1
|
Saigusa Y, Little MP, Azimzadeh O, Hamada N. Biological effects of high-LET irradiation on the circulatory system. Int J Radiat Biol 2025; 101:429-452. [PMID: 40063776 PMCID: PMC12011529 DOI: 10.1080/09553002.2025.2470947] [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] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
PURPOSE High-linear energy transfer (LET) radiation is generally thought to be more biologically effective in various tissues than low-LET radiation, but whether this also applies to the circulatory system remains unclear. We therefore reviewed biological studies about the effects of high-LET radiation on the circulatory system. CONCLUSIONS We identified 76 relevant papers (24 in vitro, 2 ex vivo, 51 in vivo, one overlapping). In vitro studies used human, bovine, porcine or chick vascular endothelial cells or cardiomyocytes, while ex vivo studies used porcine hearts. In vivo studies used mice, rats, rabbits, dogs or pigs. The types of high-LET radiation used were neutrons, α particles, heavy ions and negative pions. Most studies used a single dose, although some investigated fractionation effects. Twenty-one studies estimated the relative biological effectiveness (RBE) that ranged from 0.1 to 130, depending on radiation quality and endpoint. A meta-analysis of 6 in vitro and 8 in vivo studies (selected based on the feasibility of estimating the RBE and its uncertainty) suggested an RBE of 6.69 (95% confidence intervals (CI): 2.51, 10.88) for in vitro studies and 1.14 (95% CI: 0.91, 1.37) for in vivo studies. The meta-analysis of these 14 studies yielded an RBE of 2.88 (95% CI: 1.52, 4.25). This suggests that high-LET radiation is only slightly more effective than low-LET radiation, although substantial inter-study heterogeneity complicates interpretation. Therapeutic effects have also been reported in disease models. Further research is needed to better understand the effects on the cardiovascular system and to improve radiation protection.
Collapse
Affiliation(s)
- Yumi Saigusa
- Dosimetry Facility Management Section, Department of Nuclear Emergency Preparedness, Institute for Radiological Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-0024, Japan
| | - Mark P. Little
- Radiation Epidemiology Branch, National Cancer Institute, MD 20892-9778, USA
- Faculty of Health, Science and Technology, Oxford Brookes University, Headington Campus, OX3 0BP, UK
| | - Omid Azimzadeh
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba 270-1194, Japan
| |
Collapse
|
2
|
Nemec-Bakk AS, Sridharan V, Willey JS, Koturbash I, Williams DK, Chesal M, Patel CM, Borg AM, Reno K, Gifford G, Newhauser W, Williams J, Chancellor JC, Boerma M. Sex-specific effects on the heart from combined exposure to simulated galactic cosmic radiation and hindlimb unloading. LIFE SCIENCES IN SPACE RESEARCH 2025; 44:38-46. [PMID: 39864910 PMCID: PMC11770252 DOI: 10.1016/j.lssr.2024.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 01/28/2025]
Abstract
Future long duration space missions will expose astronauts to higher doses of galactic cosmic radiation (GCR) than those experienced on the international space station. Recent studies have demonstrated astronauts may be at risk for cardiovascular complications due to increased radiation exposure and fluid shift from microgravity. However, there is a lack of direct evidence on how the cardiovascular system is affected by GCR and microgravity since no astronauts have been exposed to exploratory mission relevant GCR doses. Therefore, we utilized a ground-based mouse model to determine the cardiovascular risks for space radiation exposure while the mice were simultaneously hindlimb suspended to mimic microgravity. 6-month-old male and female C57BL/6 mice were exposed to an absorbed dose of 0 Gy, 0.5 Gy, or 1.5 Gy simulated GCR (GCRsim) that comprised beams of 5 ions at NASA's Space Radiation Laboratory. Subcohorts of mice were hindlimb unloaded (HLU), starting 5 days before GCRsim until the completion of radiation exposure. GCRsim + HLU was performed over 8 hours (0.5 Gy) or 24 hours (1.5 Gy). After completion of GCRsim and HLU, mice were shipped to UAMS for long-term observation. Cardiac function was measured using high resolution ultrasound at 6 and 9 months after exposure. Tissues were collected after the final ultrasound and prepared for further analysis. Female mice exposed to 1.5 Gy + HLU demonstrated a significant increase in body weight compared to ground controls months after GCR exposure; however, there was no change in male body weights. Cardiac ultrasound revealed 0.5 Gy GCRsim decreased left ventricular (LV) mass, LV posterior wall thickness in diastole, and systole in males 6 months after exposure. In females, 1.5 Gy + HLU significantly increased LV posterior wall thickness in diastole and systole at 6 months. These changes in ultrasound measurements were no longer seen at 9 months. Moreover, at 9 months there was no change in total collagen content or density of the capillary network in the heart. Lastly, the combination of GCRsim and HLU influenced immune cell markers in the heart of female mice. These data suggest that combined simulated GCR and microgravity result in minor, yet statistically significant sex-dependent changes to body weight and cardiac structure.
Collapse
Affiliation(s)
- A S Nemec-Bakk
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - V Sridharan
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - J S Willey
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - I Koturbash
- Department of Environmental Health Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - D K Williams
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - M Chesal
- Department of Physics & Astronomy, Louisiana State University, Baton Rouge, Louisiana, USA
| | - C M Patel
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - A M Borg
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - K Reno
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - G Gifford
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - W Newhauser
- Department of Physics & Astronomy, Louisiana State University, Baton Rouge, Louisiana, USA
| | - J Williams
- Departments of Environmental Medicine and Radiation Oncology, University of Rochester, Medical Center, Rochester, New York, USA
| | - J C Chancellor
- Department of Physics & Astronomy, Louisiana State University, Baton Rouge, Louisiana, USA; Department of Preventive Medicine & Population Health, University of Texas Medical Branch, Galveston, Texas, USA; Outer Space Institute, University of British Columbia, Vancouver, Canada
| | - M Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
3
|
Hussain I, Ullah R, Simran BFNU, Kaur P, Kumar M, Raj R, Faraz M, Mehmoodi A, Malik J. Cardiovascular effects of long-duration space flight. Health Sci Rep 2024; 7:e2305. [PMID: 39135704 PMCID: PMC11318032 DOI: 10.1002/hsr2.2305] [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: 01/13/2024] [Revised: 06/10/2024] [Accepted: 07/24/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction Early studies exploring the physiological effects of space travel have indicated the body's capacity for reversible adaptation. However, the impact of long-duration spaceflight, exceeding 6 months, presents more intricate challenges. Effects on the Cardiovascular CV System Extended exposure to microgravity and radiation profoundly affects the CV system. Notable phenomena include fluid shifts toward the head and modified arterial pressure. These changes disrupt blood pressure regulation and elevate cardiac output. Additionally, the loss of venous compression leads to a reduction in central venous pressure. Fluid and Plasma Volume Changes The displacement of fluid from the vascular system to the interstitium, driven by baroreceptor stimulation, results in a 10%-15% decline in plasma volume. Cardiac Muscle and Hematocrit Variations Intriguingly, despite potential increases in cardiac workload, cardiac muscle atrophy and perplexing variations in hematocrit levels have been observed. The mechanism underlying atrophy appears to involve a shift in protein synthesis from the endoplasmic reticulum to the mitochondria via mortalin-mediated mechanisms. Arrhythmias and QT Interval Prolongation Instances of arrhythmias have been recurrently documented, although generally nonlethal, in both Russian and American space missions. Long-duration spaceflight has been associated with the prolongation of the QT interval, particularly in extended missions. Radiation Effects Exposure of the heart to the proton and heavy ion radiation pervasive in deep space contributes to coronary artery degeneration, augmented aortic stiffness, and carotid intima thickening through collagen-mediated processes. Moreover, it accelerates the onset of atherosclerosis and triggers proinflammatory responses. Reentry and Postflight Challenges Upon reentry, astronauts frequently experience orthostatic intolerance and altered sympathetic responses, which bear potential hazards in scenarios requiring rapid mobilization or evacuation. Conclusion Consequently, careful monitoring of these cardiac risks is imperative for forthcoming missions. While early studies illuminate the adaptability of the body to space travel's challenges, the intricacies of long-duration missions and their effects on the CV system necessitate continued investigation and vigilance to ensure astronaut health and mission success.
Collapse
Affiliation(s)
- Iqbal Hussain
- Department of Cardiovascular MedicineCardiovascular Analytics GroupIslamabadPakistan
| | - Rehmat Ullah
- Department of Cardiovascular MedicineCardiovascular Analytics GroupIslamabadPakistan
| | | | - Parvinder Kaur
- Department of Cardiovascular MedicineCardiovascular Analytics GroupIslamabadPakistan
| | - Mahendra Kumar
- Department of Cardiovascular MedicineCardiovascular Analytics GroupIslamabadPakistan
| | - Rohan Raj
- Department of Cardiovascular MedicineCardiovascular Analytics GroupIslamabadPakistan
| | - Maria Faraz
- Department of Cardiovascular MedicineCardiovascular Analytics GroupIslamabadPakistan
| | - Amin Mehmoodi
- Department of MedicineIbn e Seena HospitalKabulAfghanistan
| | - Jahanzeb Malik
- Department of Cardiovascular MedicineCardiovascular Analytics GroupIslamabadPakistan
| |
Collapse
|
4
|
Ogilvie LM, Coyle-Asbil B, Brunt KR, Petrik J, Simpson JA. Therapy-naïve malignancy causes cardiovascular disease: a state-of-the-art cardio-oncology perspective. Am J Physiol Heart Circ Physiol 2024; 326:H1515-H1537. [PMID: 38639740 DOI: 10.1152/ajpheart.00795.2023] [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: 12/22/2023] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Cardiovascular disease (CVD) and cancer are the leading causes of mortality worldwide. Although generally thought of as distinct diseases, the intersectional overlap between CVD and cancer is increasingly evident in both causal and mechanistic relationships. The field of cardio-oncology is largely focused on the cardiotoxic effects of cancer therapies (e.g., chemotherapy, radiation). Furthermore, the cumulative effects of cardiotoxic therapy exposure and the prevalence of CVD risk factors in patients with cancer lead to long-term morbidity and poor quality of life in this patient population, even when patients are cancer-free. Evidence from patients with cancer and animal models demonstrates that the presence of malignancy itself, independent of cardiotoxic therapy exposure or CVD risk factors, negatively impacts cardiac structure and function. As such, the primary focus of this review is the cardiac pathophysiological and molecular features of therapy-naïve cancer. We also summarize the strengths and limitations of preclinical cancer models for cardio-oncology research and discuss therapeutic strategies that have been tested experimentally for the treatment of cancer-induced cardiac atrophy and dysfunction. Finally, we explore an adjacent area of interest, called "reverse cardio-oncology," where the sequelae of heart failure augment cancer progression. Here, we emphasize the cross-disease communication between malignancy and the injured heart and discuss the importance of chronic low-grade inflammation and endocrine factors in the progression of both diseases.
