Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(17)2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37686516

ABSTRACT

Estimation of cancer risk among astronauts planning to undertake future deep-space missions requires understanding the quantitative and qualitative differences in radiogenic cancers after low- and high-LET radiation exposures. Previously, we reported a multifold higher RBE for high-LET radiation-induced gastrointestinal (GI) tumorigenesis in Apc1638N/+ mice. Using the same model system, i.e., Apc1638N/+ mice, here, we report qualitative differences in the cellular phenotype of low- and high-LET radiation-induced GI tumors. Stem cell (SC) phenotypes were identified using BMI1, ALDH1, CD133, DCLK1, MSI1, and LGR5 markers in low (γ-rays)- and high (56Fe)-LET radiation-induced and spontaneous tumors. We also assessed the expression of these markers in the adjacent normal mucosa. All six of these putative SC markers were shown to be overexpressed in tumors compared to the adjacent normal intestinal tissue. A differential SC phenotype for spontaneous and radiogenic intestinal tumors in Apc1638N/+ mice was observed, where the ALDH1, BMI1, CD133, MSI1, and DCLK1 expressing cells were increased, while LGR5 expressing cells were decreased in 56Fe-induced tumors compared to γ-ray-induced and spontaneous tumors. Furthermore, higher ß-catenin activation (marked by nuclear localization) was observed in 56Fe-induced tumors compared to γ and spontaneous tumors. Since differential tumor cell phenotype along with activated ß-catenin may very well affect malignant progression, our findings are relevant to understanding the higher carcinogenic risk of high-LET radiation. This study has implications for the assessment of GI-cancer risk among astronauts, as well as for the estimation of secondary cancer risk among patients receiving hadron therapy, considering that our results indicate increased stemness properties after radiation.

2.
Curr Oncol ; 30(6): 5497-5514, 2023 06 07.
Article in English | MEDLINE | ID: mdl-37366899

ABSTRACT

Ionizing radiation (IR) dose, dose rate, and linear energy transfer (LET) determine cellular DNA damage quality and quantity. High-LET heavy ions are prevalent in the deep space environment and can deposit a much greater fraction of total energy in a shorter distance within a cell, causing extensive DNA damage relative to the same dose of low-LET photon radiation. Based on the DNA damage tolerance of a cell, cellular responses are initiated for recovery, cell death, senescence, or proliferation, which are determined through a concerted action of signaling networks classified as DNA damage response (DDR) signaling. The IR-induced DDR initiates cell cycle arrest to repair damaged DNA. When DNA damage is beyond the cellular repair capacity, the DDR for cell death is initiated. An alternative DDR-associated anti-proliferative pathway is the onset of cellular senescence with persistent cell cycle arrest, which is primarily a defense mechanism against oncogenesis. Ongoing DNA damage accumulation below the cell death threshold but above the senescence threshold, along with persistent SASP signaling after chronic exposure to space radiation, pose an increased risk of tumorigenesis in the proliferative gastrointestinal (GI) epithelium, where a subset of IR-induced senescent cells can acquire a senescence-associated secretory phenotype (SASP) and potentially drive oncogenic signaling in nearby bystander cells. Moreover, DDR alterations could result in both somatic gene mutations as well as activation of the pro-inflammatory, pro-oncogenic SASP signaling known to accelerate adenoma-to-carcinoma progression during radiation-induced GI cancer development. In this review, we describe the complex interplay between persistent DNA damage, DDR, cellular senescence, and SASP-associated pro-inflammatory oncogenic signaling in the context of GI carcinogenesis.


Subject(s)
Gastrointestinal Neoplasms , Signal Transduction , Humans , Radiation, Ionizing , Cellular Senescence/physiology , DNA Damage , Gastrointestinal Neoplasms/genetics
3.
Life Sci Space Res (Amst) ; 36: 116-122, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36682820

ABSTRACT

Female astronauts inevitably exposed to galactic cosmic radiation (GCR) are considered at a greater risk for mammary cancer development. The purpose of this study is to assess the status of mammary cancer-associated preneoplasia markers after GCR and γ-ray irradiation using a mouse model of human mammary cancer. Female ApcMin/+ mice were irradiated to 50 cGy of either γ-ray (137Cs) or full-spectrum simulated galactic cosmic radiation (GCR) (33-beam), and at 110 - 120 days post-irradiation mice were euthanized, and normal-appearing mammary tissues were analyzed for histological and molecular markers of preneoplasia. Whole-mount staining, hematoxylin and eosin-based histological assessment, and Cyclin D1 immunohistochemistry (IHC) were performed to analyze ductal outgrowth and cell proliferation. Additionally, mRNA expression of known mammary preneoplasia markers (Muc1, Exo1, Foxm1, Depdc1a, Nusap1, Spp1, and Rrm2) was analyzed using qPCR, and their respective protein expression was validated using immunohistochemistry. A significant increase in ductal outgrowth and cell proliferation in mammary tissues of GCR-irradiated mice was noted which indicates a higher risk of mammary cancer, relative to γ-rays. Increased mRNA and protein expression of Spp1 was observed in the GCR group, relative to γ-rays. This study demonstrates the plausibility of Spp1 as a preneoplasia marker in the early detection of mammary cancer after space radiation exposure.


