiliation(s)
- Santosh Kumar
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
| | - Shubhankar Suman
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
| | - Bo-Hyun Moon
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
| | - Albert J Fornace
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Research Building, Room E518, 3970 Reservoir Rd., NW, Washington, DC, 20057, USA
| | - Kamal Datta
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA.
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Research Building, Room E518, 3970 Reservoir Rd., NW, Washington, DC, 20057, USA.
| |
Collapse
|
21
|
Vyas R, Kesari KK, Lukac N, Slama P, Roychoudhury S, Sisodia R. Radical scavenging activity of Chlorophytum borivilianum L. root extract and its protective role in cauda epididymal sperm integrity in Mus musculus after gamma irradiation. Front Cell Dev Biol 2023; 11:1064574. [PMID: 37025174 PMCID: PMC10070789 DOI: 10.3389/fcell.2023.1064574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 03/01/2023] [Indexed: 04/08/2023] Open
Abstract
Background: Chlorophytum borivilianum L. is a recognized herbal medicine for the management of impotency in South Asian countries. In Ayurveda, it is used for the management of multiple health conditions, including diabetes, infection, and cardiovascular diseases. Parts of the plant have been used as excellent antioxidants and scavengers of free radicals. Since oxidative stress plays an important role in spermatogenesis and fertility in male populations, this study evaluated the role of ethanolic extract of C. borivilianum roots in epididymal sperm maturation against adversities posed by ionizing gamma irradiation. Materials and methods: Antioxidant potential of C. borivilianum root extract (CRE) was evaluated through DPPH (2,2-diphenylpicrylhydrazyl) and NO (nitric oxide) scavenging assays. Four groups of healthy Swiss albino mice were constituted, which were labeled as follows: Group I: sham control, Group II: 7-day pre-treatment with 50 mg/kg CRE, Group III: 6 Gy irradiation without pre-treatment, and Group IV: 7-day pre-treatment with 50 mg/kg CRE and 6 Gy irradiation on day 7. Swiss albino mice were observed for 30 days and later sacrificed to evaluate sperm quality parameters. Results: CRE showed a remarkable antioxidant potential with IC50 values of 46.37 μg/ml and 98.39 μg/ml for DPPH and NO, respectively. A significant decline (p < 0.001) in cauda epididymal sperm count, motility, and viability was observed in Group III animals. Group IV also showed a substantial decline (p < 0.01) in all three parameters compared to Group I; nonetheless, these were significantly higher than Group III. Morphological alterations indicated a coiled and bent tail, with the presence of cytoplasmic droplets in Group III, which declined substantially in Group IV. The ultrastructure of sperm indicated higher curvature of hook in Group III than Group IV, indicating specific interferences in the sperm maturation process. Conclusion: It was concluded that pre-treatment with 50 mg/kg body weight of CRE could protect sperm during epididymal maturation against oxidative stress.
Collapse
Affiliation(s)
- Ruchi Vyas
- Department of Zoology, University of Rajasthan, Jaipur, India
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
- *Correspondence: Kavindra Kumar Kesari, ; Petr Slama, ; Shubhadeep Roychoudhury, ; Rashmi Sisodia,
| | - Norbert Lukac
- Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Petr Slama
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
- *Correspondence: Kavindra Kumar Kesari, ; Petr Slama, ; Shubhadeep Roychoudhury, ; Rashmi Sisodia,
| | - Shubhadeep Roychoudhury
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
- *Correspondence: Kavindra Kumar Kesari, ; Petr Slama, ; Shubhadeep Roychoudhury, ; Rashmi Sisodia,
| | - Rashmi Sisodia
- Department of Zoology, University of Rajasthan, Jaipur, India
- *Correspondence: Kavindra Kumar Kesari, ; Petr Slama, ; Shubhadeep Roychoudhury, ; Rashmi Sisodia,
| |
Collapse
|
22
|
Suman S, Moon BH, Datta K, Kallakury BVS, Fornace AJ. Heavy-ion radiation-induced colitis and colorectal carcinogenesis in Il10-/- mice display co-activation of β-catenin and NF-κB signaling. PLoS One 2022; 17:e0279771. [PMID: 36584137 PMCID: PMC9803147 DOI: 10.1371/journal.pone.0279771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Space radiation-induced gastrointestinal (GI) cancer risk models for future interplanetary astronauts are being developed that primarily rely on quantitative animal model studies to assess radiation-quality effects of heavy-ion space radiation exposure in relation to γ-rays. While current GI-cancer risk estimation efforts are focused on sporadic GI-cancer mouse models, emerging in-vivo data on heavy-ion radiation-induced long-term GI-inflammation are indicative of a higher but undetermined risk of GI-inflammation associated cancers, such as colitis-associated cancer (CAC). Therefore, we aimed to assess radiation quality effects on colonic inflammation, colon cancer incidence, and associated signaling events using an in-vivo CAC model i.e., Il10-/- mice. Male Il10-/- mice (8-10 weeks, n = 12/group) were irradiated with either sham, γ-rays or heavy-ions (28Si or 56Fe), and histopathological assessments for colitis and CAC were conducted at 2.5 months post-exposure. qPCR analysis for inflammation associated gene transcripts (Ptges and Tgfb1), and in-situ staining for markers of cell-proliferation (phospho-histone H3), oncogenesis (active-β-catenin, and cyclin D1), and inflammation (phospho-p65NF-κB, iNOS, and COX2) were performed. Significantly higher colitis and CAC frequency were noted after heavy-ion exposure, relative to γ and control mice. Higher CAC incidence after heavy-ion exposure was associated with greater activation of β-catenin and NF-κB signaling marked by induced expression of common downstream inflammatory (iNOS and COX2) and pro-proliferative (Cyclin D1) targets. In summary, IR-induced colitis and CAC incidence in Il10-/- mice depends on radiation quality and display co-activation of β-catenin and NF-κB signaling.
Collapse
Affiliation(s)
- Shubhankar Suman
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
- * E-mail:
| | - Bo-Hyun Moon
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Kamal Datta
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Bhaskar V. S. Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Albert J. Fornace
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, United States of America
| |
Collapse
|
23
|
Suman S, Kumar S, Kallakury BVS, Moon BH, Angdisen J, Datta K, Fornace AJ. Predominant contribution of the dose received from constituent heavy-ions in the induction of gastrointestinal tumorigenesis after simulated space radiation exposure. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:631-637. [PMID: 36167896 DOI: 10.1007/s00411-022-00997-z] [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: 04/12/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Gastrointestinal (GI) cancer risk among astronauts after encountering galactic cosmic radiation (GCR) is predicted to exceed safe permissible limits in long duration deep-space missions. Current predictions are based on relative biological effectiveness (RBE) values derived from in-vivo studies using single-ion beams, while GCR is essentially a mixed radiation field composed of protons (H), helium (He), and heavy ions. Therefore, a sequentially delivered proton (H) → Helium (He) → Oxygen (O) → Silicon (Si) beam was designed to simulate simplified-mixed-field GCR (Smf-GCR), and Apc1638N/+ mice were total-body irradiated to sham or γ (157Cs) or Smf-GCR followed by assessment of GI-tumorigenesis at 150 days post-exposure. Further, GI-tumor data from equivalent doses of heavy-ions (i.e., 0.05 Gy of O and Si) in 0.5 Gy of Smf-GCR were compared to understand the contributions of heavy-ions in GI-tumorigenesis. The Smf-GCR-induced tumor and carcinoma count were significantly greater than γ-rays, and male preponderance for GI-tumorigenesis was consistent with our earlier findings. Comparison of tumor data from Smf-GCR and equivalent doses of heavy ions revealed an association between higher GI-tumorigenesis where dose received from heavy-ions contributed to > 95% of the total GI-tumorigenic effect observed after Smf-GCR. This study provides the first experimental evidence that cancer risk after GCR exposure could largely depend on doses received from constituent heavy-ions.
