This website could be intermittent Saturday Mar 30, 2024 starting 7PM until next day 11AM Eastern Time due to server/facility maintenance. We apologize for any inconvenience.

 

Menu

 

The NASA Task Book
Advanced Search     

Project Title:  NSCOR: NASA Specialized Center of Research on Radiation Carcinogenesis Reduce
Fiscal Year: FY 2015 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 06/01/2009  
End Date: 09/30/2015  
Task Last Updated: 02/16/2017 
Download report in PDF pdf
Principal Investigator/Affiliation:   Ullrich, Robert  Ph.D. / University of Texas Medical Branch 
Address:  301 University Blvd 
Comprehensive Cancer Center, MS 1048 
Galveston , TX 77555-5302 
Email: bullrich@utmb.edu 
Phone: 409-747-1935  
Congressional District: 14 
Web:  
Organization Type: UNIVERSITY 
Organization Name: University of Texas Medical Branch 
Joint Agency:  
Comments: NOTE: PI moved to UTMB from Colorado State University in late 2008 (6/2009) 
Co-Investigator(s)
Affiliation: 
Le Beau, Michelle  University of Chicago 
Bacher, Jeff  Promega Corporation 
Yu, Yongjia  University of Texas Medical Branch 
Story, Michael  University of Texas Southwestern Medical Center at Dallas 
Bedford, Joel  Colorado State University 
Weil, Michael  Colorado State University 
Ray, F  Colorado State University 
Ding, Lianghao  University of Texas Southwestern Medical Center at Dallas 
Xie, Yang  University of Texas Southwestern Medical Center 
Key Personnel Changes / Previous PI: none
Project Information: Grant/Contract No. NNX09AM08G 
Responsible Center: NASA JSC 
Grant Monitor: Simonsen, Lisa  
Center Contact:  
lisa.c.simonsen@nasa.gov 
Unique ID: 7564 
Solicitation / Funding Source: 2008 NSCOR Space Radiation NNJ08ZSA003N 
Grant/Contract No.: NNX09AM08G 
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) Cancer:Risk of Radiation Carcinogenesis
Human Research Program Gaps: (1) Cancer-202:Evaluate the contribution of genetic background/diversity on carcinogenesis risk.
Flight Assignment/Project Notes: NOTE: End date changed to 9/30/2015 (from 5/31/2015) per NSSC information (Ed., 6/1/15)

NOTE: End date changed to 5/31/2015 per NSSC information (Ed., 3/3/14)

Task Description: The goal of this NASA Specialized Center of Research (NSCOR) is to provide the information required to develop a rational scientific basis for estimation of risks for carcinogenesis in humans from exposure to radiation during space flight. Previous results from this Program found an unexpectedly low RBE (relative biological effectiveness) value for acute myeloid leukemia (AML) induction by 1 GeV 56Fe ions. Systematic cytogenetic analyses suggested both microdosimetric factors related to the track structure of 1 GeV 56Fe ions and biological factors could account for this observation. In addition, these studies found an unexpected increase in hepatocellular carcinoma (HCC) at doses as low as 0.1 Gy of 1 GeV 56Fe ions but very little, if any, increase following gamma-ray exposure. These data suggest that processes associated with expansion and progression of initiated cells may play a more prominent role in HCC. If this is the case, it is possible that there are qualitative differences as well as quantitative in the effects of HZE (high energy) irradiations. To expand on these results and to address the overall goal of this NSCOR a series of coordinated activities will be conducted in 5 Projects and 3 Cores aimed at: (1) providing quantitative animal tumorigenesis data on the relative effectiveness of specific HZE particles and SPE (solar particle event) protons compared with gamma-rays in mouse models of AML and HCC; (2) providing a better understanding of the impact of radiation exposure on the processes involved in the initiation and in the progression of initiated cells toward the neoplastic phenotype; 3) delineating potential differences between low LET (linear energy transfer) radiation and high LET radiation such as those encountered in space travel on these processes; 4) developing links between animal data and radiation-induced effects for AML in humans; and (5) developing biologically-based modeling approaches which are critical to link these biological effects to risks in humans.

Program Overview: The Radiation Carcinogenesis NSCOR was initiated in June 2009 and builds upon results obtained in its predecessor, the Leukemogenesis NSCOR. The Radiation Carcinogenesis NSCOR consists of four projects supported by three cores. The projects and cores are briefly described below.

Project 1. Dose response relationships for induction of AML and HCC as a function of radiation quality (project leader, Dr. Robert. L. Ullrich). This project is designed to compare the effects of irradiation with gamma-rays, select HZE particles, and protons on the induction of AML and hepatocellular carcinoma (HCC) using the C3H murine model.

Project 2. Mechanisms of radiation leukemogenesis (project leader, Dr. Michael M. Weil). The goal of Project 2 is to better understand how radiation leads to AML in a murine model and to generate data for the development of a biologically based model that can be used to predict AML risks from various HZE or high energy proton exposures.

Project 3. Pathogenesis of radiation-induced hepatocellular carcinoma (project leader, Dr. Robert L. Ullrich). The overall hypothesis of this project is that the dose response is likely to reflect both quantitative as well as qualitative differences in high LET effects. This overall hypothesis will be tested in 3 specific aims:

1. Quantify the frequency and progression of preneoplastic foci (including both hyperplastic and dysplastic foci) in livers of C3H/HeNCrl mice irradiated with either 137Cs gamma-rays or HZE ions.

2. Examine irradiated liver for evidence of increased oxidative damage and alterations in the regulation of inflammatory processes.

3. Determine tumorigenic effects following HZE and gamma-ray irradiation in murine models of hepatocellular carcinoma in which secondary “promoting” events play a significant role.

Project 4. Molecular and cytogenetic targets in murine and human AML. (Project leader, Dr. Michelle Le Beau). This project is designed to develop a cytogenetic and molecular profile of human radiation-induced AML, leading to an understanding of the key events and genetic pathways involved in the pathogenesis of this disease.

Core A (Core Director, Dr. F. Andrew Ray). The Biology Core facilitates the distribution of irradiated and control animals, tissues, cells, and other biological samples to investigators. This core is also responsible for conducting the irradiations required at the various sites for all projects.

Core B (Core Director, Dr. Michael Story). The Genomics and Biostatistics core provides appropriate genomic analyses, innovative statistical modeling, simulations, and data analyses for the projects.

Core C (Core Director, Dr. Robert Ullrich). The Administrative Core provides administrative, fiscal, and management support for the Radiation Carcinogenesis NSCOR. This core also oversees the overall scientific conduct of the NSCOR and facilitates interactions between projects, core leaders, and project investigators as well as interactions with the internal and external advisors.

Research Impact/Earth Benefits: This work will provide basic information on mechanisms of carcinogenesis as well as mechanisms specific to radiation-induced cancer.

Task Progress & Bibliography Information FY2015 
Task Progress: Project 1. Dose response relationships for induction of AML and HCC as a function of radiation quality (project leader, Dr. Robert. L. Ullrich).

We completed analysis of the incidence of acute myeloid leukemia (AML) and hepatocellular carcinoma (HCC) as a function of dose in the gamma ray, 1972SPE proton, 600 MeV/n 56Fe, and 300 MeV/n 28Si groups and the data for the 600 MeV/n 56Fe and 300 MeV/n 28Si groups were published (Weil et al., 2014). While the dose responses for AML are slightly different than in our previous studies using 1 GeV/n 56Fe ions obtained in an earlier NSCOR, they confirm a relatively low RBE for induction of AML (<5). Data for HCC were also quite similar to our previous studies with 1 GeV/n 56Fe ions, confirming a very high RBE (>60) for induction of HCC.

In addition, we analyzed the frequency of metastases in each of these groups. There were significantly more metastases in the Si and Fe groups suggesting that in addition to qualitative effects, there are also qualitative differences in effects of Si and Fe ions when compared with gamma ray or protons. Studies examining effects of low dose rate protons and fractionated 600 MeV 56Fe ions on the induction of AML and HCC were also completed.

Project 2. Mechanisms of radiation leukemogenesis (project leader, Dr. Michael M. Weil).

Project 2 focused on unraveling the events that lead to radiation-induced leukemia (rAML) in a murine model. We took advantage of mice heterozygous for a Sfpi1 knockout allele on rAML susceptible and resistant backgrounds. The experimental designs and results are published in Genik et al., 2014. In the mouse, as in humans, rAML is associated with recurrent, large chromosomal deletions. In mice the radiation-induced deletion occurs on chromosome 2 and the critical gene in the deleted region is Sfpi1 (PU.1 in human nomenclature). The remaining allele of Sfpi1 is mutated in leukemias at codon 235, with a recurrent C to T transition resulting in R235C being the most common event. We found that this mutation is spontaneous, not radiation-induced. We also found that the Sfpi1 codon 235 mutation is not a rate limiting step in radiation leukemogenesis, suggesting it is present around the time of irradiation. In addition, we found evidence that loss of at least one other gene in the deleted region was required for leukemogenesis.

In further work, we found that the Sfpi1 codon 235 mutation does occur spontaneously in hematopoietic cells in unirradiated C3H mice that are susceptible to rAML, but not in C57BL/6 mice that are resistant. Furthermore, the frequency of the mutation is age dependent and corresponds to the age at exposure risk for rAML.

These results are significant to the NASA program because they support a hypothesis by Nori Nakamura (Radiat Res 2005; 163:258-265) that only some individuals are susceptible to radiation-induced leukemias, and those individuals carry cells with a pre-leukemic mutation at the time of radiation exposure. Radiation acts on the pre-leukemic cells by inducing additional mutations (e.g., a recurrent chromosomal deletion) that complete the transformation pathway. If the human correlates to the pre-leukemic mutation in Sfpi1 codon 235 and radiation-induced chromosome 2 deletion can be identified it will be possible to screen astronauts for pre-leukemic cells prior to missions, thus reducing their risk of leukemia. Based on recent results from large scale genome sequencing projects, the likely candidates are spontaneous TP53 mutations and deletions involving chromosomes 5 and/or 7.