Subject(s)
Breast Neoplasms , Cosmic Radiation , Space Flight , Female , Humans , Astronauts , Breast , Cosmic Radiation/adverse effects , Osteopontin , Animals , Mice
4.
Mol Biol Rep ; 50(3): 2067-2076, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36542238

ABSTRACT

BACKGROUND: Exposure to ionizing is known to cause persistent cellular oxidative stress and NADPH oxidase (Nox) is a major source of cellular oxidant production. Chronic oxidative stress is associated with a myriad of human diseases including gastrointestinal cancer. However, the roles of NADPH oxidase in relation of long-term oxidative stress in colonic epithelial cells after radiation exposure are yet to be clearly established. METHODS AND RESULTS: Mice were exposed either to sham or to 0.5 Gy γ radiation, and NADPH oxidase, oxidative stress, and related signaling pathways were assessed in colon samples 60 days after exposure. Radiation exposure led to increased expression of colon-specific NADPH oxidase isoform, Nox1, as well as upregulation of its modifiers such as Noxa1 and Noxo1 at the mRNA and protein level. Co-immunoprecipitation experiments showed enhanced binding of Rac1, an activator of NADPH oxidase, to Nox1. Increased 4-hydroxynonenal, 8-oxo-dG, and γH2AX along with higher protein carbonylation levels suggest increased oxidative stress after radiation exposure. Immunoblot analysis demonstrates upregulation of Ras/p38 pathway, and Gata6 and Hif1α after irradiation. Increased staining of ß-catenin, cyclinD1, and Ki67 after radiation was also observed. CONCLUSIONS: In summary, data show that exposure to a low dose of radiation was associated with upregulation of NADPH oxidase and its modifiers along with increased Ras/p38/Gata6 signaling in colon. When considered along with oxidative damage and proliferative markers, our observations suggest that the NADPH oxidase pathway could be playing a critical role in propagating long-term oxidative stress after radiation with implications for colon carcinogenesis.


Subject(s)
NADPH Oxidases , Oxidative Stress , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Colon/metabolism , Gamma Rays , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidative Stress/radiation effects , Reactive Oxygen Species/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases , ras Proteins
5.
Mol Neurobiol ; 60(1): 292-302, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36264435

ABSTRACT

The inferior colliculus (IC) is critical in initiating acoustically evoked alcohol withdrawal-induced seizures (AWSs). Recently, we reported that systemic inhibition of Ca2+ entry via the reverse mode activity of the Na+/Ca2+ exchanger (NCXrev) suppressed AWSs, suggesting remodeling of NCX expression and function, at least in the IC, the site of AWS initiation. Here, we probe putative changes in protein expression in the IC of NCX isoforms, including NCX type 1 (NCX1), 2 (NCX2), and 3 (NCX3). We also evaluated the efficacy of targeted inhibition of NCX1rev and NCX3rev activity in the IC on the occurrence and severity of AWSs using SN-6 and KB-R943, respectively. We used our well-characterized alcohol intoxication/withdrawal model associated with enhanced AWS susceptibility. IC tissues from the alcohol-treated group were collected 3 h (before the onset of AWS susceptibility), 24 h (when AWS susceptibility is maximal), and 48 h (when AWS susceptibility is resolved) following alcohol withdrawal; in comparison, IC tissues from the control-treated group were collected at 24 h after the last gavage. Analysis shows that NCX1 protein levels were markedly higher 3 and 24 h following alcohol withdrawal. However, NCX3 protein levels were only higher 3 h following alcohol withdrawal. The analysis also reveals that bilateral microinjections of SN-6 (but not KB-R7943) within the IC markedly suppressed the occurrence and severity of AWSs. Together, these findings indicate that NCX1 is a novel molecular target that may play an essential role in the pathogenesis and pathophysiology of AWSs.


Subject(s)
Alcohol Withdrawal Seizures , Alcoholism , Inferior Colliculi , Substance Withdrawal Syndrome , Rats , Animals , Alcohol Withdrawal Seizures/metabolism , Inferior Colliculi/metabolism , Sodium-Calcium Exchanger/metabolism , Calcium/metabolism
6.
PLoS One ; 17(12): e0279771, 2022.
Article in English | MEDLINE | ID: mdl-36584137

