Space radiation triggers persistent stress response, increases senescent signaling, and decreases cell migration in mouse intestine

Proliferative gastrointestinal (GI) tissue is radiation-sensitive, and heavy-ion space radiation with its high-linear energy transfer (high-LET) and higher damaging potential than low-LET γ-rays is predicted to compromise astronauts’ GI function. However, much uncertainty remains in our understandin...

Full description

Saved in:
Bibliographic Details
Published inProceedings of the National Academy of Sciences - PNAS Vol. 115; no. 42; pp. E9832 - E9841
Main Authors Kumar, Santosh, Suman, Shubhankar, Fornace, Albert J., Datta, Kamal
Format Journal Article
LanguageEnglish
Published United States National Academy of Sciences 16.10.2018
SeriesPNAS Plus
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Proliferative gastrointestinal (GI) tissue is radiation-sensitive, and heavy-ion space radiation with its high-linear energy transfer (high-LET) and higher damaging potential than low-LET γ-rays is predicted to compromise astronauts’ GI function. However, much uncertainty remains in our understanding of how heavy ions affect coordinated epithelial cell migration and extrusion, which are essential for GI homeostasis. Here we show using mouse small intestine as a model and BrdU pulse labeling that cell migration along the crypt–villus axis is persistently decreased after a low dose of heavy-ion 56Fe radiation relative to control and γ-rays. Wnt/β-catenin and its downstream EphrinB/EphB signaling are key to intestinal epithelial cell (IEC) proliferation and positioning during migration, and both are upregulated after 56Fe radiation. Conversely, factors involved in cell polarity and adhesion and cell–extracellular matrix interactions were persistently down-regulated after 56Fe irradiation—potentially altering cytoskeletal remodeling and cell extrusion. 56Fe radiation triggered a time-dependent increase in γH2AX foci and senescent cells but without a noticeable increase in apoptosis. Some senescent cells acquired the senescence-associated secretory phenotype, and this was accompanied by increased IEC proliferation, implying a role for progrowth inflammatory factors. Collectively, this study demonstrates a unique phenomenon of heavy-ion radiation-induced persistently delayed IEC migration involving chronic sublethal genotoxic and oncogenic stress-induced altered cytoskeletal dynamics, which were seen even a year later. When considered along with changes in barrier function and nutrient absorption factors as well as increased intestinal tumorigenesis, our in vivo data raise a serious concern for long-duration deep-space manned missions.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
1S.K. and S.S. contributed equally to this work.
Edited by James E. Cleaver, University of California, San Francisco, CA, and approved August 30, 2018 (received for review May 1, 2018)
Author contributions: S.K., S.S., and K.D. designed research; S.K. and S.S. performed research; S.K., S.S., and K.D. analyzed data; and S.K., S.S., A.J.F., and K.D. wrote the paper.
ISSN:0027-8424
1091-6490
DOI:10.1073/pnas.1807522115