Collapse
Affiliation(s)
- Leslie M Ogilvie
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Bridget Coyle-Asbil
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Keith R Brunt
- Department of Pharmacology, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
- IMPART Investigator Team Canada, Saint John, New Brunswick, Canada
| | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
- IMPART Investigator Team Canada, Saint John, New Brunswick, Canada
| |
Collapse
|
5
|
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
|
6
|
Pavitra E, Kancharla J, Gupta VK, Prasad K, Sung JY, Kim J, Tej MB, Choi R, Lee JH, Han YK, Raju GSR, Bhaskar L, Huh YS. The role of NF-κB in breast cancer initiation, growth, metastasis, and resistance to chemotherapy. Biomed Pharmacother 2023; 163:114822. [PMID: 37146418 DOI: 10.1016/j.biopha.2023.114822] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023] Open
Abstract
Breast cancer (BC) is the second most fatal disease and is the prime cause of cancer allied female deaths. BC is caused by aberrant tumor suppressor genes and oncogenes regulated by transcription factors (TFs) like NF-κB. NF-κB is a pro-inflammatory TF that crucially alters the expressions of various genes associated with inflammation, cell progression, metastasis, and apoptosis and modulates a network of genes that underlie tumorigenesis. Herein, we focus on NF-κB signaling pathways, its regulators, and the rationale for targeting NF-κB. This review also includes TFs that maintain NF-κB crosstalk and their roles in promoting angiogenesis and metastasis. In addition, we discuss the importance of combination therapies, resistance to treatment, and potential novel therapeutic strategies including nanomedicine that targets NF-κB.
Collapse
Affiliation(s)
- Eluri Pavitra
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea; 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea
| | - Jyothsna Kancharla
- Department of Bioscience and Biotechnology, Banasthali University, Vanasthali, Rajasthan 304022, India
| | - Vivek Kumar Gupta
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Kiran Prasad
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur- 495009, Chhattisgarh, India
| | - Ju Yong Sung
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Jigyeong Kim
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Mandava Bhuvan Tej
- Department of Health care informatics, Sacred Heart University, 5151Park Avenue, Fair fields, CT06825, USA
| | - Rino Choi
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea; Department of Materials Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Jeong-Hwan Lee
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea; Department of Materials Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Young-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea.
| | - Lvks Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur- 495009, Chhattisgarh, India.
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
7
|
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
|
8
|
Xie D, Huang Q, Zhou P. Drug Discovery Targeting Post-Translational Modifications in Response to DNA Damages Induced by Space Radiation. Int J Mol Sci 2023; 24:ijms24087656. [PMID: 37108815 PMCID: PMC10142602 DOI: 10.3390/ijms24087656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
DNA damage in astronauts induced by cosmic radiation poses a major barrier to human space exploration. Cellular responses and repair of the most lethal DNA double-strand breaks (DSBs) are crucial for genomic integrity and cell survival. Post-translational modifications (PTMs), including phosphorylation, ubiquitylation, and SUMOylation, are among the regulatory factors modulating a delicate balance and choice between predominant DSB repair pathways, such as non-homologous end joining (NHEJ) and homologous recombination (HR). In this review, we focused on the engagement of proteins in the DNA damage response (DDR) modulated by phosphorylation and ubiquitylation, including ATM, DNA-PKcs, CtIP, MDM2, and ubiquitin ligases. The involvement and function of acetylation, methylation, PARylation, and their essential proteins were also investigated, providing a repository of candidate targets for DDR regulators. However, there is a lack of radioprotectors in spite of their consideration in the discovery of radiosensitizers. We proposed new perspectives for the research and development of future agents against space radiation by the systematic integration and utilization of evolutionary strategies, including multi-omics analyses, rational computing methods, drug repositioning, and combinations of drugs and targets, which may facilitate the use of radioprotectors in practical applications in human space exploration to combat fatal radiation hazards.
Collapse
Affiliation(s)
- Dafei Xie
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing 100850, China
| | - Qi Huang
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing 100850, China
- Department of Preventive Medicine, School of Public Health, University of South China, Changsheng West Road 28th, Zhengxiang District, Hengyang 421001, China
| | - Pingkun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing 100850, China
- Department of Preventive Medicine, School of Public Health, University of South China, Changsheng West Road 28th, Zhengxiang District, Hengyang 421001, China
| |
Collapse
|
9
|
Nemec-Bakk AS, Sridharan V, Desai P, Landes RD, Hart B, Allen AR, Boerma M. Effects of Simulated 5-Ion Galactic Cosmic Radiation on Function and Structure of the Mouse Heart. Life (Basel) 2023; 13:life13030795. [PMID: 36983950 PMCID: PMC10057791 DOI: 10.3390/life13030795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
Missions into deep space will expose astronauts to the harsh space environment, and the degenerative tissue effects of space radiation are largely unknown. To assess the risks, in this study, male BALB/c mice were exposed to 500 mGy 5-ion simulated GCR (GCRsim) at the NASA Space Radiation Laboratory. In addition, male and female CD1 mice were exposed to GCRsim and administered a diet containing Transforming Growth Factor-beta (TGF-β)RI kinase (ALK5) inhibitor IPW-5371 as a potential countermeasure. An ultrasound was performed to investigate cardiac function. Cardiac tissue was collected to determine collagen deposition, the density of the capillary network, and the expression of the immune mediator toll-like receptor 4 (TLR4) and immune cell markers CD2, CD4, and CD45. In male BALB/c mice, the only significant effects of GCRsim were an increase in the CD2 and TLR4 markers. In male CD1 mice, GCRsim caused a significant increase in total collagens and a decrease in the expression of TLR4, both of which were mitigated by the TGF-β inhibitor diet. In female CD1 mice, GCRsim caused an increase in the number of capillaries per tissue area in the ventricles, which may be explained by the decrease in the left ventricular mass. However, this increase was not mitigated by TGF-β inhibition. In both male and female CD1 mice, the combination of GCRsim and TGF-β inhibition caused changes in left ventricular immune cell markers that were not seen with GCRsim alone. These data suggest that GCRsim results in minor changes to cardiac tissue in both an inbred and outbred mouse strain. While there were few GCRsim effects to be mitigated, results from the combination of GCRsim and the TGF-β inhibitor do point to a role for TGF-β in maintaining markers of immune cells in the heart after exposure to GCR.
Collapse
Affiliation(s)
- Ashley S. Nemec-Bakk
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence:
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Parth Desai
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Reid D. Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Barry Hart
- Innovation Pathways, LLC of Palo Alto, Palo Alto, CA 94301, USA
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
10
|
Pramanik J, Kumar A, Panchal L, Prajapati B. Countermeasures for Maintaining Cardiovascular Health in Space Missions. Curr Cardiol Rev 2023; 19:57-67. [PMID: 37005513 PMCID: PMC10518885 DOI: 10.2174/1573403x19666230330083225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/21/2023] [Accepted: 02/06/2023] [Indexed: 04/04/2023] Open
Abstract
During space exploration, the human body is subjected to altered atmospheric environments and gravity, exposure to radiation, sleep disturbance, and mental pressures; all these factors are responsible for cardiovascular diseases. Under microgravity, the physiological changes related to cardiovascular diseases are the cephalic fluid shift, dramatic reduction in central venous pressure, changes in blood rheology and endothelial function, cerebrovascular abnormalities, headaches, optic disc edema, intracranial hypertension, congestion of the jugular vein, facial swelling, and loss of taste. Generally, five countermeasures are used to maintain cardiovascular health (during and after space missions), including shielding, nutritional, medicinal, exercise, and artificial gravity. This article concludes with how to reduce space missions' impact on cardiovascular health with the help of various countermeasures.
Collapse
Affiliation(s)
- Jhilam Pramanik
- Department of Food Technology, ITM University, Gwalior, Madhya Pradesh, India
| | - Akash Kumar
- Department of Food Technology, SRM University, Sonipat, Haryana, India
| | - Lakshay Panchal
- Maharishi Markandeshwar Institute of Physiotherapy and Rehabilitation, Maharishi Markandeshwar University, Mullana, Haryana, India
| | - Bhupendra Prajapati
- Shree S.K. Patel College of Pharmaceutical Education and Research, Ganpat University, India
| |
Collapse
|
11
|
Weiss M, Nikisher B, Haran H, Tefft K, Adams J, Edwards JG. High throughput screen of small molecules as potential countermeasures to galactic cosmic radiation induced cellular dysfunction. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:76-87. [PMID: 36336373 DOI: 10.1016/j.lssr.2022.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/23/2022] [Accepted: 06/16/2022] [Indexed: 06/16/2023]
Abstract
Space travel increases galactic cosmic ray exposure to flight crews and this is significantly elevated once travel moves beyond low Earth orbit. This includes combinations of high energy protons and heavy ions such as 56Fe or 16O. There are distinct differences in the biological response to low-energy transfer (x-rays) or high-energy transfer (High-LET). However, given the relatively low fluence rate of exposure during flight operations, it might be possible to manage these deleterious effects using small molecules currently available. Virtually all reports to date examining small molecule management of radiation exposure are based on low-LET challenges. To that end an FDA approved drug library (725 drugs) was used to perform a high throughput screen of cultured cells following exposure to galactic cosmic radiation. The H9c2 myoblasts, ES-D3 pluripotent cells, and Hy926 endothelial cell lines were exposed to a single exposure (75 cGy) using the 5-ion GCRsim protocol developed at the NASA Space Radiation Laboratory (NSRL). Following GCR exposure cells were maintained for up to two weeks. For each drug (@10µM), a hierarchical cumulative score was developed incorporating measures of mitochondrial and cellular function, oxidant stress and cell senescence. The top 160 scores were retested following a similar protocol using 1µM of each drug. Within the 160 drugs, 33 are considered to have an anti-inflammatory capacity, while others also indirectly suppressed pro-inflammatory pathways or had noted antioxidant capacity. Lead candidates came from different drug classes that included angiotensin converting enzyme inhibitors or AT1 antagonists, COX2 inhibitors, as well as drugs mediated by histamine receptors. Surprisingly, different classes of anti-diabetic medications were observed to be useful including sulfonylureas and metformin. Using a hierarchical decision structure, we have identified several lead candidates. That no one drug or even drug class was completely successful across all parameters tested suggests the complexity of managing the consequences of galactic cosmic radiation exposure.