[Ed. note 2/1/2017: compiled from NSCOR FY2015 final progress report]

Bibliography: Description: (Last Updated: 07/25/2021) 

Show Cumulative Bibliography
 
Articles in Peer-reviewed Journals Weil MM, Ray FA, Genik PC, Yu Y, McCarthy M, Fallgren CM, Ullrich RL. "Effects of 28Si ions, 56Fe ions, and protons on the induction of murine acute myeloid leukemia and hepatocellular carcinoma." PLoS One. 2014 Aug 15;9(7):e104819. eCollection 2014. http://dx.doi.org/10.1371/journal.pone.0104819 ; PubMed PMID: 25126721 ; PubMed Central PMCID: PMC4134239 , Aug-2014
Articles in Peer-reviewed Journals Kantara C, O'Connell MR, Luthra G, Gajjar A, Sarkar S, Ullrich RL, Singh P. "Methods for detecting circulating cancer stem cells (CCSCs) as a novel approach for diagnosis of colon cancer relapse/metastasis." Lab Invest. 2015 Jan;95(1):100-12. Epub 2014 Oct 27. http://dx.doi.org/10.1038/labinvest.2014.133 ; PubMed PMID: 25347154; PubMed Central PMCID: PMC4281282 , Jan-2015
Articles in Peer-reviewed Journals Christofidou-Solomidou M, Pietrofesa RA, Arguiri E, Schweitzer KS, Berdyshev EV, McCarthy M, Corbitt A, Alwood JS, Yu Y, Globus RK, Solomides CC, Ullrich RL, Petrache I. "Space radiation-associated lung injury in a murine model." Am J Physiol Lung Cell Mol Physiol. 2015 Mar 1;308(5):L416-28. Epub 2014 Dec 19. http://dx.doi.org/10.1152/ajplung.00260.2014 ; PubMed PMID: 25526737; PubMed Central PMCID: PMC4346772 , Mar-2015
Articles in Peer-reviewed Journals Barcellos-Hoff MH, Blakely EA, Burma S, Fornace AJ Jr, Gerson S, Hlatky L, Kirsch DG, Luderer U, Shay J, Wang Y, Weil MM. "Concepts and challenges in cancer risk prediction for the space radiation environment." Life Sci Space Res. 2015 Jul;6:92-103. http://dx.doi.org/10.1016/j.lssr.2015.07.006 , Jul-2015
Articles in Peer-reviewed Journals Fleenor CJ, Higa K, Weil MM, DeGregori J. "Evolved cellular mechanisms to respond to genotoxic insults: Implications for radiation-induced hematologic malignancies." Radiat Res. 2015 Oct;184(4):341-51. Review. http://dx.doi.org/10.1667/RR14147.1 ; PubMed PMID: 26414506; PubMed Central PMCID: PMC4617554 , Oct-2015
Articles in Peer-reviewed Journals Nia AM, Shavkunov A, Ullrich RL, Emmett MR. "137Cs γ ray and 28Si irradiation induced murine hepatocellular carcinoma lipid changes in liver assessed by MALDI-MSI combined with spatial shrunken centroid clustering algorithm: A pilot study." ACS Omega. 2020 Oct 6;5(39):25164-74. https://doi.org/10.1021/acsomega.0c03047 ; PMID: 33043195; PMCID: PMC7542585 , Oct-2020
Articles in Peer-reviewed Journals Ding LH, Yu Y, Edmondson EF, Weil MM, Pop LM, McCarthy M, Ullrich RL, Story MD. "Transcriptomic analysis links hepatocellular carcinoma (HCC) in HZE ion irradiated mice to a human HCC subtype with favorable outcomes." Sci Rep. 2021 Jul 7;11(1):14052. https://doi.org/10.1038/s41598-021-93467-3 ; PMID: 34234215; PMCID: PMC8263559 , Jul-2021
Project Title:  NSCOR: NASA Specialized Center of Research on Radiation Carcinogenesis Reduce
Fiscal Year: FY 2014 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 06/01/2009  
End Date: 09/30/2015  
Task Last Updated: 08/31/2015 
Download report in PDF pdf
Principal Investigator/Affiliation:   Ullrich, Robert  Ph.D. / University of Texas Medical Branch 
Address:  301 University Blvd 
Comprehensive Cancer Center, MS 1048 
Galveston , TX 77555-5302 
Email: bullrich@utmb.edu 
Phone: 409-747-1935  
Congressional District: 14 
Web:  
Organization Type: UNIVERSITY 
Organization Name: University of Texas Medical Branch 
Joint Agency:  
Comments: NOTE: PI moved to UTMB from Colorado State University in late 2008 (6/2009) 
Co-Investigator(s)
Affiliation: 
Le Beau, Michelle  University of Chicago 
Bacher, Jeff  Promega Corporation 
Yu, Yongjia  University of Texas Medical Branch 
Story, Michael  University of Texas Southwestern Medical Center at Dallas 
Bedford, Joel  Colorado State University 
Weil, Michael  Colorado State University 
Ray, F  Colorado State University 
Ding, Lianghao  University of Texas Southwestern Medical Center at Dallas 
Xie, Yang  University of Texas Southwestern Medical Center 
Key Personnel Changes / Previous PI: none
Project Information: Grant/Contract No. NNX09AM08G 
Responsible Center: NASA JSC 
Grant Monitor: Simonsen, Lisa  
Center Contact:  
lisa.c.simonsen@nasa.gov 
Unique ID: 7564 
Solicitation / Funding Source: 2008 NSCOR Space Radiation NNJ08ZSA003N 
Grant/Contract No.: NNX09AM08G 
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) Cancer:Risk of Radiation Carcinogenesis
Human Research Program Gaps: (1) Cancer-202:Evaluate the contribution of genetic background/diversity on carcinogenesis risk.
Flight Assignment/Project Notes: NOTE: End date changed to 9/30/2015 (from 5/31/2015) per NSSC information (Ed., 6/1/15)

NOTE: End date changed to 5/31/2015 per NSSC information (Ed., 3/3/14)

Task Description: The goal of this NSCOR is to provide the information required to develop a rational scientific basis for estimation of risks for carcinogenesis in humans from exposure to radiation during space flight. Previous results from this Program found an unexpectedly low RBE value for acute myeloid leukemia (AML) induction by 1 GeV 56Fe ions. Systematic cytogenetic analyses suggested both microdosimetric factors related to the track structure of 1 GeV 56Fe ions and biological factors could account for this observation. In addition, these studies found an unexpected increase in hepatocellular carcinoma (HCC) at doses as low as 0.1 Gy of 1 GeV 56Fe ions but very little, if any, increase following gamma-ray exposure. These data suggest that processes associated with expansion and progression of initiated cells may play a more prominent role in HCC. If this is the case, it is possible that there are qualitative differences as well as quantitative in the effects of HZE irradiations. To expand on these results and to address the overall goal of this NSCOR a series of coordinated activities will conducted in 5 Projects and 3 Cores aimed at: (1) providing quantitative animal tumorigenesis data on the relative effectiveness of specific HZE particles and SPE protons compared with gamma-rays in mouse models of AML and HCC; (2) providing a better understanding of the impact of radiation exposure on the processes involved in the initiation and in the progression of initiated cells toward the neoplastic phenotype; 3) delineating potential differences between low LET radiation and high LET radiation such as those encountered in space travel on these processes; 4) developing links between animal data and radiation-induced effects for AML in humans; and (5) developing biologically-based modeling approaches which are critical to link these biological effects to risks in humans.

Program Overview: The Radiation Carcinogenesis NSCOR was initiated in June 2009 and builds upon results obtained in its predecessor, the Leukemogenesis NSCOR. The Radiation Carcinogenesis NSCOR consists of four projects supported by three cores. The projects and cores are briefly described below.

Project 1. Dose response relationships for induction of AML and HCC as a function of radiation quality (project leader, Dr. Robert. L. Ullrich). This project is designed to compare the effects of irradiation with gamma-rays, select HZE particles, and protons on the induction of AML and hepatocellular carcinoma (HCC) using the C3H murine model.

Project 2. Mechanisms of radiation leukemogenesis (project leader, Dr. Michael M. Weil). The goal of Project 2 is to better understand how radiation leads to AML in a murine model and to generate data for the development of a biologically based model that can be used to predict AML risks from various HZE or high energy proton exposures.

Project 3. Pathogenesis of radiation-induced hepatocellular carcinoma (project leader, Dr. Robert L. Ullrich). The overall hypothesis of this project is that the dose response is likely to reflect both quantitative as well as qualitative differences in high LET effects. This overall hypothesis will be tested in 3 specific aims:

1. Quantify the frequency and progression of preneoplastic foci (including both hyperplastic and dysplastic foci) in livers of C3H/HeNCrl mice irradiated with either 137Cs gamma-rays or HZE ions.

2. Examine irradiated liver for evidence of increased oxidative damage and alterations in the regulation of inflammatory processes.

3. Determine tumorigenic effects following HZE and gamma-ray irradiation in murine models of hepatocellular carcinoma in which secondary “promoting” events play a significant role.

Project 4. Molecular and cytogenetic targets in murine and human AML. (Project leader, Dr. Michelle Le Beau). This project is designed to develop a cytogenetic and molecular profile of human radiation-induced AML, leading to an understanding of the key events and genetic pathways involved in the pathogenesis of this disease.

Core A (Core Director, Dr. F. Andrew Ray). The Biology Core facilitates the distribution of irradiated and control animals, tissues, cells, and other biological samples to investigators. This core is also responsible for conducting the irradiations required at the various sites for all projects.

Core B (Core Director, Dr. Michael Story). The Genomics and Biostatistics core provides appropriate genomic analyses, innovative statistical modeling, simulations, and data analyses for the projects.

Core C (Core Director, Dr. Robert Ullrich). The Administrative Core provides administrative, fiscal and management support for the Radiation Carcinogenesis NSCOR. This core also oversees the overall scientific conduct of the NSCOR and facilitates interactions between projects, core leaders and project investigators as well as interactions with the internal and external advisors.

Research Impact/Earth Benefits: This work will provide basic information on mechanisms of carcinogenesis as well as mechanisms specific to radiation-induced cancer.

Task Progress & Bibliography Information FY2014 
Task Progress: Project 1. Dose response relationships for induction of AML and HCC as a function of radiation quality (project leader, Dr. Robert. L. Ullrich). This project is designed to compare the effects of irradiation with gamma-rays, select HZE particles, and protons on the induction of AML and hepatocellular carcinoma (HCC) using the C3H murine model. We have completed analysis of the incidence of acute myeloid leukemia (AML) and hepatocellular carcinoma (HCC) as a function of dose in the gamma ray, 1972SPE proton, 600 MeV/n 56Fe, and 300 MeV/n 28Si groups. Publication listed in Bibliography section.

Project 1 publications in process:

Weil MM, Ray FA, Genik PC, Yu Y, McCarthy M, Fallgren CM, Ullrich RL. Effects of 28Si ions, 56Fe ions, and protons on the induction of murine acute myeloid leukemia and hepatocellular carcinoma. PLoS One. Submitted April 2014.