Collapse
Affiliation(s)
- Shubhankar Suman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Research Building, Room E504, 3970 Reservoir Rd., NW, Washington, DC, 20057, USA.
| | - Santosh Kumar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Research Building, Room E504, 3970 Reservoir Rd., NW, Washington, DC, 20057, USA
| | - Bhaskar V S Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Bo-Hyun Moon
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Research Building, Room E504, 3970 Reservoir Rd., NW, Washington, DC, 20057, USA
| | - Jerry Angdisen
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Research Building, Room E504, 3970 Reservoir Rd., NW, Washington, DC, 20057, USA
| | - Kamal Datta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Research Building, Room E504, 3970 Reservoir Rd., NW, Washington, DC, 20057, USA
- Department of Biochemistry and Molecular and Cellular Biology and Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Albert J Fornace
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Research Building, Room E504, 3970 Reservoir Rd., NW, Washington, DC, 20057, USA
- Department of Biochemistry and Molecular and Cellular Biology and Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, USA
| |
Collapse
|
24
|
Suman S, Fornace AJ. Countermeasure development against space radiation-induced gastrointestinal carcinogenesis: Current and future perspectives. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:53-59. [PMID: 36336370 DOI: 10.1016/j.lssr.2022.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
A significantly higher probability of space radiation-induced gastrointestinal (GI) cancer incidence and mortality after a Mars mission has been projected using biophysical and statistical modeling approaches, and may exceed the current NASA mandated limit of less than 3% REID (risk of exposure-induced death). Since spacecraft shielding is not fully effective against heavy-ion space radiation, there is an unmet need to develop an effective medical countermeasure (MCM) strategy against heavy-ion space radiation-induced GI carcinogenesis to safeguard astronauts. In the past, we have successfully applied a GI cancer mouse model approach to understand space radiation-induced GI cancer risk and associated molecular signaling events. We have also tested several potential MCMs to safeguard astronauts during and after a prolonged space mission. In this review, we provide an updated summary of MCM testing using the GI cancer mouse model approach, lessons learned, and a perspective on the senescence signaling targeting approach for desirable protection against space radiation-induced GI carcinogenesis. Furthermore, we also discuss some of the advanced senotherapeutic candidates/combinations as a potential MCM for space radiation-induced GI carcinogenesis.
Collapse
Affiliation(s)
- Shubhankar Suman
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Research Building, Room E504, 3970 Reservoir Rd., NW, Washington D. C. 20057, USA.
| | - Albert J Fornace
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Research Building, Room E504, 3970 Reservoir Rd., NW, Washington D. C. 20057, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington D. C. 20057, USA
| |
Collapse
|
25
|
Hertel NE, Biegalski SR, Nelson VI, Nelson WA, Mukhopadhyay S, Su Z, Chan AM, Kesarwala AH, Dynan WS. Compact portable sources of high-LET radiation: Validation and potential application for galactic cosmic radiation countermeasure discovery. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:163-169. [PMID: 36336362 DOI: 10.1016/j.lssr.2022.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
Implementation of a systematic program for galactic cosmic radiation (GCR) countermeasure discovery will require convenient access to ground-based space radiation analogs. The current gold standard approach for GCR simulation is to use a particle accelerator for sequential irradiation with ion beams representing different GCR components. This has limitations, particularly for studies of non-acute responses, strategies that require robotic instrumentation, or implementation of complex in vitro models that are emerging as alternatives to animal experimentation. Here we explore theoretical and practical issues relating to a different approach to provide a high-LET radiation field for space radiation countermeasure discovery, based on use of compact portable sources to generate neutron-induced charged particles. We present modeling studies showing that DD and DT neutron generators, as well as an AmBe radionuclide-based source, generate charged particles with a linear energy transfer (LET) distribution that, within a range of biological interest extending from about 10 to 200 keV/μm, resembles the LET distribution of reference GCR radiation fields experienced in a spacecraft or on the lunar surface. We also demonstrate the feasibility of using DD neutrons to induce 53BP1 DNA double-strand break repair foci in the HBEC3-KT line of human bronchial epithelial cells, which are widely used for studies of lung carcinogenesis. The neutron-induced foci are larger and more persistent than X ray-induced foci, consistent with the induction of complex, difficult-to-repair DNA damage characteristic of exposure to high-LET (>10 keV/μm) radiation. We discuss limitations of the neutron approach, including low fluence in the low LET range (<10 keV/μm) and the absence of certain long-range features of high charge and energy particle tracks. We present a concept for integration of a compact portable source with a multiplex microfluidic in vitro culture system, and we discuss a pathway for further validation of the use of compact portable sources for countermeasure discovery.
Collapse
Affiliation(s)
- Nolan E Hertel
- G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 770 State Street, 30332-0745 Atlanta, GA, United States of America.
| | - Steven R Biegalski
- G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 770 State Street, 30332-0745 Atlanta, GA, United States of America
| | - Victoria I Nelson
- G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 770 State Street, 30332-0745 Atlanta, GA, United States of America
| | - William A Nelson
- G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 770 State Street, 30332-0745 Atlanta, GA, United States of America
| | - Sharmistha Mukhopadhyay
- G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 770 State Street, 30332-0745 Atlanta, GA, United States of America
| | - Zitong Su
- Department of Radiation Oncology and Winship Cancer Institute, Emory University School of Medicine, 1365 Clifton Road NE, 30322 Atlanta GA, United States of America; Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road NE, 30322 Atlanta GA, United States of America
| | - Alexis M Chan
- Department of Radiation Oncology and Winship Cancer Institute, Emory University School of Medicine, 1365 Clifton Road NE, 30322 Atlanta GA, United States of America; Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road NE, 30322 Atlanta GA, United States of America
| | - Aparna H Kesarwala
- Department of Radiation Oncology and Winship Cancer Institute, Emory University School of Medicine, 1365 Clifton Road NE, 30322 Atlanta GA, United States of America
| | - William S Dynan
- Department of Radiation Oncology and Winship Cancer Institute, Emory University School of Medicine, 1365 Clifton Road NE, 30322 Atlanta GA, United States of America; Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road NE, 30322 Atlanta GA, United States of America.
| |
Collapse
|
26
|
Jaylet T, Quintens R, Benotmane MA, Luukkonen J, Tanaka IB, Ibanez C, Durand C, Sachana M, Azimzadeh O, Adam-Guillermin C, Tollefsen KE, Laurent O, Audouze K, Armant O. Development of an Adverse Outcome Pathway for radiation-induced microcephaly via expert consultation and machine learning. Int J Radiat Biol 2022; 98:1752-1762. [PMID: 35947014 DOI: 10.1080/09553002.2022.2110312] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Brain development during embryogenesis and in early postnatal life is particularly complex and involves the interplay of many cellular processes and molecular mechanisms, making it extremely vulnerable to exogenous insults, including ionizing radiation (IR). Microcephaly is one of the most frequent neurodevelopmental abnormalities that is characterized by small brain size, and is often associated with intellectual deficiency. Decades of research span from epidemiological data on in utero exposure of the A-bomb survivors, to studies on animal and cellular models that allowed deciphering the most prominent molecular mechanisms leading to microcephaly. The Adverse Outcome Pathway (AOP) framework is used to organize, evaluate and portray the scientific knowledge of toxicological effects spanning different biological levels of organizations, from the initial interaction with molecular targets to the occurrence of a disease or adversity. In the present study, the framework was used in an attempt to organize the current scientific knowledge on microcephaly progression in the context of ionizing radiation (IR) exposure. This work was performed by a group of experts formed during a recent workshop organized jointly by the Multidisciplinary European Low Dose Initiative (MELODI) and the European Radioecology Alliance (ALLIANCE) associations to present the AOP approach and tools. Here we report on the development of a putative AOP for congenital microcephaly resulting from IR exposure based on discussions of the working group and we emphasize the use of a novel machine-learning approach to assist in the screening of the available literature to develop AOPs. CONCLUSION The expert consultation led to the identification of crucial biological events for the progression of microcephaly upon exposure to IR, and highlighted current knowledge gaps. The machine learning approach was successfully used to screen the existing knowledge and helped to rapidly screen the body of evidence and in particular the epidemiological data. This systematic review approach also ensured that the analysis was sufficiently comprehensive to identify the most relevant data and facilitate rapid and consistent AOP development. We anticipate that as machine learning approaches become more user-friendly through easy-to-use web interface, this would allow AOP development to become more efficient and less time consuming.