Collapse
Affiliation(s)
- M Weiss
- Department of Physiology, New York Medical College, Valhalla, New York
| | - B Nikisher
- Department of Physiology, New York Medical College, Valhalla, New York
| | - H Haran
- Department of Physiology, New York Medical College, Valhalla, New York
| | - K Tefft
- Department of Physiology, New York Medical College, Valhalla, New York
| | - J Adams
- Department of Physiology, New York Medical College, Valhalla, New York
| | - J G Edwards
- Department of Physiology, New York Medical College, Valhalla, New York.
| |
Collapse
|
12
|
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
|
13
|
Nemec-Bakk AS, Sridharan V, Landes RD, Singh P, Cao M, Dominic P, Seawright JW, Chancellor JC, Boerma M. Effects of low-dose oxygen ions on cardiac function and structure in female C57BL/6J mice. LIFE SCIENCES IN SPACE RESEARCH 2022; 32:105-112. [PMID: 35065756 PMCID: PMC8803400 DOI: 10.1016/j.lssr.2021.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 05/07/2023]
Abstract
PURPOSE Astronauts in space vehicles beyond low-Earth orbit will be exposed to high charge and energy (HZE) ions, and there is concern about potential adverse effects on the cardiovascular system. Thus far, most animal studies that assess cardiac effects of HZE particles have included only males. This study assessed the effects of oxygen ions (16O) as a representative ion of the intravehicular radiation environment on the heart of female mice. MATERIALS AND METHODS Female C57BL/6 J mice at 6 months of age were exposed to 16O (600 MeV/n) at 0.25-0.26 Gy/min to a total dose of 0, 0.1, or 0.25 Gy. Cardiac function and abdominal aorta blood velocity were measured with ultrasonography at 3, 5, 7, and 9 months after irradiation. At 2 weeks, 3 months, and 9 months, cardiac tissue was collected to assess collagen deposition and markers of immune cells. RESULTS Ultrasonography revealed increased left ventricle mass, diastolic volume and diameter but there was no change in the abdominal aorta. There was no indication of cardiac fibrosis however, a 75 kDa peptide of left ventricular collagen type III and α-smooth muscle cell actin were increased suggesting some remodeling had occurred. Left ventricular protein levels of the T-cell marker CD2 was significantly increased at all time points, while the neutrophil marker myeloperoxidase was decreased at 2 weeks and 9 months. CONCLUSIONS These results taken together suggest 16O ion exposure did not result in cardiac fibrosis or cardiac dysfunction in female mice. However, it does appear mild cardiac remodeling occurs in response to HZE radiation.
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
| | - Reid D Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Preeti Singh
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Maohua Cao
- College of Dentistry, Texas A&M University, Dallas TX, USA
| | - Paari Dominic
- Department of Medicine and Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | | - Jeffery C Chancellor
- Department of Physics & Astronomy, Louisiana State University, Baton Rouge, LA, USA; Department of Preventative Medicine & Population Health, University of Texas Medical Branch, Galveston, TX, USA; Outer Space Institute, University of British Columbia, Vancouver, CA, Canada
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
14
|
Bisserier M, Saffran N, Brojakowska A, Sebastian A, Evans AC, Coleman MA, Walsh K, Mills PJ, Garikipati VNS, Arakelyan A, Hadri L, Goukassian DA. Emerging Role of Exosomal Long Non-coding RNAs in Spaceflight-Associated Risks in Astronauts. Front Genet 2022; 12:812188. [PMID: 35111205 PMCID: PMC8803151 DOI: 10.3389/fgene.2021.812188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
During spaceflight, astronauts are exposed to multiple unique environmental factors, particularly microgravity and ionizing radiation, that can cause a range of harmful health consequences. Over the past decades, increasing evidence demonstrates that the space environment can induce changes in gene expression and RNA processing. Long non-coding RNA (lncRNA) represent an emerging area of focus in molecular biology as they modulate chromatin structure and function, the transcription of neighboring genes, and affect RNA splicing, stability, and translation. They have been implicated in cancer development and associated with diverse cardiovascular conditions and associated risk factors. However, their role on astronauts' health after spaceflight remains poorly understood. In this perspective article, we provide new insights into the potential role of exosomal lncRNA after spaceflight. We analyzed the transcriptional profile of exosomes isolated from peripheral blood plasma of three astronauts who flew on various Shuttle missions between 1998-2001 by RNA-sequencing. Computational analysis of the transcriptome of these exosomes identified 27 differentially expressed lncRNAs with a Log2 fold change, with molecular, cellular, and clinical implications.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nathaniel Saffran
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Agnieszka Brojakowska
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Angela Clare Evans
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- Department of Radiation Oncology, University of California, Davis, Sacramento, CA, United States
| | - Matthew A. Coleman
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- Department of Radiation Oncology, University of California, Davis, Sacramento, CA, United States
| | - Kenneth Walsh
- School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Paul J. Mills
- Center of Excellence for Research and Training in Integrative Health, University of California, San Diego, San Diego, CA, United States
| | - Venkata Naga Srikanth Garikipati
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Arsen Arakelyan
- Bioinformatics Group, The Institute of Molecular Biology, The National Academy of Sciences of the Republic of Armenia, Yerevan, Armenia
| | - Lahouaria Hadri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - David A. Goukassian
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
15
|
Basirun C, Ferlazzo ML, Howell NR, Liu GJ, Middleton RJ, Martinac B, Narayanan SA, Poole K, Gentile C, Chou J. Microgravity × Radiation: A Space Mechanobiology Approach Toward Cardiovascular Function and Disease. Front Cell Dev Biol 2021; 9:750775. [PMID: 34778261 PMCID: PMC8586646 DOI: 10.3389/fcell.2021.750775] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, there has been an increasing interest in space exploration, supported by the accelerated technological advancements in the field. This has led to a new potential environment that humans could be exposed to in the very near future, and therefore an increasing request to evaluate the impact this may have on our body, including health risks associated with this endeavor. A critical component in regulating the human pathophysiology is represented by the cardiovascular system, which may be heavily affected in these extreme environments of microgravity and radiation. This mini review aims to identify the impact of microgravity and radiation on the cardiovascular system. Being able to understand the effect that comes with deep space explorations, including that of microgravity and space radiation, may also allow us to get a deeper understanding of the heart and ultimately our own basic physiological processes. This information may unlock new factors to consider with space exploration whilst simultaneously increasing our knowledge of the cardiovascular system and potentially associated diseases.
Collapse
Affiliation(s)
- Carin Basirun
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Melanie L. Ferlazzo
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
- Inserm, U1296 Unit, Radiation: Defense, Health and Environment, Centre Léon Bérard, Lyon, France
| | - Nicholas R. Howell
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
- Discipline of Medical Imaging and Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Ryan J. Middleton
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - S. Anand Narayanan
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, United States
| | - Kate Poole
- EMBL Australia Node in Single Molecule Science, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Joshua Chou
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
16
|
Zhang DM, Navara R, Yin T, Szymanski J, Goldsztejn U, Kenkel C, Lang A, Mpoy C, Lipovsky CE, Qiao Y, Hicks S, Li G, Moore KMS, Bergom C, Rogers BE, Robinson CG, Cuculich PS, Schwarz JK, Rentschler SL. Cardiac radiotherapy induces electrical conduction reprogramming in the absence of transmural fibrosis. Nat Commun 2021; 12:5558. [PMID: 34561429 PMCID: PMC8463558 DOI: 10.1038/s41467-021-25730-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 08/25/2021] [Indexed: 02/08/2023] Open
Abstract
Cardiac radiotherapy (RT) may be effective in treating heart failure (HF) patients with refractory ventricular tachycardia (VT). The previously proposed mechanism of radiation-induced fibrosis does not explain the rapidity and magnitude with which VT reduction occurs clinically. Here, we demonstrate in hearts from RT patients that radiation does not achieve transmural fibrosis within the timeframe of VT reduction. Electrophysiologic assessment of irradiated murine hearts reveals a persistent supraphysiologic electrical phenotype, mediated by increases in NaV1.5 and Cx43. By sequencing and transgenic approaches, we identify Notch signaling as a mechanistic contributor to NaV1.5 upregulation after RT. Clinically, RT was associated with increased NaV1.5 expression in 1 of 1 explanted heart. On electrocardiogram (ECG), post-RT QRS durations were shortened in 13 of 19 patients and lengthened in 5 patients. Collectively, this study provides evidence for radiation-induced reprogramming of cardiac conduction as a potential treatment strategy for arrhythmia management in VT patients.
