1
|
Brauer J, Tumani M, Frey N, Lehmann LH. The cardio-oncologic burden of breast cancer: molecular mechanisms and importance of preclinical models. Basic Res Cardiol 2025; 120:91-112. [PMID: 39621070 PMCID: PMC11790711 DOI: 10.1007/s00395-024-01090-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 02/04/2025]
Abstract
Breast cancer, the most prevalent cancer affecting women worldwide, poses a significant cardio-oncological burden. Despite advancements in novel therapeutic strategies, anthracyclines, HER2 antagonists, and radiation remain the cornerstones of oncological treatment. However, each carries a risk of cardiotoxicity, though the molecular mechanisms underlying these adverse effects differ. Common mechanisms include DNA damage response, increased reactive oxygen species, and mitochondrial dysfunction, which are key areas of ongoing research for potential cardioprotective strategies. Since these mechanisms are also essential for effective tumor cytotoxicity, we explore tumor-specific effects, particularly in hereditary breast cancer linked to BRCA1 and BRCA2 mutations. These genetic variants impair DNA repair mechanisms, increase the risk of tumorigenesis and possibly for cardiotoxicity from treatments such as anthracyclines and HER2 antagonists. Novel therapies, including immune checkpoint inhibitors, are used in the clinic for triple-negative breast cancer and improve the oncological outcomes of breast cancer patients. This review discusses the molecular mechanisms underlying BRCA dysfunction and the associated pathological pathways. It gives an overview of preclinical models of breast cancer, such as genetically engineered mouse models, syngeneic murine models, humanized mouse models, and various in vitro and ex vivo systems and models to study cardiovascular side effects of breast cancer therapies. Understanding the underlying mechanism of cardiotoxicity and developing cardioprotective strategies in preclinical models are essential for improving treatment outcomes and reducing long-term cardiovascular risks in breast cancer patients.
Collapse
Affiliation(s)
- J Brauer
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - M Tumani
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - N Frey
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - L H Lehmann
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
2
|
Zong X, Zhu L, Wang Y, Wang J, Gu Y, Liu Q. Cohort Studies and Multi-omics Approaches to Low-Dose Ionizing Radiation-Induced Cardiovascular Disease: A Comprehensive Review. Cardiovasc Toxicol 2025; 25:148-165. [PMID: 39538046 DOI: 10.1007/s12012-024-09943-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
The effect of low-dose ionizing radiation exposure on the risk of cardiovascular disease (CVD) represents a significant concern in the field of radiation protection. The prevailing approach to mitigating the adverse effects of low-dose or low-dose-rate radiation does not currently incorporate the potential risk of CVD, despite the possibility that such risk may be a substantial contributor to overall health hazards. Current evidence suggests a potential association between radiation exposure and CVD; however, the overall findings remain inconclusive. This is particularly due to the uncertainty surrounding the influence of significant non-radiation risk factors on the associations reported in epidemiological studies. It is difficult to discern the underlying connection in observational epidemiology when there is substantial variation in baseline risk factors. The paucity of epidemiological research in this domain is being partially offset by the advancement of multi-omics approaches. These methods assist in identifying radiosensitive targets, comprehending underlying biological processes, and pinpointing biomarkers. This, in turn, fortifies the evidence gleaned from epidemiological studies. In this review, we delve into the body of epidemiological research pertaining to CVD induced by low-dose ionizing radiation and the application of multi-omics techniques. The integration of these two methodologies holds the promise of identifying specific molecules or biological pathways that can be employed to validate endpoints related to radiation risk assessment.
Collapse
Affiliation(s)
- Xumin Zong
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, No.238 Baidi Road, Nankai District, Tianjin, 300192, China
| | - Lin Zhu
- School of Basic Medical Sciences, Weifang Medical University, Shandong, 261000, China
| | - Yan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, No.238 Baidi Road, Nankai District, Tianjin, 300192, China
| | - Jinhan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, No.238 Baidi Road, Nankai District, Tianjin, 300192, China
| | - Yeqing Gu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, No.238 Baidi Road, Nankai District, Tianjin, 300192, China.
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, No.238 Baidi Road, Nankai District, Tianjin, 300192, China.
| |
Collapse
|
3
|
Mircea AA, Pistritu DV, Fortner A, Tanca A, Liehn EA, Bucur O. Space Travel: The Radiation and Microgravity Effects on the Cardiovascular System. Int J Mol Sci 2024; 25:11812. [PMID: 39519362 PMCID: PMC11545902 DOI: 10.3390/ijms252111812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024] Open
Abstract
Space flight modulates the functions of the cardiovascular system. The exposure to space conditions can alter the cerebral blood flow, as well as the venous return. Anemia, cardiac output changes, and increased activity of the sympathetic nervous system can also be seen. Understanding cardiac changes prepares astronauts for both better in-flight adaptations and long-term protection against cardiovascular diseases. The heart could undergo radio-degenerative effects when exposed to space radiation, increasing the risk of cardiovascular diseases in the long run. A high frequency of arrhythmias, such as ventricular/atrial premature complexes, have been reported during the Gemini and Apollo missions. Additionally, microgravity can lead to progressive degeneration of the myocytes and muscle atrophy with altered gene expression and calcium handling, along with impaired contractility. This review summarizes the potential cardiovascular effects of spaceflight and prevention measures.
Collapse
Affiliation(s)
- Andrei Alexandru Mircea
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | - Dan Valentin Pistritu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | - Andra Fortner
- Medical School, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Antoanela Tanca
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | - Elisa Anamaria Liehn
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Octavian Bucur
- Viron Molecular Medicine Institute, Boston, MA 02451, USA
- Genomics Research and Development Institute, 020021 Bucharest, Romania
| |
Collapse
|
4
|
Kussainova A, Aripova A, Ibragimova M, Bersimbaev R, Bulgakova O. Radiation-Induced miRNAs Changes and cf mtDNA Level in Trauma Surgeons: Epigenetic and Molecular Biomarkers of X-ray Exposure. Int J Mol Sci 2024; 25:8446. [PMID: 39126012 PMCID: PMC11313199 DOI: 10.3390/ijms25158446] [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: 06/26/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Exposure to ionizing radiation can result in the development of a number of diseases, including cancer, cataracts and neurodegenerative pathologies. Certain occupational groups are exposed to both natural and artificial sources of radiation as a consequence of their professional activities. The development of non-invasive biomarkers to assess the risk of exposure to ionizing radiation for these groups is of great importance. In this context, our objective was to identify epigenetic and molecular biomarkers that could be used to monitor exposure to ionizing radiation. The impact of X-ray exposure on the miRNAs profile and the level of cf mtDNA were evaluated using the RT-PCR method. The levels of pro-inflammatory cytokines in their blood were quantified using the ELISA method. A significant decrease in miR-19a-3p, miR-125b-5p and significant increase in miR-29a-3p was observed in the blood plasma of individuals exposed to X-ray. High levels of pro-inflammatory cytokines and cf mtDNA were also detected. In silico identification of potential targets of these miRNAs was conducted using MIENTURNET. VDAC1 and ALOX5 were identified as possible targets. Our study identified promising biomarkers such as miRNAs and cf mtDNA that showed a dose-dependent effect of X-ray exposure.
Collapse
Affiliation(s)
| | | | | | | | - Olga Bulgakova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (A.K.); (A.A.); (M.I.); (R.B.)
| |
Collapse
|
5
|
Ren W, Hou L, Zhang K, Chen H, Feng X, Jiang Z, Shao F, Dai J, Gao Y, He J. The sparing effect of ultra-high dose rate irradiation on the esophagus. Front Oncol 2024; 14:1442627. [PMID: 39070145 PMCID: PMC11272628 DOI: 10.3389/fonc.2024.1442627] [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: 06/02/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Background and purpose Current studies have substantiated the sparing effect of ultra-high dose rate irradiation (FLASH) in various organs including the brain, lungs, and intestines. Whether this sparing effect extends to esophageal tissue remains unexplored. This study aims to compare the different responses of esophageal tissue in histological and protein expression levels following conventional dose rate irradiation (CONV) and FLASH irradiation to ascertain the presence of a sparing effect. Methods and materials C57 female mice were randomly divided into three groups: control, CONV, and FLASH groups. The chest region of the mice in the radiation groups was exposed to a prescribed dose of 20 Gy using a modified electron linear accelerator. The CONV group received an average dose rate of 0.1 Gy/s, while the FLASH group received an average dose rate of 125 Gy/s. On the 10th day after irradiation, the mice were euthanized and their esophagi were collected for histopathological analysis. Subsequently, label-free proteomic quantification analysis was performed on esophageal tissue. The validation process involved analyzing transmission electron microscopy images and utilizing the parallel reaction monitoring method. Results Histopathology results indicated a significantly lower extent of esophageal tissue damage in the FLASH group compared to the CONV group (p < 0.05). Label-free quantitative proteomic analysis revealed that the sparing effect observed in the FLASH group may be attributed to a reduction in radiation-induced protein damage associated with mitochondrial functions, including proteins involved in the tricarboxylic acid cycle and oxidative phosphorylation, as well as a decrease in acute inflammatory responses. Conclusions Compared with CONV irradiation, a sparing effect on esophageal tissue can be observed after FLASH irradiation. This sparing effect is associated with alleviated mitochondria damage and acute inflammation.
Collapse
Affiliation(s)
- Wenting Ren
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu Hou
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ke Zhang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huan Chen
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Feng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziming Jiang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Shao
- Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianrong Dai
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yibo Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Central Laboratory and Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Mothersill C, Seymour C, Cocchetto A, Williams D. Factors Influencing Effects of Low-dose Radiation Exposure. HEALTH PHYSICS 2024; 126:296-308. [PMID: 38526248 DOI: 10.1097/hp.0000000000001816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
ABSTRACT It is now well accepted that the mechanisms induced by low-dose exposures to ionizing radiation (LDR) are different from those occurring after high-dose exposures. However, the downstream effects of these mechanisms are unclear as are the quantitative relationships between exposure, effect, harm, and risk. In this paper, we will discuss the mechanisms known to be important with an overall emphasis on how so-called "non-targeted effects" (NTE) communicate and coordinate responses to LDR. Targeted deposition of ionizing radiation energy in cells causing DNA damage is still regarded as the dominant trigger leading to all downstream events whether targeted or non-targeted. We regard this as an over-simplification dating back to formal target theory. It ignores that last 100 y of biological research into stress responses and signaling mechanisms in organisms exposed to toxic substances, including ionizing radiation. We will provide evidence for situations where energy deposition in cellular targets alone cannot be plausible as a mechanism for LDR effects. An example is where the energy deposition takes place in an organism not receiving the radiation dose. We will also discuss how effects after LDR depend more on dose rate and radiation quality rather than actual dose, which appears rather irrelevant. Finally, we will use recent evidence from studies of cataract and melanoma induction to suggest that after LDR, post-translational effects, such as protein misfolding or defects in energy metabolism or mitochondrial function, may dominate the etiology and progression of the disease. A focus on such novel pathways may open the way to successful prophylaxis and development of new biomarkers for better risk assessment after low dose exposures.
Collapse
Affiliation(s)
- Carmel Mothersill
- Department of Biology, McMaster University, 1280 Main St., Hamilton, ON, Canada L8S 4L8
| | - Colin Seymour
- Department of Biology, McMaster University, 1280 Main St., Hamilton, ON, Canada L8S 4L8
| | - Alan Cocchetto
- The National CFIDS Foundation, 285 Beach Ave., Hull, MA 02045
| | - David Williams
- Cambridge University, The Old Schools, Trinity Lane, Cambridge CB2 1TN, United Kingdom
| |
Collapse
|
7
|
Rudolf AM, Hood WR. Mitochondrial stress in the spaceflight environment. Mitochondrion 2024; 76:101855. [PMID: 38403094 DOI: 10.1016/j.mito.2024.101855] [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: 07/08/2023] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Space is a challenging environment that deregulates individual homeostasis. The main external hazards associated with spaceflight include ionizing space radiation, microgravity, isolation and confinement, distance from Earth, and hostile environment. Characterizing the biological responses to spaceflight environment is essential to validate the health risks, and to develop effective protection strategies. Mitochondria energetics is a key mechanism underpinning many physiological, ecological and evolutionary processes. Moreover, mitochondrial stress can be considered one of the fundamental features of space travel. So, we attempt to synthesize key information regarding the extensive effects of spaceflight on mitochondria. In summary, mitochondria are affected by all of the five main hazards of spaceflight at multiple levels, including their morphology, respiratory function, protein, and genetics, in various tissues and organ systems. We emphasize that investigating mitochondrial biology in spaceflight conditions should become the central focus of research on the impacts of spaceflight on human health, as this approach will help resolve numerous challenges of space health and combat several health disorders associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Agata M Rudolf
- Department of Biological Sciences, Auburn University, Auburn, AL, USA; Space Technology Centre, AGH University of Science and Technology, Krakow, Poland.
| | - Wendy R Hood
- Department of Biological Sciences, Auburn University, Auburn, AL, USA
| |
Collapse
|
8
|
Aslam MA, Ahmad H, Malik HS, Uinarni H, Karim YS, Akhmedov YM, Abdelbasset WK, Awadh SA, Abid MK, Mustafa YF, Farhood B, Sahebkar A. Radiotherapy-associated Sensorineural Hearing Loss in Pediatric Oncology Patients. Curr Med Chem 2024; 31:5351-5369. [PMID: 37190814 DOI: 10.2174/0929867330666230515112245] [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: 11/25/2022] [Revised: 03/08/2023] [Accepted: 03/27/2023] [Indexed: 05/17/2023]
Abstract
During the radiotherapeutic treatment of pediatric oncology patients, they would be at a latent risk of developing ionizing radiation-induced ototoxicity when the cochlea or auditory nerve is located within the radiation field. Sensorineural hearing loss (SNHL) is an irreversible late complication of radiotherapy, and its incidence depends on various factors such as the patient's hearing sensitivity, total radiation dose to the cochlea, radiotherapy fractionation regimen, age and chemoradiation. Importantly, this complication exhibits serious challenges to adult survivors of childhood cancer, as it has been linked to impairments in academic achievement, psychosocial development, independent living skills, and employment in the survivor population. Therefore, early detection and proper management can alleviate academic, speech, language, social, and psychological morbidity arising from hearing deficits. In the present review, we have addressed issues such as underlying mechanisms of radiation-induced SNHL, audiometric findings of pediatric cancer patients treated with radiotherapy, and management and protection measures against radiation-induced ototoxicity.