Kantara C, O'Connell MR, Luthra G, Gajjar A, Sarkar S, Ullrich RL, Singh P. Methods for detecting circulating cancer stem cells (CCSCs) as a novel approach for diagnosis of colon cancer relapse/metastasis. Lab Investigation. Submitted June 2014.

Project 2. Mechanisms of radiation leukemogenesis (project leader, Dr. Michael M. Weil). The goal of Project 2 is to better understand how radiation leads to AML in a murine model and to generate data for the development of a biologically based model that can be used to predict AML risks from various HZE or high energy proton exposures.

See Bibliography for publication in press.

Project 3. Pathogenesis of radiation-induced hepatocellular carcinoma (project leader, Drs.Robert L. Ullrich and Yongjia Yu). The overall hypothesis of this project is that the dose response is likely to reflect both quantitative as well as qualitative differences in high LET effects. We have found that HZE radiations induce a significant increase in the frequency of hepatocellular carcinomas (HCC) in certain mouse strains in comparison to gamma rays (see project 1). The underlying mechanisms remain unclear, but this observation is likely to reflect both quantitative as well as qualitative differences in high LET effects. To study the pathogenesis of HZE-induced HCC, we have been pursuing three specific aims for Project 3: 1. Quantify the frequency and progression of preneoplastic foci (including both hyperplastic and dysplastic foci) in livers of C3H/HeNCrl mice irradiated with either 137Cs gamma-rays or HZE ions; 2. Examine irradiated liver for evidence of increased oxidative damage and alterations in the regulation of inflammatory processes; 3. Determine tumorigenic effects following HZE and gamma-ray irradiation in murine models of hepatocellular carcinoma in which secondary “promoting” events play a significant role.

Project 4. Molecular and cytogenetic targets in murine and human AML. (Project leader, Dr. Michelle Le Beau). This project is designed to develop a cytogenetic and molecular profile of human radiation-induced AML, leading to an understanding of the key events and genetic pathways involved in the pathogenesis of this disease. Project 4 has been eliminated due to insufficient progress.

[Ed. note: compiled from NSCOR FY2014 annual report]

Bibliography: Description: (Last Updated: 07/25/2021) 

Show Cumulative Bibliography
 
Articles in Peer-reviewed Journals Kantara C, O'Connell M, Sarkar S, Moya S, Ullrich R, Singh P. "Curcumin promotes autophagic survival of a subset of colon cancer stem cells, which are ablated by DCLK1-siRNA." Cancer Res. 2014 May 1;74(9):2487-98. http://dx.doi.org/10.1158/0008-5472.CAN-13-3536 ; PubMed PMID: 24626093; PubMed Central PMCID: PMC4013529 , May-2014
Articles in Peer-reviewed Journals Genik PC, Vyazunova I, Steffen LS, Bacher JW, Bielefeldt-Ohmann H, McKercher S, Ullrich RL, Fallgren CM, Weil MM, Ray FA. "Leukemogenesis in heterozygous PU.1 knockout mice." Radiation Research. 2014 Sep;182(3):310-5. Epub 2014 Jul 30. http://dx.doi.org/10.1667/RR13738.1 ; PubMed PMID: 25076114 (Ed. note: previously reported in June 2015 as Accepted) , Sep-2014
Project Title:  NSCOR: NASA Specialized Center of Research on Radiation Carcinogenesis Reduce
Fiscal Year: FY 2013 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 06/01/2009  
End Date: 09/30/2015  
Task Last Updated: 04/01/2013 
Download report in PDF pdf
Principal Investigator/Affiliation:   Ullrich, Robert  Ph.D. / University of Texas Medical Branch 
Address:  301 University Blvd 
Comprehensive Cancer Center, MS 1048 
Galveston , TX 77555-5302 
Email: bullrich@utmb.edu 
Phone: 409-747-1935  
Congressional District: 14 
Web:  
Organization Type: UNIVERSITY 
Organization Name: University of Texas Medical Branch 
Joint Agency:  
Comments: NOTE: PI moved to UTMB from Colorado State University in late 2008 (6/2009) 
Co-Investigator(s)
Affiliation: 
Le Beau, Michelle  University of Chicago 
Bacher, Jeff  Promega Corporation 
Yu, Yongjia  University of Texas Medical Branch 
Story, Michael  University of Texas Southwestern Medical Center at Dallas 
Bedford, Joel  Colorado State University 
Weil, Michael  Colorado State University 
Ray, F  Colorado State University 
Ding, Lianghao  University of Texas Southwestern Medical Center at Dallas 
Xie, Yang  University of Texas Southwestern Medical Center 
Key Personnel Changes / Previous PI: none
Project Information: Grant/Contract No. NNX09AM08G 
Responsible Center: NASA JSC 
Grant Monitor: Simonsen, Lisa  
Center Contact:  
lisa.c.simonsen@nasa.gov 
Unique ID: 7564 
Solicitation / Funding Source: 2008 NSCOR Space Radiation NNJ08ZSA003N 
Grant/Contract No.: NNX09AM08G 
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) Cancer:Risk of Radiation Carcinogenesis
Human Research Program Gaps: (1) Cancer-202:Evaluate the contribution of genetic background/diversity on carcinogenesis risk.
Flight Assignment/Project Notes: NOTE: End date changed to 9/30/2015 (from 5/31/2015) per NSSC information (Ed., 6/1/15)

NOTE: End date changed to 5/31/2015 per NSSC information (Ed., 3/3/14)

Task Description: The goal of this NSCOR is to provide the information required to develop a rational scientific basis for estimation of risks for carcinogenesis in humans from exposure to radiation during space flight. Previous results from this Program found an unexpectedly low RBE value for acute myeloid leukemia (AML) induction by 1 GeV 56Fe ions. Systematic cytogenetic analyses suggested both microdosimetric factors related to the track structure of 1 GeV 56Fe ions and biological factors could account for this observation. In addition, these studies found an unexpected increase in hepatocellular carcinoma (HCC) at doses as low as 0.1 Gy of 1 GeV 56Fe ions but very little, if any, increase following gamma-ray exposure. These data suggest that processes associated with expansion and progression of initiated cells may play a more prominent role in HCC. If this is the case, it is possible that there are qualitative differences as well as quantitative in the effects of HZE irradiations. To expand on these results and to address the overall goal of this NSCOR a series of coordinated activities will conducted in 5 Projects and 3 Cores aimed at: (1) providing quantitative animal tumorigenesis data on the relative effectiveness of specific HZE particles and SPE protons compared with gamma-rays in mouse models of AML and HCC; (2) providing a better understanding of the impact of radiation exposure on the processes involved in the initiation and in the progression of initiated cells toward the neoplastic phenotype; 3) delineating potential differences between low LET radiation and high LET radiation such as those encountered in space travel on these processes; 4) developing links between animal data and radiation-induced effects for AML in humans; and (5) developing biologically-based modeling approaches which are critical to link these biological effects to risks in humans.

Program Overview: The Radiation Carcinogenesis NSCOR was initiated in June 2009 and builds upon results obtained in its predecessor, the Leukemogenesis NSCOR. The Radiation Carcinogenesis NSCOR consists of four projects supported by three cores. The projects and cores are briefly described below.

Project 1. Dose response relationships for induction of AML and HCC as a function of radiation quality (project leader, Dr. Robert. L. Ullrich). This project is designed to compare the effects of irradiation with gamma-rays, select HZE particles, and protons on the induction of AML and hepatocellular carcinoma (HCC) using the C3H murine model.

Project 2. Mechanisms of radiation leukemogenesis (project leader, Dr. Michael M. Weil). The goal of Project 2 is to better understand how radiation leads to AML in a murine model and to generate data for the development of a biologically based model that can be used to predict AML risks from various HZE or high energy proton exposures.

Project 3. Pathogenesis of radiation-induced hepatocellular carcinoma (project leader, Dr. Robert L. Ullrich). The overall hypothesis of this project is that the dose response is likely to reflect both quantitative as well as qualitative differences in high LET effects. This overall hypothesis will be tested in 3 specific aims:

1. Quantify the frequency and progression of preneoplastic foci (including both hyperplastic and dysplastic foci) in livers of C3H/HeNCrl mice irradiated with either 137Cs gamma-rays or HZE ions.

2. Examine irradiated liver for evidence of increased oxidative damage and alterations in the regulation of inflammatory processes.

3. Determine tumorigenic effects following HZE and gamma-ray irradiation in murine models of hepatocellular carcinoma in which secondary “promoting” events play a significant role.

Project 4. Molecular and cytogenetic targets in murine and human AML. (Project leader, Dr. Michelle Le Beau). This project is designed to develop a cytogenetic and molecular profile of human radiation-induced AML, leading to an understanding of the key events and genetic pathways involved in the pathogenesis of this disease.

Core A (Core Director, Dr. F. Andrew Ray). The Biology Core facilitates the distribution of irradiated and control animals, tissues, cells, and other biological samples to investigators. This core is also responsible for conducting the irradiations required at the various sites for all projects.

Core B (Core Director, Dr. Michael Story). The Genomics and Biostatistics core provides appropriate genomic analyses, innovative statistical modeling, simulations, and data analyses for the projects.

Core C (Core Director, Dr. Robert Ullrich). The Administrative Core provides administrative, fiscal and management support for the Radiation Carcinogenesis NSCOR. This core also oversees the overall scientific conduct of the NSCOR and facilitates interactions between projects, core leaders and project investigators as well as interactions with the internal and external advisors.

Research Impact/Earth Benefits: This work will provide basic information on mechanisms of carcinogenesis as well as mechanisms specific to radiation-induced cancer.

Task Progress & Bibliography Information FY2013 
Task Progress: Progress on this NSCOR has been made with respect to defining the dose response relationships for the induction of acute myeloid leukemia AML and hepatocellular carcinoma HCC using 600 MeV/n 56Fe and 350 MeV/n 28Si as well as gamma rays and SPE1972 protons. We have confirmed a low RBE for the induction of AML and a very high RBE for the induction of HCC. We have also been able to define in more depth mechanisms for the induction of radiation-induced AML in both mouse models and in cancer patients developing AML as a second cancer following treatment for a primary. Progress has also been made on defining important tissue changes in the liver that suggest that the high RBE for the induction of HCC is a result of promoting effects due to upregulation of inflammatory and cytokine pathways.