Collapse
Affiliation(s)
- Thomas Jaylet
- Université Paris Cité, T3S, Inserm UMRS 1124, Paris, France
| | - Roel Quintens
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK-CEN, Mol, Belgium
| | | | - Jukka Luukkonen
- University of Eastern Finland, Kuopio Campus, Department of Environmental and Biological Sciences, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Ignacia Braga Tanaka
- Department of Radiobiology, Institute for Environmental Sciences, 1-7 lenomae, Obuchi, Rokkasho-mura, Kamikita-gun, Aomori 039-3212, Japan
| | - Chrystelle Ibanez
- PSE-SANTE/SESANE/LRTOX Institut de Radioprotection et de Sûreté Nucléaire (IRSN), F-92262, Fontenay-aux-Roses, France
| | - Christelle Durand
- PSE-SANTE/SESANE/LRTOX Institut de Radioprotection et de Sûreté Nucléaire (IRSN), F-92262, Fontenay-aux-Roses, France
| | - Magdalini Sachana
- Organisation for Economic Co-operation and Development (OECD), Environment Health and Safety Division, 75775 CEDEX 16 Paris, France
| | - Omid Azimzadeh
- Federal Office for Radiation Protection (Bfs), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Christelle Adam-Guillermin
- PSE-SANTE/SDOS/LMDN, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Cadarache, 13115 Saint-Paul-Lez-Durance, France
| | - Knut Erik Tollefsen
- Norwegian Institute for Water Research (NIVA), Økernveien 94, N-0579, Oslo, Norway.,Norwegian University of Life Sciences (NMBU), Post box 5003, N-1432 Ås, Norway.,Centre for Environmental Radioactivity, Norwegian University of Life Sciences (NMBU), Post box 5003, N-1432 Ås, Norway
| | - Olivier Laurent
- PSE-SANTE/SESANE/LEPID, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), F-92262, Fontenay-aux-Roses, France
| | - Karine Audouze
- Université Paris Cité, T3S, Inserm UMRS 1124, Paris, France
| | - Olivier Armant
- PSE-ENV/SRTE/LECO, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Cadarache, 13115 Saint-Paul-Lez-Durance, France
| |
Collapse
|
27
|
Mukherjee S, Dutta A, Chakraborty A. The interaction of oxidative stress with MAPK, PI3/AKT, NF-κB, and DNA damage kinases influences the fate of γ-radiation-induced bystander cells. Arch Biochem Biophys 2022; 725:109302. [DOI: 10.1016/j.abb.2022.109302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/27/2022] [Accepted: 05/22/2022] [Indexed: 11/02/2022]
|
28
|
Abot A, Fried S, Cani PD, Knauf C. Reactive Oxygen Species/Reactive Nitrogen Species as Messengers in the Gut: Impact on Physiology and Metabolic Disorders. Antioxid Redox Signal 2022; 37:394-415. [PMID: 34714099 DOI: 10.1089/ars.2021.0100] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: The role of reactive oxygen/nitrogen species as "friend" or "foe" messengers in the whole body is well characterized. Depending on the concentration in the tissue considered, these molecular actors exert beneficial or deleterious impacts leading to a pathological state, as observed in metabolic disorders such as type 2 diabetes and obesity. Recent Advances: Among the tissues impacted by oxidation and inflammation in this pathological state, the intestine is a site of dysfunction that can establish diabetic symptoms, such as alterations in the intestinal barrier, gut motility, microbiota composition, and gut/brain axis communication. In the intestine, reactive oxygen/nitrogen species (from the host and/or microbiota) are key factors that modulate the transition from physiological to pathological signaling. Critical Issues: Controlling the levels of intestinal reactive oxygen/nitrogen species is a complicated balance between positive and negative impacts that is in constant equilibrium. Here, we describe the synthesis and degradation of intestinal reactive oxygen/nitrogen species and their interactions with the host. The development of novel redox-based therapeutics that alter these processes could restore intestinal health in patients with metabolic disorders. Future Directions: Deciphering the mode of action of reactive oxygen/nitrogen species in the gut of obese/diabetic patients could result in a future therapeutic strategy that combines nutritional and pharmacological approaches. Consequently, preventive and curative treatments must take into account one of the first sites of oxidative and inflammatory dysfunctions in the body, that is, the intestine. Antioxid. Redox Signal. 37, 394-415.
Collapse
Affiliation(s)
- Anne Abot
- Université Paul Sabatier, Toulouse III, INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan, Toulouse, France.,International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France
| | - Steven Fried
- Université Paul Sabatier, Toulouse III, INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan, Toulouse, France.,International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France
| | - Patrice D Cani
- International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France.,UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, WELBIO, Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Brussels, Belgium
| | - Claude Knauf
- Université Paul Sabatier, Toulouse III, INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan, Toulouse, France.,International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France
| |
Collapse
|
29
|
Huff JL, Plante I, Blattnig SR, Norman RB, Little MP, Khera A, Simonsen LC, Patel ZS. Cardiovascular Disease Risk Modeling for Astronauts: Making the Leap From Earth to Space. Front Cardiovasc Med 2022; 9:873597. [PMID: 35665268 PMCID: PMC9161032 DOI: 10.3389/fcvm.2022.873597] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Abstract
NASA has recently completed several long-duration missions to the International Space Station and is solidifying plans to return to the Moon, with an eye toward Mars and beyond. As NASA pushes the boundaries of human space exploration, the hazards of spaceflight, including space radiation, levy an increasing burden on astronaut health and performance. The cardiovascular system may be especially vulnerable due to the combined impacts of space radiation exposure, lack of gravity, and other spaceflight hazards. On Earth, the risk for cardiovascular disease (CVD) following moderate to high radiation doses is well-established from clinical, environmental, and occupational exposures (largely from gamma- and x-rays). Less is known about CVD risks associated with high-energy charged ions found in space and increasingly used in radiotherapy applications on Earth, making this a critical area of investigation for occupational radiation protection. Assessing CVD risk is complicated by its multifactorial nature, where an individual's risk is strongly influenced by factors such as family history, blood pressure, and lipid profiles. These known risk factors provide the basis for development of a variety of clinical risk prediction models (CPMs) that inform the likelihood of medical outcomes over a defined period. These tools improve clinical decision-making, personalize care, and support primary prevention of CVD. They may also be useful for individualizing risk estimates for CVD following radiation exposure both in the clinic and in space. In this review, we summarize unique aspects of radiation risk assessment for astronauts, and we evaluate the most widely used CVD CPMs for their use in NASA radiation risk assessment applications. We describe a comprehensive dual-use risk assessment framework that supports both clinical care and operational management of space radiation health risks using quantitative metrics. This approach is a first step in using personalized medicine for radiation risk assessment to support safe and productive spaceflight and long-term quality of life for NASA astronauts.
Collapse
Affiliation(s)
- Janice L. Huff
- National Aeronautics and Space Administration, Langley Research Center, Hampton, VA, United States
- *Correspondence: Janice L. Huff
| | - Ianik Plante
- KBR, Houston, TX, United States
- National Aeronautics and Space Administration, Johnson Space Center, Houston, TX, United States
| | - Steve R. Blattnig
- National Aeronautics and Space Administration, Langley Research Center, Hampton, VA, United States
| | - Ryan B. Norman
- National Aeronautics and Space Administration, Langley Research Center, Hampton, VA, United States
| | - Mark P. Little
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services (DHHS), Radiation Epidemiology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Amit Khera
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Lisa C. Simonsen
- National Aeronautics and Space Administration, NASA Headquarters, Washington, DC, United States
| | - Zarana S. Patel
- KBR, Houston, TX, United States
- National Aeronautics and Space Administration, Johnson Space Center, Houston, TX, United States
| |
Collapse
|
30
|
Vyas R, Kesari KK, Slama P, Roychoudhury S, Sisodia R. Differential Activity of Antioxidants in Testicular Tissues Following Administration of Chlorophytum borivilianum in Gamma-Irradiated Swiss Albino Mice. Front Pharmacol 2022; 12:774444. [PMID: 35111049 PMCID: PMC8802459 DOI: 10.3389/fphar.2021.774444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Oxidative stress induced by radiation causes variable expression of antioxidant enzymes in a tissue-specific manner. Testicular tissues carry out the complex process of spermatogenesis, and studies indicate that testicular damages due to irradiation require long-term recovery before complete resumption. Ionizing radiation also causes oxidative stress in tissues, leading to testicular damage. Aims and Objectives: This study measured differential expression of antioxidant enzymes following administration of C. borivilianum root extract (CRB) in response to irradiation-induced oxidative stress. The activity of various important endogenous enzymatic defense systems was evaluated and correlated for strength of association. Materials and method: Two forms of C. borivilianum (CB) extracts [CB alone and CB-silver nanoparticles (AgNPs)] were administered at a dose of 50 mg/kg body weight to Swiss albino male mice for 7 consecutive days. After that, they were irradiated with 6 Gy irradiation and further used to study various parameters of antioxidant enzymes. Results: Results indicate a significant increase in the level of glutathione (GSH) and the activity of GSH-related antioxidant enzymes in irradiated mice treated with CRE and CRE-AgNPs (silver nanoparticles biosynthesized using C. borivilianum root extract) in comparison to non-pretreated ones (groups I and II). Reciprocal elevation was observed in related enzymes, that is, glutathione S-transferase activity (GST), glutathione reductase (GR), and glutathione peroxidase activity (GPx). Elevation in the activity of catalase (CAT) and superoxide dismutase (SOD) was also evident in both the irradiated groups pretreated with CRE-AgNPs. However, expression of CAT in the CRE-treated irradiated group was similar to that of the non-treated irradiated group. Higher association among CAT-SOD, CAT-GPx, and GR-GST was observed. Conclusion: Overall, it was observed that testicular cells post-irradiation in all groups go through intense oxidative stress; however, groups pretreated with CRE or CRE-AgNPs indicated better toleration and resumption of antioxidant capacity. CRE or CRE-AgNPs pretreated non-irradiated groups mostly remained within the control range indicating stimulated expression of antioxidants.