Collapse
Affiliation(s)
- Muhammad Ammar Aslam
- Department of Emergency Medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Hassaan Ahmad
- Department of Medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Hamza Sultan Malik
- Department of Medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Herlina Uinarni
- Department of Anatomy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
- Radiologist at Pantai Indah Kapuk Hospital, Jakarta, Indonesia
| | | | - Yusuf Makhmudovich Akhmedov
- Department of Pediatric Surgery, Samarkand State Medical Institute, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, Makhtumkuli Street 103, Tashkent, 100047, Uzbekistan
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Sura A Awadh
- Department of Anesthesia, Al-Mustaqbal University, Babylon, Iraq
| | - Mohammed Kadhem Abid
- Department of Anesthesia, College of Health & medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhosein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Mehrhof F, Hüttemeister J, Tanacli R, Bock M, Bögner M, Schoenrath F, Falk V, Zips D, Hindricks G, Gerds-Li JH, Hohendanner F. Cardiac radiotherapy transiently alters left ventricular electrical properties and induces cardiomyocyte-specific ventricular substrate changes in heart failure. Europace 2023; 26:euae005. [PMID: 38193546 PMCID: PMC10803027 DOI: 10.1093/europace/euae005] [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: 11/28/2023] [Accepted: 12/28/2023] [Indexed: 01/10/2024] Open
Abstract
AIMS Ongoing clinical trials investigate the therapeutic value of stereotactic cardiac radioablation (cRA) in heart failure patients with ventricular tachycardia. Animal data indicate an effect on local cardiac conduction properties. However, the exact mechanism of cRA in patients remains elusive. Aim of the current study was to investigate in vivo and in vitro myocardial properties in heart failure and ventricular tachycardia upon cRA. METHODS AND RESULTS High-density 3D electroanatomic mapping in sinus rhythm was performed in a patient with a left ventricular assist device and repeated ventricular tachycardia episodes upon several catheter-based endocardial radio-frequency ablation attempts. Subsequent to electroanatomic mapping and cRA of the left ventricular septum, two additional high-density electroanatomic maps were obtained at 2- and 4-month post-cRA. Myocardial tissue samples were collected from the left ventricular septum during 4-month post-cRA from the irradiated and borderzone regions. In addition, we performed molecular biology and mitochondrial density measurements of tissue and isolated cardiomyocytes. Local voltage was altered in the irradiated region of the left ventricular septum during follow-up. No change of local voltage was observed in the control (i.e. borderzone) region upon irradiation. Interestingly, local activation time was significantly shortened upon irradiation (2-month post-cRA), a process that was reversible (4-month post-cRA). Molecular biology unveiled an increased expression of voltage-dependent sodium channels in the irradiated region as compared with the borderzone, while Connexin43 and transforming growth factor beta were unchanged (4-month post-cRA). Moreover, mitochondrial density was decreased in the irradiated region as compared with the borderzone. CONCLUSION Our study supports the notion of transiently altered cardiac conduction potentially related to structural and functional cellular changes as an underlying mechanism of cRA in patients with ventricular tachycardia.
Collapse
Affiliation(s)
- Felix Mehrhof
- Klinik für Radioonkologie und Strahlentherapie, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Judith Hüttemeister
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Germany, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Radu Tanacli
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Germany, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Matthias Bock
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Germany, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site, Berlin, Germany
| | - Markus Bögner
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Germany, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site, Berlin, Germany
| | - Felix Schoenrath
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site, Berlin, Germany
- Klinik für Herz-, Thorax- und Gefäßchirurgie, Deutsches Herzzentrum der Charité, Berlin, Germany
| | - Volkmar Falk
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site, Berlin, Germany
- Klinik für Herz-, Thorax- und Gefäßchirurgie, Deutsches Herzzentrum der Charité, Berlin, Germany
- Translational Cardiovascular Technologies, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Berlin, Germany
| | - Daniel Zips
- Klinik für Radioonkologie und Strahlentherapie, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Gerhard Hindricks
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Germany, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site, Berlin, Germany
| | - Jin-Hong Gerds-Li
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Germany, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Felix Hohendanner
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Germany, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site, Berlin, Germany
| |
Collapse
|
10
|
Eber J, Blondet C, Le Fevre C, Chambrelant I, Hubele F, Morel O, Antoni D, Noel G. Nuclear medicine imaging methods of early radiation-induced cardiotoxicity: a ten-year systematic review. Front Oncol 2023; 13:1240889. [PMID: 37876964 PMCID: PMC10591197 DOI: 10.3389/fonc.2023.1240889] [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: 06/15/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
Introduction Radiotherapy has significantly improved cancer survival rates, but it also comes with certain unavoidable complications. Breast and thoracic irradiation, for instance, can unintentionally expose the heart to radiation, leading to damage at the cellular level within the myocardial structures. Detecting and monitoring radiation-induced heart disease early on is crucial, and several radionuclide imaging techniques have shown promise in this regard. Method In this 10-year review, we aimed to identify nuclear medicine imaging modalities that can effectively detect early cardiotoxicity following radiation therapy. Through a systematic search on PubMed, we selected nineteen relevant studies based on predefined criteria. Results The data suggest that incidental irradiation of the heart during breast or thoracic radiotherapy can cause early metabolic and perfusion changes. Nuclear imaging plays a prominent role in detecting these subclinical effects, which could potentially serve as predictors of late cardiac complications. Discussion However, further studies with larger populations, longer follow-up periods, and specific heart dosimetric data are needed to better understand the relationship between early detection of cardiac abnormalities and radiation-induced heart disease.
Collapse
Affiliation(s)
- Jordan Eber
- Institut de Cancérologie Strasbourg Europe (ICANS), Department of Radiation Oncology, Strasbourg, France
| | - Cyrille Blondet
- Institut de Cancérologie Strasbourg Europe (ICANS), Department of Nuclear Medicine, Strasbourg, France
| | - Clara Le Fevre
- Institut de Cancérologie Strasbourg Europe (ICANS), Department of Radiation Oncology, Strasbourg, France
| | - Isabelle Chambrelant
- Institut de Cancérologie Strasbourg Europe (ICANS), Department of Radiation Oncology, Strasbourg, France
| | - Fabrice Hubele
- Institut de Cancérologie Strasbourg Europe (ICANS), Department of Nuclear Medicine, Strasbourg, France
| | - Olivier Morel
- Nouvel Hôpital Civil, Department of Cardiology, Strasbourg, France
| | - Delphine Antoni
- Institut de Cancérologie Strasbourg Europe (ICANS), Department of Radiation Oncology, Strasbourg, France
| | - Georges Noel
- Institut de Cancérologie Strasbourg Europe (ICANS), Department of Radiation Oncology, Strasbourg, France
- Strasbourg University, Institut Centre national de la recherche scientifique (CNRS), Institut Pluridisciplinaire Hubert Curien (IPHC) UMR 7178, Centre Paul Strauss, UNICANCER, Strasbourg, France
| |
Collapse
|
11
|
Azimzadeh O, Merl-Pham J, Subramanian V, Oleksenko K, Krumm F, Mancuso M, Pasquali E, Tanaka IB, Tanaka S, Atkinson MJ, Tapio S, Moertl S. Late Effects of Chronic Low Dose Rate Total Body Irradiation on the Heart Proteome of ApoE -/- Mice Resemble Premature Cardiac Ageing. Cancers (Basel) 2023; 15:3417. [PMID: 37444528 DOI: 10.3390/cancers15133417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Recent epidemiologic studies support an association between chronic low-dose radiation exposure and the development of cardiovascular disease (CVD). The molecular mechanisms underlying the adverse effect of chronic low dose exposure are not fully understood. To address this issue, we have investigated changes in the heart proteome of ApoE deficient (ApoE-/-) C57Bl/6 female mice chronically irradiated for 300 days at a very low dose rate (1 mGy/day) or at a low dose rate (20 mGy/day), resulting in cumulative whole-body doses of 0.3 Gy or 6.0 Gy, respectively. The heart proteomes were compared to those of age-matched sham-irradiated ApoE-/- mice using label-free quantitative proteomics. Radiation-induced proteome changes were further validated using immunoblotting, enzyme activity assays, immunohistochemistry or targeted transcriptomics. The analyses showed persistent alterations in the cardiac proteome at both dose rates; however, the effect was more pronounced following higher dose rates. The altered proteins were involved in cardiac energy metabolism, ECM remodelling, oxidative stress, and ageing signalling pathways. The changes in PPARα, SIRT, AMPK, and mTOR signalling pathways were found at both dose rates and in a dose-dependent manner, whereas more changes in glycolysis and ECM remodelling were detected at the lower dose rate. These data provide strong evidence for the possible risk of cardiac injury following chronic low dose irradiation and show that several affected pathways following chronic irradiation overlap with those of ageing-associated heart pathology.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Section of Radiation Biology, Federal Office of Radiation Protection (BfS), 85764 Nauenberg, Germany
| | - Juliane Merl-Pham
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Munich, Germany
| | - Vikram Subramanian
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kateryna Oleksenko
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Franziska Krumm
- Section of Radiation Biology, Federal Office of Radiation Protection (BfS), 85764 Nauenberg, Germany
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00196 Rome, Italy
| | - Emanuela Pasquali
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00196 Rome, Italy
| | - Ignacia B Tanaka
- Institute for Environmental Sciences (IES), Rokkasho, Aomori 039-3212, Japan
| | - Satoshi Tanaka
- Institute for Environmental Sciences (IES), Rokkasho, Aomori 039-3212, Japan
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- Radiation Oncology, Klinikum rechts der Isar, Technical University, 80333 Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Simone Moertl
- Section of Radiation Biology, Federal Office of Radiation Protection (BfS), 85764 Nauenberg, Germany
| |
Collapse
|
12
|
Nemec-Bakk AS, Sridharan V, Seawright JW, Nelson GA, Cao M, Singh P, Cheema AK, Singh B, Li Y, Koturbash I, Miousse IR, Ewing LE, Skinner CM, Landes RD, Lowery JD, Mao XW, Singh SP, Boerma M. Effects of proton and oxygen ion irradiation on cardiovascular function and structure in a rabbit model. LIFE SCIENCES IN SPACE RESEARCH 2023; 37:78-87. [PMID: 37087182 PMCID: PMC10122719 DOI: 10.1016/j.lssr.2023.03.008] [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: 12/05/2022] [Revised: 03/13/2023] [Accepted: 03/27/2023] [Indexed: 05/03/2023]
Abstract
PURPOSE Astronauts on missions beyond low Earth orbit will be exposed to galactic cosmic radiation, and there is concern about potential adverse cardiovascular effects. Most of the research to identify cardiovascular risk of space radiation has been performed in rodent models. To aid in the translation of research results to humans, the current study identified long-term effects of high-energy charged particle irradiation on cardiovascular function and structure in a larger non-rodent animal model. MATERIALS AND METHODS At the age of 12 months, male New Zealand white rabbits were exposed to whole-body protons (250 MeV) or oxygen ions (16O, 600 MeV/n) at a dose of 0 or 0.5 Gy and were followed for 12 months after irradiation. Ultrasonography was used to measure in vivo cardiac function and blood flow parameters at 10- and 12-months post-irradiation. At 12 months after irradiation, blood cell counts and blood chemistry values were assessed, and cardiac tissue and aorta were collected for histological as well as molecular and biochemical analyses. Plasma was used for metabolomic analysis and to quantify common markers of cardiac injury. RESULTS A small but significant decrease in the percentage of circulating lymphocytes and an increase in neutrophil percentage was seen 12 months after 0.5 Gy protons, while 16O exposure resulted in an increase in monocyte percentage. Markers of cardiac injury, cardiac troponin I (cTnI) and N-Terminal pro-B-type Natriuretic Peptide were modestly increased in the proton group, and cTnI was also increased after 16O. On the other hand, metabolomics on plasma at 12 months revealed no changes. Both types of irradiation demonstrated alterations in cardiac mitochondrial morphology and an increase in left ventricular protein levels of inflammatory cell marker CD68. However, changes in cardiac function were only mild. CONCLUSION Low dose charged particle irradiation caused mild long-term changes in inflammatory markers, cardiac function, and structure in the rabbit heart, in line with previous studies in mouse and rat models.
Collapse
Affiliation(s)
- Ashley S Nemec-Bakk
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Gregory A Nelson
- Departments of Basic Sciences and Radiation Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Maohua Cao
- College of Dentistry, Texas A&M, Dallas, TX, USA
| | | | - Amrita K Cheema
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Bhaldev Singh
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Yaoxiang Li
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Igor Koturbash
- Department of Environmental Health Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Isabelle R Miousse
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Laura E Ewing
- Natural State Laboratories and Natural State Genomics, North Little Rock, AR, USA
| | - Charles M Skinner
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Reid D Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - John D Lowery
- Department of Laboratory Animal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Xiao-Wen Mao
- Departments of Basic Sciences and Radiation Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Sharda P Singh
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
13
|
Wang KX, Ye C, Yang X, Ma P, Yan C, Luo L. New Insights into the Understanding of Mechanisms of Radiation-Induced Heart Disease. Curr Treat Options Oncol 2023; 24:12-29. [PMID: 36598620 DOI: 10.1007/s11864-022-01041-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 01/05/2023]
Abstract
OPINION STATEMENT Cancer patients who receive high-dose thoracic radiotherapy may develop radiation-induced heart disease (RIHD). The clinical presentation of RIHD comprises coronary artery atherosclerosis, valvular disease, pericarditis, cardiomyopathy, and conduction defects. These complications have significantly reduced due to the improved radiotherapy techniques. However, such methods still could not avoid heart radiation exposure. Furthermore, people who received relatively low-dose radiation exposures have exhibited significantly elevated RIHD risks in cohort studies of atomic bomb survivors and occupational exposures. The increased potential in exposure to natural and artificial ionizing radiation sources has emphasized the necessity to understand the development of RIHD. The pathological processes of RIHD include endothelial dysfunction, inflammation, fibrosis, and hypertrophy. The underlying mechanisms may involve the changes in oxidative stress, DNA damage response, telomere erosion, mitochondrial dysfunction, epigenetic regulation, circulation factors, protein post-translational modification, and metabolites. This review will discuss the recent advances in the mechanisms of RIHD at cellular and molecular levels.