Bibliography: Description: (Last Updated: 07/25/2021) 

Show Cumulative Bibliography
 
Articles in Peer-reviewed Journals Sarkar S, Kantara C, Ortiz I, Swiercz R, Kuo J, Davey R, Escobar K, Ullrich R, Singh P. "Progastrin overexpression imparts tumorigenic/metastatic potential to embryonic epithelial cells: phenotypic differences between transformed and nontransformed stem cells. " Int J Cancer. 2012 Oct 1;131(7):E1088-99. Epub 2012 May 17. http://dx.doi.org/10.1002/ijc.27615 ; PubMed PMID: 22532325 , Oct-2012
Articles in Peer-reviewed Journals Steffen LS, Bacher JW, Peng Y, Le PN, Ding LH, Genik PC, Ray FA, Bedford JS, Fallgren CM, Bailey SM, Ullrich RL, Weil MM, Story MD. "Molecular characterisation of murine acute myeloid leukaemia induced by 56Fe ion and 137Cs gamma ray irradiation." Mutagenesis. 2013 Jan;28(1):71-9. Epub 2012 Sep 17. http://dx.doi.org/10.1093/mutage/ges055 ; PubMed PMID: 22987027 , Jan-2013
Articles in Peer-reviewed Journals Bielefeldt-Ohmann H, Genik PC, Fallgren CM, Ullrich RL, Weil MM. "Animal studies of charged particle-induced carcinogenesis." Health Physics. 2012 Nov;103(5):568-76. http://dx.doi.org/10.1097/HP.0b013e318265a257 ; PubMed PMID: 23032886 , Nov-2012
Articles in Peer-reviewed Journals McNerney ME, Brown CD, Wang X, Bartom ET, Karmakar S, Bandlamudi C, Yu S, Ko J, Sandall BP, Stricker T, Anastasi J, Grossman RL, Cunningham JM, Le Beau MM, White KP. "CUX1 is a haploinsufficient myeloid tumor suppressor on chromosome 7 frequently inactivated in acute myeloid leukemia." Blood. 2013 Feb 7;121(6):975-83. Epub 2012 Dec 3. http://dx.doi.org/10.1182/blood-2012-04-426965 ; PubMed PMID: 23212519 , Feb-2013
Articles in Peer-reviewed Journals Peng Y, Nagasawa H, Warner C, Bedford JS. "Genetic susceptibility: radiation effects relevant to space travel." Health Phys. 2012 Nov;103(5):607-20. http://dx.doi.org/10.1097/HP.0b013e31826945b9 ; PubMed PMID: 23032891 , Nov-2012
Project Title:  NSCOR: NASA Specialized Center of Research on Radiation Carcinogenesis Reduce
Fiscal Year: FY 2012 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 06/01/2009  
End Date: 05/31/2014  
Task Last Updated: 06/04/2012 
Download report in PDF pdf
Principal Investigator/Affiliation:   Ullrich, Robert  Ph.D. / University of Texas Medical Branch 
Address:  301 University Blvd 
Comprehensive Cancer Center, MS 1048 
Galveston , TX 77555-5302 
Email: bullrich@utmb.edu 
Phone: 409-747-1935  
Congressional District: 14 
Web:  
Organization Type: UNIVERSITY 
Organization Name: University of Texas Medical Branch 
Joint Agency:  
Comments: NOTE: PI moved to UTMB from Colorado State University in late 2008 (6/2009) 
Co-Investigator(s)
Affiliation: 
Le Beau, Michelle  University of Chicago 
Bacher, Jeff  Promega Corporation 
Yu, Yongjia  University of Texas Medical Branch 
Story, Michael  University of Texas Southwestern Medical Center at Dallas 
Bedford, Joel  Colorado State University 
Weil, Michael  Colorado State University 
Ray, F  Colorado State University 
Ding, Lianghao  University of Texas Southwestern Medical Center at Dallas 
Xie, Yang  University of Texas Southwestern Medical Center 
Key Personnel Changes / Previous PI: none
Project Information: Grant/Contract No. NNX09AM08G 
Responsible Center: NASA JSC 
Grant Monitor: Cucinott1a, Francis  
Center Contact: 281-483-0968 
noaccess@nasa.gov 
Unique ID: 7564 
Solicitation / Funding Source: 2008 NSCOR Space Radiation NNJ08ZSA003N 
Grant/Contract No.: NNX09AM08G 
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) Cancer:Risk of Radiation Carcinogenesis
Human Research Program Gaps: (1) Cancer-202:Evaluate the contribution of genetic background/diversity on carcinogenesis risk.
Task Description: The goal of this NSCOR is to provide the information required to develop a rational scientific basis for estimation of risks for carcinogenesis in humans from exposure to radiation during space flight. Previous results from this Program found an unexpectedly low RBE value for acute myeloid leukemia (AML) induction by 1 GeV 56Fe ions. Systematic cytogenetic analyses suggested both microdosimetric factors related to the track structure of 1 GeV 56Fe ions and biological factors could account for this observation. In addition, these studies found an unexpected increase in hepatocellular carcinoma (HCC) at doses as low as 0.1 Gy of 1 GeV 56Fe ions but very little, if any, increase following gamma-ray exposure. These data suggest that processes associated with expansion and progression of initiated cells may play a more prominent role in HCC. If this is the case, it is possible that there are qualitative differences as well as quantitative in the effects of HZE irradiations. To expand on these results and to address the overall goal of this NSCOR a series of coordinated activities will conducted in 5 Projects and 3 Cores aimed at: (1) providing quantitative animal tumorigenesis data on the relative effectiveness of specific HZE particles and SPE protons compared with gamma-rays in mouse models of AML and HCC; (2) providing a better understanding of the impact of radiation exposure on the processes involved in the initiation and in the progression of initiated cells toward the neoplastic phenotype; 3) delineating potential differences between low LET radiation and high LET radiation such as those encountered in space travel on these processes; 4) developing links between animal data and radiation-induced effects for AML in humans; and (5) developing biologically-based modeling approaches which are critical to link these biological effects to risks in humans.

Program Overview: The Radiation Carcinogenesis NSCOR was initiated in June 2009 and builds upon results obtained in its predecessor, the Leukemogenesis NSCOR. The Radiation Carcinogenesis NSCOR consists of four projects supported by three cores. The projects and cores are briefly described below.

Project 1. Dose response relationships for induction of AML and HCC as a function of radiation quality (project leader, Dr. Robert. L. Ullrich). This project is designed to compare the effects of irradiation with gamma-rays, select HZE particles, and protons on the induction of AML and hepatocellular carcinoma (HCC) using the C3H murine model.

Project 2. Mechanisms of radiation leukemogenesis (project leader, Dr. Michael M. Weil). The goal of Project 2 is to better understand how radiation leads to AML in a murine model and to generate data for the development of a biologically based model that can be used to predict AML risks from various HZE or high energy proton exposures.

Project 3. Pathogenesis of radiation-induced hepatocellular carcinoma (project leader, Dr. Robert L. Ullrich). The overall hypothesis of this project is that the dose response is likely to reflect both quantitative as well as qualitative differences in high LET effects. This overall hypothesis will be tested in 3 specific aims:

1. Quantify the frequency and progression of preneoplastic foci (including both hyperplastic and dysplastic foci) in livers of C3H/HeNCrl mice irradiated with either 137Cs gamma-rays or HZE ions.

2. Examine irradiated liver for evidence of increased oxidative damage and alterations in the regulation of inflammatory processes.

3. Determine tumorigenic effects following HZE and gamma-ray irradiation in murine models of hepatocellular carcinoma in which secondary “promoting” events play a significant role.

Project 4. Molecular and cytogenetic targets in murine and human AML. (Project leader, Dr. Michelle Le Beau). This project is designed to develop a cytogenetic and molecular profile of human radiation-induced AML, leading to an understanding of the key events and genetic pathways involved in the pathogenesis of this disease.

Core A (Core Director, Dr. F. Andrew Ray). The Biology Core facilitates the distribution of irradiated and control animals, tissues, cells, and other biological samples to investigators. This core is also responsible for conducting the irradiations required at the various sites for all projects.

Core B (Core Director, Dr. Michael Story). The Genomics and Biostatistics core provides appropriate genomic analyses, innovative statistical modeling, simulations, and data analyses for the projects.

Core C (Core Director, Dr. Robert Ullrich). The Administrative Core provides administrative, fiscal and management support for the Radiation Carcinogenesis NSCOR. This core also oversees the overall scientific conduct of the NSCOR and facilitates interactions between projects, core leaders and project investigators as well as interactions with the internal and external advisors.

Research Impact/Earth Benefits: This work will provide basic information on mechanisms of carcinogenesis as well as mechanisms specific to radiation-induced cancer.

Task Progress & Bibliography Information FY2012 
Task Progress: Annual Progress Report/ Robert L. Ullrich, PhD. Reporting Period: May 11, 2011-May 31, 2012.

Program Overview: The Radiation Carcinogenesis NSCOR consists of four projects supported by three cores. The projects and cores are briefly described below. Details of progress for each project during this reporting period follow this description.

Project 1. Dose response relationships for induction of AML and HCC as a function of radiation quality (project leader, Dr. Robert. L. Ullrich). This project is designed to compare the effects of irradiation with gamma-rays, select HZE particles, and protons on the induction of AML and hepatocellular carcinoma (HCC) using the C3H murine model.

Project 2. Mechanisms of radiation leukemogenesis (project leader, Dr. Michael M. Weil). The goal of Project 2 is to better understand how radiation leads to AML in a murine model and to generate data for the development of a biologically based model that can be used to predict AML risks from various HZE or high energy proton exposures.

Project 3. Pathogenesis of radiation-induced hepatocellular carcinoma (project leader, Drs. Robert L. Ullrich and Yongjia Yu). The overall hypothesis of this project is that the dose response is likely to reflect both quantitative as well as qualitative differences in high LET effects.

Project 4. Molecular and cytogenetic targets in murine and human AML. (Project leader, Dr. Michelle Le Beau). This project is designed to develop a cytogenetic and molecular profile of human radiation-induced AML, leading to an understanding of the key events and genetic pathways involved in the pathogenesis of this disease.

Core A (Core Director, Dr. F. Andrew Ray). The Biology Core facilitates the distribution of irradiated and control animals, tissues, cells, and other biological samples to investigators. This core is also responsible for conducting the irradiations required at the various sites for all projects.

Core B (Core Director, Dr. Michael Story). The Genomics and Biostatistics core provides appropriate genomic analyses, innovative statistical modeling, simulations, and data analyses for the projects.

Core C (Core Director, Dr. Robert Ullrich). The Administrative Core provides administrative, fiscal and management support for the Radiation Carcinogenesis NSCOR. This core also oversees the overall scientific conduct of the NSCOR and facilitates interactions between projects, core leaders and project investigators as well as interactions with the internal and external advisors.