Collapse
Affiliation(s)
- David M Zhang
- Center for Noninvasive Cardiac Radioablation, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Rachita Navara
- Center for Noninvasive Cardiac Radioablation, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Tiankai Yin
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Jeffrey Szymanski
- Department of Radiation Oncology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Uri Goldsztejn
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Camryn Kenkel
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Adam Lang
- Department of Pathology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Cedric Mpoy
- Department of Radiation Oncology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Catherine E Lipovsky
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Developmental Biology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Yun Qiao
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Stephanie Hicks
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Gang Li
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Kaitlin M S Moore
- Center for Noninvasive Cardiac Radioablation, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Carmen Bergom
- Center for Noninvasive Cardiac Radioablation, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Radiation Oncology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Buck E Rogers
- Department of Radiation Oncology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Clifford G Robinson
- Center for Noninvasive Cardiac Radioablation, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Radiation Oncology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Phillip S Cuculich
- Center for Noninvasive Cardiac Radioablation, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Radiation Oncology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Julie K Schwarz
- Center for Noninvasive Cardiac Radioablation, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
- Department of Radiation Oncology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA
| | - Stacey L Rentschler
- Center for Noninvasive Cardiac Radioablation, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA.
- Department of Medicine, Cardiovascular Division, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University in St. Louis, School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
17
|
Kumar A, Tahimic CGT, Almeida EAC, Globus RK. Spaceflight Modulates the Expression of Key Oxidative Stress and Cell Cycle Related Genes in Heart. Int J Mol Sci 2021; 22:9088. [PMID: 34445793 PMCID: PMC8396460 DOI: 10.3390/ijms22169088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022] Open
Abstract
Spaceflight causes cardiovascular changes due to microgravity-induced redistribution of body fluids and musculoskeletal unloading. Cardiac deconditioning and atrophy on Earth are associated with altered Trp53 and oxidative stress-related pathways, but the effects of spaceflight on cardiac changes at the molecular level are less understood. We tested the hypothesis that spaceflight alters the expression of key genes related to stress response pathways, which may contribute to cardiovascular deconditioning during extended spaceflight. Mice were exposed to spaceflight for 15 days or maintained on Earth (ground control). Ventricle tissue was harvested starting ~3 h post-landing. We measured expression of select genes implicated in oxidative stress pathways and Trp53 signaling by quantitative PCR. Cardiac expression levels of 37 of 168 genes tested were altered after spaceflight. Spaceflight downregulated transcription factor, Nfe2l2 (Nrf2), upregulated Nox1 and downregulated Ptgs2, suggesting a persistent increase in oxidative stress-related target genes. Spaceflight also substantially upregulated Cdkn1a (p21) and cell cycle/apoptosis-related gene Myc, and downregulated the inflammatory response gene Tnf. There were no changes in apoptosis-related genes such as Trp53. Spaceflight altered the expression of genes regulating redox balance, cell cycle and senescence in cardiac tissue of mice. Thus, spaceflight may contribute to cardiac dysfunction due to oxidative stress.
Collapse
Affiliation(s)
- Akhilesh Kumar
- Space Biosciences Division, NASA Ames Research Center, Mail Stop 288-2, Moffett Field, CA 94035, USA; (A.K.); (E.A.C.A.)
| | | | - Eduardo A. C. Almeida
- Space Biosciences Division, NASA Ames Research Center, Mail Stop 288-2, Moffett Field, CA 94035, USA; (A.K.); (E.A.C.A.)
| | - Ruth K. Globus
- Space Biosciences Division, NASA Ames Research Center, Mail Stop 288-2, Moffett Field, CA 94035, USA; (A.K.); (E.A.C.A.)
| |
Collapse
|
18
|
Garikipati VNS, Arakelyan A, Blakely EA, Chang PY, Truongcao MM, Cimini M, Malaredy V, Bajpai A, Addya S, Bisserier M, Brojakowska A, Eskandari A, Khlgatian MK, Hadri L, Fish KM, Kishore R, Goukassian DA. Long-Term Effects of Very Low Dose Particle Radiation on Gene Expression in the Heart: Degenerative Disease Risks. Cells 2021; 10:387. [PMID: 33668521 PMCID: PMC7917872 DOI: 10.3390/cells10020387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/27/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
Compared to low doses of gamma irradiation (γ-IR), high-charge-and-energy (HZE) particle IR may have different biological response thresholds in cardiac tissue at lower doses, and these effects may be IR type and dose dependent. Three- to four-month-old female CB6F1/Hsd mice were exposed once to one of four different doses of the following types of radiation: γ-IR 137Cs (40-160 cGy, 0.662 MeV), 14Si-IR (4-32 cGy, 260 MeV/n), or 22Ti-IR (3-26 cGy, 1 GeV/n). At 16 months post-exposure, animals were sacrificed and hearts were harvested and archived as part of the NASA Space Radiation Tissue Sharing Forum. These heart tissue samples were used in our study for RNA isolation and microarray hybridization. Functional annotation of twofold up/down differentially expressed genes (DEGs) and bioinformatics analyses revealed the following: (i) there were no clear lower IR thresholds for HZE- or γ-IR; (ii) there were 12 common DEGs across all 3 IR types; (iii) these 12 overlapping genes predicted various degrees of cardiovascular, pulmonary, and metabolic diseases, cancer, and aging; and (iv) these 12 genes revealed an exclusive non-linear DEG pattern in 14Si- and 22Ti-IR-exposed hearts, whereas two-thirds of γ-IR-exposed hearts revealed a linear pattern of DEGs. Thus, our study may provide experimental evidence of excess relative risk (ERR) quantification of low/very low doses of full-body space-type IR-associated degenerative disease development.
Collapse
Affiliation(s)
- Venkata Naga Srikanth Garikipati
- Department of Emergency Medicine, Dorothy M Davis Heart and Lung Research Institute, Wexner Medical School, The Ohio State University, Columbus, OH 43210, USA;
| | - Arsen Arakelyan
- Bioinformatics Group, The Institute of Molecular Biology, The National Academy of Sciences of the Republic of Armenia, Yerevan 0014, Armenia;
- PathVerse, Yerevan 0014, Armenia
| | | | | | - May M. Truongcao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - Maria Cimini
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - Vandana Malaredy
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - Anamika Bajpai
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - Sankar Addya
- Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Agnieszka Brojakowska
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Abrisham Eskandari
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Mary K. Khlgatian
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Kenneth M. Fish
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - David. A. Goukassian
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| |
Collapse
|
19
|
Meerman M, Bracco Gartner TCL, Buikema JW, Wu SM, Siddiqi S, Bouten CVC, Grande-Allen KJ, Suyker WJL, Hjortnaes J. Myocardial Disease and Long-Distance Space Travel: Solving the Radiation Problem. Front Cardiovasc Med 2021; 8:631985. [PMID: 33644136 PMCID: PMC7906998 DOI: 10.3389/fcvm.2021.631985] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Radiation-induced cardiovascular disease is a well-known complication of radiation exposure. Over the last few years, planning for deep space missions has increased interest in the effects of space radiation on the cardiovascular system, as an increasing number of astronauts will be exposed to space radiation for longer periods of time. Research has shown that exposure to different types of particles found in space radiation can lead to the development of diverse cardiovascular disease via fibrotic myocardial remodeling, accelerated atherosclerosis and microvascular damage. Several underlying mechanisms for radiation-induced cardiovascular disease have been identified, but many aspects of the pathophysiology remain unclear. Existing pharmacological compounds have been evaluated to protect the cardiovascular system from space radiation-induced damage, but currently no radioprotective compounds have been approved. This review critically analyzes the effects of space radiation on the cardiovascular system, the underlying mechanisms and potential countermeasures to space radiation-induced cardiovascular disease.
Collapse
Affiliation(s)
- Manon Meerman
- Division Heart and Lung, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tom C L Bracco Gartner
- Division Heart and Lung, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jan Willem Buikema
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sean M Wu
- Division of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Sailay Siddiqi
- Department of Cardiothoracic Surgery, Radboud University, Nijmegen, Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Technical University Eindhoven, Eindhoven, Netherlands
| | | | - Willem J L Suyker
- Division Heart and Lung, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jesper Hjortnaes
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands.,Division Heart and Lung, Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
20
|
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: 72] [Impact Index Per Article: 18.0] [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
|
21
|
Davis CM, Allen AR, Bowles DE. Consequences of space radiation on the brain and cardiovascular system. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:180-218. [PMID: 33902387 DOI: 10.1080/26896583.2021.1891825] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Staying longer in outer space will inevitably increase the health risks of astronauts due to the exposures to galactic cosmic rays and solar particle events. Exposure may pose a significant hazard to space flight crews not only during the mission but also later, when slow-developing adverse effects could finally become apparent. The body of literature examining ground-based outcomes in response to high-energy charged-particle radiation suggests differential effects in response to different particles and energies. Numerous animal and cellular models have repeatedly demonstrated the negative effects of high-energy charged-particle on the brain and cognitive function. However, research on the role of space radiation in potentiating cardiovascular dysfunction is still in its early stages. This review summarizes the available data from studies using ground-based animal models to evaluate the response of the brain and heart to the high-energy charged particles of GCR and SPE, addresses potential sex differences in these effects, and aims to highlight gaps in the current literature for future study.
Collapse
Affiliation(s)
- Catherine M Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Dawn E Bowles
- Division of Surgical Sciences, Department of Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
22
|
Yuan R, Sun Z, Cai J, Yang X, Zhang W, Wu C, Shen Y, Yin A, Wang X, Cai X, Fu X, Shen L, He B. A Novel Anticancer Therapeutic Strategy to Target Autophagy Accelerates Radiation-Associated Atherosclerosis. Int J Radiat Oncol Biol Phys 2020; 109:540-552. [PMID: 32942003 DOI: 10.1016/j.ijrobp.2020.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/13/2020] [Accepted: 09/06/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Autophagy inhibition is a novel therapeutic strategy suggested for patients with advanced cancer, especially those who have undergone radiation therapy. In the present study, we investigated whether autophagy inhibitors accelerate the progression of radiation-associated atherosclerosis (RAA). METHODS AND MATERIALS Eight-week-old apolipoprotein (ApoE-/-) mice were fed a Western diet, and their left common carotid arteries were partially ligated to induce atherogenesis. Four weeks later, local ionizing radiation (IR) at a dose of 5 or 10 Gy was used to induce RAA in the left common carotid artery. After another 4 weeks, severe plaque burden associated with increased macrophage infiltration and lipid deposition, reduced smooth muscle cells, and decreased collagen expression was observed. In addition, these changes occurred in a dose-dependent manner. Improved autophagic flux caused by IR was observed in both macrophages of the atherosclerotic plaque and peritoneal macrophages in vitro. The inhibition of autophagic flux by chloroquine (50 mg/kg/d) further accelerated the progression of RAA in the left common carotid arteries of ApoE-/- mice. Furthermore, chloroquine treatment exacerbated IR-induced p65 nuclear translocation, IκBα degradation, and transcription of nuclear factor-κB (NF-κB) target genes in peritoneal macrophages. CONCLUSIONS IR promotes atherogenesis and increases autophagic flux. In addition, autophagy inhibition by chloroquine accelerates the progression of RAA lesions by stimulating NF-κB-mediated inflammatory responses in macrophages.