Collapse
Affiliation(s)
- Kai-Xuan Wang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Cong Ye
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Xu Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Ping Ma
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Chen Yan
- Department of Rheumatology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang City, Jiangxi Province, 330006, People's Republic of China.
| | - Lan Luo
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China.
| |
Collapse
|
14
|
Azimzadeh O, Moertl S, Ramadan R, Baselet B, Laiakis EC, Sebastian S, Beaton D, Hartikainen JM, Kaiser JC, Beheshti A, Salomaa S, Chauhan V, Hamada N. Application of radiation omics in the development of adverse outcome pathway networks: an example of radiation-induced cardiovascular disease. Int J Radiat Biol 2022; 98:1722-1751. [PMID: 35976069 DOI: 10.1080/09553002.2022.2110325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Epidemiological studies have indicated that exposure of the heart to doses of ionizing radiation as low as 0.5 Gy increases the risk of cardiac morbidity and mortality with a latency period of decades. The damaging effects of radiation to myocardial and endothelial structures and functions have been confirmed radiobiologically at high dose, but much less is known at low dose. Integration of radiation biology and epidemiology data is a recommended approach to improve the radiation risk assessment process. The adverse outcome pathway (AOP) framework offers a comprehensive tool to compile and translate mechanistic information into pathological endpoints which may be relevant for risk assessment at the different levels of a biological system. Omics technologies enable the generation of large volumes of biological data at various levels of complexity, from molecular pathways to functional organisms. Given the quality and quantity of available data across levels of biology, omics data can be attractive sources of information for use within the AOP framework. It is anticipated that radiation omics studies could improve our understanding of the molecular mechanisms behind the adverse effects of radiation on the cardiovascular system. In this review, we explored the available omics studies on radiation-induced cardiovascular disease (CVD) and their applicability to the proposed AOP for CVD. RESULTS The results of 80 omics studies published on radiation-induced CVD over the past 20 years have been discussed in the context of the AOP of CVD proposed by Chauhan et al. Most of the available omics data on radiation-induced CVD are from proteomics, transcriptomics, and metabolomics, whereas few datasets were available from epigenomics and multi-omics. The omics data presented here show great promise in providing information for several key events of the proposed AOP of CVD, particularly oxidative stress, alterations of energy metabolism, extracellular matrix and vascular remodeling. CONCLUSIONS The omics data presented here shows promise to inform the various levels of the proposed AOP of CVD. However, the data highlight the urgent need of designing omics studies to address the knowledge gap concerning different radiation scenarios, time after exposure and experimental models. This review presents the evidence to build a qualitative omics-informed AOP and provides views on the potential benefits and challenges in using omics data to assess risk-related outcomes.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Simone Moertl
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Raghda Ramadan
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Bjorn Baselet
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Evagelia C Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | | | | | - Jaana M Hartikainen
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
| | - Jan Christian Kaiser
- Helmholtz Zentrum München, Institute of Radiation Medicine (HMGU-IRM), 85764 Neuherberg, Germany
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Sisko Salomaa
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vinita Chauhan
- Environmental Health Science Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Komae, Tokyo 201-8511, Japan
| |
Collapse
|
15
|
Ortiz de Choudens S, Sparapani R, Narayanan J, Lohr N, Gao F, Fish BL, Zielonka M, Gasperetti T, Veley D, Beyer A, Olson J, Jacobs ER, Medhora M. Lisinopril Mitigates Radiation-Induced Mitochondrial Defects in Rat Heart and Blood Cells. Front Oncol 2022; 12:828177. [PMID: 35311118 PMCID: PMC8924663 DOI: 10.3389/fonc.2022.828177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/31/2022] [Indexed: 12/20/2022] Open
Abstract
The genetic bases and disparate responses to radiotherapy are poorly understood, especially for cardiotoxicity resulting from treatment of thoracic tumors. Preclinical animal models such as the Dahl salt-sensitive (SS) rat can serve as a surrogate model for salt-sensitive low renin hypertension, common to African Americans, where aldosterone contributes to hypertension-related alterations of peripheral vascular and renal vascular function. Brown Norway (BN) rats, in comparison, are a normotensive control group, while consomic SSBN6 with substitution of rat chromosome 6 (homologous to human chromosome 14) on an SS background manifests cardioprotection and mitochondrial preservation to SS rats after injury. In this study, 2 groups from each of the 3 rat strains had their hearts irradiated (8 Gy X 5 fractions). One irradiated group was treated with the ACE-inhibitor lisinopril, and a separate group in each strain served as nonirradiated controls. Radiation reduced cardiac end diastolic volume by 9-11% and increased thickness of the interventricular septum (11-16%) and left ventricular posterior wall (14-15%) in all 3 strains (5-10 rats/group) after 120 days. Lisinopril mitigated the increase in posterior wall thickness. Mitochondrial function was measured by the Seahorse Cell Mitochondrial Stress test in peripheral blood mononuclear cells (PBMC) at 90 days. Radiation did not alter mitochondrial respiration in PBMC from BN or SSBN6. However, maximal mitochondrial respiration and spare capacity were reduced by radiation in PBMC from SS rats (p=0.016 and 0.002 respectively, 9-10 rats/group) and this effect was mitigated by lisinopril (p=0.04 and 0.023 respectively, 9-10 rats/group). Taken together, these results indicate injury to the heart by radiation in all 3 strains of rats, although the SS rats had greater susceptibility for mitochondrial dysfunction. Lisinopril mitigated injury independent of genetic background.
Collapse
Affiliation(s)
| | - Rodney Sparapani
- Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, United States.,Cardiovascular Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jayashree Narayanan
- Department of Radiation Oncology, Froedtert & the Medical College of Wisconsin, Milwaukee WI, United States
| | - Nicole Lohr
- Cardiovascular Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States
| | - Feng Gao
- Department of Radiation Oncology, Froedtert & the Medical College of Wisconsin, Milwaukee WI, United States
| | - Brian L Fish
- Department of Radiation Oncology, Froedtert & the Medical College of Wisconsin, Milwaukee WI, United States
| | - Monika Zielonka
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tracy Gasperetti
- Department of Radiation Oncology, Froedtert & the Medical College of Wisconsin, Milwaukee WI, United States
| | - Dana Veley
- Department of Radiation Oncology, Froedtert & the Medical College of Wisconsin, Milwaukee WI, United States
| | - Andreas Beyer
- Cardiovascular Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jessica Olson
- Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, United States.,Cardiovascular Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States
| | - Elizabeth R Jacobs
- Cardiovascular Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Pulmonary Medicine, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Research Service, Veterans Affairs, Zablocki VA Medical Center (VAMC), Milwaukee, WI, United States
| | - Meetha Medhora
- Department of Radiation Oncology, Froedtert & the Medical College of Wisconsin, Milwaukee WI, United States.,Cardiovascular Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Pulmonary Medicine, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Research Service, Veterans Affairs, Zablocki VA Medical Center (VAMC), Milwaukee, WI, United States
| |
Collapse
|
16
|
Read GH, Bailleul J, Vlashi E, Kesarwala AH. Metabolic response to radiation therapy in cancer. Mol Carcinog 2022; 61:200-224. [PMID: 34961986 PMCID: PMC10187995 DOI: 10.1002/mc.23379] [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: 08/11/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 11/11/2022]
Abstract
Tumor metabolism has emerged as a hallmark of cancer and is involved in carcinogenesis and tumor growth. Reprogramming of tumor metabolism is necessary for cancer cells to sustain high proliferation rates and enhanced demands for nutrients. Recent studies suggest that metabolic plasticity in cancer cells can decrease the efficacy of anticancer therapies by enhancing antioxidant defenses and DNA repair mechanisms. Studying radiation-induced metabolic changes will lead to a better understanding of radiation response mechanisms as well as the identification of new therapeutic targets, but there are few robust studies characterizing the metabolic changes induced by radiation therapy in cancer. In this review, we will highlight studies that provide information on the metabolic changes induced by radiation and oxidative stress in cancer cells and the associated underlying mechanisms.
Collapse
Affiliation(s)
- Graham H. Read
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Justine Bailleul
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| | - Aparna H. Kesarwala
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
17
|
Azimzadeh O, Subramanian V, Sievert W, Merl-Pham J, Oleksenko K, Rosemann M, Multhoff G, Atkinson MJ, Tapio S. Activation of PPARα by Fenofibrate Attenuates the Effect of Local Heart High Dose Irradiation on the Mouse Cardiac Proteome. Biomedicines 2021; 9:biomedicines9121845. [PMID: 34944662 PMCID: PMC8698387 DOI: 10.3390/biomedicines9121845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/18/2021] [Accepted: 12/02/2021] [Indexed: 01/13/2023] Open
Abstract
Radiation-induced cardiovascular disease is associated with metabolic remodeling in the heart, mainly due to the inactivation of the transcription factor peroxisome proliferator-activated receptor alpha (PPARα), thereby inhibiting lipid metabolic enzymes. The objective of the present study was to investigate the potential protective effect of fenofibrate, a known agonist of PPARα on radiation-induced cardiac toxicity. To this end, we compared, for the first time, the cardiac proteome of fenofibrate- and placebo-treated mice 20 weeks after local heart irradiation (16 Gy) using label-free proteomics. The observations were further validated using immunoblotting, enzyme activity assays, and ELISA. The analysis showed that fenofibrate restored signalling pathways that were negatively affected by irradiation, including lipid metabolism, mitochondrial respiratory chain, redox response, tissue homeostasis, endothelial NO signalling and the inflammatory status. The results presented here indicate that PPARα activation by fenofibrate attenuates the cardiac proteome alterations induced by irradiation. These findings suggest a potential benefit of fenofibrate administration in the prevention of cardiovascular diseases, following radiation exposure.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Section Radiation Biology, Federal Office for Radiation Protection, 85764 Neuherberg, Germany
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Correspondence: ; Tel.: +49-030/18333-2242
| | - Vikram Subramanian
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa, IA 52242, USA
| | - Wolfgang Sievert
- Department of Radiation Oncology, Campus Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany; (W.S.); (G.M.)
- Central Institute for Translational Cancer Research-TranslaTUM, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Juliane Merl-Pham
- Research Unit Protein Science, Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, 80939 Munich, Germany;
| | - Kateryna Oleksenko
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
| | - Michael Rosemann
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
| | - Gabriele Multhoff
- Department of Radiation Oncology, Campus Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany; (W.S.); (G.M.)
- Central Institute for Translational Cancer Research-TranslaTUM, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Michael J. Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Chair of Radiation Biology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| |
Collapse
|
18
|
Chen C, Liu S, Zhao C, Wang R, Yu N, Long X, Wang Y, Yang F, Sun J, Lu ZL, Xia Y, Jiang Y, Yang M. Activity of keloids evaluated by multimodal photoacoustic/ultrasonic imaging system. PHOTOACOUSTICS 2021; 24:100302. [PMID: 34540586 PMCID: PMC8441086 DOI: 10.1016/j.pacs.2021.100302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/25/2021] [Accepted: 09/06/2021] [Indexed: 06/13/2023]
Abstract
Multiple objective assessments have been used to assess the activity of keloids to compare different therapeutic regimens and facilitate the best individual treatment choice for patients, but none of them are standardized. A multimodal photoacoustic/ultrasonic (PA/US) imaging system, including photoacoustic imaging, elastography, ultra-micro-angiography, and conventional US technologies (gray scale US, color Doppler US, and power Doppler US), was applied to evaluate keloids by a radiologist. Growing stages were defined by patients, and Vancouver Scar Scale (VSS) was assessed by a plastic surgeon. A comprehensive model based on multimodal ultrasound parameters (poor-echo pattern, high vascular density, decreased elasticity, and low SO2 within the keloid) and VSS might be a potential indicator of active keloids, comparing with VSS alone. The multimodal PA/US imaging system could be a promising technique for keloids assessment.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Ultrasonography, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sirui Liu
- Department of Ultrasonography, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenyang Zhao
- Department of Ultrasonography, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruojiao Wang
- Department of Ultrasonography, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nanze Yu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Long
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Youbin Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Yang
- Mindray Bio-Medical Electronics Co., Ltd., Shenzhen, China
| | - Jie Sun
- Mindray Bio-Medical Electronics Co., Ltd., Shenzhen, China
| | - Zhao Ling Lu
- Mindray North American Innovation Center, San Jose, CA, United States
| | - Yu Xia
- Department of Ultrasonography, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Jiang
- Department of Ultrasonography, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Yang
- Department of Ultrasonography, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
19
|
McCastlain K, Howell CR, Welsh CE, Wang Z, Wilson CL, Mulder HL, Easton J, Mertens AC, Zhang J, Yasui Y, Hudson MM, Robison LL, Kundu M, Ness KK. The Association of Mitochondrial Copy Number With Sarcopenia in Adult Survivors of Childhood Cancer. J Natl Cancer Inst 2021; 113:1570-1580. [PMID: 33871611 PMCID: PMC8562958 DOI: 10.1093/jnci/djab084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/07/2021] [Accepted: 04/14/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Adult childhood cancer survivors are at risk for frailty, including low muscle mass and weakness (sarcopenia). Using peripheral blood mitochondrial DNA copy number (mtDNAcn) as a proxy for functional mitochondria, this study describes cross-sectional associations between mtDNAcn and sarcopenia among survivors. METHODS Among 1762 adult childhood cancer survivors (51.6% male; median age = 29.4 years, interquartile range [IQR] = 23.3-36.8), with a median of 20.6 years from diagnosis (IQR = 15.2-28.2), mtDNAcn estimates were derived from whole-genome sequencing. A subset was validated by quantitative polymerase chain reaction and evaluated cross-sectionally using multivariable logistic regression for their association with sarcopenia, defined by race-, age-, and sex-specific low lean muscle mass or weak grip strength. All statistical tests were 2-sided. RESULTS The prevalence of sarcopenia was 27.0%, higher among female than male survivors (31.5% vs 22.9%; P < .001) and associated with age at diagnosis; 51.7% of survivors with sarcopenia were diagnosed ages 4-13 years (P = .01). Sarcopenia was most prevalent (39.0%) among central nervous system tumor survivors. Cranial radiation (odds ratio [OR] = 1.84, 95% confidence interval [CI] = 1.32 to 2.59) and alkylating agents (OR = 1.34, 95% CI = 1.04 to 1.72) increased, whereas glucocorticoids decreased odds (OR = 0.72, 95% CI = 0.56 to 0.93) of sarcopenia. mtDNAcn decreased with age (β = -0.81, P = .002) and was higher among female survivors (β = 9.23, P = .01) and among survivors with a C allele at mt.204 (β = -17.9, P = .02). In adjusted models, every standard deviation decrease in mtDNAcn increased the odds of sarcopenia 20% (OR = 1.20, 95% CI = 1.07 to 1.34). CONCLUSIONS A growing body of evidence supports peripheral blood mtDNAcn as a biomarker for adverse health outcomes; however, this study is the first to report an association between mtDNAcn and sarcopenia among childhood cancer survivors.