Project 1

The first cohort of the non-irradiated controls reached 800 days on 11/12/2011. In addition, the first cohort of the Fe and Si irradiated animals also reached 800 days and euthanized on 11/12/2011. Remaining animals in each group are being analyzed histopathologically as they are euthanized.

Project 2

Project 2 in the Radiation Carcinogenesis NSCOR is focused on developing a biological model for radiation-induced murine acute myeloid leukemia (rAML) development and determining the effect of radiation dose and quality on each leukemogenic step. The murine model of radiation-induced AML is particularly well suited for this project because the molecular and cytogenetic events involved are becoming increasingly well characterized and AML susceptible and resistant mouse strains are available allowing the use of genetic approaches.

The critical initiating step in murine radiation-induced AML is the biallelic loss of the PU.1 tumor suppressor gene in a hematopoietic cell at the appropriate differentiation stage. The actual differentiation stage of target cell for leukemic transformation (leukemia-initiating cell) is unknown, though it is likely the hematopoietic stem cell (HSC), common myeloid progenitor (CMP) or granulocyte/macrophage progenitor (GMP). We have also found evidence of microsatellite instability (MSI) in radiogenic AMLs, though we don’t know if it is necessary for leukemogenesis. We are developing experiments that will identify the leukemia initiating cell, determine the mechanism(s) leading to PU.1 loss, and determine the role of microsatellite instability in radiation-induced AML leukemogenesis. Our progress over the past year in assay development and completing specific experiments is described below. To place the assays into their experimental contexts, please see last year’s progress report.

Apoptosis in Irradiated Hematopoietic Stem and Progenitor Cells from CBA and C57BL/6 Mice. Several hypotheses have been advanced to explain murine strain differences in susceptibility to radiogenic AML. One possibility is that radiation damaged leukemia initiating cells in susceptible strains are less likely to undergo apoptosis than those in resistant strains. The likely cells at risk for leukemogenic transformation are hematopoietic stem or progenitor cells (HSPC) so over the last year we have quantified apoptosis levels in HSPC cell subpopulations in irradiated CBA/CaJ and C57BL/6J mice.

Mice were irradiated to the whole body with 3 Gy of 137Cs gamma-rays at a dose rate of 1.17 Gy/min. Apoptosis in various bone marrow cell subpopulations was assayed at several post-irradiation time intervals. The irradiations were timed so that the bone marrow cells were always harvested at 11:00 A.M.. To avoid cage effects or day to day variation in the assay, mice for each time point were irradiated on at least two separate days and mice within a single cage were split among at least two time points. At varying times post-irradiation mice were euthanized by CO2 inhalation and their bone marrow cells were collected in RPMI 1640 (Hyclone Thermo Scientific, Logan, Utah). The cells were disaggregated and the cell suspension was treated for 3 minutes with ACK buffer (150 mM NH4Cl, 10 mM KHCO3, and 0.1 mM Na2EDTA) to lyse red blood cells. Cells were plated at approximately 1-2 x 10(6) cells per well in a 96-well plate in FACs buffer (PBS with 2% fetal bovine serum and 0.05% sodium azide) or 1 x 10(7) cells in FACs buffer per snap cap tube. Prior to immunostaining, non-specific antibody binding was blocked by addition of normal mouse serum and unlabeled anti-Fc gammaRIII antibody (clone 24.G2, eBioscience, San Diego, CA), except for wells in which labeled anti-Fc gammaRII/RIII was used, in which case just mouse serum was added. Staining with indirectly and directly conjugated antibodies was done in FACS buffer at room temperature followed by washing. Secondary streptavidin conjugates were added to the cells, incubated at room temperature in FACs buffer, and the cells washed. For assessment of apoptosis, Pacific blue-labeled Annexin V (BD Horizon, San Diego, CA) was added to each well and incubated for 20 minutes at room temperature prior to analysis. In addition, either propidium iodine (PI) or 7-AAD was added to each sample to analyze live verses dead cells. All flow cytometry was done using a Cyan ADP flow cytometer (Beckman Coulter, Fort Collins, CO). Data was analyzed using FlowJo (Tree Star, Inc, Ashland, OR) software. The bone marrow cells were immunophenotyped with anti-Fc gammaRII/III PE (clone 93), anti-ckit/CD117 APC-eFluor 780 (clone ACK2), anti-CD127-biotin and PE (IL-7R alpha, clone A7R34), anti-Sca1 APC (clone D7), anti-CD150 PerCP-eFluor 710 (clone mShad150), anti-CD90.1 APC –eFluor780 (Thy1, clone HIS51), anti-ckit FITC or eFluor 780 (clone 2B8), anti-CD135 PE (Flt-3, clone A2F10), (all from eBioscience), Mouse Lineage Mixture-biotin (Lin, Invitrogen, Camarillo, CA) and anti-CD34 FITC (clone RAM34, BD/Pharmingen).

Common myeloid progenitor cells (CMP) were identified as Lin- IL7R alpha- Sca1- ckit+ CD34+ Fc alphaRII/III lo; granulocyte macrophage progenitors (GMP) as Lin- IL7R alpha- Sca1- ckit+ CD34+ Fc alphaRII/III hi; megakaryocyte-erythroid progenitors (MEP) cells as Lin- IL7R alpha- Sca1- CD34- ckit+ Fc alphaRII/III lo and common lymphoid progenitors (CLP) as Lin- IL7R alpha+ Thy1- Sca1 lo ckit lo.

We initially defined hematopoetic stem cells (HSCs) Lin- IL7R- Sca1+ ckit+/hi CD34- CD150+ CD135+. However, it soon became clear that CBA hematopoietic cells lack Sca1 expression, an observation recently reported by Papathanasiou and colleagues (1). Therefore we modified the panel by inclusion of anti-CD48 (eBioscience) and defined HSC as Lin- IL7R- ckit+/hi CD34- CD150+ CD135+ CD48-.

For CMPs, GMPs, MEPs, and CLPs, between 500,000 – 1,000,000 total bone marrow events were collected per sample. Due to the rarity of the HSC population (predicted as 1:10,000-1:15,000), we collected approximately 10 million events per sample.

The mice were irradiated with the maximally leukemic dose of 3 Gy and at various times afterwards their bone marrow cells were collected for simultaneous flow cytometric analysis of their differentiation stage and apoptosis status. Apoptosis levels were low for all of the subpopulations and did not differ consistently between the rAML sensitive CBA mice and the rAML resistant C57BL/6 mice. These results suggest that strain differences in susceptible to radiation-induced apoptosis do not account strain differences in susceptibility to rAML.

Chromosome 2 Engineered Mice. Previously we described the rationale and technology for making a panel of mice with chromosome 2 deletions. We isolated three correctly targeted ES cell clones and over the past year we tested one of these clones for germline competency (that is, a clone that generates chimeric founders that can father pups carrying the engineered allele). We found germline competency to be extremely low and have now begun to test the other two target ES cell clones. If one of these turns out to be suitable it will be irradiated and a panel of subclones with chromosome 2 deletions of varying sizes will be established. Selected subclones will be used to generate chromosome engineered mice that will be monitored for the development of AML.

Development of Additional rAML Mouse Models. We are developing or continuing to develop three additional mouse models to study the events involved in radiation leukemogenesis. As described in the previous progress report we are transgressing a green fluorescence protein (GFP) gene that is expressed in hematopoietic cells from C57BL/6-Tg(UBC-GFP)30Scha/J transgenic mice onto a CBA/CaJ background by repetitive backcrossing to the CBA/CaJ strain with selection for the transgene. The mice will be used to study the effects of irradiated stromal cells on leukemogenesis. We are currently on backcross generation N5.

A second mouse model we are constructing is a PU.1 R235C knock-in mouse. This knock-in should be particularly susceptible to rAML and will be useful in uncovering the events in addition to PU.1 activation that are required for radiation leukemogenesis.

At our last NSCOR external advisory panel meeting, we were directed to generate PU.1+/- Ddb2+/- knockout mice and follow them for leukemogenesis. The rationale for this experiment is that while a large deletion on chromosome 2 containing the PU.1 gene leads to AML, simply knocking out one allele of the PU.1 gene does not. This suggests that hemizygous loss of another gene in the deleted region contributes to leukemogenesis, perhaps by facilitating mutation of the remaining PU.1 allele. Ddb2, a DNA damage recognition gene involved in DNA repair, is in the region of chromosome 2 that is commonly deleted in rAML and consequently is a good candidate. If it facilitates the R235 mutation we would anticipate the mice would develop AML.

Development of an Assay for Rare PU.1 R235C Mutations. In order to determine when the PU.1 R235C mutation occurs during radiation leukemogenesis and the cells that are affected we need to develop an assay that can detect that mutation in one cell among many cells not carrying the mutation. We are developing an assay in which the wild type allele is removed from the DNA sample being assayed by digestion with a restriction endonuclease (BbvI) that recognizes the wild type allele but not the mutant allele. The mutant allele is further enriched by COLD-PCR techniques and identified by high-resolution melt analysis. We tested this approach with mixtures of DNA that contain known amounts of the mutant allele from an AML cell line. The assay can detect mutations that are present in the starting mix at <1%. The next step in assay development will be to perform the COLD-PCR step as small pool PCR which should give a detection sensitivity on the order of 2 mutant alleles in 105 wildtype alleles per 384-well plate.

Reference List

1. P. Papathanasiou, R. Tunningley, D. R. Pattabiraman, P. Ye, T. J. Gonda, B. Whittle, A. E. Hamilton, S. O. Cridland, R. Lourie and others, A recessive screen for genes regulating hematopoietic stem cells. Blood 116, 5849-5858 (2010).

Project 3

In the fall of 2010, 60 mice were irradiated for Project 3. Our original design was to perform serial sacrifice studies for liver tissue analysis at 6, 9, and 12 months after irradiation following 3 Gy of gamma rays or 0.1 Gy of 1 GeV Iron based on our previous studies. Surprisingly, we are observing liver tumors much earlier than expected. As a result we added a sacrifice time point at 3 months post irradiation. Based on our results we may add a 1 month time point.

We have added analysis of liver tumors in the main groups as well. These include livers from non-tumor bearing animals. Tumor and adjacent tissues in tumor bearing animals and in tumors that have metastasized are undergoing analysis. To date metastases have only been found in heavy ion irradiated mice. We have also begun to examine state-of-the art 3-dimensional imaging of tumors and surrounding stroma. Also we are exploring techniques for transplantation of hepatocytes into livers of other mice. With this technique clonal outgrowths can be identified and analyzed. This also allows us to examine effects of irradiation of the host liver on cells from donor.