Collapse
Affiliation(s)
- Ruosen Yuan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhe Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jiali Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxiao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weifeng Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Caizhe Wu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yejiao Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Anwen Yin
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xuwei Cai
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaolong Fu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
23
|
Sridharan V, Seawright JW, Landes RD, Cao M, Singh P, Davis CM, Mao XW, Singh SP, Zhang X, Nelson GA, Boerma M. Effects of single-dose protons or oxygen ions on function and structure of the cardiovascular system in male Long Evans rats. LIFE SCIENCES IN SPACE RESEARCH 2020; 26:62-68. [PMID: 32718688 PMCID: PMC7387753 DOI: 10.1016/j.lssr.2020.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 05/03/2023]
Abstract
PURPOSE Studies are required to determine whether exposures to radiation encountered during manned missions in deep space may have adverse effects on the cardiovascular system. Most of the prior studies on effects of simulated space radiation on the heart and vasculature have been performed in mouse models. To provide data from a second animal species, two studies were performed to assess effects of high-energy charged particle radiation on the heart and abdominal aorta in a rat model. MATERIALS AND METHODS In study A, male Long Evans rats were exposed to whole-body protons (250 MeV, 0.5 Gy) or oxygen ions (16O, 600 MeV/n, 0.5 Gy), and ultrasonography was used to measure in vivo cardiac function and blood flow parameters at 3, 5, 9 and 12 months after radiation, followed by tissue collection at 12 months. In study B, male Long Evans rats were exposed to 16O (1 GeV/n, 0.01-0.25 Gy), and hearts collected at 6 to 7 and 12 months for histology and western-blots. RESULTS Both protons (250 MeV) and 16O (600 MeV/n) caused a decrease in left ventricular posterior wall thickness at 3-5 months, but did not change echocardiographic measures of cardiac function. In Pulsed-wave Doppler assessment of the abdominal aorta, an increase was seen in mean velocity, peak velocity, and velocity time integral at 12 months after 16O (600 MeV/n), suggesting a change in vascular function. There were no significant changes in histopathology or histological quantification of total collagens in heart or aorta. On the other hand, an increase was seen in a 75 kDa peptide of collagen type III in the left ventricle of rats exposed to protons (250 MeV) and 16O (600 MeV/n and 1 GeV/n), suggesting that radiation caused remodeling of existing collagens in the heart. 16O (600 MeV/n and 1 GeV/n) caused increases in left ventricular protein levels of immune cell markers CD2, CD4, CD8, and CD68. CONCLUSION A single low dose of whole body protons or 16O in male Long Evans rats did not change cardiac function or induce gross pathological changes in the heart or aorta, but induced mild changes in vascular function and remodeling of existing collagens in the heart. Altogether, studies in prior mouse models and the current work in rats indicate minor changes in cardiac function and structure after a low dose of single-ion radiation.
Collapse
Affiliation(s)
- Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock 72205, AR, USA
| | - John W Seawright
- McLennan Community College, Waco, TX, formerly at the Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock 72205, AR, USA
| | - Reid D Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock 72205, AR, USA
| | - Maohua Cao
- College of Dentistry, Texas A&M University, Dallas, TX; formerly at the Division o f Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock 72205, AR, USA
| | - Preeti Singh
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock 72205, AR, USA
| | - Catherine M Davis
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiao-Wen Mao
- Department 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
| | - Xin Zhang
- Department of Pharmacodynamics, University of Florida at Gainesville, Gainesville, FL, USA
| | - Gregory A Nelson
- Department of Basic Sciences and Radiation Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock 72205, AR, USA.
| |
Collapse
|
24
|
Vernice NA, Meydan C, Afshinnekoo E, Mason CE. Long-term spaceflight and the cardiovascular system. PRECISION CLINICAL MEDICINE 2020; 3:284-291. [PMID: 33391848 PMCID: PMC7757439 DOI: 10.1093/pcmedi/pbaa022] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 02/01/2023] Open
Abstract
While early investigations into the physiological effects of spaceflight suggest the body's ability to reversibly adapt, the corresponding effects of long-term spaceflight (>6 months) are much less conclusive. Prolonged exposure to microgravity and radiation yields profound effects on the cardiovascular system, including a massive cephalad fluid translocation and altered arterial pressure, which attenuate blood pressure regulatory mechanisms and increase cardiac output. Also, central venous pressure decreases as a result of the loss of venous compression. The stimulation of baroreceptors by the cephalad shift results in an approximately 10%–15% reduction in plasma volume, with fluid translocating from the vascular lumen to the interstitium. Despite possible increases in cardiac workload, myocyte atrophy and notable, yet unexplained, alterations in hematocrit have been observed. Atrophy is postulated to result from shunting of protein synthesis from the endoplasmic reticulum to the mitochondria via mortalin-mediated action. While data are scarce regarding their causative agents, arrhythmias have been frequently reported, albeit sublethal, during both Russian and American expeditions, with QT interval prolongation observed in long, but not short duration, spaceflight. Exposure of the heart to the proton and heavy ion radiation of deep space has also been shown to result in coronary artery degeneration, aortic stiffness, carotid intima thickening via collagen-mediated action, accelerated atherosclerosis, and induction of a pro-inflammatory state. Upon return, long-term spaceflight frequently results in orthostatic intolerance and altered sympathetic responses, which can prove hazardous should any rapid mobilization or evacuation be required, and indicates that these cardiac risks should be especially monitored for future missions.
Collapse
Affiliation(s)
- Nicholas A Vernice
- The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY 10021, USA
| | - Cem Meydan
- The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY 10021, USA
| | - Ebrahim Afshinnekoo
- The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY 10021, USA
| | - Christopher E Mason
- The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY 10021, USA
| |
Collapse
|
25
|
Cole AJ, Iyengar M, Panesso-Gómez S, O'Hayer P, Chan D, Delgoffe GM, Aird KM, Yoon E, Bai S, Buckanovich RJ. NFATC4 promotes quiescence and chemotherapy resistance in ovarian cancer. JCI Insight 2020; 5:131486. [PMID: 32182216 DOI: 10.1172/jci.insight.131486] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 03/11/2020] [Indexed: 12/30/2022] Open
Abstract
Development of chemotherapy resistance is a major problem in ovarian cancer. One understudied mechanism of chemoresistance is the induction of quiescence, a reversible nonproliferative state. Unfortunately, little is known about regulators of quiescence. Here, we identify the master transcription factor nuclear factor of activated T cells cytoplasmic 4 (NFATC4) as a regulator of quiescence in ovarian cancer. NFATC4 is enriched in ovarian cancer stem-like cells and correlates with decreased proliferation and poor prognosis. Treatment of cancer cells with cisplatin resulted in NFATC4 nuclear translocation and activation of the NFATC4 pathway, while inhibition of the pathway increased chemotherapy response. Induction of NFATC4 activity resulted in a marked decrease in proliferation, G0 cell cycle arrest, and chemotherapy resistance, both in vitro and in vivo. Finally, NFATC4 drove a quiescent phenotype in part via downregulation of MYC. Together, these data identify NFATC4 as a driver of quiescence and a potential new target to combat chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Alexander J Cole
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mangala Iyengar
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Santiago Panesso-Gómez
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick O'Hayer
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel Chan
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Greg M Delgoffe
- Tumor Microenvironment Center, UPMC Hillman Cancer Center; and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katherine M Aird
- Department of Cellular & Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Michigan, USA
| | - Shoumei Bai
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ronald J Buckanovich
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
Transcriptome Analysis of Hypertrophic Heart Tissues from Murine Transverse Aortic Constriction and Human Aortic Stenosis Reveals Key Genes and Transcription Factors Involved in Cardiac Remodeling Induced by Mechanical Stress. DISEASE MARKERS 2019; 2019:5058313. [PMID: 31772688 PMCID: PMC6854968 DOI: 10.1155/2019/5058313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/20/2019] [Accepted: 09/17/2019] [Indexed: 11/18/2022]
Abstract
Background Mechanical stress-induced cardiac remodeling that results in heart failure is characterized by transcriptional reprogramming of gene expression. However, a systematic study of genomic changes involved in this process has not been performed to date. To investigate the genomic changes and underlying mechanism of cardiac remodeling, we collected and analyzed DNA microarray data for murine transverse aortic constriction (TAC) and human aortic stenosis (AS) from the Gene Expression Omnibus database and the European Bioinformatics Institute. Methods and Results The differential expression genes (DEGs) across the datasets were merged. The Venn diagrams showed that the number of intersections for early and late cardiac remodeling was 74 and 16, respectively. Gene ontology and protein–protein interaction network analysis showed that metabolic changes, cell differentiation and growth, cell cycling, and collagen fibril organization accounted for a great portion of the DEGs in the TAC model, while in AS patients' immune system signaling and cytokine signaling displayed the most significant changes. The intersections between the TAC model and AS patients were few. Nevertheless, the DEGs of the two species shared some common regulatory transcription factors (TFs), including SP1, CEBPB, PPARG, and NFKB1, when the heart was challenged by applied mechanical stress. Conclusions This study unravels the complex transcriptome profiles of the heart tissues and highlighting the candidate genes involved in cardiac remodeling induced by mechanical stress may usher in a new era of precision diagnostics and treatment in patients with cardiac remodeling.