Collapse
Affiliation(s)
- Kelly McCastlain
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Carrie R Howell
- Department of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Catherine E Welsh
- Department of Mathematics & Computer Science, Rhodes College, Memphis, TN, USA
| | - Zhaoming Wang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Carmen L Wilson
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Heather L Mulder
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ann C Mertens
- Aflac Cancer & Blood Disorders Center at Children’s Healthcare of Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Melissa M Hudson
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mondira Kundu
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
20
|
Fu X, Tang J, Wen P, Huang Z, Najafi M. Redox interactions-induced cardiac toxicity in cancer therapy. Arch Biochem Biophys 2021; 708:108952. [PMID: 34097901 DOI: 10.1016/j.abb.2021.108952] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023]
Abstract
Cancer patients undergoing radiotherapy, chemotherapy, or targeted cancer therapy are exposed to the risk of several side effects because of the heavy production of ROS by ionizing radiation or some chemotherapy drugs. Damages to DNA, mitochondria, membrane and other organelles within normal tissue cells such as cardiomyocytes and endothelial cells lead to the release of some toxins which are associated with triggering inflammatory cells to release several types of cytokines, chemokines, ROS, and RNS. The release of some molecules following radiotherapy or chemotherapy stimulates reduction/oxidation (redox) reactions. Redox reactions cause remarkable changes in the level of reactive oxygen species (ROS) and reactive nitrogen species (RNS). Excessive production of ROS and RNS or suppression of antioxidant defense enzymes leads to damage to critical macromolecules, which may continue for long times. Increased levels of some cytokines and oxidative injury are hallmarks of heart injury following cancer therapy. Redox reactions may be involved in several heart disorders such as fibrosis, cardiomyopathy, and endothelium injury. In the current review, we explain the cellular and molecular mechanisms of redox interactions following radiotherapy, chemotherapy, and targeted cancer therapy. Afterward, we explain the evidence of the involvement of redox reactions in heart diseases.
Collapse
Affiliation(s)
- Xiao Fu
- College of Basic Medicine, Shaoyang University, Shaoyang, 422000, China
| | - Juan Tang
- College of Basic Medicine, Shaoyang University, Shaoyang, 422000, China
| | - Ping Wen
- College of Basic Medicine, Shaoyang University, Shaoyang, 422000, China
| | - Zezhi Huang
- Shaoyang Key Laboratory of Molecular Biology Diagnosis, Shaoyang, 422000, China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
21
|
Abstract
PURPOSE The aim of this article is to describe the technical development in proteomics during the last two decades with the focus on its use in radiation biology. It is written from a subjective point of view and aims not to be a scientific review of the subject. CONCLUSION Proteomics is a fast developing technique and it has already contributed greatly to our understanding of biological mechanisms following radiation exposure. Novel proteomics approaches can be used in adequately designed cellular and animal experiments and above all in big clinical trials to investigate effects of ionizing radiation in the future.
Collapse
Affiliation(s)
- Soile Tapio
- Institute of Radiation Biology and Institute for Biological and Medical Imaging, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
22
|
Sheikholeslami S, Aryafar T, Abedi-Firouzjah R, Banaei A, Dorri-Giv M, Zamani H, Ataei G, Majdaeen M, Farhood B. The role of melatonin on radiation-induced pneumonitis and lung fibrosis: A systematic review. Life Sci 2021; 281:119721. [PMID: 34146555 DOI: 10.1016/j.lfs.2021.119721] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/19/2021] [Accepted: 06/07/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Pneumonitis and lung fibrosis, as the most common compliances of lung irradiation, can affect the quality of life. The use of radio-protective agents can ameliorate these injuries. This study aimed to review the potential protective role of melatonin in the treatment of radiation-induced Pneumonitis and lung fibrosis. METHODS The current systematic study was conducted based on PRISMA guidelines to identify relevant literature on " the effect of melatonin on radiation-induced pneumonitis and lung fibrosis" in the electronic databases of Web of Science, Embase, PubMed, and Scopus up to January 2021. Eighty-one articles were screened in accordance with the inclusion and exclusion criteria of the study. Finally, eight articles were included in this systematic review. RESULTS The finding showed that the lung irradiation-induced pneumonitis and lung fibrosis. The co-treatment with melatonin could alleviate these compliances through its anti-oxidant and anti-inflammatory actions. Melatonin through upregulation of some enzymes such as catalase, superoxide dismutase, glutathione, NADPH oxidases 2 and 4, dual oxidases 1 and 2, and also downregulation of malondialdehyde reduced oxidative stress following lung radiation. Moreover, melatonin through its anti-inflammatory effects, can attenuate the increased levels of nuclear factor kappa B, tumor necrosis factor alpha, transforming growth factor beta 1, SMAD2, interleukin (IL)-4, IL-4 receptor-a1 (IL4ra1), and IL-1 beta following lung radiation. The histological damages induced by ionizing radiation were also alleviated by co-treatment with melatonin. CONCLUSION According to the obtained results, it was found that melatonin can have anti-pneumonitis and anti-fibrotic following lung irradiation.
Collapse
Affiliation(s)
- Sahar Sheikholeslami
- Department of Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Tayebeh Aryafar
- Department of Radiation Sciences, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Amin Banaei
- Department of Medical Physics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoumeh Dorri-Giv
- Nuclear Medicine Research Center, Department of Nuclear Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Zamani
- Department of Medical Physics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Gholamreza Ataei
- Department of Radiology Technology, Faculty of Paramedical Sciences, Babol University of Medical Science, Babol, Iran
| | - Mehrsa Majdaeen
- Department of Radiotherapy and Oncology, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
23
|
Sheikholeslami S, Khodaverdian S, Dorri-Giv M, Mohammad Hosseini S, Souri S, Abedi-Firouzjah R, Zamani H, Dastranj L, Farhood B. The radioprotective effects of alpha-lipoic acid on radiotherapy-induced toxicities: A systematic review. Int Immunopharmacol 2021; 96:107741. [PMID: 33989970 DOI: 10.1016/j.intimp.2021.107741] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/16/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE Radiation therapy is one of the main cancer treatment modalities applied in 50-70% of cancer patients. Despite the many advantages of this treatment, such as non-invasiveness, organ-preservation, and spatiotemporal flexibility in tumor targeting, it can lead to complications in irradiated healthy cells/tissues. In this regard, the use of radio-protective agents can alleviate radiation-induced complications. This study aimed to review the potential role of alpha-lipoic acid in the prevention/reduction of radiation-induced toxicities on healthy cells/tissues. METHODS A systematic search was performed following PRISMA guidelines to identify relevant literature on the "role of alpha-lipoic acid in the treatment of radiotherapy-induced toxicity" in the electronic databases of Web of Science, Embase, PubMed, and Scopus up to January 2021. Based on the inclusion and exclusion criteria of the present study, 278 articles were screened. Finally, 29 articles were included in this systematic review. RESULTS The obtained results showed that in experimental in vivo models, the radiation-treated groups had decreased survival rate and body weight compared to the control groups. It was also found that radiation can induce mild to severe toxicities on gastrointestinal, circulatory, reproductive, central nervous, respiratory, endocrine, exocrine systems, etc. However, the use of alpha-lipoic acid could alleviate the radiation-induced toxicities in most cases. This radio-protective agent exerts its effects through mechanisms of anti-oxidant, anti-apoptosis, anti-inflammatory, and so on. CONCLUSION According to the obtained results, it can be mentioned that co-treatment of alpha-lipoic acid with radiotherapy ameliorates the radiation-induced toxicities in healthy cells/tissues.
Collapse
Affiliation(s)
- Sahar Sheikholeslami
- Department of Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shaghayegh Khodaverdian
- Department of Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoumeh Dorri-Giv
- Nuclear Medicine Research Center, Department of Nuclear Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyyed Mohammad Hosseini
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shokoufeh Souri
- Department of Medical Physics, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Hamed Zamani
- Department of Medical Physics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Leila Dastranj
- Department of Physics, Hakim Sabzevari Universuty, Sabzevar, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan. Iran.
| |
Collapse
|
24
|
Little MP, Azizova TV, Hamada N. Low- and moderate-dose non-cancer effects of ionizing radiation in directly exposed individuals, especially circulatory and ocular diseases: a review of the epidemiology. Int J Radiat Biol 2021; 97:782-803. [PMID: 33471563 PMCID: PMC10656152 DOI: 10.1080/09553002.2021.1876955] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/24/2020] [Accepted: 01/09/2021] [Indexed: 01/29/2023]
Abstract
PURPOSE There are well-known correlations between high and moderate doses (>0.5 Gy) of ionizing radiation exposure and circulatory system damage, also between radiation and posterior subcapsular cataract. At lower dose correlations with circulatory disease are emerging in the Japanese atomic bomb survivors and in some occupationally exposed groups, and are still to some extent controversial. Heterogeneity in excess relative risks per unit dose in epidemiological studies at low (<0.1 Gy) and at low-moderate (>0.1 Gy, <0.5 Gy) doses may result from confounding and other types of bias, and effect modification by established risk factors. There is also accumulating evidence of excess cataract risks at lower dose and low dose rate in various cohorts. Other ocular endpoints, specifically glaucoma and macular degeneration have been little studied. In this paper, we review recent epidemiological findings, and also discuss some of the underlying radiobiology of these conditions. We briefly review some other types of mainly neurological nonmalignant disease in relation to radiation exposure. CONCLUSIONS We document statistically significant excess risk of the major types of circulatory disease, specifically ischemic heart disease and stroke, in moderate- or low-dose exposed groups, with some not altogether consistent evidence suggesting dose-response non-linearity, particularly for stroke. However, the patterns of risk reported are not straightforward. We also document evidence of excess risks at lower doses/dose-rates of posterior subcapsular and cortical cataract in the Chernobyl liquidators, US Radiologic Technologists and Russian Mayak nuclear workers, with fundamentally linear dose-response. Nuclear cataracts are less radiogenic. For other ocular endpoints, specifically glaucoma and macular degeneration there is very little evidence of effects at low doses; radiation-associated glaucoma has been documented only for doses >5 Gy, and so has the characteristics of a tissue reaction. There is some evidence of neurological detriment following low-moderate dose (∼0.1-0.2 Gy) radiation exposure in utero or in early childhood.
Collapse
Affiliation(s)
- Mark P Little
- Radiation Epidemiology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Tamara V Azizova
- Clinical Department, Southern Urals Biophysics Institute, Ozyorsk, Ozyorsk Chelyabinsk Region, Russia
| | - Nobuyuki Hamada
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Komae, Tokyo, Japan
| |
Collapse
|
25
|
Tapio S, Little MP, Kaiser JC, Impens N, Hamada N, Georgakilas AG, Simar D, Salomaa S. Ionizing radiation-induced circulatory and metabolic diseases. ENVIRONMENT INTERNATIONAL 2021; 146:106235. [PMID: 33157375 PMCID: PMC10686049 DOI: 10.1016/j.envint.2020.106235] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/09/2020] [Accepted: 10/08/2020] [Indexed: 05/23/2023]
Abstract
Risks to health are the prime consideration in all human situations of ionizing radiation exposure and therefore of relevance to radiation protection in all occupational, medical, and public exposure situations. Over the past few decades, advances in therapeutic strategies have led to significant improvements in cancer survival rates. However, a wide range of long-term complications have been reported in cancer survivors, in particular circulatory diseases and their major risk factors, metabolic diseases. However, at lower levels of exposure, the evidence is less clear. Under real-life exposure scenarios, including radiotherapy, radiation effects in the whole organism will be determined mainly by the response of normal tissues receiving relatively low doses, and will be mediated and moderated by systemic effects. Therefore, there is an urgent need for further research on the impact of low-dose radiation. In this article, we review radiation-associated risks of circulatory and metabolic diseases in clinical, occupational or environmental exposure situations, addressing epidemiological, biological, risk modelling, and systems biology aspects, highlight the gaps in knowledge and discuss future directions to address these gaps.