Project 4

The overall goal of Project 4 is to develop a cytogenetic and molecular profile of human radiation-induced myeloid neoplasms (t-MN), and to elucidate the key events and genetic pathways involved in the pathogenesis of this disease. To this end, we proposed to profile the genetic alterations in radiation-induced t-MN by (1) Cytogenetic analysis; (2) High-density SNP array analysis of copy number alterations and loss of heterozygosity; (3) Gene expression profiling; (4) Analysis of promoter methylation; and (5) miRNA expression profiling. As described below, we have elected to replace some of the above technologies with transcriptome and whole genome sequencing.

Exposure to ionizing radiation is associated with the development of therapy-related myeloid neoplasms (t-MN). Several distinct clinical and cytogenetic subtypes of t-MN have been recognized and found to be closely associated various cytotoxic therapies, such as alkylating agents, anti-metabolites, and DNA topoisomerase-II inhibitors. However, the characteristics of t-MN that follow radiation therapy (RT) alone are not as clear. To characterize this subgroup of t-MN, we examined the clinical, epidemiologic, and cytogenetic characteristics of 72 cases of t-MN in patients who received RT alone for an antecedent disease at the University of Chicago.

Sixty-five (90%) patients had had a primary solid tumor, 5 had had a primary hematologic malignancy (4 Hodgkin lymphoma), and 2 had received RT for non-malignant disorders (acne; hydatiform mole). Prostate and testicular cancers were the predominant primary tumors (39%). Breast (21%) and gynecological cancers (18%, including ovarian, cervical, endometrial, and vaginal cancers) were also common. There were 36 women and 36 men; 51 patients were white (71%), 10 were African American (14%), 1 was an Asian American (1%), and race/ethnicity was unrecorded in the remaining ten patients.

The median time from diagnosis of t-MN to death was 0.9 years (95% confidence interval (CI), 0.6-1.2 years). Sixty-one patients had one or more clonal cytogenetic abnormality (85%) at diagnosis of t-MN; 11 (15%) had no detectable abnormality. Most common (39 patients, 54%) were loss or deletion of chromosome 5 (n=19), 7 (n=9), or both (n=11). Eleven patients (15%) had recurring balanced translocations previously reported in t-MN; 3 with inv(16), 4 with t(15;17), and 3 with t(21q22). Eleven patients (15%) had other clonal abnormalities. Survival according to cytogenetic subgroups was determined. Patients with a normal cytogenetic pattern or recurring balanced translocations survived longer (log rank, p=0.008). t-MN with clonal abnormalities of chromosome 5, 7, or both had a poor median survival of 0.6 years (95% CI, 0.3-1.0 years) compared to other cytogenetic groups. There was no association between age at diagnosis of t-MN and overall survival. We reviewed treatment records for 47 patients who received therapy for t- MN -- either supportive care, chemotherapy alone, or chemotherapy with hematopoietic cell transplant. There was a trend towards better survival with higher intensity treatment, but it was not statistically significant (p=0.065).

In summary, our analysis revealed that t-MN following RT alone bears clinical and cytogenetic similarities to alkylator-associated t-MN with frequent clonal abnormalities of chromosomes 5 and/or 7, relatively long latency of 5-10 years, and poor outcomes even with intensive therapy. However, some patients who develop t-MN after RT alone have recurring, balanced chromosomal translocations or normal karyotypes. These patients have a shorter latency, better response to anti-leukemia treatment, and longer survival. This work was reported at the 2011 annual meeting of the American Society of Hematology.

To determine the genetic profile for t-MN, we performed transcriptome sequencing on 22 patient samples, and SNP array analysis on 35 samples, half of which were characterized by -7/del(7q) (collaboration with Dr. Kevin White, University of Chicago). Given the high frequency of -7/del(7q), in radiation-induced t-MN, and the adverse prognosis associated with this cytogenetic subgroup, we focused initially on the analysis of this subgroup of t-MN. Copy number analysis identified a 2.17 Mb commonly deleted segment on chromosome band 7q22.1, containing CUX1, a lineage-determining transcription factor normally expressed in hematopoietic stem cells. In addition, CUX1 was disrupted by a translocation resulting in an out-of-frame, chimeric fusion transcript in one patient. Moreover, CUX1 was expressed at haploinsufficient levels in t-MN with a -7/del(7q). To test the tumor suppressor activity of CUX1, the Drosophila homologue, CUT, was knocked-down in Drosophila melanogaster hemocytes, which led to melanotic tumor formation and increased numbers of hemocytes (the Drosophila equivalent of myeloid leukemia). These data suggest that CUX1 is a conserved tumor suppressor associated with -7/del(7q). To extend this work to radiation-induced t-MN, we have identified 13 cases from the series described above for which we have cryopreserved leukemia samples (germline tissue is available in a subset of these cases). We have extracted DNA and submitted samples from 5 cases to the Beijing Institute for Genomics for whole genome sequencing, and anticipate receiving the raw data for analysis within three months. Our research plan for the next year is to analyze additional radiation-induced t-MN samples depending the results of the initial series, and to perform functional analysis of candidate genes identified from these analyses.

Core A: Irradiation and Tissue Acquisition. Core A personnel have irradiated all mice for project 1. Animals are monitored over an 800 day time period and moribund and tumor bearing animals are euthanized. All animals are removed for necropsy based on clinical signs and symptoms of AML or HCC, other tumors, or when moribund. When animals reach 800 days post-irradiation they are being euthanized and examined for tumors. The first of the non-irradiated controls group reached 800 days on 11/12/2011. The first cohort of the Fe and Si irradiated animals also reached 800 days and euthanized on 11/12/2011. Other groups will reach 800 days at various times throughout the next two years.

Core B: Genomics. Data Analytics: All genomic/epigenomic data can now be placed in a user accessible database from remote locations. As part of data integration probe alignments can now be performed across 7 gene expression platforms with miRNA platforms (Illumina, Exiqon) ready shortly. The next generation sequencing data analysis pipeline is also running, including Chip-seq, DNA-seq, RNA-seq and shRNA-seq are operational. Joint modeling (Xie et al, Stat in Med, 2010) is available for integration of DNA copy number variation (CNV) and gene expression. We could consider developing a Carcinogenesis database and placing it on the UTSW cluster. That cluster is available now for outside access. It consists of a 36 CPU cluster and 84 Tbytes of storage with tape backup. Various statistical tools reside within the database confines which would be available for users to interrogate data.

Leukemia Genomics: To date 12 CBA splenic tumors have been examined for CNV. A common deletion of 34 Mb has been identified with very little in the way of CNV in other regions of the genome. This 34 Mb region contains Pu.1 (Sfpi1) and some 20 other genes and one miRNA (miR-130a). There are also two zinc fingers, the MADD gene (MAP-kinase activating death domain) and DDB2 (DNA damage binding protein 2), a gene associated with DNA repair. It must be kept in mind that these are single copy losses and gene expression will confirm a gene dosage effect. Our early gene expression analysis showed that PU.1 as actually upregulated over 2-fold. This comparison was against CD34+ cells and it was suggested that at the annual review that these were not necessarily the most appropriate control cell to use. We are in the process of obtaining CD117+ cells, which the review panel suggested was a more appropriate control. In common with the human AML analysis we also see a 20-fold reduction in the EGR1 gene, however, with all of our gene expression analysis we await the appropriate control (CD117+). It was suggested by the external advisory panel that also examine miRNA in the leukemia cells generated from C3H mice. This is an expensive goal and it may require some re-budgeting or re-prioritization.

Hepatocellular Carcinoma Genomics: Samples for gene expression, CNV, miRNA and methylation are being acquired. We will await the capture of all samples before we begin the appropriate analyses. This will be in the next grant year.

Core C Administrative Core. Core C ordered all animals for irradiations described above and planned and coordinated the NSCOR annual meeting on September 24th and 25th.

Bibliography: Description: (Last Updated: 07/25/2021) 

Show Cumulative Bibliography
 
Abstracts for Journals and Proceedings Weil MM, Ray A, Bacher JW, Bedford JS, Steffen LS, Ding L, Bielefeldt-Ohmann H, Genik PC, Yu Y, McCarthy M, Fallgren CM, Story MD, Ullrich RL. "The Radiation Carcinogenesis NSCOR." To be presented at the 23rd Annual NASA Space Radiation Investigators’ Workshop, Durham, NC, July 8–11, 2012.

Program and abstracts. 23rd Annual NASA Space Radiation Investigators’ Workshop, Durham, NC, July 8–11, 2012. In press, as of June 2012. , Jun-2012

Abstracts for Journals and Proceedings Weil MM, Ray A, Bacher JW, Story MD, Bedford JS, Le Beau MM, Ullrich RL. "The Radiation Carcinogenesis NSCOR." Presented at the 22nd Annual Space Radiation Investigators' Workshop, League City, TX, September 18-21, 2011.

22nd Annual Space Radiation Investigators' Workshop, League City, TX, September 18-21, 2011. http://www.dsls.usra.edu/meetings/radiation2011/pdf/7063.pdf , Sep-2011

