Menu

 

The NASA Task Book
Advanced Search     

Project Title:  Proteomic Signatures of Space Radiation Induced Cardiovascular Degeneration Reduce
Images: icon  Fiscal Year: FY 2021 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 05/12/2016  
End Date: 05/11/2022  
Task Last Updated: 11/26/2021 
Download report in PDF pdf
Principal Investigator/Affiliation:   Bowles, Dawn  Ph.D. / Duke University 
Address:  Department of Surgery 
Msrb1 Room 401B, DUMC 2642 
Durham , NC 27710-0001 
Email: dawn.bowles@duke.edu 
Phone: 919-668-1947  
Congressional District:
Web:  
Organization Type: UNIVERSITY 
Organization Name: Duke University 
Joint Agency:  
Comments:  
Co-Investigator(s)
Affiliation: 
Abraham, Dennis  M.D. Duke University 
Kidane, Yared  Ph.D. Wyle Laboratories, Inc. 
Mao, Lan  M.D. Duke University 
Moseley, Martin  Ph.D. Duke University 
Key Personnel Changes / Previous PI: November 2021 report: Dr. Mark Dewhirst retired and is no longer CoInvestigator on the project.
Project Information: Grant/Contract No. NNX16AK20G 
Responsible Center: NASA JSC 
Grant Monitor: Elgart, Robin  
Center Contact: 281-244-0596 (o)/832-221-4576 (m) 
shona.elgart@nasa.gov 
Unique ID: 10855 
Solicitation / Funding Source: 2014-15 HERO NNJ14ZSA001N-RADIATION. Appendix D: Ground-Based Studies in Space Radiobiology 
Grant/Contract No.: NNX16AK20G 
Project Type: GROUND 
Flight Program:  
TechPort: No 
No. of Post Docs:
No. of PhD Candidates:
No. of Master's Candidates:  
No. of Bachelor's Candidates:  
No. of PhD Degrees:  
No. of Master's Degrees:  
No. of Bachelor's Degrees:  
Human Research Program Elements: (1) SR:Space Radiation
Human Research Program Risks: (1) Cardiovascular:Risk of Cardiovascular Adaptations Contributing to Adverse Mission Performance and Health Outcomes
Human Research Program Gaps: (1) CV-102:Determine whether space radiation induces cardiovascular structural and functional changes and/or oxidative stress & damage (OSaD)/inflammation, that can contribute to development of disease.
Flight Assignment/Project Notes: NOTE: End date changed to 5/11/2022 per L. Barnes-Moten/JSC and NSSC information (Ed., 5/10/21)

NOTE: End date changed to 5/11/2021 per NSSC information (Ed., 8/12/20)

Task Description: Radiation damage and the cell’s attempt to repair it triggers a myriad of signal transduction pathways which alter gene, and ultimately, protein expression. Space radiation may affect biomolecules, cellular processes, and ultimately the cellular protein content (the proteome) differently than radiation present on Earth. Epidemiological analysis of terrestrial radiation exposure indicates that single high- or multiple low-dose radiation exposure can culminate in a wide array of cardiac injury and malfunction over time. Based on terrestrial data, it is believed that cardiovascular disorders may develop in astronauts from exposure to the space radiation environment. Indeed, a recent study by Yan et al. (2014), found that a single full body exposure to a low dose of proton or iron particle radiation, which somewhat mimics the space radiation environment, was sufficient to induce a significant, long term, negative effect on murine cardiovascular function. In this proposal, we take advantage of our expertise with bioinformatics analysis of cardiovascular proteomic data sets and murine cardiovascular physiology to evaluate the consequences of low dose, chronic space radiation, or mixed field space radiation on the dynamics of the cardiac proteome and to understand how the radiation induced changes relate to cardiovascular function. In doing so, we will extend Yan et al.’s work by identifying a proteomic signature that predicts the development of permanent cardiovascular degeneration from a single low dose space radiation exposure. Further, we seek to evaluate whether the proteomic signatures differ when mice experience repeated exposures of space-like radiation or mixed field space radiation. This information will lead to a mechanistic understanding of the altered cellular and molecular processes contributing to the development of cardiovascular dysfunction at the organ and organismal level in scenarios better mimicking the space radiation environment. This information is needed to predict, monitor, and prevent cardiac damage during long term space flight.

Reference: Yan, X., et al., Cardiovascular risks associated with low dose ionizing particle radiation. PLoS One, 2014. 9(10): p. e110269.

Research Impact/Earth Benefits: Limited information is known regarding the impact of chronic low level radiation on cardiovascular molecular biology and function both terrestrially and during extended space exploration. Our research is expected to provide information in regards to terrestrial and astronaut heath. Innovative technologies that may arise from our studies may include novel biomarkers predictive of cardiovascular susceptibility to chronic low level radiation as well as countermeasures that may be employed both on Earth as well as during space exploration.

Task Progress & Bibliography Information FY2021 
Task Progress: Radiation and follow up -- Approximately 920 C57b/6 male mice have been irradiated and undergone a thorough cardiovascular assessment (some over an 18-month period post radiation).

Cardiac function -- Functionally, in contrast to other forms of radiation, a single exposure to 5 ion Galactic Cosmic Ray Simulation (GCRSim) appears to have significant effects on clinically relevant parameters of cardiac function.

Tissue repository-- a large tissue repository covering a range of organs has been established. Tissues have been shared with other investigators as well as used in our proteomics studies.

Proteomics-- Bioinformatics analyses reveals both commonly as well as uniquely affected biological pathways across radiation types and doses. Notably, GCR exhibits highly perturbed cornification and keratinization pathways as well as mitochondrial pathways modulated by phosphorylation. In agreement with numerous other studies, perturbation of mitochondrial processes and cellular components was affected across most of the radiation conditions evaluated.