Collapse
Affiliation(s)
- Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health GmbH, Neuherberg, Germany.
| | - Mark P Little
- Radiation Epidemiology Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), MD, USA
| | - Jan Christian Kaiser
- Institute of Radiation Medicine, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Nathalie Impens
- Institute of Environment, Health and Safety, Biosphere Impact Studies, SCK•CEN, Mol, Belgium
| | - Nobuyuki Hamada
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Tokyo, Japan
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece
| | - David Simar
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Sisko Salomaa
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
26
|
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
|
27
|
Klein K, He K, Younes AI, Barsoumian HB, Chen D, Ozgen T, Mosaffa S, Patel RR, Gu M, Novaes J, Narayanan A, Cortez MA, Welsh JW. Role of Mitochondria in Cancer Immune Evasion and Potential Therapeutic Approaches. Front Immunol 2020; 11:573326. [PMID: 33178201 PMCID: PMC7596324 DOI: 10.3389/fimmu.2020.573326] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/18/2020] [Indexed: 11/13/2022] Open
Abstract
The role of mitochondria in cancer formation and progression has been studied extensively, but much remains to be understood about this complex relationship. Mitochondria regulate many processes that are known to be altered in cancer cells, from metabolism to oxidative stress to apoptosis. Here, we review the evolving understanding of the role of mitochondria in cancer cells, and highlight key evidence supporting the role of mitochondria in cancer immune evasion and the effects of mitochondria-targeted antitumor therapy. Also considered is how knowledge of the role of mitochondria in cancer can be used to design and improve cancer therapies, particularly immunotherapy and radiation therapy. We further offer critical insights into the mechanisms by which mitochondria influence tumor immune responses, not only in cancer cells but also in immune cells. Given the central role of mitochondria in the complex interactions between cancer and the immune system, high priority should be placed on developing rational strategies to address mitochondria as potential targets in future preclinical and clinical studies. We believe that targeting mitochondria may provide additional opportunities in the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Katherine Klein
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,McGovern Medical School at UTHealth, Houston, TX, United States
| | - Kewen He
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Ahmed I Younes
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dawei Chen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Tugce Ozgen
- Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sara Mosaffa
- Department of Molecular Biosciences, The University of Texas at Austin, Houston, TX, United States
| | - Roshal R Patel
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Meidi Gu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jose Novaes
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, The Bronx, NY, United States
| | - Aarthi Narayanan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
28
|
Azimzadeh O, Azizova T, Merl-Pham J, Blutke A, Moseeva M, Zubkova O, Anastasov N, Feuchtinger A, Hauck SM, Atkinson MJ, Tapio S. Chronic Occupational Exposure to Ionizing Radiation Induces Alterations in the Structure and Metabolism of the Heart: A Proteomic Analysis of Human Formalin-Fixed Paraffin-Embedded (FFPE) Cardiac Tissue. Int J Mol Sci 2020; 21:ijms21186832. [PMID: 32957660 PMCID: PMC7555548 DOI: 10.3390/ijms21186832] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 12/23/2022] Open
Abstract
Epidemiological studies on workers employed at the Mayak plutonium enrichment plant have demonstrated an association between external gamma ray exposure and an elevated risk of ischemic heart disease (IHD). In a previous study using fresh-frozen post mortem samples of the cardiac left ventricle of Mayak workers and non-irradiated controls, we observed radiation-induced alterations in the heart proteome, mainly downregulation of mitochondrial and structural proteins. As the control group available at that time was younger than the irradiated group, we could not exclude age as a confounding factor. To address this issue, we have now expanded our study to investigate additional samples using archival formalin-fixed paraffin-embedded (FFPE) tissue. Importantly, the control group studied here is older than the occupationally exposed (>500 mGy) group. Label-free quantitative proteomics analysis showed that proteins involved in the lipid metabolism, sirtuin signaling, mitochondrial function, cytoskeletal organization, and antioxidant defense were the most affected. A histopathological analysis elucidated large foci of fibrotic tissue, myocardial lipomatosis and lymphocytic infiltrations in the irradiated samples. These data highlight the suitability of FFPE material for proteomics analysis. The study confirms the previous results emphasizing the role of adverse metabolic changes in the radiation-associated IHD. Most importantly, it excludes age at the time of death as a confounding factor.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany; (N.A.); (M.J.A.); (S.T.)
- Correspondence: ; Tel.: +49-89-3187-3887
| | - Tamara Azizova
- Southern Urals Biophysics Institute (SUBI), Russian Federation, 456780 Ozyorsk, Russia; (T.A.); (M.M.); (O.Z.)
| | - Juliane Merl-Pham
- Helmholtz Zentrum München—German Research Centre for Environmental Health, Research Unit Protein Science, 80939 Munich, Germany; (J.M.-P.); (S.M.H.)
| | - Andreas Blutke
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Research Unit Analytical Pathology, 85764 Neuherberg, Germany; (A.B.); (A.F.)
| | - Maria Moseeva
- Southern Urals Biophysics Institute (SUBI), Russian Federation, 456780 Ozyorsk, Russia; (T.A.); (M.M.); (O.Z.)
| | - Olga Zubkova
- Southern Urals Biophysics Institute (SUBI), Russian Federation, 456780 Ozyorsk, Russia; (T.A.); (M.M.); (O.Z.)
| | - Natasa Anastasov
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany; (N.A.); (M.J.A.); (S.T.)
| | - Annette Feuchtinger
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Research Unit Analytical Pathology, 85764 Neuherberg, Germany; (A.B.); (A.F.)
| | - Stefanie M. Hauck
- Helmholtz Zentrum München—German Research Centre for Environmental Health, Research Unit Protein Science, 80939 Munich, Germany; (J.M.-P.); (S.M.H.)
| | - Michael J. Atkinson
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany; (N.A.); (M.J.A.); (S.T.)
- Chair of Radiation Biology, Technical University of Munich, 81675 Munich, Germany
| | - Soile Tapio
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany; (N.A.); (M.J.A.); (S.T.)
| |
Collapse
|
29
|
Fu S, Li Z, Xiao L, Hu W, Zhang L, Xie B, Zhou Q, He J, Qiu Y, Wen M, Peng Y, Gao J, Tan R, Deng Y, Weng L, Sun LQ. Glutamine Synthetase Promotes Radiation Resistance via Facilitating Nucleotide Metabolism and Subsequent DNA Damage Repair. Cell Rep 2020; 28:1136-1143.e4. [PMID: 31365859 DOI: 10.1016/j.celrep.2019.07.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/12/2019] [Accepted: 06/27/2019] [Indexed: 01/10/2023] Open
Abstract
Radiation resistance is a critical problem in radiotherapy for cancer. Radiation kills tumor cells mainly through causing DNA damage. Thus, efficiency of DNA damage repair is one of the most important factors that limits radiotherapy efficacy. Glutamine physiologically functions to generate protein and nucleotides. Here, we study the impact of glutamine metabolism on cancer therapeutic responses, in particular under irradiation-induced stress. We show that radiation-resistant cells possessed low glycolysis, mitochondrial respiration, and TCA cycle but high glutamine anabolism. Transcriptome analyses revealed that glutamine synthetase (GS), an enzyme catalyzing glutamate and ammonia to glutamine, was responsible for the metabolic alteration. ChIP and luciferase reporter assays revealed that GS could be transcriptionally regulated by STAT5. Knockdown of GS delayed DNA repair, weakened nucleotide metabolism, and enhanced radiosensitivity both in vitro and in vivo. Our data show that GS links glutamine metabolism to radiotherapy response through fueling nucleotide synthesis and accelerating DNA repair.
Collapse
Affiliation(s)
- Shujun Fu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China
| | - Zhi Li
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China; Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China
| | - Lanbo Xiao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha 410008, China
| | - Wenfeng Hu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China
| | - Lu Zhang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China
| | - Bowen Xie
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China
| | - Qin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Junju He
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yanfang Qiu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ming Wen
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China
| | - Yanni Peng
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Gao
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rong Tan
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China; Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China
| | - Yuezhen Deng
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China; Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China
| | - Liang Weng
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China; Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China
| | - Lun-Quan Sun
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China; Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China; National Clinical Research Center for Gerontology, Changsha 410008, China.
| |
Collapse
|
30
|
Yahyapour R, Amini P, Saffar H, Motevaseli E, Farhood B, Pooladvand V, Shabeeb D, Musa AE, Najafi M. Protective Effect of Metformin, Resveratrol and Alpha-lipoic Acid on Radiation- Induced Pneumonitis and Fibrosis: A Histopathological Study. Curr Drug Res Rev 2020; 11:111-117. [PMID: 31875783 DOI: 10.2174/2589977511666191018180758] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/22/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Radiation-induced pneumonitis and fibrosis are the most common side effects of chest radiotherapy. They result from massive and chronic production of Reactive Oxygen Species (ROS), inhibition of antioxidant enzymes as well as the release of several inflammatory mediators. In this study, we aimed to detect the radioprotective effects of metformin (as inhibitor of mitochondrial ROS), resveratrol (as stimulator of antioxidant defense enzymes) and alpha-lipoic acid (as direct antioxidant) for alleviating radiation-induced pneumonitis and fibrosis. METHODS 80 Male Mice were randomly allotted to eight groups which include G1: control; G2: resveratrol; G3: alpha-lipoic acid; G4: metformin; G5: radiation; G6: radiation plus resveratrol; G7: radiation plus alpha-lipoic acid; G8: radiation plus metformin. Drugs' doses were as follows: 100 mg/kg metformin, 200 mg/kg resveratrol and 200 mg/kg alpha-lipoic acid. Irradiation with a single radiation dose of 18 Gy was performed using a cobalt-60 (60Co) gamma-ray source. After 80 days, all mice were sacrificed and their lung tissues evaluated for morphological changes using histopathological markers. RESULTS Irradiation led to acute pneumonitis including infiltration of inflammatory cells and damages to alveolar and vascular, as well as mild fibrosis. Metformin, alpha-lipoic acid and resveratrol were able to reduce pneumonitis and overcome radiation-induced fibrosis. CONCLUSION All agents could protect against radiation-induced lung injury moderately. It is possible that administering higher doses of these drugs over a long period of time could give better radioprotection of the lung.
Collapse
Affiliation(s)
- Rasoul Yahyapour
- School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Hana Saffar
- Clinical and Anatomical Pathologist at Tehran University of Medical Science, Imam Khomeini Hospital Complex, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Vahid Pooladvand
- Department of Sciences, Faculty of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences, Tehran, Iran.,Department of Physics, Federal University of Technology, Minna, Nigeria
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
31
|
Davidson CQ, Tharmalingam S, Niccoli S, Nemec-Bakk A, Khurana S, Murray A, Tai TC, Boreham DR, Khaper N, Lees SJ. Dose threshold for radiation induced fetal programming in a mouse model at 4 months of age: Hepatic expression of genes and proteins involved in glucose metabolism and glucose uptake in brown adipose tissue. PLoS One 2020; 15:e0231650. [PMID: 32315370 PMCID: PMC7173787 DOI: 10.1371/journal.pone.0231650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/27/2020] [Indexed: 01/21/2023] Open
Abstract
Exposure to ionizing radiation contributing to negative health outcomes is a widespread concern. However, the impact of low dose and sub-lethal dose radiation (SLDR) exposures remain contentious, particularly in pregnant women who represent a vulnerable group. The fetal programming hypothesis states that an adverse in utero environment or stress during development of an embryo or fetus can result in permanent physiologic changes often resulting in progressive metabolic dysfunction with age. To assess changes in gene expression profiles of glucose/insulin signaling and lipid metabolism caused by radiation exposure in utero, pregnant C57Bl/6J mice were irradiated using a dose response ranging from low dose to SLDR and compared to a Sham-irradiated group. mRNA expression analysis in 16 week old offspring (n = 84) revealed that genes involved in metabolic function including glucose metabolism, insulin signaling and lipid metabolism were unaffected by prenatal radiation exposures up to 300 mGy. However, female offspring of dams exposed to 1000 mGy had upregulated expression of genes contributing to insulin resistance and gluconeogenesis. In a second cohort of mice, the effects of SLDR on fetal programming of hepatic SOCS3 and PEPCK protein expression were assessed. 4 month old female offspring of dams irradiated at 1000 mGy had: 1) increased liver weights, 2) increased hepatic expression of proteins involved in glucose metabolism and 3) increased 18F-fluorodeoxyglucose (FDG) uptake in interscapular brown adipose tissue (IBAT) measured by positron emission tomography (PET) (n = 25). The results of this study indicate that prenatal radiation exposure does not affect metabolic function up to 300 mGy and 1000 mGy may be a threshold dose for sex-specific alterations in glucose uptake and hepatic gene and protein expression of SOCS3, PEPCK, PPARGC1A and PPARGC1B. These findings suggest that SLDR doses alter glucose uptake in IBAT and hepatic gene and protein expression of offspring and these changes may progress with age.
Collapse
Affiliation(s)
| | - Sujeenthar Tharmalingam
- Division of Medical Sciences, Northern Ontario School of Medicine, Laurentian University, Sudbury, Ontario, Canada
| | - Sarah Niccoli
- Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada
| | - Ashley Nemec-Bakk
- Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada
| | - Sandhya Khurana
- Division of Medical Sciences, Northern Ontario School of Medicine, Laurentian University, Sudbury, Ontario, Canada
| | - Alyssa Murray
- Division of Medical Sciences, Northern Ontario School of Medicine, Laurentian University, Sudbury, Ontario, Canada
| | - T. C. Tai
- Division of Medical Sciences, Northern Ontario School of Medicine, Laurentian University, Sudbury, Ontario, Canada
| | - Douglas R. Boreham
- Division of Medical Sciences, Northern Ontario School of Medicine, Laurentian University, Sudbury, Ontario, Canada
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Neelam Khaper
- Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada
- Division of Medical Sciences, Northern Ontario School of Medicine, Lakehead University, Thunder Bay, Ontario, Canada
| | - Simon J. Lees
- Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada
- Division of Medical Sciences, Northern Ontario School of Medicine, Lakehead University, Thunder Bay, Ontario, Canada
- * E-mail:
| |
Collapse
|
32
|
Schlaak RA, Frei A, SenthilKumar G, Tsaih SW, Wells C, Mishra J, Flister MJ, Camara AKS, Bergom C. Differences in Expression of Mitochondrial Complexes Due to Genetic Variants May Alter Sensitivity to Radiation-Induced Cardiac Dysfunction. Front Cardiovasc Med 2020; 7:23. [PMID: 32195269 PMCID: PMC7066205 DOI: 10.3389/fcvm.2020.00023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/11/2020] [Indexed: 01/02/2023] Open
Abstract
Radiation therapy is received by over half of all cancer patients. However, radiation doses may be constricted due to normal tissue side effects. In thoracic cancers, including breast and lung cancers, cardiac radiation is a major concern in treatment planning. There are currently no biomarkers of radiation-induced cardiotoxicity. Complex genetic modifiers can contribute to the risk of radiation-induced cardiotoxicities, yet these modifiers are largely unknown and poorly understood. We have previously reported the SS (Dahl salt-sensitive/Mcwi) rat strain is a highly sensitized model of radiation-induced cardiotoxicity compared to the more resistant Brown Norway (BN) rat strain. When rat chromosome 3 from the resistant BN rat strain is substituted into the SS background (SS.BN3 consomic), it significantly attenuates radiation-induced cardiotoxicity, demonstrating inherited genetic variants on rat chromosome 3 modify radiation sensitivity. Genes involved with mitochondrial function were differentially expressed in the hearts of SS and SS.BN3 rats 1 week after radiation. Here we further assessed differences in mitochondria-related genes between the sensitive SS and resistant SS.BN3 rats. We found mitochondrial-related gene expression differed in untreated hearts, while no differences in mitochondrial morphology were seen 1 week after localized heart radiation. At 12 weeks after localized cardiac radiation, differences in mitochondrial complex protein expression in the left ventricles were seen between the SS and SS.BN3 rats. These studies suggest that differences in mitochondrial gene expression caused by inherited genetic variants may contribute to differences in sensitivity to cardiac radiation.