Collapse
Affiliation(s)
- Ruchi Vyas
- Department of Zoology, S.S Jain Subodh PG College, Jaipur, India.,Department of Zoology, University of Rajasthan, Jaipur, India
| | | | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | | | - Rashmi Sisodia
- Department of Zoology, University of Rajasthan, Jaipur, India
| |
Collapse
|
31
|
Malo ME, Frank C, Khokhoev E, Gorbunov A, Dontsov A, Garg R, Dadachova E. Mitigating effects of sublethal and lethal whole-body gamma irradiation in a mouse model with soluble melanin. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2022; 42:011508. [PMID: 35037901 DOI: 10.1088/1361-6498/ac3dcf] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/25/2021] [Indexed: 05/27/2023]
Abstract
The field of radiation countermeasures is growing, however, currently there are no effective and non-toxic compounds which could be administered orally to the individuals post exposure to high doses of ionising radiation. The pigment melanin is ubiquitous through all kingdoms of life and provides selective advantage under radiation stress through its role as a chemical and physical shield, and its capacity to respond and react to exposures. Soluble allomelanin was administered to mice following whole-body exposure to lethal or sublethal doses of gamma radiation to determine its capacity to mitigate the effects of acute radiation syndrome, and its utility as a radiation countermeasure. Allomelanin has shown a trend to improve survival post an 8 Gy sublethal radiation exposure when administered up to 48 h post-irradiation. Furthermore, it improved median and overall survival to a 10 Gy lethal radiation exposure, specifically when administered at 24 h post-irradiation. Histological analysis on the jejunum region of the small intestine of this treatment group indicated that alterations of the mucosal and submucosal architecture, and disruption of the lymphatic system associated with lethal radiation exposure were mitigated when allomelanin was administered at 24 h post-irradiation. Based on this work soluble allomelanin derived from a fungal source could serve as an easily sourced, cost-effective, and viable countermeasure to accidental radiation exposure and merits further investigation.
Collapse
Affiliation(s)
- Mackenzie E Malo
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Connor Frank
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | | | | | - Alexander Dontsov
- Emanuel Institute of Biochemical Physics, Russian Academy of Science, Moscow, Russia
| | - Ravendra Garg
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
32
|
Mhatre SD, Iyer J, Puukila S, Paul AM, Tahimic CGT, Rubinstein L, Lowe M, Alwood JS, Sowa MB, Bhattacharya S, Globus RK, Ronca AE. Neuro-consequences of the spaceflight environment. Neurosci Biobehav Rev 2021; 132:908-935. [PMID: 34767877 DOI: 10.1016/j.neubiorev.2021.09.055] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 08/03/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022]
Abstract
As human space exploration advances to establish a permanent presence beyond the Low Earth Orbit (LEO) with NASA's Artemis mission, researchers are striving to understand and address the health challenges of living and working in the spaceflight environment. Exposure to ionizing radiation, microgravity, isolation and other spaceflight hazards pose significant risks to astronauts. Determining neurobiological and neurobehavioral responses, understanding physiological responses under Central Nervous System (CNS) control, and identifying putative mechanisms to inform countermeasure development are critically important to ensuring brain and behavioral health of crew on long duration missions. Here we provide a detailed and comprehensive review of the effects of spaceflight and of ground-based spaceflight analogs, including simulated weightlessness, social isolation, and ionizing radiation on humans and animals. Further, we discuss dietary and non-dietary countermeasures including artificial gravity and antioxidants, among others. Significant future work is needed to ensure that neural, sensorimotor, cognitive and other physiological functions are maintained during extended deep space missions to avoid potentially catastrophic health and safety outcomes.
Collapse
Affiliation(s)
- Siddhita D Mhatre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; KBR, Houston, TX, 77002, USA; COSMIAC Research Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Janani Iyer
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA
| | - Stephanie Puukila
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA; Flinders University, Adelaide, Australia
| | - Amber M Paul
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; KBR, Houston, TX, 77002, USA; Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Linda Rubinstein
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA
| | - Moniece Lowe
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Blue Marble Space Institute of Science, Seattle, WA, 98154, USA
| | - Joshua S Alwood
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Marianne B Sowa
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Sharmila Bhattacharya
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - April E Ronca
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Wake Forest Medical School, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
33
|
The effects of real and simulated microgravity on cellular mitochondrial function. NPJ Microgravity 2021; 7:44. [PMID: 34750383 PMCID: PMC8575887 DOI: 10.1038/s41526-021-00171-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 10/07/2021] [Indexed: 11/22/2022] Open
Abstract
Astronauts returning from space shuttle missions or the International Space Station have been diagnosed with various health problems such as bone demineralization, muscle atrophy, cardiovascular deconditioning, and vestibular and sensory imbalance including visual acuity, altered metabolic and nutritional status, and immune system dysregulation. These health issues are associated with oxidative stress caused by a microgravity environment. Mitochondria are a source of reactive oxygen species (ROS). However, the molecular mechanisms through which mitochondria produce ROS in a microgravity environment remain unclear. Therefore, this review aimed to explore the mechanism through which microgravity induces oxidative damage in mitochondria by evaluating the expression of genes and proteins, as well as relevant metabolic pathways. In general, microgravity-induced ROS reduce mitochondrial volume by mainly affecting the efficiency of the respiratory chain and metabolic pathways. The impaired respiratory chain is thought to generate ROS through premature electron leakage in the electron transport chain. The imbalance between ROS production and antioxidant defense in mitochondria is the main cause of mitochondrial stress and damage, which leads to mitochondrial dysfunction. Moreover, we discuss the effects of antioxidants against oxidative stress caused by the microgravity environment space microgravity in together with simulated microgravity (i.e., spaceflight or ground-based spaceflight analogs: parabolic flight, centrifugal force, drop towers, etc.). Further studies should be taken to explore the effects of microgravity on mitochondrial stress-related diseases, especially for the development of new therapeutic drugs that can help increase the health of astronauts on long space missions.
Collapse
|
34
|
Suman S, Kumar S, Moon BH, Angdisen J, Kallakury BVS, Datta K, Fornace AJ. Effects of dietary aspirin on high-LET radiation-induced prostaglandin E2 levels and gastrointestinal tumorigenesis in Apc 1638N/+ mice. LIFE SCIENCES IN SPACE RESEARCH 2021; 31:85-91. [PMID: 34689954 PMCID: PMC9808916 DOI: 10.1016/j.lssr.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/18/2021] [Accepted: 09/03/2021] [Indexed: 05/21/2023]
Abstract
Inevitable exposure to high-LET ionizing radiation (IR) present in galactic cosmic radiation (GCR) could enhance gastrointestinal (GI) cancer incidence among astronauts undertaking deep space exploration and GI-cancer mortality has been predicted to far exceed NASA's limit of < 3% REID (Radiation exposure-induced death) from cancer. Therefore, the development of countermeasure agents against high-LET radiation-induced GI cancer is needed to safeguard astronauts during and after an outer space mission. The cyclooxygenase-2/prostaglandin E2 (COX2/PGE2) mediated activation of pro-inflammatory and oncogenic signaling has been reported to play an important role in persistent inflammation and GI-tumorigenesis after high-LET radiation exposure. Therefore, aspirin, a well-known inhibitor of the COX/PGE2 pathway, was evaluated as a potential countermeasure against 28Si-induced PGE2 and tumorigenesis in Apc1638N/+, a murine model of human GI-cancer. Animals were fed either standard or aspirin supplemented diet (75, 150, or 300 mg/day of human equivalent dose) starting at the age of 4 weeks and continued till the end of the study, while mice were exposed to 28Si-ions (300 MeV/n; 69 keV/μm) at the age of 8 weeks. Serum PGE2 level, GI tumor size (>2mm2), number, and cluster (>5 adjoining tumors) were analyzed at 150 days post-exposure. Aspirin led to a significant reduction in PGE2 in a dose-dependent manner but did not reduce 28Si-induced GI tumorigenesis even at the highest (300 mg/day) dose. In summary, this study suggests that aspirin could reduce high-LET IR-induced pro-inflammatory PGE2 levels, however, lacks the ability to reduce high-LET IR-induced GI tumorigenesis in Apc1638N/+ mice.