We are currently working on four papers based on our findings. The first paper is an evaluation of cardiac function focusing on GCR mice. This paper is in revision at iscience. The second paper is focusing on our GCR proteomics findings. This paper is still in draft format but relevant findings are noted in this report. The third paper is an evaluation of cardiac function looking across all doses of gamma, oxygen, and iron. The 4th paper focuses on proteomics findings across gamma, oxygen, and iron.

Bibliography: Description: (Last Updated: 07/11/2023) 

Show Cumulative Bibliography
 
Abstracts for Journals and Proceedings Bowles DE, Kidane Y, Lee F, Bishawi M, Thompson JW, Moseley MA, Foster MW, Abraham DA, Glass C, Blocker S, Wang C, Johnson A, Rockman H, Mao L, Slaba T, Dewhirts MW. "Proteomics Signatures of Space Radiation Induced Cardiovascular Degeneration." 2021 NASA Human Research Program Investigators’ Workshop, Virtual, February 1-4, 2021.

Abstracts. 2021 NASA Human Research Program Investigators’ Workshop, Virtual, February 1-4, 2021. , Feb-2021

Articles in Peer-reviewed Journals Davis CM, Allen AR, Bowles DE. "Consequences of space radiation on the brain and cardiovascular system." J Environ Sci Health C Toxicol Carcinog. 2021 Apr 27;39(2):180-218. https://doi.org/10.1080/26896583.2021.1891825 ; PMID: 33902387 , Apr-2021
Project Title:  Proteomic Signatures of Space Radiation Induced Cardiovascular Degeneration Reduce
Images: icon  Fiscal Year: FY 2020 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 05/12/2016  
End Date: 05/11/2022  
Task Last Updated: 04/12/2020 
Download report in PDF pdf
Principal Investigator/Affiliation:   Bowles, Dawn  Ph.D. / Duke University 
Address:  Department of Surgery 
Msrb1 Room 401B, DUMC 2642 
Durham , NC 27710-0001 
Email: dawn.bowles@duke.edu 
Phone: 919-668-1947  
Congressional District:
Web:  
Organization Type: UNIVERSITY 
Organization Name: Duke University 
Joint Agency:  
Comments:  
Co-Investigator(s)
Affiliation: 
Abraham, Dennis  M.D. Duke University 
Kidane, Yared  Ph.D. Wyle Laboratories, Inc. 
Mao, Lan  M.D. Duke University 
Dewhirst, Mark  D.V.M., Ph.D. Duke University 
Moseley, Martin  Ph.D. Duke University 
Project Information: Grant/Contract No. NNX16AK20G 
Responsible Center: NASA JSC 
Grant Monitor: Simonsen, Lisa  
Center Contact:  
lisa.c.simonsen@nasa.gov 
Unique ID: 10855 
Solicitation / Funding Source: 2014-15 HERO NNJ14ZSA001N-RADIATION. Appendix D: Ground-Based Studies in Space Radiobiology 
Grant/Contract No.: NNX16AK20G 
Project Type: GROUND 
Flight Program:  
TechPort: No 
No. of Post Docs:
No. of PhD Candidates:
No. of Master's Candidates:  
No. of Bachelor's Candidates:  
No. of PhD Degrees:  
No. of Master's Degrees:  
No. of Bachelor's Degrees:  
Human Research Program Elements: (1) SR:Space Radiation
Human Research Program Risks: (1) Cardiovascular:Risk of Cardiovascular Adaptations Contributing to Adverse Mission Performance and Health Outcomes
Human Research Program Gaps: (1) CV-102:Determine whether space radiation induces cardiovascular structural and functional changes and/or oxidative stress & damage (OSaD)/inflammation, that can contribute to development of disease.
Flight Assignment/Project Notes: NOTE: End date changed to 5/11/2022 per L. Barnes-Moten/JSC and NSSC information (Ed., 5/10/21)

NOTE: End date changed to 5/11/2021 per NSSC information (Ed., 8/12/20)

Task Description: Radiation damage and the cell’s attempt to repair it triggers a myriad of signal transduction pathways which alter gene, and ultimately, protein expression. Space radiation may affect biomolecules, cellular processes, and ultimately the cellular protein content (the proteome) differently than radiation present on Earth. Epidemiological analysis of terrestrial radiation exposure indicates that single high- or multiple low-dose radiation exposure can culminate in a wide array of cardiac injury and malfunction over time. Based on terrestrial data, it is believed that cardiovascular disorders may develop in astronauts from exposure to the space radiation environment. Indeed, a recent study by Yan et al. (2014), found that a single full body exposure to a low dose of proton or iron particle radiation, which somewhat mimics the space radiation environment, was sufficient to induce a significant, long term, negative effect on murine cardiovascular function. In this proposal, we take advantage of our expertise with bioinformatics analysis of cardiovascular proteomic data sets and murine cardiovascular physiology to evaluate the consequences of low dose, chronic space radiation, or mixed field space radiation on the dynamics of the cardiac proteome and to understand how the radiation induced changes relate to cardiovascular function. In doing so, we will extend Yan et al.’s work by identifying a proteomic signature that predicts the development of permanent cardiovascular degeneration from a single low dose space radiation exposure. Further, we seek to evaluate whether the proteomic signatures differ when mice experience repeated exposures of space-like radiation or mixed field space radiation. This information will lead to a mechanistic understanding of the altered cellular and molecular processes contributing to the development of cardiovascular dysfunction at the organ and organismal level in scenarios better mimicking the space radiation environment. This information is needed to predict, monitor, and prevent cardiac damage during long term space flight.

Reference: Yan, X., et al., Cardiovascular risks associated with low dose ionizing particle radiation. PLoS One, 2014. 9(10): p. e110269.

