Cargando…

Long-Term Differential Changes in Mouse Intestinal Metabolomics after γ and Heavy Ion Radiation Exposure

Tissue consequences of radiation exposure are dependent on radiation quality and high linear energy transfer (high-LET) radiation, such as heavy ions in space is known to deposit higher energy in tissues and cause greater damage than low-LET γ radiation. While radiation exposure has been linked to i...

Descripción completa

Detalles Bibliográficos
Autores principales: Cheema, Amrita K., Suman, Shubhankar, Kaur, Prabhjit, Singh, Rajbir, Fornace, Albert J., Datta, Kamal
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3903607/
https://www.ncbi.nlm.nih.gov/pubmed/24475228
http://dx.doi.org/10.1371/journal.pone.0087079
_version_ 1782301120875462656
author Cheema, Amrita K.
Suman, Shubhankar
Kaur, Prabhjit
Singh, Rajbir
Fornace, Albert J.
Datta, Kamal
author_facet Cheema, Amrita K.
Suman, Shubhankar
Kaur, Prabhjit
Singh, Rajbir
Fornace, Albert J.
Datta, Kamal
author_sort Cheema, Amrita K.
collection PubMed
description Tissue consequences of radiation exposure are dependent on radiation quality and high linear energy transfer (high-LET) radiation, such as heavy ions in space is known to deposit higher energy in tissues and cause greater damage than low-LET γ radiation. While radiation exposure has been linked to intestinal pathologies, there are very few studies on long-term effects of radiation, fewer involved a therapeutically relevant γ radiation dose, and none explored persistent tissue metabolomic alterations after heavy ion space radiation exposure. Using a metabolomics approach, we report long-term metabolomic markers of radiation injury and perturbation of signaling pathways linked to metabolic alterations in mice after heavy ion or γ radiation exposure. Intestinal tissues (C57BL/6J, female, 6 to 8 wks) were analyzed using ultra performance liquid chromatography coupled with electrospray quadrupole time-of-flight mass spectrometry (UPLC-QToF-MS) two months after 2 Gy γ radiation and results were compared to an equitoxic (56)Fe (1.6 Gy) radiation dose. The biological relevance of the metabolites was determined using Ingenuity Pathway Analysis, immunoblots, and immunohistochemistry. Metabolic profile analysis showed radiation-type-dependent spatial separation of the groups. Decreased adenine and guanosine and increased inosine and uridine suggested perturbed nucleotide metabolism. While both the radiation types affected amino acid metabolism, the (56)Fe radiation preferentially altered dipeptide metabolism. Furthermore, (56)Fe radiation caused upregulation of ‘prostanoid biosynthesis’ and ‘eicosanoid signaling’, which are interlinked events related to cellular inflammation and have implications for nutrient absorption and inflammatory bowel disease during space missions and after radiotherapy. In conclusion, our data showed for the first time that metabolomics can not only be used to distinguish between heavy ion and γ radiation exposures, but also as a radiation-risk assessment tool for intestinal pathologies through identification of biomarkers persisting long after exposure.
format Online
Article
Text
id pubmed-3903607
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-39036072014-01-28 Long-Term Differential Changes in Mouse Intestinal Metabolomics after γ and Heavy Ion Radiation Exposure Cheema, Amrita K. Suman, Shubhankar Kaur, Prabhjit Singh, Rajbir Fornace, Albert J. Datta, Kamal PLoS One Research Article Tissue consequences of radiation exposure are dependent on radiation quality and high linear energy transfer (high-LET) radiation, such as heavy ions in space is known to deposit higher energy in tissues and cause greater damage than low-LET γ radiation. While radiation exposure has been linked to intestinal pathologies, there are very few studies on long-term effects of radiation, fewer involved a therapeutically relevant γ radiation dose, and none explored persistent tissue metabolomic alterations after heavy ion space radiation exposure. Using a metabolomics approach, we report long-term metabolomic markers of radiation injury and perturbation of signaling pathways linked to metabolic alterations in mice after heavy ion or γ radiation exposure. Intestinal tissues (C57BL/6J, female, 6 to 8 wks) were analyzed using ultra performance liquid chromatography coupled with electrospray quadrupole time-of-flight mass spectrometry (UPLC-QToF-MS) two months after 2 Gy γ radiation and results were compared to an equitoxic (56)Fe (1.6 Gy) radiation dose. The biological relevance of the metabolites was determined using Ingenuity Pathway Analysis, immunoblots, and immunohistochemistry. Metabolic profile analysis showed radiation-type-dependent spatial separation of the groups. Decreased adenine and guanosine and increased inosine and uridine suggested perturbed nucleotide metabolism. While both the radiation types affected amino acid metabolism, the (56)Fe radiation preferentially altered dipeptide metabolism. Furthermore, (56)Fe radiation caused upregulation of ‘prostanoid biosynthesis’ and ‘eicosanoid signaling’, which are interlinked events related to cellular inflammation and have implications for nutrient absorption and inflammatory bowel disease during space missions and after radiotherapy. In conclusion, our data showed for the first time that metabolomics can not only be used to distinguish between heavy ion and γ radiation exposures, but also as a radiation-risk assessment tool for intestinal pathologies through identification of biomarkers persisting long after exposure. Public Library of Science 2014-01-27 /pmc/articles/PMC3903607/ /pubmed/24475228 http://dx.doi.org/10.1371/journal.pone.0087079 Text en © 2014 Cheema et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Cheema, Amrita K.
Suman, Shubhankar
Kaur, Prabhjit
Singh, Rajbir
Fornace, Albert J.
Datta, Kamal
Long-Term Differential Changes in Mouse Intestinal Metabolomics after γ and Heavy Ion Radiation Exposure
title Long-Term Differential Changes in Mouse Intestinal Metabolomics after γ and Heavy Ion Radiation Exposure
title_full Long-Term Differential Changes in Mouse Intestinal Metabolomics after γ and Heavy Ion Radiation Exposure
title_fullStr Long-Term Differential Changes in Mouse Intestinal Metabolomics after γ and Heavy Ion Radiation Exposure
title_full_unstemmed Long-Term Differential Changes in Mouse Intestinal Metabolomics after γ and Heavy Ion Radiation Exposure
title_short Long-Term Differential Changes in Mouse Intestinal Metabolomics after γ and Heavy Ion Radiation Exposure
title_sort long-term differential changes in mouse intestinal metabolomics after γ and heavy ion radiation exposure
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3903607/
https://www.ncbi.nlm.nih.gov/pubmed/24475228
http://dx.doi.org/10.1371/journal.pone.0087079
work_keys_str_mv AT cheemaamritak longtermdifferentialchangesinmouseintestinalmetabolomicsaftergandheavyionradiationexposure
AT sumanshubhankar longtermdifferentialchangesinmouseintestinalmetabolomicsaftergandheavyionradiationexposure
AT kaurprabhjit longtermdifferentialchangesinmouseintestinalmetabolomicsaftergandheavyionradiationexposure
AT singhrajbir longtermdifferentialchangesinmouseintestinalmetabolomicsaftergandheavyionradiationexposure
AT fornacealbertj longtermdifferentialchangesinmouseintestinalmetabolomicsaftergandheavyionradiationexposure
AT dattakamal longtermdifferentialchangesinmouseintestinalmetabolomicsaftergandheavyionradiationexposure