Collapse
Affiliation(s)
- Shubhankar Suman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Santosh Kumar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Bo-Hyun Moon
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jerry Angdisen
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | - Kamal Datta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Biochemistry and Molecular & Cellular Biology and Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Albert J Fornace
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Biochemistry and Molecular & Cellular Biology and Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
35
|
Vorhees CV, Vatner RE, Williams MT. Review of Conventional and High Dose Rate Brain Radiation (FLASH): Neurobehavioural, Neurocognitive and Assessment Issues in Rodent Models. Clin Oncol (R Coll Radiol) 2021; 33:e482-e491. [PMID: 34548203 PMCID: PMC10114147 DOI: 10.1016/j.clon.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/29/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Ionising radiation causes secondary tumours and/or enduring cognitive deficits, especially in children. Proton radiotherapy reduces exposure of the developing brain in children but may still cause some lasting effects. Recent observations show that ultra-high dose rate radiation treatment (≥40 Gy/s), called the FLASH effect, is equally effective at tumour control but less damaging to surrounding tissue compared with conventional dose rate protons (0.03-3 Gy/s). Most studies on the FLASH effect in brain and other tissues with different radiation modalities (electron and photon radiation), show FLASH benefits in these preclinical rodent models, but the data are limited, especially for proton FLASH, including for dose, dose rate and neurochemical and neurobehavioural outcomes. Tests of neurocognitive outcomes have been limited despite clinical evidence that this is the area of greatest concern. The FLASH effect in the context of proton exposure is promising, but a more systematic and comprehensive approach to outcomes is needed.
Collapse
Affiliation(s)
- C V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, Ohio, USA.
| | - R E Vatner
- Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, Ohio, USA; Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - M T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, Ohio, USA
| |
Collapse
|
36
|
Barnette BL, Yu Y, Ullrich RL, Emmett MR. Mitochondrial Effects in the Liver of C57BL/6 Mice by Low Dose, High Energy, High Charge Irradiation. Int J Mol Sci 2021; 22:ijms222111806. [PMID: 34769236 PMCID: PMC8584048 DOI: 10.3390/ijms222111806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 12/19/2022] Open
Abstract
Galactic cosmic rays are primarily composed of protons (85%), helium (14%), and high charge/high energy ions (HZEs) such as 56Fe, 28Si, and 16O. HZE exposure is a major risk factor for astronauts during deep-space travel due to the possibility of HZE-induced cancer. A systems biology integrated omics approach encompassing transcriptomics, proteomics, lipidomics, and functional biochemical assays was used to identify microenvironmental changes induced by HZE exposure. C57BL/6 mice were placed into six treatment groups and received the following irradiation treatments: 600 MeV/n 56Fe (0.2 Gy), 1 GeV/n 16O (0.2 Gy), 350 MeV/n 28Si (0.2 Gy), 137Cs (1.0 Gy) gamma rays, 137Cs (3.0 Gy) gamma rays, and sham irradiation. Left liver lobes were collected at 30, 60, 120, 270, and 360 days post-irradiation. Analysis of transcriptomic and proteomic data utilizing ingenuity pathway analysis identified multiple pathways involved in mitochondrial function that were altered after HZE irradiation. Lipids also exhibited changes that were linked to mitochondrial function. Molecular assays for mitochondrial Complex I activity showed significant decreases in activity after HZE exposure. HZE-induced mitochondrial dysfunction suggests an increased risk for deep space travel. Microenvironmental and pathway analysis as performed in this research identified possible targets for countermeasures to mitigate risk.
Collapse
Affiliation(s)
- Brooke L. Barnette
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA;
| | - Yongjia Yu
- Department of Radiation Oncology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA;
| | - Robert L. Ullrich
- The Radiation Effects Research Foundation (RERF), Hiroshima 732-0815, Japan;
| | - Mark R. Emmett
- Department of Radiation Oncology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA;
- Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA
- Correspondence: ; Tel.: +1-(409)-747-1943
| |
Collapse
|
37
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Shabeeb D, Musa AE. Resveratrol as an Adjuvant for Normal Tissues Protection and Tumor Sensitization. Curr Cancer Drug Targets 2021; 20:130-145. [PMID: 31738153 DOI: 10.2174/1568009619666191019143539] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/12/2019] [Accepted: 07/22/2019] [Indexed: 12/24/2022]
Abstract
Cancer is one of the most complicated diseases in present-day medical science. Yearly, several studies suggest various strategies for preventing carcinogenesis. Furthermore, experiments for the treatment of cancer with low side effects are ongoing. Chemotherapy, targeted therapy, radiotherapy and immunotherapy are the most common non-invasive strategies for cancer treatment. One of the most challenging issues encountered with these modalities is low effectiveness, as well as normal tissue toxicity for chemo-radiation therapy. The use of some agents as adjuvants has been suggested to improve tumor responses and also alleviate normal tissue toxicity. Resveratrol, a natural flavonoid, has attracted a lot of attention for the management of both tumor and normal tissue responses to various modalities of cancer therapy. As an antioxidant and anti-inflammatory agent, in vitro and in vivo studies show that it is able to mitigate chemo-radiation toxicity in normal tissues. However, clinical studies to confirm the usage of resveratrol as a chemo-radioprotector are lacking. In addition, it can sensitize various types of cancer cells to both chemotherapy drugs and radiation. In recent years, some clinical studies suggested that resveratrol may have an effect on inducing cancer cell killing. Yet, clinical translation of resveratrol has not yielded desirable results for the combination of resveratrol with radiotherapy, targeted therapy or immunotherapy. In this paper, we review the potential role of resveratrol for preserving normal tissues and sensitization of cancer cells in combination with different cancer treatment modalities.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48175-861, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed E Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| |
Collapse
|
38
|
Mohamed HA, Said RS. Coenzyme Q10 attenuates inflammation and fibrosis implicated in radiation enteropathy through suppression of NF-kB/TGF-β/MMP-9 pathways. Int Immunopharmacol 2021; 92:107347. [PMID: 33418245 DOI: 10.1016/j.intimp.2020.107347] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 01/01/2023]
Abstract
Radiation enteropathy is one the most common clinical issue for patients receiving radiotherapy for abdominal/pelvic tumors which severely affect the quality of life of cancer patients due to dysplastic lesions (ischemia, ulcer, or fibrosis) that aggravate the radiation damage. Herein, this study demonstrated the prophylactic role of coenzyme Q10 (CoQ10), a powerful antioxidant, against radiotherapy-induced gastrointestinal injury. Male Sprague Dawley rats were divided into four groups: group 1 was defined as control, and group 2 was the irradiated group. Group 3 and 4 were CoQ10 control and radiation plus CoQ10 groups, respectively. CoQ10 (10 mg/kg) was orally administered for 10 days before 10 Gy whole-body radiation and was continued for 4 days post-irradiation. CoQ10 administration protected rats delivered a lethal dose of ϒ-radiation from changes in crypt-villus structures and promoted regeneration of the intestinal epithelium. CoQ10 attenuated radiation-induced oxidative stress by decreasing lipid peroxidation and increasing the antioxidant enzyme catalase activity and reduced glutathione level. CoQ10 also counteracts inflammatory response mediated after radiation exposure through downregulating intestinal NF-ĸB expression which subsequently decreased the level of inflammatory cytokine IL-6 and the expression of COX-2. Radiation-induced intestinal fibrosis confirmed via Masson's trichrome staining occurred through upregulating transforming growth factor (TGF)-β1 and matrix metalloproteinase (MMP)-9 expression, while CoQ10 administration significantly diminishes these effects which further confirmed the anti-fibrotic property of CoQ10. Therefore, CoQ10 is a promising radioprotector that could prevent intestinal complications and enhance the therapeutic ratio of radiotherapy in patients with pelvic tumors through suppressing the NF-kB/TGF-β1/MMP-9 signaling pathway.