Research Impact/Earth Benefits: Limited information is known regarding the impact of chronic low level radiation on cardiovascular molecular biology and function both terrestrially and during extended space exploration. Our research is expected to provide information in regards to terrestrial and astronaut heath. Innovative technologies that may arise from our studies may include novel biomarkers predictive of cardiovascular susceptibility to chronic low level radiation as well as countermeasures that may be employed both on Earth as well as during space exploration.

Task Progress & Bibliography Information FY2020 
Task Progress: The long-term objective of this study is to identify molecular mechanism(s) underlying space radiation induced cardiovascular dysfunction. Our hypothesis is that the onset and magnitude of cardiac dysfunction (phenome) can be predicted by cardiac proteomic signatures arising from injury produced by distinct space radiation exposures. To address this hypothesis, we will define the cardiac phenome-proteome after iron, oxygen, and gamma radiation exposure.

Five month old male C57Bl6 mice (Jackson Laboratories) underwent transthoracic echocardiograms at Duke University Medical Center to establish baseline cardiac function. Subsequently, mice were subjected to single fully body irradiation at Brookhaven National Laboratories (BNL) under the following conditions: a) gamma (50-400cGy), b) 16O (15-150cGy/ 600 MeV/n), c) 56Fe (15-150cGy 1 GeV/n), d) Galactic Cosmic Radiation (GCR) (150cGy). All radiation groups included sham irradiated control animals. A subset of these animals underwent comprehensive cardiac structural and functional evaluation at 9-12 months post radiation including a) serial transthoracic echocardiograms capturing systolic and diastolic parameters, b) terminal pressure volume PV loop hemodynamic assessments, c) cardiac MRI, (d) blood pressure assessment, and e) cardiac and aortic tissue histology and immunohistochemistry. All echocardiograms were performed on awake animals, and were independently interpreted by two clinicians in a blinded fashion. Cardiac MRIs were performed using a 7.0 T Bruker Biospec small animal MRI scanner. Approximately 1000 mice were irradiated during five different BNL campaigns.

Non-invasive echocardiography and cardiac MRI did not demonstrate gross differences on systolic and diastolic function in GCR treated mice in comparison to controls. However, invasive pressure-volume loop hemodynamic analyses demonstrated that GCR treated mice, in comparison to controls, have a significant reduction in load independent (Preload Recruitable Stroke Work) and load dependent measures of cardiac systolic function (Cardiac Output and Stroke Volume). The differences in systolic function are partly explained by GCR induced increases in arterial elastance, suggesting that GCR exposure increases vascular resistance and afterload. The rise in afterload is associated with elastic fiber thickening, degeneration and disruption in aortic tissue, identified by histology that is present in GCR irradiated mice and absent in age-matched controls.

Quantitative mass spectrometry was performed on peptides (total proteome) as well as phosphopeptides (phosphoproteome) obtained from digested protein homogenates from the hearts of male C57B6 mice that had undergone radiation exposure at the NASA Radiation Science Laboratory at Brookhaven National Laboratories. These mice were subjected to single fully body irradiation at 6 months of age under the following conditions: a) 56Fe (50 cGy; n=3; obtained 3 month post radiation, b) 16O (50 cGy 600 MeV/n; n=3; obtained 7 months post radiation), c) 56Fe (50 cGy 1 GeV/n; n=3; obtained 7 months post radiation), and d) GCR (150 cGy, n=6; obtained 8 months post radiation). All radiation groups included sham irradiated control animals. Bioinformatics analysis of mass spectrometry data sets include: a) identification of significantly expressed proteins for each does and radiation types compared to corresponding control samples, (b) pathway enrichment analysis using functional annotation data obtained from Gene Ontology, KEGG (Kyoto Encyclopedia of Genes and Genomes), Reactome, and Molecular Signature Database (MSigDB), and c) identification of protein and pathway interaction modules that distinguish molecular signature of the various radiation types.

From the hearts of gamma, 56Fe, and 16O irradiated mice, 4,381 proteins and 5435 phosphopeptides were quantified. Similarly, from hearts from GCR irradiated mice, 4,650 proteins and 7,002 phosphopeptides (mapping to 303 phosphoproteins) were quantified. Pathways commonly influenced by all radiation types evaluated include mitochondrial gene expression and translation and fatty acid catabolic processes and oxidation. Pathways uniquely modulated by GCR involve cornification and keratinization.

Our data suggests that, in contrast to other forms of radiation, a single exposure to GCR increases afterload by injuring aortic architecture, and causes pronounced defects in load dependent cardiac systolic function. The cornification and keratinization processes observed in heart tissue from GCR irradiated animals may be contributing to the cardiovascular functional decline observed by hemodynamic and MRI analyses of these mice. Further characterization of the proteins involved in these processes may serve as targets of GCR countermeasures.

Bibliography: Description: (Last Updated: 07/11/2023) 