Articles in Peer-reviewed Journals Chen M, Xie Y, Story M. "An exponential-gamma convolution model for background correction of Illumina BeadArray data." Communications in Statistics - Theory and Methods. 2011 Sep;40(17):3055-69. http://dx.doi.org/10.1080/03610921003797753 , Sep-2011
Project Title:  NSCOR: NASA Specialized Center of Research on Radiation Carcinogenesis Reduce
Fiscal Year: FY 2011 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 06/01/2009  
End Date: 05/31/2014  
Task Last Updated: 05/16/2011 
Download report in PDF pdf
Principal Investigator/Affiliation:   Ullrich, Robert  Ph.D. / University of Texas Medical Branch 
Address:  301 University Blvd 
Comprehensive Cancer Center, MS 1048 
Galveston , TX 77555-5302 
Email: bullrich@utmb.edu 
Phone: 409-747-1935  
Congressional District: 14 
Web:  
Organization Type: UNIVERSITY 
Organization Name: University of Texas Medical Branch 
Joint Agency:  
Comments: NOTE: PI moved to UTMB from Colorado State University in late 2008 (6/2009) 
Co-Investigator(s)
Affiliation: 
Le Beau, Michelle  University of Chicago 
Bacher, Jeff  Promega Corporation 
Yu, Yongjia  University of Texas Medical Branch 
Story, Michael  University of Texas Southwestern Medical Center at Dallas 
Bedford, Joel  Colorado State University 
Weil, Michael  Colorado State University 
Ray, F  Colorado State University 
Ding, Lianghao  University of Texas Southwestern Medical Center at Dallas 
Xie, Yang  University of Texas Southwestern Medical Center 
Key Personnel Changes / Previous PI: none
Project Information: Grant/Contract No. NNX09AM08G 
Responsible Center: NASA JSC 
Grant Monitor: Cucinott1a, Francis  
Center Contact: 281-483-0968 
noaccess@nasa.gov 
Unique ID: 7564 
Solicitation / Funding Source: 2008 NSCOR Space Radiation NNJ08ZSA003N 
Grant/Contract No.: NNX09AM08G 
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) Cancer:Risk of Radiation Carcinogenesis
Human Research Program Gaps: (1) Cancer-202:Evaluate the contribution of genetic background/diversity on carcinogenesis risk.
Task Description: The goal of this NSCOR is to provide the information required to develop a rational scientific basis for estimation of risks for carcinogenesis in humans from exposure to radiation during space flight. Previous results from this Program found an unexpectedly low RBE value for acute myeloid leukemia (AML) induction by 1 GeV 56Fe ions. Systematic cytogenetic analyses suggested both microdosimetric factors related to the track structure of 1 GeV 56Fe ions and biological factors could account for this observation. In addition, these studies found an unexpected increase in hepatocellular carcinoma (HCC) at doses as low as 0.1 Gy of 1 GeV 56Fe ions but very little, if any, increase following gamma-ray exposure. These data suggest that processes associated with expansion and progression of initiated cells may play a more prominent role in HCC. If this is the case, it is possible that there are qualitative differences as well as quantitative in the effects of HZE irradiations. To expand on these results and to address the overall goal of this NSCOR a series of coordinated activities will conducted in 5 Projects and 3 Cores aimed at: (1) providing quantitative animal tumorigenesis data on the relative effectiveness of specific HZE particles and SPE protons compared with gamma-rays in mouse models of AML and HCC; (2) providing a better understanding of the impact of radiation exposure on the processes involved in the initiation and in the progression of initiated cells toward the neoplastic phenotype; 3) delineating potential differences between low LET radiation and high LET radiation such as those encountered in space travel on these processes; 4) developing links between animal data and radiation-induced effects for AML in humans; and (5) developing biologically-based modeling approaches which are critical to link these biological effects to risks in humans.

Program Overview: The Radiation Carcinogenesis NSCOR was initiated in June 2009 and builds upon results obtained in its predecessor, the Leukemogenesis NSCOR. The Radiation Carcinogenesis NSCOR consists of four projects supported by three cores. The projects and cores are briefly described below.

Project 1. Dose response relationships for induction of AML and HCC as a function of radiation quality (project leader, Dr. Robert. L. Ullrich). This project is designed to compare the effects of irradiation with gamma-rays, select HZE particles, and protons on the induction of AML and hepatocellular carcinoma (HCC) using the C3H murine model.

Project 2. Mechanisms of radiation leukemogenesis (project leader, Dr. Michael M. Weil). The goal of Project 2 is to better understand how radiation leads to AML in a murine model and to generate data for the development of a biologically based model that can be used to predict AML risks from various HZE or high energy proton exposures.

Project 3. Pathogenesis of radiation-induced hepatocellular carcinoma (project leader, Dr. Robert L. Ullrich). The overall hypothesis of this project is that the dose response is likely to reflect both quantitative as well as qualitative differences in high LET effects. This overall hypothesis will be tested in 3 specific aims:

1. Quantify the frequency and progression of preneoplastic foci (including both hyperplastic and dysplastic foci) in livers of C3H/HeNCrl mice irradiated with either 137Cs gamma-rays or HZE ions.

2. Examine irradiated liver for evidence of increased oxidative damage and alterations in the regulation of inflammatory processes.

3. Determine tumorigenic effects following HZE and gamma-ray irradiation in murine models of hepatocellular carcinoma in which secondary “promoting” events play a significant role.

Project 4. Molecular and cytogenetic targets in murine and human AML. (Project leader, Dr. Michelle Le Beau). This project is designed to develop a cytogenetic and molecular profile of human radiation-induced AML, leading to an understanding of the key events and genetic pathways involved in the pathogenesis of this disease.

Core A (Core Director, Dr. F. Andrew Ray). The Biology Core facilitates the distribution of irradiated and control animals, tissues, cells, and other biological samples to investigators. This core is also responsible for conducting the irradiations required at the various sites for all projects.

Core B (Core Director, Dr. Michael Story). The Genomics and Biostatistics core provides appropriate genomic analyses, innovative statistical modeling, simulations, and data analyses for the projects.

Core C (Core Director, Dr. Robert Ullrich). The Administrative Core provides administrative, fiscal and management support for the Radiation Carcinogenesis NSCOR. This core also oversees the overall scientific conduct of the NSCOR and facilitates interactions between projects, core leaders and project investigators as well as interactions with the internal and external advisors.

Research Impact/Earth Benefits: This work will provide basic information on mechanisms of carcinogenesis as well as mechanisms specific to radiation-induced cancer.

Task Progress & Bibliography Information FY2011 
Task Progress: Project 1 involves irradiating large numbers of mice with 350 MeV 28Si ions, 600 MeV 56Fe ions, 1 GeV 56Fe ions, 137Cs gamma-rays, or protons (acute and low dose rate exposures mimicking the 1972 Solar Particle Event), and monitoring these mice for morbidity until they are 800 days of age. The mice are being necropsied and any tumors that arise characterized by histopathology. Because of the monitoring period, the immediate goal at the start of funding was to begin irradiating mice at NSRL as quickly as possible. To date, 3,800 mice have been irradiated.

For Project 2 we are developing experiments that will identify the leukemia initiating cell, determine the mechanism(s) leading to PU.1 loss, and determine the role of microsatellite instability in radiation-induced AML leukemogenesis. We are engineering a panel of mice with different sized chromosome 2 deletions encompassing PU.1 and various adjacent genes.

A multiplex for detection of microsatellite mutations in C3H mice has been constructed to test AML and HCC samples for evidence of MSI at the single cell level. The multiplex consists of five endogenous polyA mononucleotide repeats, including mBat-55d, mBat-56g, mBat-56j, mBat-57d, and mBat-59j. Similar assays have been developed for CBA and C57BL/6 strains, allowing us to compare MSI status in mice from any of these backgrounds. The software for analyzing MSI results has been improved to accommodate the need for high sample throughput and is now mostly automated. We are also examining mismatch repair (MMR) expression. Quantitative RT-PCR assays have been developed for Msh2, Msh3, Msh5, Msh6, Mlh1, and Pms2 genes along with a panel of mouse reference genes ActB, B2m, Gapdh, Hmbs, Hprt1, Rp113a, Sdha and Tbp. In addition, immunofluorescence assays for mouse Msh2, Msh3, Msh6 and Mlh1 are currently being optimized to test bone marrow and spleen samples for MMR protein expression.

In the fall of 2010, 60 mice were irradiated for Project 3 which is designed to examine the pathogenesis of heavy ion induced hepatocellular carcinoma. Our original design was to perform serial sacrifice studies for liver tissue analysis at 6, 9, and 12 months after irradiation following 3 Gy of gamma rays or 0.1 Gy of 1 GeV Iron based on our previous studies. Surprisingly, we are observing liver tumors much earlier than expected. As a result we added a sacrifice time point at 3 months post irradiation. Based on our results we may add a 1 month time point. Below is a list of dates for serial sacrifice.

The goal of Project 4 is to develop a cytogenetic and molecular profile of human radiation-induced AML, and to elucidate the key events and genetic pathways involved in the pathogenesis of this disease. To this end, we are profiling the genetic alterations in radiation-induced t-AML by (1) Cytogenetic analysis; (2) High-density SNP array analysis of copy number alterations (CNAs) and loss of heterozygosity; (3) Gene expression profiling; (4) Analysis of promoter methylation; and (5) miRNA expression profiling. To identify somatically acquired genetic CNAs in RT-induced t-AML, we are using a combination of high resolution copy number analysis using the Affymetrix Genome-Wide human SNP Array 6.0 platform (resolution of <5 kb), and targeted resequencing. Importantly, matched germline samples are available for most patients, allowing us to determine definitively if a CNA identified is somatically acquired. To date, we have extracted and hybridized DNA from 8 RT-induced t-AMLs. Analysis of recurrent CNAs by GISTIC identified several amplified (21q22.2) or deleted (5p13.3; 5q31.1 and 17p11.2) regions.

Core A personnel started large scale mouse irradiations at the earliest possible time after notification that the NSCOR had been funded. The first groups of mice were irradiated in November 2009 at the NSRL facility at BNL. As of March 2011, 3,800 mice have been irradiated and are currently being monitored daily for AML and HCC at UTMB Galveston. 350 MeV 28Si, 600 MeV 56Fe, 1GeV 56Fe, 137Cs gamma-Rays, and unirradiated controls were all irradiated at the NSRL facility at the Brookhaven National Laboratory. 100 mice will be irradiated with Acute Protons this May. An additional 500 C3H mice will be irradiated with 350 MeV 28Si in the spring of 2012, thus completing the acute irradiations for Project 1 tumorigenicity studies.

This year core B concentrated on concluding the expression analysis of radiation-induced AML samples. While we analyzed the data based upon both bone marrow cells and CD34+ cells last year, the final pathology had not been concluded. Interestingly, a handful of samples changed designation which meant the gene expression analysis and the aCGH had to be done. We now consider the pathology final as well as the genomic analysis. In addition to the gene expression and aCGH analysis we re-examined earlier data on DNA methylation where the Nimblegen methylation platform was used. Interestingly, there was little overall change in the gene expression data. AML and CD34+ or bone marrow cells all segregated into clusters of like samples. Some specific genes did drop out of our analysis as being differentially expressed. This was predominantly because the probes were no longer considered as valid for their gene target. This was especially true for genes associated with mis-match repair. While there were individual genes within the mis-match repair family differentially expressed, overall the pathway itself was not differentially expressed. Pu.1 is seen modestly upregulated even though one copy of the gene has been eliminated through chromosomal loss.

The aCGH analysis reveals 3 very modest regions as minimally deleted which affect a handful of genes including Pu.1 for which we are investigating. Other than the massive deletion of one copy of chromosome 2 was expected, and seen, there are few other chromosomal rearrangements to speak of. The methylation data was re-examined because of the more robust analysis tools available now that we did not have originally. Although there is nothing that stands out as far as specific genes whose promoters are methylated, we have identified a number of genes whose 3’ regions are highly methylated. This in fact, may be regulatory mechanism for miRNA expression and will be followed up on.