Collapse
Affiliation(s)
- Rachel A Schlaak
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anne Frei
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Gopika SenthilKumar
- Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Shirng-Wern Tsaih
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Clive Wells
- Electron Microscope Facility, Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jyotsna Mishra
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michael J Flister
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Amadou K S Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Carmen Bergom
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
33
|
Relationship Between Changes in Myocardial F-18 Fluorodeoxyglucose Uptake and Radiation Dose After Adjuvant Three-Dimensional Conformal Radiotherapy in Patients with Breast Cancer. J Clin Med 2020; 9:jcm9030666. [PMID: 32131475 PMCID: PMC7141354 DOI: 10.3390/jcm9030666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/24/2020] [Accepted: 02/28/2020] [Indexed: 12/28/2022] Open
Abstract
This study aimed to assess the relationship between radiation dose and changes in the irradiated myocardial F-18 fluorodeoxyglucose (FDG) uptake after radiotherapy (RT) in breast cancer patients. The data of 55 patients with left and 48 patients with right breast cancer who underwent curative surgical resection and adjuvant three-dimensional conformal RT and staging (PET1), post-adjuvant chemotherapy (PET2), post-RT (PET3), and surveillance (PET4) FDG positron emission tomography/computed tomography (PET/CT) were retrospectively reviewed. The median interval between PET1 and curative surgical resection, between the end of adjuvant chemotherapy and PET2, between the end of RT and PET3, and between the end of RT and PET4 were five days, 13 days, 132 days, and 353 days, respectively. The myocardial-to-blood pool uptake ratio was measured in all patients. For patients with left breast cancer, the 30 Gy- (30 Gy) and 47.5 Gy-irradiated myocardium-to-low-irradiated myocardium (47.5 Gy) FDG uptake ratios were additionally measured. There were no differences in the myocardial-to-blood pool uptake ratios between left and right breast cancer on all PET scans. For left breast cancer, higher 30 Gy and 47.5 Gy uptake ratios were observed on PET3 than on PET1 and PET2. Both uptake ratios decreased on PET4 compared to PET3, but, were still higher compared to PET1. On PET3 and PET4, the 47.5 Gy were higher than the 30 Gy uptake ratios, while there were no differences between them on PET1 and PET2. Although the whole myocardium FDG uptake showed no significant change, the irradiated myocardium FDG uptake significantly increased after RT and was related to radiation dose to the myocardium in breast cancer patients. These results might be an imaging evidence that supports the increased risk of heart disease after RT in patients with left breast cancer.
Collapse
|
34
|
Livingston K, Schlaak RA, Puckett LL, Bergom C. The Role of Mitochondrial Dysfunction in Radiation-Induced Heart Disease: From Bench to Bedside. Front Cardiovasc Med 2020; 7:20. [PMID: 32154269 PMCID: PMC7047199 DOI: 10.3389/fcvm.2020.00020] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/05/2020] [Indexed: 12/25/2022] Open
Abstract
Radiation is a key modality in the treatment of many cancers; however, it can also affect normal tissues adjacent to the tumor, leading to toxic effects. Radiation to the thoracic region, such as that received as part of treatment for breast and lung cancer, can result in incidental dose to the heart, leading to cardiac dysfunction, such as pericarditis, coronary artery disease, ischemic heart disease, conduction defects, and valvular dysfunction. The underlying mechanisms for these morbidities are currently being studied but are not entirely understood. There has been increasing focus on the role of radiation-induced mitochondrial dysfunction and the ensuing impact on various cardiac functions in both preclinical models and in humans. Cardiomyocyte mitochondria are critical to cardiac function, and mitochondria make up a substantial part of a cardiomyocyte's volume. Mitochondrial dysfunction can also alter other cell types in the heart. This review summarizes several factors related to radiation-induced mitochondrial dysfunction in cardiomyocytes and endothelial cells. These factors include mitochondrial DNA mutations, oxidative stress, alterations in various mitochondrial function-related transcription factors, and apoptosis. Through improved understanding of mitochondria-dependent mechanisms of radiation-induced heart dysfunction, potential therapeutic targets can be developed to assist in prevention and treatment of radiation-induced heart damage.
Collapse
Affiliation(s)
- Katie Livingston
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Rachel A Schlaak
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lindsay L Puckett
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Carmen Bergom
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States.,Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
35
|
McDonald JT, Stainforth R, Miller J, Cahill T, da Silveira WA, Rathi KS, Hardiman G, Taylor D, Costes SV, Chauhan V, Meller R, Beheshti A. NASA GeneLab Platform Utilized for Biological Response to Space Radiation in Animal Models. Cancers (Basel) 2020; 12:E381. [PMID: 32045996 PMCID: PMC7072278 DOI: 10.3390/cancers12020381] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Ionizing radiation from galactic cosmic rays (GCR) is one of the major risk factors that will impact the health of astronauts on extended missions outside the protective effects of the Earth's magnetic field. The NASA GeneLab project has detailed information on radiation exposure using animal models with curated dosimetry information for spaceflight experiments. Methods: We analyzed multiple GeneLab omics datasets associated with both ground-based and spaceflight radiation studies that included in vivo and in vitro approaches. A range of ions from protons to iron particles with doses from 0.1 to 1.0 Gy for ground studies, as well as samples flown in low Earth orbit (LEO) with total doses of 1.0 mGy to 30 mGy, were utilized. Results: From this analysis, we were able to identify distinct biological signatures associating specific ions with specific biological responses due to radiation exposure in space. For example, we discovered changes in mitochondrial function, ribosomal assembly, and immune pathways as a function of dose. Conclusions: We provided a summary of how the GeneLab's rich database of omics experiments with animal models can be used to generate novel hypotheses to better understand human health risks from GCR exposures.
Collapse
Affiliation(s)
| | - Robert Stainforth
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, ON K1A-1C1, Canada; (R.S.); (V.C.)
| | - Jack Miller
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA;
| | - Thomas Cahill
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.)
| | - Willian A. da Silveira
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.)
| | - Komal S. Rathi
- Department of Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Gary Hardiman
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.)
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Deanne Taylor
- Department of Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- The Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- The Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sylvain V. Costes
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA;
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, ON K1A-1C1, Canada; (R.S.); (V.C.)
| | - Robert Meller
- Department of Neurobiology and Pharmacology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Afshin Beheshti
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA;
| |
Collapse
|
36
|
Barlaz Us S, Vezir O, Yildirim M, Bayrak G, Yalin S, Balli E, Yalin AE, Çömelekoğlu Ü. Protective effect of N-acetyl cysteine against radiotherapy-induced cardiac damage. Int J Radiat Biol 2020; 96:661-670. [PMID: 31990607 DOI: 10.1080/09553002.2020.1721605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Purpose: Although radiotherapy (RT) is an important component of cancer treatment, it induces adverse tissue reactions in the around of cancer tissue. Therefore, radioprotectives are needed to protect normal tissues. The aim of this study was to investigate the radioprotective effect of N-acetylcysteine (NAC) on RT-induced cardiac damage in rats for the acute term.Materials and methods: The animals were divided into four groups. The rats in control group were injected with saline for 7 d; the rats in NAC group were injected NAC at dose of 240 mg/kg d for 7 d; the rats in RT group were injected with saline for 7 d plus was irradiated 1 h after the last injection and the rats in NAC + RT group were injected with NAC for 7 d and irradiated 1 h after the last NAC dose. The electrocardiogram was recorded and evaluated PR interval, QRS duration, QT interval, T wave alterations and heart rate. Serum interleukin-4, interleukin-6, tumor necrosis factor-alpha, interleukin 1 beta, galectin-3 levels and creatine kinase and creatine kinase isoenzyme-MB activities were determined in all groups. Also, tissue malondialdehyde (MDA) and nitric oxide levels, superoxide dismutase, catalase and glutathione peroxidase activities were determined. In addition, histological changes of heart were evaluated. All measurements were performed 24 h after RT.Results: In the RT group, findings supporting cardiac injury were observed in the electrocardiogram. Also, cytokine levels and oxidative stress were significantly increased. Pretreatment of rats with NAC ameliorated cardiac injury induced by RT.Conclusions: Our findings suggested that NAC may be a potential radioprotector which is capable of preventing cardiac damage.
Collapse
Affiliation(s)
- Songul Barlaz Us
- Department of Radiation Oncology, Mersin University, Mersin, Turkey
| | - Ozden Vezir
- Department of Cardiovascular Surgery, Mersin City Hospital, Mersin, Turkey
| | - Metin Yildirim
- Department of Biochemistry, Mersin University, Mersin, Turkey
| | - Gülsen Bayrak
- Department of Histology-Embryology, Mersin University, Mersin, Turkey
| | - Serap Yalin
- Department of Biochemistry, Mersin University, Mersin, Turkey
| | - Ebru Balli
- Department of Histology-Embryology, Mersin University, Mersin, Turkey
| | | | | |
Collapse
|
37
|
Amorim NML, Kee A, Coster ACF, Lucas C, Bould S, Daniel S, Weir JM, Mellett NA, Barbour J, Meikle PJ, Cohn RJ, Turner N, Hardeman EC, Simar D. Irradiation impairs mitochondrial function and skeletal muscle oxidative capacity: significance for metabolic complications in cancer survivors. Metabolism 2020; 103:154025. [PMID: 31765667 DOI: 10.1016/j.metabol.2019.154025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND Metabolic complications are highly prevalent in cancer survivors treated with irradiation but the underlying mechanisms remain unknown. METHODS Chow or high fat-fed C57Bl/6J mice were irradiated (6Gy) before investigating the impact on whole-body or skeletal muscle metabolism and profiling their lipidomic signature. Using a transgenic mouse model (Tg:Pax7-nGFP), we isolated muscle progenitor cells (satellite cells) and characterised their metabolic functions. We recruited childhood cancer survivors, grouped them based on the use of total body irradiation during their treatment and established their lipidomic profile. RESULTS In mice, irradiation delayed body weight gain and impaired fat pads and muscle weights. These changes were associated with impaired whole-body fat oxidation in chow-fed mice and altered ex vivo skeletal muscle fatty acid oxidation, potentially due to a reduction in oxidative fibres and reduced mitochondrial enzyme activity. Irradiation led to fasting hyperglycaemia and impaired glucose uptake in isolated skeletal muscles. Cultured satellite cells from irradiated mice showed decreased fatty acid oxidation and reduced glucose uptake, recapitulating the host metabolic phenotype. Irradiation resulted in a remodelling of lipid species in skeletal muscles, with the extensor digitorum longus muscle being particularly affected. A large number of lipid species were reduced, with several of these species showing a positive correlation with mitochondrial enzymes activity. In cancer survivors exposed to irradiation, we found a similar decrease in systemic levels of most lipid species, and lipid species that increased were positively correlated with insulin resistance (HOMA-IR). CONCLUSION Irradiation leads to long-term alterations in body composition, and lipid and carbohydrate metabolism in skeletal muscle, and affects muscle progenitor cells. Such changes result in persistent impairment of metabolic functions, providing a new mechanism for the increased prevalence of metabolic diseases reported in irradiated individuals. In this context, changes in the lipidomic signature in response to irradiation could be of diagnostic value.
Collapse
Affiliation(s)
- Nadia M L Amorim
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Anthony Kee
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Adelle C F Coster
- School of Mathematics and Statistics, UNSW Sydney, Sydney, Australia
| | - Christine Lucas
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Sarah Bould
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Sara Daniel
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Jacquelyn M Weir
- Metabolomics Laboratory, Baker IDI, Heart and Diabetes Institute, Melbourne, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker IDI, Heart and Diabetes Institute, Melbourne, Australia
| | - Jayne Barbour
- Mitochondrial Bioenergetics Lab, Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker IDI, Heart and Diabetes Institute, Melbourne, Australia
| | - Richard J Cohn
- School of Women's and Children's Health, UNSW Sydney, Randwick, Australia; Kids Cancer Centre, Sydney Children's Hospital Network, Randwick, Australia
| | - Nigel Turner
- Mitochondrial Bioenergetics Lab, Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Edna C Hardeman
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia.
| | - David Simar
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney, Australia.
| |
Collapse
|
38
|
Gramatyka M, Sokół M. Radiation metabolomics in the quest of cardiotoxicity biomarkers: the review. Int J Radiat Biol 2020; 96:349-359. [PMID: 31976800 DOI: 10.1080/09553002.2020.1704299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Purpose: Ionizing radiation is a risk factor to the whole organism, including the heart. Cardiac damage is considered to be a late effect of radiation exposure. While the acute cardiotoxicity of high doses is well characterized, the knowledge about nature and magnitude of the cardiac risk following lower doses exposure is incomplete. It has been shown that the cardiotoxic effects of radiation are source-, dose- and time-dependent. This paper provides an overview on these dependencies with regard to the molecular responses at the cellular and tissue levels. Main focus is put on the Nuclear Magnetic Resonance (NMR)-based and Mass Spectrometry (MS)-based metabolomic approaches in search of toxicity markers of relatively small doses of radiation.Conclusions: Available literature indicates that radiation exposure affects metabolites associated with: energy production, degradation of proteins and cell membranes, expression of proteins and stress response. Such effects are common for both animal and human studies. However, the specific metabolic response depends on several factors, including the examined organ. Radiation metabolomics can be used to explain the mechanisms of development of radiation-induced heart disease and to find an organ-specific biomarker of radiation exposure. The main aim of this review was to collect the information on the human cardiotoxicity biomarkers. In addition it also summarizes results of the studies on the metabolic responses to ionizing radiation for other organs, as well as the comparative data concerning animal studies.