Show Cumulative Bibliography
 
Articles in Peer-reviewed Journals Brown ZD, Bishawi M, Roan J-N, Lee F, Nevo A, Watson M, Bowles DE. "The use of an inexpensive Processing Aid Device (the Mouse PAD) to facilitate rodent tissue banking." Biotechniques. 2020 Jul;69(1):364-8. Epub 2020 May 18. https://doi.org/10.2144/btn-2019-0069 ; PMID: 32418443 , Jul-2020
Project Title:  Proteomic Signatures of Space Radiation Induced Cardiovascular Degeneration Reduce
Images: icon  Fiscal Year: FY 2019 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 05/12/2016  
End Date: 05/11/2020  
Task Last Updated: 03/13/2019 
Download report in PDF pdf
Principal Investigator/Affiliation:   Bowles, Dawn  Ph.D. / Duke University 
Address:  Department of Surgery 
Msrb1 Room 401B, DUMC 2642 
Durham , NC 27710-0001 
Email: dawn.bowles@duke.edu 
Phone: 919-668-1947  
Congressional District:
Web:  
Organization Type: UNIVERSITY 
Organization Name: Duke University 
Joint Agency:  
Comments:  
Co-Investigator(s)
Affiliation: 
Abraham, Dennis  M.D. Duke University 
Kidane, Yared  Ph.D. Wyle Laboratories, Inc. 
Mao, Lan  M.D. Duke University 
Dewhirst, Mark  D.V.M., Ph.D. Duke University 
Moseley, Martin  Ph.D. Duke University 
Project Information: Grant/Contract No. NNX16AK20G 
Responsible Center: NASA JSC 
Grant Monitor: Simonsen, Lisa  
Center Contact:  
lisa.c.simonsen@nasa.gov 
Unique ID: 10855 
Solicitation / Funding Source: 2014-15 HERO NNJ14ZSA001N-RADIATION. Appendix D: Ground-Based Studies in Space Radiobiology 
Grant/Contract No.: NNX16AK20G 
Project Type: GROUND 
Flight Program:  
TechPort: No 
No. of Post Docs:
No. of PhD Candidates:
No. of Master's Candidates:  
No. of Bachelor's Candidates:  
No. of PhD Degrees:  
No. of Master's Degrees:  
No. of Bachelor's Degrees:  
Human Research Program Elements: (1) SR:Space Radiation
Human Research Program Risks: (1) Cardiovascular:Risk of Cardiovascular Adaptations Contributing to Adverse Mission Performance and Health Outcomes
Human Research Program Gaps: (1) CV-102:Determine whether space radiation induces cardiovascular structural and functional changes and/or oxidative stress & damage (OSaD)/inflammation, that can contribute to development of disease.
Task Description: Radiation damage and the cell’s attempt to repair it triggers a myriad of signal transduction pathways which alter gene, and ultimately, protein expression. Space radiation may affect biomolecules, cellular processes, and ultimately the cellular protein content (the proteome) differently than radiation present on Earth. Epidemiological analysis of terrestrial radiation exposure indicates that single high- or multiple low-dose radiation exposure can culminate in a wide array of cardiac injury and malfunction over time. Based on terrestrial data, it is believed that cardiovascular disorders may develop in astronauts from exposure to the space radiation environment. Indeed, a recent study by Yan et al. (2014), found that a single full body exposure to a low dose of proton or iron particle radiation, which somewhat mimics the space radiation environment, was sufficient to induce a significant, long term, negative effect on murine cardiovascular function. In this proposal, we take advantage of our expertise with bioinformatics analysis of cardiovascular proteomic data sets and murine cardiovascular physiology to evaluate the consequences of low dose, chronic space radiation, or mixed field space radiation on the dynamics of the cardiac proteome and to understand how the radiation induced changes relate to cardiovascular function. In doing so, we will extend Yan et al.’s work by identifying a proteomic signature that predicts the development of permanent cardiovascular degeneration from a single low dose space radiation exposure. Further, we seek to evaluate whether the proteomic signatures differ when mice experience repeated exposures of space-like radiation or mixed field space radiation. This information will lead to a mechanistic understanding of the altered cellular and molecular processes contributing to the development of cardiovascular dysfunction at the organ and organismal level in scenarios better mimicking the space radiation environment. This information is needed to predict, monitor, and prevent cardiac damage during long term space flight.

Reference: Yan, X., et al., Cardiovascular risks associated with low dose ionizing particle radiation. PLoS One, 2014. 9(10): p. e110269.

Research Impact/Earth Benefits: Limited information is known regarding the impact of chronic low level radiation on cardiovascular molecular biology and function both terrestrially and during extended space exploration. Our research is expected to provide information in regards to terrestrial and astronaut heath. Innovative technologies that may arise from our studies may include novel biomarkers predictive of cardiovascular susceptibility to chronic low level radiation as well as countermeasures that may be employed both on Earth as well as during space exploration.

Task Progress & Bibliography Information FY2019 
Task Progress: Five trips to Brookhaven National Laboratory (BNL) for mice experimentation have been made for this grant (Fall 2016, Spring 2017, Fall 2017, Summer 2018, and Fall 2018). For Fall 2016 studies 110 Male C57B6 mice were acquired from Jackson Laboratories. These mice were shipped to Duke University Medical Center where at 5 months of age they underwent transthoracic echocardiograms to establish baseline cardiac function. Parameters evaluated included (a) M-mode (done in both long and short axis), (b) Septal and posterior wall width in diastole, (c) End diastolic dimension and end systolic dimension, (d) aortic valve velocity, and (e) aortic ejection time (all measured and averaged over 3 consecutive beats). Echocardiogram images were also acquired for measurement of diastolic dysfunction and strain.

Mice were shipped to the NASA Space Radiation Laboratory (NSRL) at Brookhaven National Laboratories where they were subjected to single full body irradiation at 6 months of age under the following conditions: a) gamma (50cGy, 100cGy, 200cGy), b) 16O (15cGy, 25cGy, 50cGy/ 600 MeV/n), c) 56Fe (15cGy, 25cGy, 50cGy 1 GeV/n). There were a total of 20 sham irradiated control animals who traveled to NSRL. Evaluations include: a) serial transthoracic echocardiograms capturing all above parameters, b) terminal pressure volume loop hemodynamic assessments. Overall conclusions from the Fall 2016 experiment were that no significant changes in any echo measurement at any dose or rad type compared to control at any time point were observed. However across all groups including controls aging related decrements in function were observed as expected. Caveats were that this was essentially a pilot study (n=10 per group). There was also excessive early fighting among mice. The study was terminated at 9M post rad with the exception of a small group of animals which were held to 1 year post radiation when pressure volume (PV) loops were obtained. There was no significant difference in invasive hemodynamic measurements at 1 year post radiation. Again, small # of animals was examined in this manner.