Collapse
|
30
|
Ma CX, Zhao XK, Li YD. New therapeutic insights into radiation-induced myocardial fibrosis. Ther Adv Chronic Dis 2019; 10:2040622319868383. [PMID: 31448071 PMCID: PMC6689916 DOI: 10.1177/2040622319868383] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy (RT) for the treatment of thoracic tumors causes radiation-induced heart disease (RIHD). Radiation-induced myocardial fibrosis (RIMF) is both an acute and chronic stage of RIHD, depending on the specific pathology, and is thought to be a major risk factor for adverse myocardial remodeling and vascular changes. With the use of more three-dimensional conformal radiation regimens and early screenings and diagnoses for RIMF, the incidence of RIHD is declining, but it still must be carefully investigated to minimize the mortality and morbidity of patients with thoracic malignancies after RT treatment. Effective methods for preventing RIMF involve a decrease in the direct radiation dose in the heart, and early screening and diagnosis. Medications remain as a useful adjunct for preventing or treating RIMF. This review mainly discusses the cellular and molecular mechanisms underlying RIMF, and new therapeutic drugs that can potentially be developed from this knowledge.
Collapse
Affiliation(s)
- Cheng-Xu Ma
- Gansu University of Chinese Medicine, Lanzhou, PR China
| | - Xin-Ke Zhao
- Department of Interventional Section, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, PR China
| | - Ying-Dong Li
- Gansu University of Chinese Medicine, Lanzhou, 730000, PR China
| |
Collapse
|
31
|
Beheshti A, McDonald JT, Miller J, Grabham P, Costes SV. GeneLab Database Analyses Suggest Long-Term Impact of Space Radiation on the Cardiovascular System by the Activation of FYN Through Reactive Oxygen Species. Int J Mol Sci 2019; 20:ijms20030661. [PMID: 30717456 PMCID: PMC6387434 DOI: 10.3390/ijms20030661] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/17/2022] Open
Abstract
Space radiation has recently been considered a risk factor for astronauts’ cardiac health. As an example, for the case of how to query and identify datasets within NASA’s GeneLab database and demonstrate the database utility, we used an unbiased systems biology method for identifying key genes/drivers for the contribution of space radiation on the cardiovascular system. This knowledge can contribute to designing appropriate experiments targeting these specific pathways. Microarray data from cardiomyocytes of male C57BL/6 mice followed-up for 28 days after exposure to 900 mGy of 1 GeV proton or 150 mGy of 1 GeV/n 56Fe were compared to human endothelial cells (HUVECs) cultured for 7 days on the International Space Station (ISS). We observed common molecular pathways between simulated space radiation and HUVECs flown on the ISS. The analysis suggests FYN is the central driver/hub for the cardiovascular response to space radiation: the known oxidative stress induced immediately following radiation would only be transient and would upregulate FYN, which in turn would reduce reactive oxygen species (ROS) levels, protecting the cardiovascular system. The transcriptomic signature of exposure to protons was also much closer to the spaceflight signature than 56Fe’s signature. To our knowledge, this is the first time GeneLab datasets were utilized to provide potential biological indications that the majority of ions on the ISS are protons, clearly illustrating the power of omics analysis. More generally, this work also demonstrates how to combine animal radiation studies done on the ground and spaceflight studies to evaluate human risk in space.
Collapse
Affiliation(s)
- Afshin Beheshti
- WYLE Labs, NASA Ames Research Center, Moffett Field CA 94035, USA.
| | - J Tyson McDonald
- Department of Physics, Hampton University, Hampton, VA 23668 USA.
| | - Jack Miller
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Peter Grabham
- Center for Radiological Research, Columbia University, New York, NY 10032, USA.
| | - Sylvain V Costes
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA.
| |
Collapse
|
32
|
Seawright JW, Sridharan V, Landes RD, Cao M, Singh P, Koturbash I, Mao XW, Miousse IR, Singh SP, Nelson GA, Hauer-Jensen M, Boerma M. Effects of low-dose oxygen ions and protons on cardiac function and structure in male C57BL/6J mice. LIFE SCIENCES IN SPACE RESEARCH 2019; 20:72-84. [PMID: 30797436 PMCID: PMC6391741 DOI: 10.1016/j.lssr.2019.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 05/07/2023]
Abstract
PURPOSE Astronauts traveling beyond low-Earth orbit will be exposed to high linear-energy transfer charged particles. Because there is concern about the adverse effects of space radiation on the cardiovascular system, this study assessed cardiac function and structure and immune cell infiltration in a mouse model of charged-particle irradiation. MATERIALS AND METHODS Male C57BL/6 J mice were exposed to oxygen ions (16O, 600 MeV/n at 0.25-0.26 Gy/min to a total dose of 0, 0.05, 0.1, 0.25, or 1 Gy), protons (150 MeV, 0.35-0.55 Gy/min to 0, 0.5, or 1 Gy), or protons (150 MeV, 0.5 Gy) followed by 16O (600 MeV/n, 0.1 Gy). Separate groups of mice received 137Cs γ-rays (1 Gy/min to 0, 0.5, 1, or 3 Gy) as a reference. Cardiac function and blood velocity were measured with ultrasonography at 3, 5, 7, and 9 months after irradiation. At 2 weeks, 3 months, and 9 months, cardiac tissue was collected to assess apoptosis, tissue remodeling, and markers of immune cells. RESULTS Ejection fraction and fractional shortening decreased at 3 and 7 months after 16O. These parameters did not change in mice exposed to γ-rays, protons, or protons followed by 16O. Each of the radiation exposures caused only small increases in cleaved caspase-3 and numbers of apoptotic nuclei. Changes in the levels of α-smooth muscle cell actin and a 75-kDa peptide of collagen type III in the left ventricle suggested tissue remodeling, but there was no significant change in total collagen deposition at 2 weeks, 3 months, and 9 months. Increases in protein amounts of cluster of differentiation (CD)2, CD68, and CD45 as measured with immunoblots at 2 weeks, 3 months, and 9 months after exposure to protons or 16O alone suggested immune cell infiltration. For type III collagen, CD2 and CD68, the efficacy in inducing protein abundance of CD2, CD68, and CD45 was 16O > protons > γ-rays > protons followed by 16O. CONCLUSIONS Low-dose, high-energy charged-particle irradiation caused mild changes in cardiac function and tissue remodeling in the mouse.
Collapse
Affiliation(s)
- John W Seawright
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock, AR 72205, USA
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock, AR 72205, USA
| | - Reid D Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Maohua Cao
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock, AR 72205, USA
| | - Preeti Singh
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock, AR 72205, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Xiao-Wen Mao
- Department of Basic Sciences and Radiation Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sharda P Singh
- Department of Internal Medicine, Texas Tech Health Sciences Center, Lubbock, TX, USA
| | - Gregory A Nelson
- Department of Basic Sciences and Radiation Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock, AR 72205, USA
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham Slot 522-10, Little Rock, AR 72205, USA.
| |
Collapse
|
33
|
Li Z, Zhang X, Guo Z, Zhong Y, Wang P, Li J, Li Z, Liu P. SIRT6 Suppresses NFATc4 Expression and Activation in Cardiomyocyte Hypertrophy. Front Pharmacol 2019; 9:1519. [PMID: 30670969 PMCID: PMC6331469 DOI: 10.3389/fphar.2018.01519] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022] Open
Abstract
NFATc4, a member from the Nuclear Factor of Activated T cells (NFATs) transcription factor family, plays a pivotal role in the development of cardiac hypertrophy. NFATc4 is dephosphorylated by calcineurin and translocated from the cytoplasm to the nucleus to regulate the expression of hypertrophic genes, like brain natriuretic polypeptide (BNP). The present study identified SIRT6, an important subtype of NAD+ dependent class III histone deacetylase, to be a negative regulator of NFATc4 in cardiomyocyte hypertrophy. In phenylephrine (PE)-induced hypertrophic cardiomyocyte model, overexpression of SIRT6 by adenovirus infection or by plasmid transfection repressed the protein and mRNA expressions of NFATc4, elevated its phosphorylation level, prevented its nuclear accumulation, subsequently suppressed its transcriptional activity and downregulated its target gene BNP. By contrast, mutant of SIRT6 without deacetylase activity (H133Y) did not demonstrate these effects, suggesting that the inhibitory effect of SIRT6 on NFATc4 was dependent on its deacetylase activity. Moreover, the effect of SIRT6 overexpression on repressing BNP expression was reversed by NFATc4 replenishment, whereas the effect of SIRT6 deficiency on upregulating BNP was recovered by NFATc4 silencing. Mechanistically, interactions between SIRT6 and NFATc4 might possibly facilitate the deacetylation of NFATc4 by SIRT6, thereby preventing the activation of NFATc4. In conclusion, the present study reveals that SIRT6 suppresses the expression and activation of NFATc4. These findings provide more evidences of the anti-hypertrophic effect of SIRT6 and suggest SIRT6 as a potential therapeutic target for cardiac hypertrophy.
Collapse
Affiliation(s)
- Zhenzhen Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaoying Zhang
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Shaanxi, China
| | - Zhen Guo
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yao Zhong
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Department of Cardiology, Third People's Hospital of Dongguan, Dongguan, China
| | - Panxia Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jingyan Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhuoming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peiqing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
34
|
Hellweg CE, Chishti AA, Diegeler S, Spitta LF, Henschenmacher B, Baumstark-Khan C. Molecular Signaling in Response to Charged Particle Exposures and its Importance in Particle Therapy. Int J Part Ther 2018; 5:60-73. [PMID: 31773020 PMCID: PMC6871585 DOI: 10.14338/ijpt-18-00016.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/13/2018] [Indexed: 12/17/2022] Open
Abstract
Energetic, charged particles elicit an orchestrated DNA damage response (DDR) during their traversal through healthy tissues and tumors. Complex DNA damage formation, after exposure to high linear energy transfer (LET) charged particles, results in DNA repair foci formation, which begins within seconds. More protein modifications occur after high-LET, compared with low-LET, irradiation. Charged-particle exposure activates several transcription factors that are cytoprotective or cytodestructive, or that upregulate cytokine and chemokine expression, and are involved in bystander signaling. Molecular signaling for a survival or death decision in different tumor types and healthy tissues should be studied as prerequisite for shaping sensitizing and protective strategies. Long-term signaling and gene expression changes were found in various tissues of animals exposed to charged particles, and elucidation of their role in chronic and late effects of charged-particle therapy will help to develop effective preventive measures.