Bibliography: Description: (Last Updated: 07/25/2021) 

Show Cumulative Bibliography
 
Articles in Peer-reviewed Journals Fabre KM, Ramaiah L, Dregalla RC, Desaintes C, Weil MM, Bailey SM, Ullrich RL. "Murine Prkdc polymorphisms impact DNA-PKcs function." Radiat Res. 2011 Apr;175(4):493-500. Epub 2011 Jan 25. PMID: 21265624 , Apr-2011
Project Title:  NSCOR: NASA Specialized Center of Research on Radiation Carcinogenesis Reduce
Fiscal Year: FY 2010 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 06/01/2009  
End Date: 05/31/2014  
Task Last Updated: 04/19/2010 
Download report in PDF pdf
Principal Investigator/Affiliation:   Ullrich, Robert  Ph.D. / University of Texas Medical Branch 
Address:  301 University Blvd 
Comprehensive Cancer Center, MS 1048 
Galveston , TX 77555-5302 
Email: bullrich@utmb.edu 
Phone: 409-747-1935  
Congressional District: 14 
Web:  
Organization Type: UNIVERSITY 
Organization Name: University of Texas Medical Branch 
Joint Agency:  
Comments: NOTE: PI moved to UTMB from Colorado State University in late 2008 (6/2009) 
Co-Investigator(s)
Affiliation: 
Le Beau, Michelle  University of Chicago 
Bacher, Jeff  Promega Corporation 
Yu, Yongjia  University of Texas Medical Branch 
Story, Michael  University of Texas Southwestern Medical Center at Dallas 
Bedford, Joel  Colorado State University 
Weil, Michael  Colorado State University 
Ray, F  Colorado State University 
Ding, Lianghao  University of Texas Southwestern Medical Center at Dallas 
Xie, Yang  University of Texas Southwestern Medical Center 
Project Information: Grant/Contract No. NNX09AM08G 
Responsible Center: NASA JSC 
Grant Monitor: Cucinott1a, Francis  
Center Contact: 281-483-0968 
noaccess@nasa.gov 
Unique ID: 7564 
Solicitation / Funding Source: 2008 NSCOR Space Radiation NNJ08ZSA003N 
Grant/Contract No.: NNX09AM08G 
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) Cancer:Risk of Radiation Carcinogenesis
Human Research Program Gaps: (1) Cancer-202:Evaluate the contribution of genetic background/diversity on carcinogenesis risk.
Task Description: The goal of this NSCOR is to provide the information required to develop a rational scientific basis for estimation of risks for carcinogenesis in humans from exposure to radiation during space flight. Previous results from this Program found an unexpectedly low RBE value for acute myeloid leukemia (AML) induction by 1 GeV 56Fe ions. Systematic cytogenetic analyses suggested both microdosimetric factors related to the track structure of 1 GeV 56Fe ions and biological factors could account for this observation. In addition, these studies found an unexpected increase in hepatocellular carcinoma (HCC) at doses as low as 0.1 Gy of 1 GeV 56Fe ions but very little, if any, increase following gamma-ray exposure. These data suggest that processes associated with expansion and progression of initiated cells may play a more prominent role in HCC. If this is the case, it is possible that there are qualitative differences as well as quantitative in the effects of HZE irradiations. To expand on these results and to address the overall goal of this NSCOR a series of coordinated activities will conducted in 5 Projects and 3 Cores aimed at: (1) providing quantitative animal tumorigenesis data on the relative effectiveness of specific HZE particles and SPE protons compared with gamma-rays in mouse models of AML and HCC; (2) providing a better understanding of the impact of radiation exposure on the processes involved in the initiation and in the progression of initiated cells toward the neoplastic phenotype; 3) delineating potential differences between low LET radiation and high LET radiation such as those encountered in space travel on these processes; 4) developing links between animal data and radiation-induced effects for AML in humans; and (5) developing biologically-based modeling approaches which are critical to link these biological effects to risks in humans.

Program Overview: The Radiation Carcinogenesis NSCOR was initiated in June 2009 and builds upon results obtained in its predecessor, the Leukemogenesis NSCOR. The Radiation Carcinogenesis NSCOR consists of four projects supported by three cores. The projects and cores are briefly described below.

Project 1. Dose response relationships for induction of AML and HCC as a function of radiation quality (project leader, Dr. Robert. L. Ullrich). This project is designed to compare the effects of irradiation with gamma-rays, select HZE particles, and protons on the induction of AML and hepatocellular carcinoma (HCC) using the C3H murine model.

Project 2. Mechanisms of radiation leukemogenesis (project leader, Dr. Michael M. Weil). The goal of Project 2 is to better understand how radiation leads to AML in a murine model and to generate data for the development of a biologically based model that can be used to predict AML risks from various HZE or high energy proton exposures.

Project 3. Pathogenesis of radiation-induced hepatocellular carcinoma (project leader, Dr. Robert L. Ullrich). The overall hypothesis of this project is that the dose response is likely to reflect both quantitative as well as qualitative differences in high LET effects. This overall hypothesis will be tested in 3 specific aims:

1. Quantify the frequency and progression of preneoplastic foci (including both hyperplastic and dysplastic foci) in livers of C3H/HeNCrl mice irradiated with either 137Cs gamma-rays or HZE ions.

2. Examine irradiated liver for evidence of increased oxidative damage and alterations in the regulation of inflammatory processes.

3. Determine tumorigenic effects following HZE and gamma-ray irradiation in murine models of hepatocellular carcinoma in which secondary “promoting” events play a significant role.

Project 4. Molecular and cytogenetic targets in murine and human AML. (Project leader, Dr. Michelle Le Beau). This project is designed to develop a cytogenetic and molecular profile of human radiation-induced AML, leading to an understanding of the key events and genetic pathways involved in the pathogenesis of this disease.

Core A (Core Director, Dr. F. Andrew Ray). The Biology Core facilitates the distribution of irradiated and control animals, tissues, cells, and other biological samples to investigators. This core is also responsible for conducting the irradiations required at the various sites for all projects.

Core B (Core Director, Dr. Michael Story). The Genomics and Biostatistics core provides appropriate genomic analyses, innovative statistical modeling, simulations, and data analyses for the projects.

Core C (Core Director, Dr. Robert Ullrich). The Administrative Core provides administrative, fiscal and management support for the Radiation Carcinogenesis NSCOR. This core also oversees the overall scientific conduct of the NSCOR and facilitates interactions between projects, core leaders and project investigators as well as interactions with the internal and external advisors.

Research Impact/Earth Benefits: This work will provide basic information on mechanisms of carcinogenesis as well as mechanisms specific to radiation-induced cancer

Task Progress & Bibliography Information FY2010 
Task Progress: This is the first year of a 5 year program. As described in the grant, the major effort for this year has been irradiation of animals. So far more than 1300 animals have been irradiated at BNL with more scheduled for later in 2010.

Bibliography: Description: (Last Updated: 07/25/2021) 

Show Cumulative Bibliography
 
 None in FY 2010
Project Title:  NSCOR: NASA Specialized Center of Research on Radiation Carcinogenesis Reduce
Fiscal Year: FY 2009 
Division: Human Research 
Research Discipline/Element:
HRP SR:Space Radiation
Start Date: 06/01/2009  
End Date: 05/31/2014  
Task Last Updated: 08/07/2009 
Download report in PDF pdf
Principal Investigator/Affiliation:   Ullrich, Robert  Ph.D. / University of Texas Medical Branch 
Address:  301 University Blvd 
Comprehensive Cancer Center, MS 1048 
Galveston , TX 77555-5302 
Email: bullrich@utmb.edu 
Phone: 409-747-1935  
Congressional District: 14 
Web:  
Organization Type: UNIVERSITY 
Organization Name: University of Texas Medical Branch 
Joint Agency:  
Comments: NOTE: PI moved to UTMB from Colorado State University in late 2008 (6/2009) 
Co-Investigator(s)
Affiliation: 
Le Beau, Michelle  University of Chicago 
Bacher, Jeff  Promega Corporation 
Yu, Yongjia  University of Texas Medical Branch 
Hogan, Christopher  University of Colorado Denver 
Story, Michael  University of Texas Southwestern Medical Center at Dallas 
Bedford, Joel  Colorado State University 
Weil, Michael  Colorado State University 
Ray, F  Colorado State University 
Ding, Lianghao  University of Texas Southwestern Medical Center at Dallas 
Xie, Yang  University of Texas Southwestern Medical Center 
Borak, Thomas  Colorado State University 
Brenner, David  Columbia University 
Project Information: Grant/Contract No. NNX09AM08G 
Responsible Center: NASA JSC 
Grant Monitor:  
Center Contact:   
Unique ID: 7564 
Solicitation / Funding Source: 2008 NSCOR Space Radiation NNJ08ZSA003N 
Grant/Contract No.: NNX09AM08G 
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) Cancer:Risk of Radiation Carcinogenesis
Human Research Program Gaps: (1) Cancer-202:Evaluate the contribution of genetic background/diversity on carcinogenesis risk.
Task Description: The goal of this NSCOR is to provide the information required to develop a rational scientific basis for estimation of risks for carcinogenesis in humans from exposure to radiation during space flight. Previous results from this Program found an unexpectedly low RBE value for acute myeloid leukemia (AML) induction by 1 GeV 56Fe ions. Systematic cytogenetic analyses suggested both microdosimetric factors related to the track structure of 1 GeV 56Fe ions and biological factors could account for this observation. In addition, these studies found an unexpected increase in hepatocellular carcinoma (HCC) at doses as low as 0.1 Gy of 1 GeV 56Fe ions but very little, if any, increase following gamma-ray exposure. These data suggest that processes associated with expansion and progression of initiated cells may play a more prominent role in HCC. If this is the case, it is possible that there are qualitative differences as well as quantitative in the effects of HZE irradiations. To expand on these results and to address the overall goal of this NSCOR a series of coordinated activities will conducted in 5 Projects and 3 Cores aimed at: (1) providing quantitative animal tumorigenesis data on the relative effectiveness of specific HZE particles and SPE protons compared with gamma-rays in mouse models of AML and HCC; (2) providing a better understanding of the impact of radiation exposure on the processes involved in the initiation and in the progression of initiated cells toward the neoplastic phenotype; 3) delineating potential differences between low LET radiation and high LET radiation such as those encountered in space travel on these processes; 4) developing links between animal data and radiation-induced effects for AML in humans; and (5) developing biologically-based modeling approaches which are critical to link these biological effects to risks in humans.

Research Impact/Earth Benefits:

Task Progress & Bibliography Information FY2009 
Task Progress: New project for FY2009.

Bibliography: Description: (Last Updated: 07/25/2021) 

Show Cumulative Bibliography
 
 None in FY 2009