Collapse
Affiliation(s)
- Heba A Mohamed
- Department of Drug Radiation Research, National Center for Radiation Research & Technology, Atomic Energy Authority, Cairo, Egypt
| | - Riham S Said
- Department of Drug Radiation Research, National Center for Radiation Research & Technology, Atomic Energy Authority, Cairo, Egypt.
| |
Collapse
|
39
|
|
40
|
Dissmore T, DeMarco AG, Jayatilake M, Girgis M, Bansal S, Li Y, Mehta K, Sridharan V, Gill K, Bansal S, Tyburski JB, Cheema AK. Longitudinal metabolic alterations in plasma of rats exposed to low doses of high linear energy transfer radiation. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:219-233. [PMID: 33902389 PMCID: PMC9896584 DOI: 10.1080/26896583.2020.1865027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Astronauts embarking on deep space missions are at high risk of long-term exposure to low doses of high linear energy transfer (LET) radiation, which can contribute to the development of cancer and multiple degenerative diseases. However, long term effects of exposure to low doses of high LET radiation in plasma metabolite profiles have not been elucidated. We utilized an untargeted metabolomics and lipidomics approach to analyze plasma obtained from adult male Long Evans rats to determine the longitudinal effects of low-dose proton and low-dose oxygen ion whole-body irradiation on metabolic pathways. Our findings reveal that radiation exposure induced modest changes in the metabolic profiles in plasma, 7 months after exposure. Furthermore, we identified some common metabolite dysregulations between protons and oxygen ions, which may indicate a similar mechanism of action for both radiation types.
Collapse
Affiliation(s)
- Tixieanna Dissmore
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Andrew G DeMarco
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC, USA
| | - Meth Jayatilake
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Michael Girgis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Shivani Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Khyati Mehta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kirandeep Gill
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Sunil Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | | | - Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
41
|
Lee HJ, Lee SH, Lee JH, Kim Y, Seong KM, Jin YW, Min KJ. Role of Commensal Microbes in the γ-Ray Irradiation-Induced Physiological Changes in Drosophila melanogaster. Microorganisms 2020; 9:microorganisms9010031. [PMID: 33374132 PMCID: PMC7824294 DOI: 10.3390/microorganisms9010031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/19/2020] [Accepted: 12/22/2020] [Indexed: 12/29/2022] Open
Abstract
Ionizing radiation induces biological/physiological changes and affects commensal microbes, but few studies have examined the relationship between the physiological changes induced by irradiation and commensal microbes. This study investigated the role of commensal microbes in the γ-ray irradiation-induced physiological changes in Drosophila melanogaster. The bacterial load was increased in 5 Gy irradiated flies, but irradiation decreased the number of operational taxonomic units. The mean lifespan of conventional flies showed no significant change by irradiation, whereas that of axenic flies was negatively correlated with the radiation dose. γ-Ray irradiation did not change the average number of eggs in both conventional and axenic flies. Locomotion of conventional flies was decreased after 5 Gy radiation exposure, whereas no significant change in locomotion activity was detected in axenic flies after irradiation. γ-Ray irradiation increased the generation of reactive oxygen species in both conventional and axenic flies, but the increase was higher in axenic flies. Similarly, the amounts of mitochondria were increased in irradiated axenic flies but not in conventional flies. These results suggest that axenic flies are more sensitive in their mitochondrial responses to radiation than conventional flies, and increased sensitivity leads to a reduced lifespan and other physiological changes in axenic flies.
Collapse
Affiliation(s)
- Hwa-Jin Lee
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (H.-J.L.); (S.-H.L.); (J.-H.L.)
| | - Shin-Hae Lee
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (H.-J.L.); (S.-H.L.); (J.-H.L.)
| | - Ji-Hyeon Lee
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (H.-J.L.); (S.-H.L.); (J.-H.L.)
| | - Yongjoong Kim
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea; (Y.K.); (K.M.S.); (Y.W.J.)
| | - Ki Moon Seong
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea; (Y.K.); (K.M.S.); (Y.W.J.)
| | - Young Woo Jin
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea; (Y.K.); (K.M.S.); (Y.W.J.)
| | - Kyung-Jin Min
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (H.-J.L.); (S.-H.L.); (J.-H.L.)
- Correspondence:
| |
Collapse
|
42
|
Broxmeyer HE, Liu Y, Kapur R, Orschell CM, Aljoufi A, Ropa JP, Trinh T, Burns S, Capitano ML. Fate of Hematopoiesis During Aging. What Do We Really Know, and What are its Implications? Stem Cell Rev Rep 2020; 16:1020-1048. [PMID: 33145673 PMCID: PMC7609374 DOI: 10.1007/s12015-020-10065-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 12/11/2022]
Abstract
There is an ongoing shift in demographics such that older persons will outnumber young persons in the coming years, and with it age-associated tissue attrition and increased diseases and disorders. There has been increased information on the association of the aging process with dysregulation of hematopoietic stem (HSC) and progenitor (HPC) cells, and hematopoiesis. This review provides an extensive up-to date summary on the literature of aged hematopoiesis and HSCs placed in context of potential artifacts of the collection and processing procedure, that may not be totally representative of the status of HSCs in their in vivo bone marrow microenvironment, and what the implications of this are for understanding aged hematopoiesis. This review covers a number of interactive areas, many of which have not been adequately explored. There are still many unknowns and mechanistic insights to be elucidated to better understand effects of aging on the hematopoietic system, efforts that will take multidisciplinary approaches, and that could lead to means to ameliorate at least some of the dysregulation of HSCs and HPCs associated with the aging process. Graphical Abstract.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, 950 West Walnut Street, R2-302, Indianapolis, IN, 46202-5181, USA.
| | - Yan Liu
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Reuben Kapur
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christie M Orschell
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arafat Aljoufi
- Department of Microbiology and Immunology, Indiana University School of Medicine, 950 West Walnut Street, R2-302, Indianapolis, IN, 46202-5181, USA
| | - James P Ropa
- Department of Microbiology and Immunology, Indiana University School of Medicine, 950 West Walnut Street, R2-302, Indianapolis, IN, 46202-5181, USA
| | - Thao Trinh
- Department of Microbiology and Immunology, Indiana University School of Medicine, 950 West Walnut Street, R2-302, Indianapolis, IN, 46202-5181, USA
| | - Sarah Burns
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maegan L Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, 950 West Walnut Street, R2-302, Indianapolis, IN, 46202-5181, USA.
| |
Collapse
|
43
|
Dong S, Lyu X, Yuan S, Wang S, Li W, Chen Z, Yu H, Li F, Jiang Q. Oxidative stress: A critical hint in ionizing radiation induced pyroptosis. RADIATION MEDICINE AND PROTECTION 2020. [DOI: 10.1016/j.radmp.2020.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
44
|
Maan K, Tyagi R, Dutta A, Bakhshi R, Rana P. Comparative metabolic profiles of total and partial body radiation exposure in mice using an untargeted metabolomics approach. Metabolomics 2020; 16:124. [PMID: 33245511 DOI: 10.1007/s11306-020-01742-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022]
Abstract
INTRODUCTION A large scale population exposure to ionizing radiation during intentional or unintentional nuclear accidents undoubtedly generates a complex scenario with partial-body as well as total-body irradiated victims. A high throughput technique based rapid assessment method is an urgent necessity for stratification of exposed subjects independent of whether exposure is uniform total-body or non-homogenous partial-body. OBJECTIVE Here, we used Nuclear Magnetic Resonance (NMR) based metabolomics approach to compare and identify candidate metabolites differentially expressed in total and partially irradiated mice model. METHODS C57BL/6 male mice (8-10 weeks) were irradiated total-body or locally to thoracic, hind limb or abdominal regions with 10 Gy of gamma radiation. Urine samples collected at 24 h post irradiation were examined using high resolution NMR spectroscopy and the datasets were analysed using multivariate analysis. RESULTS Multivariate and metabolic pathway analysis in urine samples collected at 24 h post-radiation exhibited segregation of all irradiated groups from controls. Metabolites associated with energy metabolism, gut flora metabolism and taurine were common to partial and total-body irradiation, thus making them potential candidates for radiation exposure. Nevertheless, a distinct metabolic pattern was observed in partial-body exposed groups with maximum changes observed in the hind limb region indicating differential tissue associated radiation sensitivity. The organ-specific changes may provide an early warning regarding the physiological system at risk after radiation injury. CONCLUSION The study affirms potentiality of metabolite markers and comparative analysis could be an important piece of information for an integrated solution to a complex research question in terms of radiation biomarkers.
