In the event of a radiation disaster or attack, high dose radiation exposure to humans causes Acute Radiation Syndrome (ARS). The hematopoietic system and gastrointestinal (GI) tract are among the most vulnerable tissues to radiation injury due to the presence of cycling stem cells that are sensitive to radiation. Loss of regenerative capacity in these organs leads to lethal toxicity. For example, damage to the bone marrow from total body irradiation leads to the hematopoietic syndrome. High doses of radiation to the abdomen results in GI syndrome characterized by damage to the intestinal epithelium, loss of mucosal barrier, and sepsis. While, medical countermeasures to mitigate the hematopoietic syndrome are available, there are currently no FDA approved therapies to mitigate the GI syndrome. We have demonstrated that p53 mediated expression of the cell cycle inhibitor p21 in GI epithelial cells is critical for survival of the GI syndrome. High p21 expression prevents damaged cells from progressing through mitosis and succumbing to mitotic catastrophe. In contrast, radiation kills p21-low, fast cycling LGR5+ GI stem cells in the base of the crypts. When these cells are lost, regeneration of the intestinal epithelium is dependent on a slow-cycling, and thus radioresistant, cell population that resides higher in the crypt. Therefore, identifying the GI epithelial cell type that is preserved by high p21 expression and capable of supporting tissue regeneration following injury will be important for developing mitigation strategies for GI syndrome. Recently published single cell RNA sequencing data from irradiated small intestine epithelial cells in mice identified Clu+ revival stem cells as slow cycling cells with high p21 levels that survive 24 hours and proliferate after high dose irradiation. Clu+ cells are capable of regenerating LGR5+ stem cells after injury and loss of this cell type sensitizes mice to GI syndrome lethality. Clu+ cells rely on the Yap1/Hippo signaling axis for maintenance and therefore we hypothesize that modulation of this pathway will mitigate the GI syndrome. In addition, Dclk1+ tuft cells were also identified to have high p21 expression and survive high dose radiation. These cells are chemosensory and secretory cells that support renewal of the GI epithelium following irradiation by maintaining the stem cell niche. We hypothesize that p53 regulation of p21 in these cell types is critical to intestinal recovery following radiation injury and that targeting these cells for mitigation is a promising strategy. We propose to dissect the mechanisms by which Clu+ revival stem cells and Dclk1+ tuft cells function to promote renewal of the GI epithelium after radiation injury using sophisticated genetically engineered mouse models. We will use targeted approaches with small molecule drugs in vivo and CRISPR/Cas9 screening methods in organoid cultures to identify novel mitigator targets that function specifically in these cell types to reduce lethal GI toxicity.

Public Health Relevance

Statement The proposed research is relevant to public health because (1) it aims to develop novel countermeasures against radiation with efficacy at 24 hours after radiation exposure and (2) it aims to define the mechanisms of action of the countermeasures which would provide data necessary for FDA approval of a drug according to the Animal Rule. Therefore, this research has the potential to lead to the approval of a new mitigator of the acute radiation syndrome, which is relevant to the part of the National Institute of Allergy and Infectious Disease?s mission to support research that develops countermeasures against radiation injury.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Program--Cooperative Agreements (U19)
Project #
2U19AI067798-16
Application #
9940009
Study Section
Special Emphasis Panel (ZAI1)
Project Start
Project End
Budget Start
2020-08-01
Budget End
2021-07-31
Support Year
16
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Duke University
Department
Type
DUNS #
044387793
City
Durham
State
NC
Country
United States
Zip Code
27705
Cline, John Mark; Dugan, Greg; Bourland, John Daniel et al. (2018) Post-Irradiation Treatment with a Superoxide Dismutase Mimic, MnTnHex-2-PyP5+, Mitigates Radiation Injury in the Lungs of Non-Human Primates after Whole-Thorax Exposure to Ionizing Radiation. Antioxidants (Basel) 7:
Farris, Michael; McTyre, Emory R; Okoukoni, Catherine et al. (2018) Cortical Thinning and Structural Bone Changes in Non-Human Primates after Single-Fraction Whole-Chest Irradiation. Radiat Res 190:63-71
Naqvi, Ibtehaj; Gunaratne, Ruwan; McDade, Jessica E et al. (2018) Polymer-Mediated Inhibition of Pro-invasive Nucleic Acid DAMPs and Microvesicles Limits Pancreatic Cancer Metastasis. Mol Ther 26:1020-1031
Ghandhi, Shanaz A; Turner, Helen C; Shuryak, Igor et al. (2018) Whole thorax irradiation of non-human primates induces persistent nuclear damage and gene expression changes in peripheral blood cells. PLoS One 13:e0191402
Castle, Katherine D; Daniel, Andrea R; Moding, Everett J et al. (2018) Mice Lacking RIP3 Kinase are not Protected from Acute Radiation Syndrome. Radiat Res 189:627-633
Fanning, K M; Pfisterer, B; Davis, A T et al. (2017) Changes in microvascular density differentiate metabolic health outcomes in monkeys with prior radiation exposure and subsequent skeletal muscle ECM remodeling. Am J Physiol Regul Integr Comp Physiol 313:R290-R297
Swanson, Karen V; Junkins, Robert D; Kurkjian, Cathryn J et al. (2017) A noncanonical function of cGAMP in inflammasome priming and activation. J Exp Med 214:3611-3626
Kurkjian, Cathryn J; Guo, Hao; Montgomery, Nathan D et al. (2017) The Toll-Like Receptor 2/6 Agonist, FSL-1 Lipopeptide, Therapeutically Mitigates Acute Radiation Syndrome. Sci Rep 7:17355
Racioppi, Luigi; Lento, William; Huang, Wei et al. (2017) Calcium/calmodulin-dependent kinase kinase 2 regulates hematopoietic stem and progenitor cell regeneration. Cell Death Dis 8:e3076
Himburg, Heather A; Doan, Phuong L; Quarmyne, Mamle et al. (2017) Dickkopf-1 promotes hematopoietic regeneration via direct and niche-mediated mechanisms. Nat Med 23:91-99

Showing the most recent 10 out of 197 publications