Collapse
Affiliation(s)
- Christine E. Hellweg
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| | - Arif Ali Chishti
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
- The Karachi Institute of Biotechnology and Genetic Engineering, University of Karachi, Karachi, Pakistan
| | - Sebastian Diegeler
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| | - Luis F. Spitta
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| | - Bernd Henschenmacher
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| | - Christa Baumstark-Khan
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| |
Collapse
|
35
|
Hu Y, Xia W, Hou M. Macrophage migration inhibitory factor serves a pivotal role in�the regulation of radiation-induced cardiac senescencethrough rebalancing the microRNA-34a/sirtuin 1 signaling pathway. Int J Mol Med 2018; 42:2849-2858. [PMID: 30226567 DOI: 10.3892/ijmm.2018.3838] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/10/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yiwang Hu
- Department of Colorectal Surgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Wenzheng Xia
- Department of Neurosurgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Meng Hou
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
36
|
Beheshti A, Miller J, Kidane Y, Berrios D, Gebre SG, Costes SV. NASA GeneLab Project: Bridging Space Radiation Omics with Ground Studies. Radiat Res 2018; 189:553-559. [DOI: 10.1667/rr15062.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Afshin Beheshti
- Wyle Labs, NASA Ames Research Center, Moffett Field, California, 94035
| | - Jack Miller
- Lawrence Berkeley National Laboratory, Berkeley, California, 94720
| | - Yared Kidane
- Wyle Labs, NASA Ames Research Center, Moffett Field, California, 94035
| | - Daniel Berrios
- USRA, NASA Ames Research Center, Moffett Field, Calfornia 94035
| | - Samrawit G. Gebre
- Wyle Labs, NASA Ames Research Center, Moffett Field, California, 94035
| | - Sylvain V. Costes
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, California 94035
| |
Collapse
|
37
|
Metabolic Pathway Genes Associated with Susceptibility Genes to Coronary Artery Disease. Int J Genomics 2018; 2018:9025841. [PMID: 29607312 PMCID: PMC5828413 DOI: 10.1155/2018/9025841] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/15/2017] [Accepted: 12/04/2017] [Indexed: 12/16/2022] Open
Abstract
Coronary artery disease (CAD) is one of the leading threats to global health. Previous research has proven that metabolic pathway disorders, such as high blood lipids and diabetes, are one of the risk factors that mostly cause CAD. However, the crosstalk between metabolic pathways and CAD was mostly studied on physiology processes by analyzing a single gene function. A canonical correlation analysis was used to identify the metabolic pathways, which were integrated as a unit to coexpress with CAD susceptibility genes, and to resolve additional metabolic factors that are related to CAD. Seven pathways, including citrate cycle, ubiquinone, terpenoid quinone biosynthesis, and N-glycan biosynthesis, were identified as an integrated unit coexpressed with CAD genes. These pathways could not be revealed as a coexpressed pathway through traditional methods as each single gene has weak correlation. Furthermore, sets of genes in these pathways were candidate markers for diagnosis and detection from patients' serum.
Collapse
|
38
|
Sylvester CB, Abe JI, Patel ZS, Grande-Allen KJ. Radiation-Induced Cardiovascular Disease: Mechanisms and Importance of Linear Energy Transfer. Front Cardiovasc Med 2018; 5:5. [PMID: 29445728 PMCID: PMC5797745 DOI: 10.3389/fcvm.2018.00005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/09/2018] [Indexed: 12/24/2022] Open
Abstract
Radiation therapy (RT) in the form of photons and protons is a well-established treatment for cancer. More recently, heavy charged particles have been used to treat radioresistant and high-risk cancers. Radiation treatment is known to cause cardiovascular disease (CVD) which can occur acutely during treatment or years afterward in the form of accelerated atherosclerosis. Radiation-induced cardiovascular disease (RICVD) can be a limiting factor in treatment as well as a cause of morbidity and mortality in successfully treated patients. Inflammation plays a key role in both acute and chronic RICVD, but the underling pathophysiology is complex, involving DNA damage, reactive oxygen species, and chronic inflammation. While understanding of the molecular mechanisms of RICVD has increased, the growing number of patients receiving RT warrants further research to identify individuals at risk, plans for prevention, and targets for the treatment of RICVD. Research on RICVD is also relevant to the National Aeronautics and Space Administration (NASA) due to the prevalent space radiation environment encountered by astronauts. NASA's current research on RICVD can both contribute to and benefit from concurrent work with cell and animal studies informing radiotoxicities resulting from cancer therapy. This review summarizes the types of radiation currently in clinical use, models of RICVD, current knowledge of the mechanisms by which they cause CVD, and how this knowledge might apply to those exposed to various types of radiation.
Collapse
Affiliation(s)
- Christopher B Sylvester
- Department of Bioengineering, Rice University, Houston, TX, United States.,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Jun-Ichi Abe
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zarana S Patel
- Science and Space Operations, KBRwyle, Houston, TX, United States
| | | |
Collapse
|
39
|
Nikitaki Z, Holá M, Donà M, Pavlopoulou A, Michalopoulos I, Angelis KJ, Georgakilas AG, Macovei A, Balestrazzi A. Integrating plant and animal biology for the search of novel DNA damage biomarkers. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 775:21-38. [DOI: 10.1016/j.mrrev.2018.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/08/2018] [Accepted: 01/16/2018] [Indexed: 12/11/2022]
|
40
|
Abstract
National space agencies and private corporations aim at an extended presence of humans in space in the medium to long term. Together with currently suboptimal technology, microgravity and cosmic rays raise health concerns about deep-space exploration missions. Both of these physical factors affect the cardiovascular system, whose gravity-dependence is pronounced. Heart and vascular function are, therefore, susceptible to substantial changes in weightlessness. The altered cardiovascular function in space causes physiological problems in the postflight period. A compromised cardiovascular system can be excessively vulnerable to space radiation, synergistically resulting in increased damage. The space radiation dose is significantly lower than in patients undergoing radiotherapy, in whom cardiac damage is well-documented following cancer therapy in the thoracic region. Nevertheless, epidemiological findings suggest an increased risk of late cardiovascular disease even with low doses of radiation. Moreover, the peculiar biological effectiveness of heavy ions in cosmic rays might increase this risk substantially. However, whether radiation-induced cardiovascular effects have a threshold at low doses is still unclear. The main countermeasures to mitigate the effect of the space environment on cardiac function are physical exercise, antioxidants, nutraceuticals, and radiation shielding.
Collapse
|
41
|
Fujishiro A, Miura Y, Iwasa M, Fujii S, Sugino N, Andoh A, Hirai H, Maekawa T, Ichinohe T. Effects of acute exposure to low-dose radiation on the characteristics of human bone marrow mesenchymal stromal/stem cells. Inflamm Regen 2017; 37:19. [PMID: 29259718 PMCID: PMC5725824 DOI: 10.1186/s41232-017-0049-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/10/2017] [Indexed: 12/26/2022] Open
Abstract
Background In recent years, increasing attention has been paid to the effects of low-dose irradiation on human health. We examined whether low-dose irradiation affected the functions of mesenchymal stromal/stem cells (MSCs), which are tissue/organ-supportive stem cells, derived from bone marrow (BM). Methods Normal human BM-MSCs from five healthy individuals were used in this study. Culture-expanded BM-MSCs were exposed to 0.1 gray (Gy) of γ-radiation (Cesium-137) at a rate of 0.8 Gy/min (Ir-MSCs), and their expansion, multi-differentiation, and hematopoiesis-supportive capabilities were investigated. Results The expansion of BM-MSCs was transiently delayed after low-dose γ-irradiation compared with that of non-irradiated BM-MSCs (non-Ir-MSCs) in two out of five lots. Adipogenic and osteogenic differentiation capabilities were not significantly affected by low-dose irradiation, although one lot of BM-MSCs tended to have transiently reduced differentiation. When human BM hematopoietic stem/progenitor cells (HPCs) were co-cultured with Ir-MSCs, the generation of CD34+CD38+ cells from HPCs was enhanced compared with that in co-cultures with non-Ir-MSCs in two out of five lots. The mRNA expression level of interleukin (IL)-6 was increased and those of stem cell factor (SCF) and fms-related tyrosine kinase 3 ligand (Flt3L) were decreased in the affected lots of Ir-MSCs. In the other three lots of BM-MSCs, a cell growth delay, enhanced generation of CD34+CD38+ cells from HPCs in co-culture, and a combination of increased expression of IL-6 and decreased expression of SCF and Flt3L were not observed. Of note, the characteristics of these affected Ir-MSCs recovered to a similar level as those of non-Ir-MSCs following culture for 3 weeks. Conclusions Our results suggest that acute exposure to low-dose (0.1 Gy) radiation can transiently affect the functional characteristics of human BM-MSCs.