Collapse
Affiliation(s)
- Kiran Maan
- Metabolomics Research Facility, Division of Behavioral Neuroscience, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
- Department of Biomedical Science, Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, Delhi, India
| | - Ritu Tyagi
- Metabolomics Research Facility, Division of Behavioral Neuroscience, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Ajaswrata Dutta
- Division of Radiation Biodosimetry, Department of Radiation Epigenetics, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Radhika Bakhshi
- Department of Biomedical Science, Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, Delhi, India
| | - Poonam Rana
- Metabolomics Research Facility, Division of Behavioral Neuroscience, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India.
| |
Collapse
|
45
|
Biological Effects of Scattered Versus Scanned Proton Beams on Normal Tissues in Total Body Irradiated Mice: Survival, Genotoxicity, Oxidative Stress and Inflammation. Antioxidants (Basel) 2020; 9:antiox9121170. [PMID: 33255388 PMCID: PMC7761103 DOI: 10.3390/antiox9121170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Side effects of proton therapy are poorly studied. Moreover, the differences in the method of dose delivery on normal tissues are not taken into account when proton beams are scanned instead of being scattered. We proposed here to study the effects of both modalities of proton beam delivery on blood; skin; lung and heart in a murine model. In that purpose; C57BL/6 mice were total body irradiated by 190.6 MeV proton beams either by Double Scattering (DS) or by Pencil Beam Scanning (PBS) in the plateau phase before the Bragg Peak. Mouse survival was evaluated. Blood and organs were removed three months after irradiation. Biomarkers of genotoxicity; oxidative stress and inflammation were measured. Proton irradiation was shown to increase lymphocyte micronucleus frequency; lung superoxide dismutase activity; erythrocyte and skin glutathione peroxidase activity; erythrocyte catalase activity; lung; heart and skin oxidized glutathione level; erythrocyte and lung lipid peroxidation and erythrocyte protein carbonylation even 3 months post-irradiation. When comparing both methods of proton beam delivery; mouse survival was not different. However, PBS significantly increased lymphocyte micronucleus frequency; erythrocyte glutathione peroxidase activity and heart oxidized glutathione level compared to DS. These results point out the necessity to take into account the way of delivering dose in PT as it could influence late side effects.
Collapse
|
46
|
Patel ZS, Brunstetter TJ, Tarver WJ, Whitmire AM, Zwart SR, Smith SM, Huff JL. Red risks for a journey to the red planet: The highest priority human health risks for a mission to Mars. NPJ Microgravity 2020; 6:33. [PMID: 33298950 PMCID: PMC7645687 DOI: 10.1038/s41526-020-00124-6] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
NASA's plans for space exploration include a return to the Moon to stay-boots back on the lunar surface with an orbital outpost. This station will be a launch point for voyages to destinations further away in our solar system, including journeys to the red planet Mars. To ensure success of these missions, health and performance risks associated with the unique hazards of spaceflight must be adequately controlled. These hazards-space radiation, altered gravity fields, isolation and confinement, closed environments, and distance from Earth-are linked with over 30 human health risks as documented by NASA's Human Research Program. The programmatic goal is to develop the tools and technologies to adequately mitigate, control, or accept these risks. The risks ranked as "red" have the highest priority based on both the likelihood of occurrence and the severity of their impact on human health, performance in mission, and long-term quality of life. These include: (1) space radiation health effects of cancer, cardiovascular disease, and cognitive decrements (2) Spaceflight-Associated Neuro-ocular Syndrome (3) behavioral health and performance decrements, and (4) inadequate food and nutrition. Evaluation of the hazards and risks in terms of the space exposome-the total sum of spaceflight and lifetime exposures and how they relate to genetics and determine the whole-body outcome-will provide a comprehensive picture of risk profiles for individual astronauts. In this review, we provide a primer on these "red" risks for the research community. The aim is to inform the development of studies and projects with high potential for generating both new knowledge and technologies to assist with mitigating multisystem risks to crew health during exploratory missions.
Collapse
Affiliation(s)
- Zarana S Patel
- KBR, Houston, TX, USA.
- NASA Lyndon B. Johnson Space Center, Houston, TX, USA.
| | | | | | | | - Sara R Zwart
- NASA Lyndon B. Johnson Space Center, Houston, TX, USA
- University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Scott M Smith
- NASA Lyndon B. Johnson Space Center, Houston, TX, USA
| | | |
Collapse
|
47
|
Suman S, Jaruga P, Dizdaroglu M, Fornace AJ, Datta K. Heavy ion space radiation triggers ongoing DNA base damage by downregulating DNA repair pathways. LIFE SCIENCES IN SPACE RESEARCH 2020; 27:27-32. [PMID: 34756227 DOI: 10.1016/j.lssr.2020.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 06/13/2023]
Abstract
Long-duration space missions outside low earth orbit will expose astronauts to a cumulative dose of high-energy particle radiation especially to highly damaging heavy ion radiation, which poses considerable risk to astronauts' health. The purpose of the current study was to quantitatively identify oxidatively induced DNA base modifications and assess status of the repair pathways involved in removing the modified bases in mouse intestinal cells after exposure to γ-rays and iron radiation. Mice (C57BL/6J; 6 to 8 weeks; female) were exposed to 0.5 Gy of either γ-rays or iron radiation and control mice were sham-irradiated. Intestinal tissues were collected 2 months after radiation. DNA base lesions were measured using gas chromatography-tandem mass spectrometry with isotope‑dilution. Base excision repair (BER) and nucleotide excision repair (NER) pathways were assessed using PCR and immunoblotting. Effects of iron radiation were compared to γ-rays and sham-irradiated controls. Exposure to iron radiation resulted in significantly higher levels of several DNA base lesions relative to control animals and those exposed to γ radiation. Assessment of BER and NER showed downregulation of pathway factors both at the RNA as well as at the protein levels. Our results not only provide important insight into DNA damage pattern in intestinal cells in response to iron radiation, but they also confirm our previous immunohistochemistry data on oxidatively induced DNA damage. We suggest that downregulation of the BER and NER pathways is contributing to ongoing DNA base damages long time after radiation exposure and has implications for chronic diseases including gastrointestinal diseases after heavy ion radiation exposure during space travel.
Collapse
Affiliation(s)
- Shubhankar Suman
- Department of Biochemistry and Molecular & Cellular Biology, Lombardi Comprehensive Cancer Center, Georgetown University, Research Building, Room E518, 3970 Reservoir Rd., NW, Washington, DC 20057, USA
| | - Pawel Jaruga
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Miral Dizdaroglu
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Albert J Fornace
- Department of Biochemistry and Molecular & Cellular Biology, Lombardi Comprehensive Cancer Center, Georgetown University, Research Building, Room E518, 3970 Reservoir Rd., NW, Washington, DC 20057, USA
| | - Kamal Datta
- Department of Biochemistry and Molecular & Cellular Biology, Lombardi Comprehensive Cancer Center, Georgetown University, Research Building, Room E518, 3970 Reservoir Rd., NW, Washington, DC 20057, USA.