A larger group of mice were evaluated in Spring 2018. Energies and doses are the following: Gamma (50cGy, 100cGy, 200cGy), 16Ox 600 MeV/n (15cGy, 25cGy, 50cGy), 56Fe 1GeV/n (15cGy, 25cGy, 50cGy). There was a larger number of animals per group in this experiment. Echocardiographic images have been obtained for each of the animals (if they were alive at the time of Echo acquisition) at the following time points: Pre-irradiation, 1 week post, 1M post, 2M post, 3M post, 4M post, 7M post, 9M post. Pressure volume (PV) loops were acquired on 30 animals 9 M post radiation. At the time of experiment termination, multiple organs were processed and biobanked for ‘omics analyses and future collaboration.

Overall, the echo reads for Spring 2017 are ongoing. We had a computer hard drive malfunction and lost a large amount of analyzed data. Therefore, no definite conclusions can be made yet with this larger cohort of animals. The PV loop analysis focused on 56Fe 50cGy, 1 year post IR with 10-15 per group. No statistically significant differences were observed in these measurements. However, a trend in several measurements was observed which suggests the possibility of diastolic dysfunction development. Additional PV loops have been done from animals in Fall 2017 experiment but have yet to be analyzed. Between Fall 2016 and Spring 2017 experiments we have developed an extensive biorepository of tissues for molecular analyses and for collaborations.

For the NSRL Fall 2017 experiment the same energies and doses from the two previous experiments were used. Echocardiographic images have been obtained for each of the animals at the following time points: Pre-irradiation, 1M post, 2M post, 3M post, 4M post, 7M post, 9M post, 16M post. PV loops were acquired on 97 animals 16M post radiation. All of these echos and PV loops are yet to be analyzed. At the time of animal sacrifice, multiple organs were processed and biobanked for ‘omics analyses and future collaboration. A subset of animals were AHA labeled for mass spec proteomics analyses. Brain regions from a subset of animals (59) were processed by Andrew Wryobek for funded tissue sharing FLAGSHIP grant in March 2019. NSRL Summer 2018 study examined GCR (galactic comic radiation) sim 5 ion (150 cGy). Echocardiographic images have been obtained for each of the animals at the following time points: Pre-irradiation, 1M post, 3M post, 7M post, 8M post. Echos are planned for 11 and 17M post irradiation in May 2019 and Nov 2019, respectively. PV loops are planned for November 2019. 12 animals have been labeled with AHA and submitted to Duke Proteomics Core for dynamic, abundance, and phosphoproteomics analyses. Echos are currently being read for this cohort. Data is still preliminary.

Outline of the study design for the Group 5 mice irradiated at NSRL Fall 2018. Energies and doses are the following: Gamma (300cGy, 400cGy), 16Ox (75cGy, 150cGy) 600 MeV/n, 56Fe (75cGy, 150cGy) 1 GeV/n, GCR sim 5 ion (150 cGy). Echocardiographic images have been obtained for each of the animals (if they were alive at the time of Echo acquisition) at the following time points: Pre-irradiation, 2M post, 3M post, 4M post. Echos are planned 7M, 9M, 12M, and 18M post irradiation. PV loops are planned for May 2019.

Bibliography: Description: (Last Updated: 07/11/2023) 

Show Cumulative Bibliography
 
Articles in Peer-reviewed Journals Brown ZD, Bishawi M, Bowles DE. "Using proteomics approaches to understand mechanisms underlying low LET or GCR radiation-induced cardiovascular disease." THREE. 2018 May 15. https://three.jsc.nasa.gov/articles/Proteomics_Bowles.pdf , May-2018
Project Title:  Proteomic Signatures of Space Radiation Induced Cardiovascular Degeneration Reduce
Images: icon  Fiscal Year: FY 2018 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 05/12/2016  
End Date: 05/11/2020  
Task Last Updated: 03/13/2018 
Download report in PDF pdf
Principal Investigator/Affiliation:   Bowles, Dawn  Ph.D. / Duke University 
Address:  Department of Surgery 
Msrb1 Room 401B, DUMC 2642 
Durham , NC 27710-0001 
Email: dawn.bowles@duke.edu 
Phone: 919-668-1947  
Congressional District:
Web:  
Organization Type: UNIVERSITY 
Organization Name: Duke University 
Joint Agency:  
Comments:  
Co-Investigator(s)
Affiliation: 
Abraham, Dennis  M.D. Duke University 
Kidane, Yared  Ph.D. Wyle Laboratories, Inc. 
Mao, Lan  M.D. Duke University 
Dewhirst, Mark  D.V.M., Ph.D. Duke University 
Moseley, Martin  Ph.D. Duke University 
Project Information: Grant/Contract No. NNX16AK20G 
Responsible Center: NASA JSC 
Grant Monitor: Simonsen, Lisa  
Center Contact:  
lisa.c.simonsen@nasa.gov 
Unique ID: 10855 
Solicitation / Funding Source: 2014-15 HERO NNJ14ZSA001N-RADIATION. Appendix D: Ground-Based Studies in Space Radiobiology 
Grant/Contract No.: NNX16AK20G 
Project Type: GROUND 
Flight Program:  
TechPort: No 
No. of Post Docs:  
No. of PhD Candidates:
No. of Master's Candidates:  
No. of Bachelor's Candidates:  
No. of PhD Degrees:  
No. of Master's Degrees:  
No. of Bachelor's Degrees:  
Human Research Program Elements: (1) SR:Space Radiation
Human Research Program Risks: (1) Cardiovascular:Risk of Cardiovascular Adaptations Contributing to Adverse Mission Performance and Health Outcomes
Human Research Program Gaps: (1) CV-102:Determine whether space radiation induces cardiovascular structural and functional changes and/or oxidative stress & damage (OSaD)/inflammation, that can contribute to development of disease.
Task Description: Radiation damage and the cell’s attempt to repair it triggers a myriad of signal transduction pathways which alter gene, and ultimately, protein expression. Space radiation may affect biomolecules, cellular processes, and ultimately the cellular protein content (the proteome) differently than radiation present on Earth. Epidemiological analysis of terrestrial radiation exposure indicates that single high- or multiple low-dose radiation exposure can culminate in a wide array of cardiac injury and malfunction over time. Based on terrestrial data, it is believed that cardiovascular disorders may develop in astronauts from exposure to the space radiation environment. Indeed, a recent study by Yan et al. (2014), found that a single full body exposure to a low dose of proton or iron particle radiation, which somewhat mimics the space radiation environment, was sufficient to induce a significant, long term, negative effect on murine cardiovascular function. In this proposal, we take advantage of our expertise with bioinformatics analysis of cardiovascular proteomic data sets and murine cardiovascular physiology to evaluate the consequences of low dose, chronic space radiation, or mixed field space radiation on the dynamics of the cardiac proteome and to understand how the radiation induced changes relate to cardiovascular function. In doing so, we will extend Yan et al.’s work by identifying a proteomic signature that predicts the development of permanent cardiovascular degeneration from a single low dose space radiation exposure. Further, we seek to evaluate whether the proteomic signatures differ when mice experience repeated exposures of space-like radiation or mixed field space radiation. This information will lead to a mechanistic understanding of the altered cellular and molecular processes contributing to the development of cardiovascular dysfunction at the organ and organismal level in scenarios better mimicking the space radiation environment. This information is needed to predict, monitor, and prevent cardiac damage during long term space flight.