Collapse
Affiliation(s)
- Aya Fujishiro
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan.,Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Setatsukinowacho, Otsu, Shiga 520-2192 Japan
| | - Yasuo Miura
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan.,Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minamiku, Hiroshima, 734-8553 Japan
| | - Masaki Iwasa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan.,Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Setatsukinowacho, Otsu, Shiga 520-2192 Japan
| | - Sumie Fujii
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan.,Department of Hematology/Oncology, Graduate School for Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Noriko Sugino
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan.,Department of Hematology/Oncology, Graduate School for Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Akira Andoh
- Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Setatsukinowacho, Otsu, Shiga 520-2192 Japan
| | - Hideyo Hirai
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Taira Maekawa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minamiku, Hiroshima, 734-8553 Japan
| |
Collapse
|
42
|
Sasi SP, Yan X, Zuriaga-Herrero M, Gee H, Lee J, Mehrzad R, Song J, Onufrak J, Morgan J, Enderling H, Walsh K, Kishore R, Goukassian DA. Different Sequences of Fractionated Low-Dose Proton and Single Iron-Radiation-Induced Divergent Biological Responses in the Heart. Radiat Res 2017; 188:191-203. [PMID: 28613990 DOI: 10.1667/rr14667.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Deep-space travel presents risks of exposure to ionizing radiation composed of a spectrum of low-fluence protons (1H) and high-charge and energy (HZE) iron nuclei (e.g., 56Fe). When exposed to galactic cosmic rays, each cell in the body may be traversed by 1H every 3-4 days and HZE nuclei every 3-4 months. The effects of low-dose sequential fractionated 1H or HZE on the heart are unknown. In this animal model of simulated ionizing radiation, middle-aged (8-9 months old) male C57BL/6NT mice were exposed to radiation as follows: group 1, nonirradiated controls; group 2, three fractionated doses of 17 cGy 1H every other day (1H × 3); group 3, three fractionated doses of 17 cGy 1H every other day followed by a single low dose of 15 cGy 56Fe two days after the final 1H dose (1H × 3 + 56Fe); and group 4, a single low dose of 15 cGy 56Fe followed (after 2 days) by three fractionated doses of 17 cGy 1H every other day (56Fe + 1H × 3). A subgroup of mice from each group underwent myocardial infarction (MI) surgery at 28 days postirradiation. Cardiac structure and function were assessed in all animals at days 7, 14 and 28 after MI surgery was performed. Compared to the control animals, the treatments that groups 2 and 3 received did not induce negative effects on cardiac function or structure. However, compared to all other groups, the animals in group 4, showed depressed left ventricular (LV) functions at 1 month with concomitant enhancement in cardiac fibrosis and induction of cardiac hypertrophy signaling at 3 months. In the irradiated and MI surgery groups compared to the control group, the treatments received by groups 2 and 4 did not induce negative effects at 1 month postirradiation and MI surgery. However, in group 3 after MI surgery, there was a 24% increase in mortality, significant decreases in LV function and a 35% increase in post-infarction size. These changes were associated with significant decreases in the angiogenic and cell survival signaling pathways. These data suggest that fractionated doses of radiation induces cellular and molecular changes that result in depressed heart functions both under basal conditions and particularly after myocardial infarction.
Collapse
Affiliation(s)
- Sharath P Sasi
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts
| | - Xinhua Yan
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts.,b Tufts University School of Medicine, Boston, Massachusetts
| | - Marian Zuriaga-Herrero
- f Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Hannah Gee
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts
| | - Juyong Lee
- c Calhoun Cardiology Center, University of Connecticut Health Center, Farmington, Connecticut
| | - Raman Mehrzad
- d Steward Carney Hospital, Dorchester, Massachusetts
| | - Jin Song
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts
| | - Jillian Onufrak
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts
| | - James Morgan
- b Tufts University School of Medicine, Boston, Massachusetts.,d Steward Carney Hospital, Dorchester, Massachusetts
| | - Heiko Enderling
- e Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Walsh
- f Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Raj Kishore
- 7 Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - David A Goukassian
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts.,f Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts.,7 Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
Boerma M, Sridharan V, Mao XW, Nelson GA, Cheema AK, Koturbash I, Singh SP, Tackett AJ, Hauer-Jensen M. Effects of ionizing radiation on the heart. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 770:319-327. [PMID: 27919338 DOI: 10.1016/j.mrrev.2016.07.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 12/20/2022]
Abstract
This article provides an overview of studies addressing effects of ionizing radiation on the heart. Clinical studies have identified early and late manifestations of radiation-induced heart disease, a side effect of radiation therapy to tumors in the chest when all or part of the heart is situated in the radiation field. Studies in preclinical animal models have contributed to our understanding of the mechanisms by which radiation may injure the heart. More recent observations in human subjects suggest that ionizing radiation may have cardiovascular effects at lower doses than was previously thought. This has led to examinations of low-dose photons and low-dose charged particle irradiation in animal models. Lastly, studies have started to identify non-invasive methods for detection of cardiac radiation injury and interventions that may prevent or mitigate these adverse effects. Altogether, this ongoing research should increase our knowledge of biological mechanisms of cardiovascular radiation injury, identify non-invasive biomarkers for early detection, and potential interventions that may prevent or mitigate these adverse effects.
Collapse
Affiliation(s)
- Marjan Boerma
- University of Arkansas for Medical Sciences, Division of Radiation Health, Little Rock, AR, United States.
| | - Vijayalakshmi Sridharan
- University of Arkansas for Medical Sciences, Division of Radiation Health, Little Rock, AR, United States
| | - Xiao-Wen Mao
- Loma Linda University, Department of Basic Sciences, Loma Linda, CA, United States
| | - Gregory A Nelson
- Loma Linda University, Department of Basic Sciences, Loma Linda, CA, United States
| | - Amrita K Cheema
- Georgetown University Medical Center, Departments of Oncology and Biochemistry, Molecular and Cellular Biology, Washington, DC, United States
| | - Igor Koturbash
- University of Arkansas for Medical Sciences, Department of Environment and Occupational Health, Little Rock, AR, United States
| | - Sharda P Singh
- University of Arkansas for Medical Sciences, Department of Pharmacology and Toxicology, Little Rock, AR, United States
| | - Alan J Tackett
- University of Arkansas for Medical Sciences, Department of Biochemistry and Molecular Biology, Little Rock, AR, United States
| | - Martin Hauer-Jensen
- University of Arkansas for Medical Sciences, Division of Radiation Health, Little Rock, AR, United States; Central Arkansas Veterans Healthcare System, Surgical Service, Little Rock, AR, United States
| |
Collapse
|
44
|
Natarajan M, Aravindan N, Sprague EA, Mohan S. Hemodynamic Flow-Induced Mechanotransduction Signaling Influences the Radiation Response of the Vascular Endothelium. Radiat Res 2016; 186:175-88. [PMID: 27387860 DOI: 10.1667/rr14410.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Hemodynamic shear stress is defined as the physical force exerted by the continuous flow of blood in the vascular system. Endothelial cells, which line the inner layer of blood vessels, sense this physiological force through mechanotransduction signaling and adapt to maintain structural and functional homeostasis. Hemodynamic flow, shear stress and mechanotransduction signaling are, therefore, an integral part of endothelial pathophysiology. Although this is a well-established concept in the cardiovascular field, it is largely dismissed in studies aimed at understanding radiation injury to the endothelium and subsequent cardiovascular complications. We and others have reported on the differential response of the endothelium when the cells are under hemodynamic flow shear compared with static culture. Further, we have demonstrated significant differences in the gene expression of static versus shear-stressed irradiated cells in four key pathways, reinforcing the importance of shear stress in understanding radiation injury of the endothelium. This article further emphasizes the influence of hemodynamic shear stress and the associated mechanotransduction signaling on physiological functioning of the vascular endothelium and underscores its significance in understanding radiation injury to the vasculature and associated cardiac complications. Studies of radiation effect on endothelial biology and its implication on cardiotoxicity and vascular complications thus far have failed to highlight the significance of these factors. Factoring in these integral parts of the endothelium will enhance our understanding of the contribution of the endothelium to radiation biology. Without such information, the current approaches to studying radiation-induced injury to the endothelium and its consequences in health and disease are limited.
Collapse
Affiliation(s)
| | - Natarajan Aravindan
- c Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Eugene A Sprague
- b Medicine University of Texas Health Science Center, San Antonio, Texas 78229; and
| | | |
Collapse
|
45
|
Ramadan SS, Sridharan V, Koturbash I, Miousse IR, Hauer-Jensen M, Nelson GA, Boerma M. A priming dose of protons alters the early cardiac cellular and molecular response to (56)Fe irradiation. LIFE SCIENCES IN SPACE RESEARCH 2016; 8:8-13. [PMID: 26948008 PMCID: PMC4782196 DOI: 10.1016/j.lssr.2015.12.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/28/2015] [Accepted: 12/08/2015] [Indexed: 05/07/2023]
Abstract
PURPOSE Recent evidence suggests that the heart may be injured by ionizing radiation at lower doses than was previously thought. This raises concerns about the cardiovascular risks from exposure to radiation during space travel. Since space travel is associated with exposure to both protons from solar particle events and heavy ions from galactic cosmic rays, we here examined the effects of a "priming" dose of protons on the cardiac cellular and molecular response to a "challenge" dose of (56)Fe in a mouse model. METHODS Male C57BL/6 mice at 10 weeks of age were exposed to sham-irradiation, 0.1 Gy of protons (150 MeV), 0.5 Gy of (56)Fe (600 MeV/n), or 0.1 Gy of protons 24 hours prior to 0.5 Gy of (56)Fe. Hearts were obtained at 7 days post-irradiation and western-blots were used to determine protein markers of cardiac remodeling, inflammatory infiltration, and cell death. RESULTS Exposure to (56)Fe caused an increase in expression of α-smooth muscle cell actin, collagen type III, the inflammatory cell markers mast cell tryptase, CD2 and CD68, the endothelial glycoprotein thrombomodulin, and cleaved caspase 3. Of all proteins investigated, protons at a dose of 0.1 Gy induced a small increase only in cleaved caspase 3 levels. On the other hand, exposure to protons 24 hours before (56)Fe prevented all of the responses to (56)Fe. CONCLUSIONS This study shows that a low dose of protons may prime the heart to respond differently to a subsequent challenge dose of heavy ions. Further investigation is required to identify responses at additional time points, consequences for cardiac function, threshold dose levels, and mechanisms by which a proton priming dose may alter the response to heavy ions.
Collapse
Affiliation(s)
- Samy S Ramadan
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Surgical Service, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Gregory A Nelson
- Departments of Basic Sciences and Radiation Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|