Collapse
Affiliation(s)
- Michalina Gramatyka
- Department of Medical Physics, Maria Sklodowska-Curie Memorial Center and Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Maria Sokół
- Department of Medical Physics, Maria Sklodowska-Curie Memorial Center and Institute of Oncology Gliwice Branch, Gliwice, Poland
| |
Collapse
|
39
|
Gramatyka M, Boguszewicz ᴌ, Ciszek M, Gabryś D, Kulik R, Sokół M. Metabolic changes in mice cardiac tissue after low-dose irradiation revealed by 1H NMR spectroscopy. JOURNAL OF RADIATION RESEARCH 2020; 61:14-26. [PMID: 31840756 PMCID: PMC6976729 DOI: 10.1093/jrr/rrz079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/22/2019] [Accepted: 03/01/2019] [Indexed: 05/08/2023]
Abstract
Ionizing radiation may cause cardiotoxicity not only at high, but even at low (considered as harmless) doses, yet the molecular mechanisms of the heart's response to low doses are not clear. In this work, we used high-resolution nuclear magnetic resonance (NMR) spectroscopy to detect the early and late effects of radiation on the metabolism of murine hearts. The hearts of C57Bl/6NCrl female mice were irradiated in vivo with single 0.2 Gy or 2 Gy doses using 6 MV photons, then tissues were collected 48 h and 20 weeks after exposure. The most distinct changes in the profile of polar metabolites were detected 48 h after irradiation with 2 Gy, and included increased levels of pantothenate and glutamate as well as decreased levels of alanine, malonate, acetylcarnitine, glycine and adenosine. Significant effects of the 2 Gy dose were also observed 20 weeks after irradiation and included decreased levels of glutamine and acetylcarnitine when compared with age-matched controls. Moreover, several differences were observed between hearts irradiated with 2 Gy and analyzed either 48 h or 20 weeks after the exposure, which included changes in levels of acetylcarnitine, alanine, glycine, glutamate, glutamine, formate, myo-inositol and trimethylamine. No statistically significant effects induced by the 0.2 Gy dose were observed 20 weeks after irradiation. In general, radiation-affected compounds were associated with energy metabolism, fatty acid beta-oxidation, oxidative stress and damage to cell structures. At the same time, radiation-related effects were not detected at the level of tissue histology, which indicated a higher sensitivity of metabolomics-based tests for cardiac tissue response to radiation.
Collapse
Affiliation(s)
- Michalina Gramatyka
- Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101 Gliwice, Poland
| | - ᴌukasz Boguszewicz
- Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101 Gliwice, Poland
| | - Mateusz Ciszek
- Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101 Gliwice, Poland
| | - Dorota Gabryś
- Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101 Gliwice, Poland
| | - Roland Kulik
- Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101 Gliwice, Poland
| | - Maria Sokół
- Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101 Gliwice, Poland
| |
Collapse
|
40
|
Zeng ZM, Du HY, Xiong L, Zeng XL, Zhang P, Cai J, Huang L, Liu AW. BRCA1 protects cardiac microvascular endothelial cells against irradiation by regulating p21-mediated cell cycle arrest. Life Sci 2020; 244:117342. [PMID: 31978450 DOI: 10.1016/j.lfs.2020.117342] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/12/2020] [Accepted: 01/20/2020] [Indexed: 12/14/2022]
Abstract
AIMS Microvascular endothelial cell dysfunction is a leading cause of radiation-induced heart disease (RIHD). BRCA1 plays an important role in DNA damage repair. The study aims to explore the effect of BRCA1 in endothelial cells involved in RIHD. MATERIALS AND METHODS BRCA1 and p21 expression were detected in human umbilical vein endothelial cells (HUVECs) and in mouse heart tissue after irradiation exposure. The effects of BRCA1 on cell proliferation, cell cycle and radiosensitivity were determined in HUVECs with overexpression and knockdown of BRCA1. A mouse model of RIHD was established. Heart damage was detected in C57BL/6J mice and endothelial cell specific knockout BRCA1 mice (EC-BRCA1-/-). KEY FINDINGS BRCA1 and p21 expression was significantly increased both in vitro and vivo response to irradiation. BRCA1 overexpression in endothelial cells enhanced cell growth and G1/S phase arrest, and the opposite results were observed in BRCA1 knockdown endothelial cells. BRCA1 downregulated endothelial cell cycle-related genes cyclin A, cyclin D1, cyclin E and p-Rb through increasing p21 expression, and HUVECs with BRCA1 gene knockdown were more sensitive to radiation. In vivo, a decrease in cardiac microvascular density, as well as cardiomyocyte hypoxia and apoptosis were observed in a time-dependent manner. EC-BRCA1-/- mice were more prone to severe RIHD than EC-BRCA1+/- mice after 16Gy radiation exposure due to endothelial dysfunction caused by loss of BRCA1, and p21 was declined in EC-BRCA1-/- mice heart. SIGNIFICANCE These findings indicate that BRCA1 plays a protective role in RIHD by regulating endothelial cell cycle arrest mediated by p21 signal.
Collapse
Affiliation(s)
- Zhi-Min Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
| | - Hai-Yang Du
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
| | - Le Xiong
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
| | - Xiao-Li Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Peng Zhang
- Department of Radiation Oncology, Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, PR China
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Long Huang
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China.
| | - An-Wen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China.
| |
Collapse
|
41
|
Zou B, Schuster JP, Niu K, Huang Q, Rühle A, Huber PE. Radiotherapy-induced heart disease: a review of the literature. PRECISION CLINICAL MEDICINE 2019; 2:270-282. [PMID: 35693876 PMCID: PMC8985808 DOI: 10.1093/pcmedi/pbz025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 11/20/2022] Open
Abstract
Radiotherapy as one of the four pillars of cancer therapy plays a critical role in the multimodal treatment of thoracic cancers. Due to significant improvements in overall cancer survival, radiotherapy-induced heart disease (RIHD) has become an increasingly recognized adverse reaction which contributes to major radiation-associated toxicities including non-malignant death. This is especially relevant for patients suffering from diseases with excellent prognosis such as breast cancer or Hodgkin's lymphoma, since RIHD may occur decades after radiotherapy. Preclinical studies have enriched our knowledge of many potential mechanisms by which thoracic radiotherapy induces heart injury. Epidemiological findings in humans reveal that irradiation might increase the risk of cardiac disease at even lower doses than previously assumed. Recent preclinical studies have identified non-invasive methods for evaluation of RIHD. Furthermore, potential options preventing or at least attenuating RIHD have been developed. Ongoing research may enrich our limited knowledge about biological mechanisms of RIHD, identify non-invasive early detection biomarkers and investigate potential treatment options that might attenuate or prevent these unwanted side effects. Here, we present a comprehensive review about the published literature regarding clinical manifestation and pathological alterations in RIHD. Biological mechanisms and treatment options are outlined, and challenges in RIHD treatment are summarized.
Collapse
Affiliation(s)
- Bingwen Zou
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, Heidelberg 69120, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Julius Philipp Schuster
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, Heidelberg 69120, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Kerun Niu
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Qianyi Huang
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, Heidelberg 69120, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
- Heidelberg Institute for Radiation Oncology (HIRO) and National Center for Radiation Oncology (NCRO), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Peter Ernst Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, Heidelberg 69120, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
- Heidelberg Institute for Radiation Oncology (HIRO) and National Center for Radiation Oncology (NCRO), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| |
Collapse
|
42
|
The Use of DMS-MS for the Quantitative Analysis of Acylcarnitines. Methods Mol Biol 2019. [PMID: 31729655 DOI: 10.1007/978-1-0716-0030-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Differential mobility spectrometry (DMS) is capable of separating molecules based on their size and shape. When coupled to mass spectrometry (MS), DMS reduces chemical background and enhances signal-to-noise (S/N) ratio. Flow injection analysis (FIA) is a technique used to introduce samples into the source of the DMS-MS platform. Here we describe the application of FIA-DMS-MS/MS for the analysis of urinary acylcarnitine species. More than 20 acylcarnitine species can be detected and quantified during a single FIA-DMS-MS/MS acquisition.
Collapse
|
43
|
Barjaktarovic Z, Merl-Pham J, Braga-Tanaka I, Tanaka S, Hauck SM, Saran A, Mancuso M, Atkinson MJ, Tapio S, Azimzadeh O. Hyperacetylation of Cardiac Mitochondrial Proteins Is Associated with Metabolic Impairment and Sirtuin Downregulation after Chronic Total Body Irradiation of ApoE -/- Mice. Int J Mol Sci 2019; 20:ijms20205239. [PMID: 31652604 PMCID: PMC6829468 DOI: 10.3390/ijms20205239] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/17/2019] [Accepted: 10/19/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic exposure to low-dose ionizing radiation is associated with an increased risk of cardiovascular disease. Alteration in energy metabolism has been suggested to contribute to radiation-induced heart pathology, mitochondrial dysfunction being a hallmark of this disease. The goal of this study was to investigate the regulatory role of acetylation in heart mitochondria in the long-term response to chronic radiation. ApoE-deficient C57Bl/6J mice were exposed to low-dose-rate (20 mGy/day) gamma radiation for 300 days, resulting in a cumulative total body dose of 6.0 Gy. Heart mitochondria were isolated and analyzed using quantitative proteomics. Radiation-induced proteome and acetylome alterations were further validated using immunoblotting, enzyme activity assays, and ELISA. In total, 71 proteins showed peptides with a changed acetylation status following irradiation. The great majority (94%) of the hyperacetylated proteins were involved in the TCA cycle, fatty acid oxidation, oxidative stress response and sirtuin pathway. The elevated acetylation patterns coincided with reduced activity of mitochondrial sirtuins, increased the level of Acetyl-CoA, and were accompanied by inactivation of major cardiac metabolic regulators PGC-1 alpha and PPAR alpha. These observations suggest that the changes in mitochondrial acetylation after irradiation is associated with impairment of heart metabolism. We propose a novel mechanism involved in the development of late cardiac damage following chronic irradiation.
Collapse
Affiliation(s)
- Zarko Barjaktarovic
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
- Agency for Medicines and Medical Devices of Montenegro, 81000 Podgorica, Montenegro.
| | - Juliane Merl-Pham
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 80939 München, Germany.
| | | | - Satoshi Tanaka
- Institute for Environmental Sciences (IES), Rokkasho, Aomori 039-3213, Japan.
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 80939 München, Germany.
| | - Anna Saran
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 76 00196 Rome, Italy.
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 76 00196 Rome, Italy.
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
- Chair of Radiation Biology, Technical University Munich, 80333 Munich, Germany.
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| | - Omid Azimzadeh
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
44
|
Lacombe J, Brengues M, Mangé A, Bourgier C, Gourgou S, Pèlegrin A, Ozsahin M, Solassol J, Azria D. Quantitative proteomic analysis reveals AK2 as potential biomarker for late normal tissue radiotoxicity. Radiat Oncol 2019; 14:142. [PMID: 31399108 PMCID: PMC6688300 DOI: 10.1186/s13014-019-1351-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 08/01/2019] [Indexed: 12/27/2022] Open
Abstract
Background Biomarkers for predicting late normal tissue toxicity to radiotherapy are necessary to personalize treatments and to optimize clinical benefit. Many radiogenomic studies have been published on this topic. Conversely, proteomics approaches are not much developed, despite their advantages. Methods We used the isobaric tags for relative and absolute quantitation (iTRAQ) proteomic approach to analyze differences in protein expression levels in ex-vivo irradiated (8 Gy) T lymphocytes from patients with grade ≥ 2 radiation-induced breast fibrosis (grade ≥ 2 bf+) and patients with grade < 2 bf + after curative intent radiotherapy. Patients were selected from two prospective clinical trials (COHORT and PHRC 2005) and were used as discovery and confirmation cohorts. Results Among the 1979 quantified proteins, 23 fulfilled our stringent biological criteria. Immunoblotting analysis of four of these candidate proteins (adenylate kinase 2, AK2; annexin A1; heat shock cognate 71 kDa protein; and isocitrate dehydrogenase 2) confirmed AK2 overexpression in 8 Gy-irradiated T lymphocytes from patients with grade ≥ 2 bf + compared with patients with grade < 2 bf+. As these candidate proteins are involved in oxidative stress regulation, we also evaluated radiation-induced reactive oxygen species (ROS) production in peripheral blood mononuclear cells from patients with grade ≥ 2 bf + and grade < 2 bf+. Total ROS level, and especially superoxide anion level, increased upon ex-vivo 8 Gy-irradiation in all patients. Analysis of NADPH oxidases (NOXs), a major source of superoxide ion in the cell, showed a significant increase of NOX4 mRNA and protein levels after irradiation in both patient groups. Conversely, only NOX4 mRNA level was significantly different between groups (grade ≥ 2 bf + and grade < 2 bf+). Conclusion These findings identify AK2 as a potential radiosensitivity candidate biomarker. Overall, our proteomic approach highlights the important role of oxidative stress in late radiation-induced toxicity, and paves the way for additional studies on NOXs and superoxide ion metabolism. Electronic supplementary material The online version of this article (10.1186/s13014-019-1351-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jérôme Lacombe
- IRCM, INSERM, University Montpellier, ICM, Montpellier, France
| | - Muriel Brengues
- IRCM, INSERM, University Montpellier, ICM, Montpellier, France
| | - Alain Mangé
- IRCM, INSERM, University Montpellier, ICM, Montpellier, France
| | - Céline Bourgier
- IRCM, INSERM, University Montpellier, ICM, Montpellier, France
| | | | - André Pèlegrin
- IRCM, INSERM, University Montpellier, ICM, Montpellier, France
| | | | - Jérôme Solassol
- IRCM, INSERM, University Montpellier, ICM, Montpellier, France.,Department of Pathology and Onco-Biology, CHU Montpellier, Montpellier, France
| | - David Azria
- IRCM, INSERM, University Montpellier, ICM, Montpellier, France. .,Department of Radiation Oncology, ICM, 34298, Montpellier Cedex 5, France.