Yan, X., et al., Cardiovascular risks associated with low dose ionizing particle radiation. PLoS One, 2014. 9(10): p. e110269.

Research Impact/Earth Benefits: Limited information is known regarding the impact of chronic low level radiation on cardiovascular molecular biology and function both terrestrially and during extended space exploration. Our research is expected to provide information in regards to terrestrial and astronaut heath. Innovative technologies that may arise from our studies may include novel biomarkers predictive of cardiovascular susceptibility to chronic low level radiation as well as countermeasures that may be employed both on Earth as well as during space exploration.

Task Progress & Bibliography Information FY2018 
Task Progress: Five trips to Brookhaven National Laboratory (BNL) for mice experimentation have been planned for this grant (Fall 2016, Spring 2017, Fall 2017, Summer 2018, and Fall 2018). Thus far three trips have occurred (Fall 2016, Spring 2017, and Fall 2017).

Male C57B6 mice are purchased from Jackson Laboratories. Mice are shipped to Duke University Medical Center where at 5 months of age they undergo transthoracic echocardiograms to establish baseline cardiac function (pre IR echo). Parameters evaluated included (a) M-mode (done in both long and short axis), (b) Septal and posterior wall width in diastole, (c) End diastolic dimension and end systolic dimension, (d) aortic valve velocity, and (e) aortic ejection time (all measured and averaged over 3 consecutive beats). Echocardiogram images acquired are also evaluated for measurement of diastolic dysfunction and strain.

Mice are then shipped to the NASA Space Radiation Laboratory (NSRL) at BNL where they are subjected to single full body irradiation at 6 months of age under the following conditions: a) gamma (50cGy, 100cGy, 200cGy), b) 16O (15cGy, 25cGy, 50cGy/ 600 MeV/n), c) 56Fe (15cGy, 25cGy, 50cGy 1 GeV/n). Control (sham irradiated) animals also travel back and forth from Duke to NRSL and back to Duke in order to experience identical stressors as the irradiated mice. Evaluations include: a) serial transthoracic echocardiograms capturing all above parameters. These are done at 1, 2, 3, 7, and 9 months post irradiation; b) terminal pressure volume loop hemodynamic assessments; and c) mass spectrometry based proteomics assessments (quantitative, dynamic, and post-translational modification proteomics) of the cardiac proteome.

All studies are ongoing. Therefore, the data set is still preliminary and conclusions presented in this report should be considered preliminary.

To date, we have not observed a significant difference in any parameter evaluated that allows us to make a statement that cardiovascular function is affected at any time point, radiation type, or dose post irradiation compared to control sham irradiated control mice.

Bibliography: Description: (Last Updated: 07/11/2023) 

Show Cumulative Bibliography
 
Abstracts for Journals and Proceedings Bishawi M, Brown Z, Roan J, Bishawi M, Lee F, Abraham D, Isaac D, Mao L, Slaba T, Kidane Y, Thompson JW, Moseley MA, Truskey G, Dewhirst MW, Bowles DE. "Proteomic Signatures of Space Radiation Induced Cardiovascular Degeneration." 2018 NASA Human Research Program Investigators’ Workshop, Galveston, TX, January 22-25, 2018.

2018 NASA Human Research Program Investigators’ Workshop, Galveston, TX, January 22-25, 2018. , Jan-2018

Abstracts for Journals and Proceedings Brown ZD, Bishawi M, Feger BJ, Carnell LS, Blattnig S, Bowles DE. "A Systematic Literature Review of Radiation-Induced Cardiovascular Disease." 2018 NASA Human Research Program Investigators’ Workshop, Galveston, TX, January 22-25, 2018.