| |
Collapse
|
48
|
Squire T, Ryan A, Bernard S. Radioprotective effects of induced astronaut torpor and advanced propulsion systems during deep space travel. LIFE SCIENCES IN SPACE RESEARCH 2020; 26:105-113. [PMID: 32718676 DOI: 10.1016/j.lssr.2020.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Human metabolic suppression is not a new concept, with 1950s scientific literature and movies demonstrating its potential use for deep space travel (Hock, 1960). An artificially induced state of metabolic suppression in the form of torpor would improve the amount of supplies required and therefore lessen weight and fuel required for missions to Mars and beyond (Choukèr et al., 2019). Transfer habitats for human stasis to Mars have been conceived (Bradford et al., 2018). Evidence suggests that animals, when hibernating, demonstrate relative radioprotection compared to their awake state. Experiments have also demonstrated relative radioprotection in conditions of hypothermia as well as during sleep (Bellesi et al., 2016 and Andersen et al., 2009). Circadian rhythm disrupted cells also appear to be more susceptible to radiation damage compared to those that are under a rhythmic control (Dakup et al., 2018). An induced torpor state for astronauts on deep space missions may provide a biological radioprotective state due to a decreased metabolism and hypothermic conditions. A regular enforced circadian rhythm might further limit DNA damage from radiation. The As Low As Reasonably Achievable (A.L.A.R.A.) radiation protection concept defines time, distance and shielding as ways to decrease radiation exposure. Whilst distance cannot be altered in space and shielding either passively or actively may be beneficial, time of exposure may be drastically decreased with improved propulsion systems. Whilst chemical propulsion systems have superior thrust to other systems, they lack high changes in velocity and fuel efficiency which can be achieved with nuclear or electric based propulsion systems. Radiation toxicity could be limited by reduced transit times, combined with the radioprotective effects of enforced circadian rhythms during a state of torpor or hibernation. OBJECTIVES 1. Investigate how the circadian clock and body temperature may contribute to radioprotection during human torpor on deep space missions. 2. Estimate radiation dose received by astronauts during a transit to Mars with varying propulsion systems. METHODS We simulated three types of conditions to investigate the potential radioprotective effect of the circadian clock and decreased temperature on cells being exposed to radiation such that may be the case during astronaut torpor. These conditions were: - Circadian clock strength: strong vs weak. - Light exposure: dark-dark vs light-dark cycle - Body temperature: 37C vs hypothermia vs torpor. We estimated transit times for a mission to Mars from Earth utilizing chemical, nuclear and electrical propulsion systems. Transit times were generated using the General Mission Analysis Tool (GMAT) and Matlab. These times were then input into the National Aeronautics and Space Administration (NASA) Online Tool for the Assessment of Radiation In Space (OLTARIS) computer simulator to estimate doses received by an astronaut for the three propulsion methods. RESULTS Our simulation demonstrated an increase in radioprotection with decreasing temperature. The greatest degree of radioprotection was shown in cells that maintained a strong circadian clock during torpor. This was in contrast to relatively lower radioprotection in cells with a weak clock during normothermia. We were also able to demonstrate that if torpor weakened the circadian clock, a protective effect could be partially restored by an external drive such as lighting schedules to aid entrainment i.e.: Blue light exposure for periods of awake and no light for rest times For the propulsion simulation, estimated transit times from Earth to Mars were 258 days for chemical propulsion with 165.9mSv received, 209 days for nuclear propulsion with 134.4mSv received and 80 days for electrical propulsion with 51.4mSv received. CONCLUSION A state of torpor for astronauts on deep space missions may not only improve weight, fuel and storage requirements but also provide a potential biological radiation protection strategy. Moreover, maintaining a controlled circadian rhythm during torpor conditions may aid radioprotection. In the not too distant future, propulsion techniques will be improved to limit transit time and hence decrease radiation dose to astronauts. Limiting exposure time and enhancing physiological radioprotection during transit could provide superior radioprotection benefits compared with active and passive radiation shielding strategies alone.
Collapse
Affiliation(s)
- T Squire
- The Canberra Hospital, Department of Radiation Oncology. Garran. Australian Capital Territory, Australia; University of Notre Dame Australia, School of Medicine. Darlinghurst, New South Wales, Australia.
| | - A Ryan
- University of Sydney, Applied and Plasma Physics Research Group. School of Aerospace Mechanical and Mechatronic Engineering, Camperdown, NSW 2006. Australia
| | - S Bernard
- Université de Lyon. CNRS UMR5208 Institut Camille Jordan. Villeurbanne, France & Inria Grenoble, France
| |
Collapse
|
49
|
Kumar S, Suman S, Fornace AJ, Datta K. Intestinal stem cells acquire premature senescence and senescence associated secretory phenotype concurrent with persistent DNA damage after heavy ion radiation in mice. Aging (Albany NY) 2020; 11:4145-4158. [PMID: 31239406 PMCID: PMC6629005 DOI: 10.18632/aging.102043] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/17/2019] [Indexed: 12/19/2022]
Abstract
Heavy ion radiation, prevalent in outer space and relevant for radiotherapy, is densely ionizing and poses risk to stem cells that are key to intestinal homeostasis. Currently, the molecular spectrum of heavy ion radiation-induced perturbations in intestinal stem cells (ISCs), that could trigger intestinal pathologies, remains largely unexplored. The Lgr5-EGFP-IRES-creERT mice were exposed to 50 cGy of iron radiation. Mice were euthanized 60 d after exposure and ISCs were sorted using fluorescence activated cell sorting. Reactive oxygen species (ROS) and mitochondrial superoxide were measured using fluorescent probes. Since DNA damage is linked to senescence and senescent cells acquire senescence-associated secretory phenotype (SASP), we stained ISCs for both senescence markers p16, p21, and p19 as well as SASP markers IL6, IL8, and VEGF. Due to potential positive effects of SASP on proliferation, we also stained for PCNA. Data show increased ROS and ongoing DNA damage, by staining for γH2AX, and 53BP1, along with accumulation of senescence markers. Results also showed increased SASP markers in senescent cells. Collectively, our data suggest that heavy-ion-induced chronic stress and ongoing DNA damage is promoting SASP in a fraction of the ISCs, which has implications for gastrointestinal function, inflammation, and carcinogenesis in astronauts and patients.
Collapse
Affiliation(s)
- Santosh Kumar
- Department of Biochemistry and Molecular & Cellular Biology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Shubhankar Suman
- Department of Biochemistry and Molecular & Cellular Biology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Albert J Fornace
- Department of Biochemistry and Molecular & Cellular Biology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Kamal Datta
- Department of Biochemistry and Molecular & Cellular Biology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
50
|
Shukla PK, Meena AS, Gangwar R, Szabo E, Balogh A, Chin Lee S, Vandewalle A, Tigyi G, Rao R. LPAR2 receptor activation attenuates radiation-induced disruption of apical junctional complexes and mucosal barrier dysfunction in mouse colon. FASEB J 2020; 34:11641-11657. [PMID: 32654268 DOI: 10.1096/fj.202000544r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 01/13/2023]
Abstract
The tight junction (TJ) and barrier function of colonic epithelium is highly sensitive to ionizing radiation. We evaluated the effect of lysophosphatidic acid (LPA) and its analog, Radioprotein-1, on γ-radiation-induced colonic epithelial barrier dysfunction using Caco-2 and m-ICC12 cell monolayers in vitro and mice in vivo. Mice were subjected to either total body irradiation (TBI) or partial body irradiation (PBI-BM5). Intestinal barrier function was assessed by analyzing immunofluorescence localization of TJ proteins, mucosal inulin permeability, and plasma lipopolysaccharide (LPS) levels. Oxidative stress was analyzed by measuring protein thiol oxidation and antioxidant mRNA. In Caco-2 and m-ICC12 cell monolayers, LPA attenuated radiation-induced redistribution of TJ proteins, which was blocked by a Rho-kinase inhibitor. In mice, TBI and PBI-BM5 disrupted colonic epithelial tight junction and adherens junction, increased mucosal permeability, and elevated plasma LPS; TJ disruption by TBI was more severe in Lpar2-/- mice compared to wild-type mice. RP1, administered before or after irradiation, alleviated TBI and PBI-BM5-induced TJ disruption, barrier dysfunction, and endotoxemia accompanied by protein thiol oxidation and downregulation of antioxidant gene expression, cofilin activation, and remodeling of the actin cytoskeleton. These data demonstrate that LPAR2 receptor activation prevents and mitigates γ-irradiation-induced colonic mucosal barrier dysfunction and endotoxemia.
Collapse
Affiliation(s)
- Pradeep K Shukla
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Avtar S Meena
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ruchika Gangwar
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Erzsebet Szabo
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Andrea Balogh
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sue Chin Lee
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Alain Vandewalle
- INSERM U773, Centre de Recherche Biomédicale, Bichat-Beaujon, CRB3, UFR de Médecine, Paris Cedex 18, France
| | - Gabor Tigyi
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - RadhaKrishna Rao
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|