| |
Collapse
|
45
|
Kiang JG, Smith JT, Anderson MN, Umali MV, Ho C, Zhai M, Lin B, Jiang S. A novel therapy, using Ghrelin with pegylated G-CSF, inhibits brain hemorrhage from ionizing radiation or combined radiation injury. PHARMACY & PHARMACOLOGY INTERNATIONAL JOURNAL 2019; 7:133-145. [PMID: 34368440 PMCID: PMC8341084 DOI: 10.15406/ppij.2019.07.00243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Medical treatment becomes challenging when complicated injuries arise from secondary reactive metabolic and inflammatory products induced by initial acute ionizing radiation injury (RI) or when combined with subsequent trauma insult(s) (CI). With such detrimental effects on many organs, CI exacerbates the severity of primary injuries and decreases survival. Previously, in a novel study, we reported that ghrelin therapy significantly improved survival after CI. This study aimed to investigate whether brain hemorrhage induced by RI and CI could be inhibited by ghrelin therapy with pegylated G-CSF (i.e., Neulasta®, an FDA-approved drug). B6D2F1 female mice were exposed to 9.5 Gy 60Co-γ-radiation followed by 15% total-skin surface wound. Several endpoints were measured at several days. Brain hemorrhage and platelet depletion were observed in RI and CI mice. Brain hemorrhage severity was significantly higher in CI mice than in RI mice. Ghrelin therapy with pegylated G-CSF reduced the severity in brains of both RI and CI mice. RI and CI did not alter PARP and NF-κB but did significantly reduce PGC-1α and ghrelin receptors; the therapy, however, was able to partially recover ghrelin receptors. RI and CI significantly increased IL-6, KC, Eotaxin, G-CSF, MIP-2, MCP-1, MIP-1α, but significantly decreased IL-2, IL-9, IL-10, MIG, IFN-γ, and PDGF-bb; the therapy inhibited these changes. RI and CI significantly reduced platelet numbers, cellular ATP levels, NRF1/2, and AKT phosphorylation. The therapy significantly mitigated these CI-induced changes and reduced p53-mdm2 mediated caspase-3 activation. Our data are the first to support the view that Ghrelin therapy with pegylated G-CSF is potentially a novel therapy for treating brain hemorrhage after RI and CI.
Collapse
Affiliation(s)
- J G Kiang
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, USA
| | - J T Smith
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - M N Anderson
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - M V Umali
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - C Ho
- Department of Biochemistry, University of California, USA
| | - M Zhai
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - B Lin
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - S Jiang
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| |
Collapse
|
46
|
Bons J, Macron C, Aude-Garcia C, Vaca-Jacome SA, Rompais M, Cianférani S, Carapito C, Rabilloud T. A Combined N-terminomics and Shotgun Proteomics Approach to Investigate the Responses of Human Cells to Rapamycin and Zinc at the Mitochondrial Level. Mol Cell Proteomics 2019; 18:1085-1095. [PMID: 31154437 PMCID: PMC6553941 DOI: 10.1074/mcp.ra118.001269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/14/2019] [Indexed: 12/19/2022] Open
Abstract
All but thirteen mammalian mitochondrial proteins are encoded by the nuclear genome, translated in the cytosol and then imported into the mitochondria. For a significant proportion of the mitochondrial proteins, import is coupled with the cleavage of a presequence called the transit peptide, and the formation of a new N-terminus. Determination of the neo N-termini has been investigated by proteomic approaches in several systems, but generally in a static way to compile as many N-termini as possible. In the present study, we have investigated how the mitochondrial proteome and N-terminome react to chemical stimuli that alter mitochondrial metabolism, namely zinc ions and rapamycin. To this end, we have used a strategy that analyzes both internal and N-terminal peptides in a single run, the dN-TOP approach. We used these two very different stressors to sort out what could be a generic response to stress and what is specific to each of these stressors. Rapamycin and zinc induced different changes in the mitochondrial proteome. However, convergent changes to key mitochondrial enzymatic activities such as pyruvate dehydrogenase, succinate dehydrogenase and citrate synthase were observed for both treatments. Other convergent changes were seen in components of the N-terminal processing system and mitochondrial proteases. Investigations into the generation of neo-N-termini in mitochondria showed that the processing system is robust, as indicated by the lack of change in neo N-termini under the conditions tested. Detailed analysis of the data revealed that zinc caused a slight reduction in the efficiency of the N-terminal trimming system and that both treatments increased the degradation of mitochondrial proteins. In conclusion, the use of this combined strategy allowed a detailed analysis of the dynamics of the mitochondrial N-terminome in response to treatments which impact the mitochondria.
Collapse
Affiliation(s)
- Joanna Bons
- From the ‡Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Charlotte Macron
- From the ‡Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Catherine Aude-Garcia
- §Chemistry and Biology of Metals, Univ. Grenoble Alpes, CNRS UMR5249, CEA, BIG-LCBM, 38000 Grenoble, France
| | - Sebastian Alvaro Vaca-Jacome
- From the ‡Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Magali Rompais
- From the ‡Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Sarah Cianférani
- From the ‡Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Christine Carapito
- From the ‡Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France;
| | - Thierry Rabilloud
- §Chemistry and Biology of Metals, Univ. Grenoble Alpes, CNRS UMR5249, CEA, BIG-LCBM, 38000 Grenoble, France
| |
Collapse
|
47
|
Effect of low dose gamma rays on certain essential metals and oxidative stress in different rat organs. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2019. [DOI: 10.1016/j.jrras.2013.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
48
|
Abdel-Magied N, Abdel-Aziz N, Shedid SM, Ahmed AG. Modulating effect of tiron on the capability of mitochondrial oxidative phosphorylation in the brain of rats exposed to radiation or manganese toxicity. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:12550-12562. [PMID: 30848428 DOI: 10.1007/s11356-019-04594-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 02/18/2019] [Indexed: 06/09/2023]
Abstract
The brain is an important organ rich in mitochondria and more susceptible to oxidative stress. Tiron (sodium 4,5-dihydroxybenzene-1,3-disulfonate) is a potent antioxidant. This study aims to evaluate the effect of tiron on the impairment of brain mitochondria induced by exposure to radiation or manganese (Mn) toxicity. We assessed the capability of oxidative phosphorylation (OXPHOS) through determination of mitochondrial redox state, the activity of electron transport chain (ETC), and Krebs cycle as well as the level of adenosine triphosphate (ATP) production. Rats were exposed to 7 Gy of γ-rays or injected i.p. with manganese chloride (100 mg/kg), then treated with tiron (471 mg/kg) for 7 days. The results showed that tiron treatment revealed positive modulation on the mitochondrial redox state manifested by a marked decrease of hydrogen peroxide (H2O2), malondialdehyde (MDA), and total nitrate/nitrite (NOx) associated with a significant increase in total antioxidant capacity (TAC), glutathione (GSH) content, manganese superoxide dismutase (MnSOD), and glutathione peroxidase (GPx) activities. Moreover, tiron can increase the activity of ETC through preventing the depletion in the activity of mitochondrial complexes (I, II, III, and IV), an elevation of coenzyme Q10 (CoQ10) and cytochrome c (Cyt-c) levels. Additionally, tiron showed a noticeable increase in mitochondrial aconitase (mt-aconitase) activity as the major component of Krebs cycle to maintain a high level of ATP production. Tiron also can restore mitochondrial metal homeostasis through positive changes in the levels of calcium (Ca), iron (Fe), Mn, and copper (Cu). It can be concluded that tiron may be used as a good mitigating agent to attenuate the harmful effects on the brain through the inhibition of mitochondrial injury post-exposure to radiation or Mn toxicity.
Collapse
Affiliation(s)
- Nadia Abdel-Magied
- Department of Radiation Biology, Atomic Energy Authority, National Center for Radiation Research and Technology (NCRRT), 3st Ahmed Elzomer, P.O. Box 29, Nasr City, Cairo, Egypt.
| | - Nahed Abdel-Aziz
- Department of Radiation Biology, Atomic Energy Authority, National Center for Radiation Research and Technology (NCRRT), 3st Ahmed Elzomer, P.O. Box 29, Nasr City, Cairo, Egypt
| | - Shereen M Shedid
- Department of Radiation Biology, Atomic Energy Authority, National Center for Radiation Research and Technology (NCRRT), 3st Ahmed Elzomer, P.O. Box 29, Nasr City, Cairo, Egypt
| | - Amal G Ahmed
- Department of Radiation Biology, Atomic Energy Authority, National Center for Radiation Research and Technology (NCRRT), 3st Ahmed Elzomer, P.O. Box 29, Nasr City, Cairo, Egypt
| |
Collapse
|
49
|
Soloviev AI, Kizub IV. Mechanisms of vascular dysfunction evoked by ionizing radiation and possible targets for its pharmacological correction. Biochem Pharmacol 2018; 159:121-139. [PMID: 30508525 DOI: 10.1016/j.bcp.2018.11.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
Abstract
Ionizing radiation (IR) leads to a variety of the cardiovascular diseases, including the arterial hypertension. A number of studies have demonstrated that blood vessels represent important target for IR, and the endothelium is one of the most vulnerable components of the vascular wall. IR causes an inhibition of nitric oxide (NO)-mediated endothelium-dependent vasodilatation and generation of reactive oxygen (ROS) and nitrogen (RNS) species trigger this process. Inhibition of NO-mediated vasodilatation could be due to endothelial NO synthase (eNOS) down-regulation, inactivation of endothelium-derived NO, and abnormalities in diffusion of NO from the endothelial cells (ECs) leading to a decrease in NO bioavailability. Beside this, IR suppresses endothelial large conductance Ca2+-activated K+ channels (BKCa) activity, which control NO synthesis. IR also leads to inhibition of the BKCa current in vascular smooth muscle cells (SMCs) which is mediated by protein kinase C (PKC). On the other hand, IR-evoked enhanced vascular contractility may result from PKC-mediated increase in SMCs myofilament Ca2+ sensitivity. Also, IR evokes vascular wall inflammation and atherosclerosis development. Vascular function damaged by IR can be effectively restored by quercetin-filled phosphatidylcholine liposomes and mesenchymal stem cells injection. Using RNA-interference technique targeted to different PKC isoforms can also be a perspective approach for pharmacological treatment of IR-induced vascular dysfunction.
Collapse
Affiliation(s)
- Anatoly I Soloviev
- Department of Pharmacology of Cellular Signaling Systems and Experimental Therapy, Institute of Pharmacology and Toxicology, National Academy of Medical Sciences of Ukraine, 14 Eugene Pottier Street, Kiev 03068, Ukraine
| | - Igor V Kizub
- Department of Pharmacology, New York Medical College, 15 Dana Road, Valhalla 10595, NY, United States.
| |
Collapse
|
50
|
Cheema AK, Byrum SD, Sharma NK, Altadill T, Kumar VP, Biswas S, Balgley BM, Hauer-Jensen M, Tackett AJ, Ghosh SP. Proteomic Changes in Mouse Spleen after Radiation-Induced Injury and its Modulation by Gamma-Tocotrienol. Radiat Res 2018; 190:449-463. [PMID: 30070965 PMCID: PMC6297072 DOI: 10.1667/rr15008.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gamma-tocotrienol (GT3), a naturally occurring vitamin E isomer, a promising radioprotector, has been shown to protect mice against radiation-induced hematopoietic and gastrointestinal injuries. We analyzed changes in protein expression profiles of spleen tissue after GT3 treatment in mice exposed to gamma radiation to gain insights into the molecular mechanism of radioprotective efficacy. Male CD2F1 mice, 12-to-14 weeks old, were treated with either vehicle or GT3 at 24 h prior to 7 Gy total-body irradiation. Nonirradiated vehicle, nonirradiated GT3 and age-matched naïve animals were used as controls. Blood and tissues were harvested on days 0, 1, 2, 4, 7, 10 and 14 postirradiation. High-resolution mass-spectrometry-based radioproteomics was used to identify differentially expressed proteins in spleen tissue with or without drug treatment. Subsequent bioinformatic analyses helped delineate molecular markers of biological pathways and networks regulating the cellular radiation responses in spleen. Our results show a robust alteration in spleen proteomic profiles including upregulation of the Wnt signaling pathway and actin-cytoskeleton linked proteins in mediating the radiation injury response in spleen. Furthermore, we show that 24 h pretreatment with GT3 attenuates radiation-induced hematopoietic injury in the spleen by modulating various cell signaling proteins. Taken together, our results show that the radioprotective effects of GT3 are mediated, via alleviation of radiation-induced alterations in biochemical pathways, with wide implications on overall hematopoietic injury.
Collapse
Affiliation(s)
- Amrita K. Cheema
- Departments of Oncology, Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Stephanie D. Byrum
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - Neel Kamal Sharma
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| | - Tatiana Altadill
- Departments of Oncology, Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC
- Institut d’Investigacio Biomedica de Bellvitge (IDIBELL), Gynecological Department, Vall Hebron University Hospital, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Vidya P. Kumar
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| | - Shukla Biswas
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| | | | - Martin Hauer-Jensen
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - Alan J. Tackett
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - Sanchita P. Ghosh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| |
Collapse
|