2018 NASA Human Research Program Investigators’ Workshop, Galveston, TX, January 22-25, 2018. , Jan-2018

Project Title:  Proteomic Signatures of Space Radiation Induced Cardiovascular Degeneration Reduce
Images: icon  Fiscal Year: FY 2017 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 05/12/2016  
End Date: 05/11/2020  
Task Last Updated: 09/26/2017 
Download report in PDF pdf
Principal Investigator/Affiliation:   Bowles, Dawn  Ph.D. / Duke University 
Address:  Department of Surgery 
Msrb1 Room 401B, DUMC 2642 
Durham , NC 27710-0001 
Email: dawn.bowles@duke.edu 
Phone: 919-668-1947  
Congressional District:
Web:  
Organization Type: UNIVERSITY 
Organization Name: Duke University 
Joint Agency:  
Comments:  
Co-Investigator(s)
Affiliation: 
Abraham, Dennis  M.D. Duke University 
Kidane, Yared  Ph.D. Wyle Laboratories, Inc. 
Mao, Lan  M.D. Duke University 
Dewhirst, Mark  D.V.M., Ph.D. Duke University 
Moseley, Martin  Ph.D. Duke University 
Project Information: Grant/Contract No. NNX16AK20G 
Responsible Center: NASA JSC 
Grant Monitor: Simonsen, Lisa  
Center Contact:  
lisa.c.simonsen@nasa.gov 
Unique ID: 10855 
Solicitation / Funding Source: 2014-15 HERO NNJ14ZSA001N-RADIATION. Appendix D: Ground-Based Studies in Space Radiobiology 
Grant/Contract No.: NNX16AK20G 
Project Type: GROUND 
Flight Program:  
TechPort: No 
No. of Post Docs:  
No. of PhD Candidates:
No. of Master's Candidates:  
No. of Bachelor's Candidates:  
No. of PhD Degrees:  
No. of Master's Degrees:  
No. of Bachelor's Degrees:  
Human Research Program Elements: (1) SR:Space Radiation
Human Research Program Risks: (1) Cardiovascular:Risk of Cardiovascular Adaptations Contributing to Adverse Mission Performance and Health Outcomes
Human Research Program Gaps: (1) CV-102:Determine whether space radiation induces cardiovascular structural and functional changes and/or oxidative stress & damage (OSaD)/inflammation, that can contribute to development of disease.
Task Description: Radiation damage and the cell’s attempt to repair it triggers a myriad of signal transduction pathways which alter gene, and ultimately, protein expression. Space radiation may affect biomolecules, cellular processes, and ultimately the cellular protein content (the proteome) differently than radiation present on Earth. Epidemiological analysis of terrestrial radiation exposure indicates that single high- or multiple low-dose radiation exposure can culminate in a wide array of cardiac injury and malfunction over time. Based on terrestrial data, it is believed that cardiovascular disorders may develop in astronauts from exposure to the space radiation environment. Indeed, a recent study by Yan et al. (2014), found that a single full body exposure to a low dose of proton or iron particle radiation, which somewhat mimics the space radiation environment, was sufficient to induce a significant, long term, negative effect on murine cardiovascular function. In this proposal, we take advantage of our expertise with bioinformatics analysis of cardiovascular proteomic data sets and murine cardiovascular physiology to evaluate the consequences of low dose, chronic space radiation, or mixed field space radiation on the dynamics of the cardiac proteome and to understand how the radiation induced changes relate to cardiovascular function. In doing so, we will extend Yan et al.’s work by identifying a proteomic signature that predicts the development of permanent cardiovascular degeneration from a single low dose space radiation exposure. Further, we seek to evaluate whether the proteomic signatures differ when mice experience repeated exposures of space-like radiation or mixed field space radiation. This information will lead to a mechanistic understanding of the altered cellular and molecular processes contributing to the development of cardiovascular dysfunction at the organ and organismal level in scenarios better mimicking the space radiation environment. This information is needed to predict, monitor, and prevent cardiac damage during long term space flight.

Yan, X., et al., Cardiovascular risks associated with low dose ionizing particle radiation. PLoS One, 2014. 9(10): p. e110269.

Research Impact/Earth Benefits: Limited information is known regarding the impact of chronic low level radiation on cardiovascular molecular biology and function both terrestrially and during extended space exploration. Our research is expected to provide information in regards to terrestrial and astronaut heath. Innovative technologies that may arise from our studies may include novel biomarkers predictive of cardiovascular susceptibility to chronic low level radiation as well as countermeasures that may be employed both on Earth as well as during space exploration.

Task Progress & Bibliography Information FY2017 
Task Progress: Studies are ongoing. One trip to Brookhaven National Laboratory (BNL) was made in Nov 2016. Male C57B6 mice acquired from Jackson Laboratories were shipped to Duke University Medical Center where at 5 months of age they underwent transthoracic echocardiograms to establish baseline cardiac function. Parameters evaluated included (a) M-mode (done in both long and short axis), (b) Septal and posterior wall width in diastole, (c) End diastolic dimension and end systolic dimension, (d) aortic valve velocity, and (e) aortic ejection time (all measured and averaged over 3 consecutive beats). Echocardiogram images were also acquired for measurement of diastolic dysfunction and strain. Mice were shipped to the NASA Radiation Science Laboratory at Brookhaven National Laboratories where they were subjected to single fully body irradiation at 6 months of age under the following conditions: a) gamma (50cGy, 100cGy, 200cGy), b) 16O (15cGy, 25cGy, 50cGy/ 600 MeV/n), c) 56Fe (15cGy, 25cGy, 50cGy 1 GeV/n). All radiation groups included sham irradiated control animals. Studies of these mice are ongoing. Evaluations include: a) serial transthoracic echocardiograms capturing all above parameters, b) terminal pressure volume loop hemodynamic assessments, and c) mass spectrometry based proteomics assessments (quantitative, dynamic, and post-translational modification proteomics) of the cardiac proteome.

A second trip to BNL will occur in April 2017. A larger group of mice will be administered the same radiation exposures. The exposures will be single fully body irradiation at 6 months of age under the following conditions: a) gamma (50cGy, 100cGy, 200cGy), b) 16O (15cGy, 25cGy, 50cGy/ 600 MeV/n), c) 56Fe (15cGy, 25cGy, 50cGy 1 GeV/n). All radiation groups include sham irradiated control animals. All animals will be monitored as described above. Evaluations include: a) serial transthoracic echocardiograms capturing all above parameters, b) terminal pressure volume loop hemodynamic assessments, and c) mass spectrometry based proteomics assessments (quantitative, dynamic, and post-translational modification proteomics) of the cardiac proteome.

Bibliography: Description: (Last Updated: 07/11/2023) 

Show Cumulative Bibliography
 
Abstracts for Journals and Proceedings Bishawi M, Isaac D, Abraham D, Mao L, Slaba T, Kidane Y, Kuchibhatla M, Thompson JS, Moseley MA, Dewhirst MW, Bowles DE. "Proteomic signatures of space radiation induced cardiovascular degeneration." Poster presentation at 2017 NASA Human Research Program Investigators’ Workshop, Galveston, TX, January 23-26, 2017.

2017 NASA Human Research Program Investigators’ Workshop, Galveston, TX, January 23-26, 2017. , Feb-2017

Abstracts for Journals and Proceedings Isaac D, Bishawi M, Mishra R, Watson M, Slaba T, Dewhirst MW, Joshi M, Wu H, Bowles DE. "Tissue biobanking to understand molecular signatures of space radiation induced tissue degeneration." Oral Presentation at ISBER 2017 Meeting, Toronto, Canada, May 9-12, 2017.

ISBER 2017 Meeting, Toronto, Canada, May 9-12, 2017. , May-2017

Project Title:  Proteomic Signatures of Space Radiation Induced Cardiovascular Degeneration Reduce
Images: icon  Fiscal Year: FY 2016 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 05/12/2016  
End Date: 05/11/2020  
Task Last Updated: 06/22/2016 
Download report in PDF pdf
Principal Investigator/Affiliation:   Bowles, Dawn  Ph.D. / Duke University 
Address:  Department of Surgery 
Msrb1 Room 401B, DUMC 2642 
Durham , NC 27710-0001 
Email: dawn.bowles@duke.edu 
Phone: 919-668-1947  
Congressional District:
Web:  
Organization Type: UNIVERSITY 
Organization Name: Duke University 
Joint Agency:  
Comments:  
Co-Investigator(s)
Affiliation: 
Abraham, Dennis  M.D. Duke University 
Kidane, Yared  Ph.D. Wyle Laboratories, Inc. 
Mao, Lan  M.D. Duke University 
Dewhirst, Mark  D.V.M., Ph.D. Duke University 
Moseley, Martin  Ph.D. Duke University 
Project Information: Grant/Contract No. NNX16AK20G 
Responsible Center: NASA JSC 
Grant Monitor: Simonsen, Lisa  
Center Contact:  
lisa.c.simonsen@nasa.gov 
Unique ID: 10855 
Solicitation / Funding Source: 2014-15 HERO NNJ14ZSA001N-RADIATION. Appendix D: Ground-Based Studies in Space Radiobiology 
Grant/Contract No.: NNX16AK20G 
Project Type: GROUND 
Flight Program:  
TechPort: No 
No. of Post Docs:  
No. of PhD Candidates:  
No. of Master's Candidates:  
No. of Bachelor's Candidates:  
No. of PhD Degrees:  
No. of Master's Degrees:  
No. of Bachelor's Degrees:  
Human Research Program Elements: (1) SR:Space Radiation
Human Research Program Risks: (1) Cardiovascular:Risk of Cardiovascular Adaptations Contributing to Adverse Mission Performance and Health Outcomes
Human Research Program Gaps: (1) CV-102:Determine whether space radiation induces cardiovascular structural and functional changes and/or oxidative stress & damage (OSaD)/inflammation, that can contribute to development of disease.
Task Description: Radiation damage and the cell’s attempt to repair it triggers a myriad of signal transduction pathways which alter gene, and ultimately, protein expression. Space radiation may affect biomolecules, cellular processes, and ultimately the cellular protein content (the proteome) differently than radiation present on Earth. Epidemiological analysis of terrestrial radiation exposure indicates that single high- or multiple low-dose radiation exposure can culminate in a wide array of cardiac injury and malfunction over time. Based on terrestrial data, it is believed that cardiovascular disorders may develop in astronauts from exposure to the space radiation environment. Indeed, a recent study by Yan et al. (2014), found that a single full body exposure to a low dose of proton or iron particle radiation, which somewhat mimics the space radiation environment, was sufficient to induce a significant, long term, negative effect on murine cardiovascular function. In this proposal, we take advantage of our expertise with bioinformatics analysis of cardiovascular proteomic data sets and murine cardiovascular physiology to evaluate the consequences of low dose, chronic space radiation, or mixed field space radiation on the dynamics of the cardiac proteome and to understand how the radiation induced changes relate to cardiovascular function. In doing so, we will extend Yan et al.’s work by identifying a proteomic signature that predicts the development of permanent cardiovascular degeneration from a single low dose space radiation exposure. Further, we seek to evaluate whether the proteomic signatures differ when mice experience repeated exposures of space-like radiation or mixed field space radiation. This information will lead to a mechanistic understanding of the altered cellular and molecular processes contributing to the development of cardiovascular dysfunction at the organ and organismal level in scenarios better mimicking the space radiation environment. This information is needed to predict, monitor, and prevent cardiac damage during long term space flight.

Yan, X., et al., Cardiovascular risks associated with low dose ionizing particle radiation. PLoS One, 2014. 9(10): p. e110269.

Research Impact/Earth Benefits:

Task Progress & Bibliography Information FY2016 
Task Progress: New project for FY2016.

Bibliography: Description: (Last Updated: 07/11/2023) 

Show Cumulative Bibliography
 
 None in FY 2016