ABSTRACT

Space radiation-induced gastrointestinal (GI) cancer risk models for future interplanetary astronauts are being developed that primarily rely on quantitative animal model studies to assess radiation-quality effects of heavy-ion space radiation exposure in relation to γ-rays. While current GI-cancer risk estimation efforts are focused on sporadic GI-cancer mouse models, emerging in-vivo data on heavy-ion radiation-induced long-term GI-inflammation are indicative of a higher but undetermined risk of GI-inflammation associated cancers, such as colitis-associated cancer (CAC). Therefore, we aimed to assess radiation quality effects on colonic inflammation, colon cancer incidence, and associated signaling events using an in-vivo CAC model i.e., Il10-/- mice. Male Il10-/- mice (8-10 weeks, n = 12/group) were irradiated with either sham, γ-rays or heavy-ions (28Si or 56Fe), and histopathological assessments for colitis and CAC were conducted at 2.5 months post-exposure. qPCR analysis for inflammation associated gene transcripts (Ptges and Tgfb1), and in-situ staining for markers of cell-proliferation (phospho-histone H3), oncogenesis (active-ß-catenin, and cyclin D1), and inflammation (phospho-p65NF-κB, iNOS, and COX2) were performed. Significantly higher colitis and CAC frequency were noted after heavy-ion exposure, relative to γ and control mice. Higher CAC incidence after heavy-ion exposure was associated with greater activation of ß-catenin and NF-κB signaling marked by induced expression of common downstream inflammatory (iNOS and COX2) and pro-proliferative (Cyclin D1) targets. In summary, IR-induced colitis and CAC incidence in Il10-/- mice depends on radiation quality and display co-activation of ß-catenin and NF-κB signaling.


Subject(s)
Colitis , Colonic Neoplasms , Gastritis , Neoplasms, Radiation-Induced , Mice , Male , Animals , NF-kappa B/metabolism , Cyclin D1/genetics , beta Catenin/genetics , beta Catenin/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Colitis/chemically induced , Carcinogenesis , Colonic Neoplasms/pathology , Inflammation/complications , Neoplasms, Radiation-Induced/genetics , Gastritis/complications , Mice, Inbred C57BL , Disease Models, Animal
7.
Radiat Environ Biophys ; 61(4): 631-637, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36167896

ABSTRACT

Gastrointestinal (GI) cancer risk among astronauts after encountering galactic cosmic radiation (GCR) is predicted to exceed safe permissible limits in long duration deep-space missions. Current predictions are based on relative biological effectiveness (RBE) values derived from in-vivo studies using single-ion beams, while GCR is essentially a mixed radiation field composed of protons (H), helium (He), and heavy ions. Therefore, a sequentially delivered proton (H) → Helium (He) → Oxygen (O) → Silicon (Si) beam was designed to simulate simplified-mixed-field GCR (Smf-GCR), and Apc1638N/+ mice were total-body irradiated to sham or γ (157Cs) or Smf-GCR followed by assessment of GI-tumorigenesis at 150 days post-exposure. Further, GI-tumor data from equivalent doses of heavy-ions (i.e., 0.05 Gy of O and Si) in 0.5 Gy of Smf-GCR were compared to understand the contributions of heavy-ions in GI-tumorigenesis. The Smf-GCR-induced tumor and carcinoma count were significantly greater than γ-rays, and male preponderance for GI-tumorigenesis was consistent with our earlier findings. Comparison of tumor data from Smf-GCR and equivalent doses of heavy ions revealed an association between higher GI-tumorigenesis where dose received from heavy-ions contributed to > 95% of the total GI-tumorigenic effect observed after Smf-GCR. This study provides the first experimental evidence that cancer risk after GCR exposure could largely depend on doses received from constituent heavy-ions.


Subject(s)
Cosmic Radiation , Heavy Ions , Neoplasms, Radiation-Induced , Radiation Exposure , Space Flight , Mice , Male , Animals , Heavy Ions/adverse effects , Helium , Cosmic Radiation/adverse effects , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Carcinogenesis , Protons
8.
Heliyon ; 8(1): e08691, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35028468

ABSTRACT

Low-LET photon radiation-induced persistent alterations in bone marrow (BM) cells are well documented in total-body irradiated (TBI) rodents and also among radiotherapy patients. However, the late effects of protons and high-LET heavy-ion radiation on BM cells and its implications in osteoclastogenesis are not fully understood. Therefore, C57BL6/J female mice (8 weeks; n = 10/group) were irradiated to sham, and 1 Gy of the proton (0.22 keV/µm), or high-LET 56Fe-ions (148 keV/µm) and at 60 d post-exposure, mice were sacrificed and femur sections were obtained for histological, cellular and molecular analysis. Cell proliferation (PCNA), cell death (active caspase-3), senescence (p16), osteoclast (RANK), osteoblast (OPG), osteoblast progenitor (c-Kit), and osteoclastogenesis-associated secretory factors (like RANKL) were assessed using immunostaining. While no change in cell proliferation and apoptosis between control and irradiated groups was noted, the number of BM megakaryocytes was significantly reduced in irradiated mice at 60 d post-exposure. A remarkable increase in p16 positive cells indicated a persistent increase in cell senescence, whereas increased RANKL/OPG ratio, reductions in the number of osteoblast progenitor cells, and osteocalcin provided clear evidence that exposure to both proton and 56Fe-ions promotes pro-osteoclastogenic activity in BM. Among irradiated groups, 56Fe-induced alterations in the BM cellularity and osteoclastogenesis were significantly greater than the protons that demonstrated a radiation quality-dependent effect. This study has implications in understanding the role of IR-induced late changes in the BM cells and its involvement in bone degeneration among deep-space astronauts, and also in patients undergoing proton or heavy-ion radiotherapy.

9.
Life Sci Space Res (Amst) ; 31: 85-91, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34689954

ABSTRACT

Inevitable exposure to high-LET ionizing radiation (IR) present in galactic cosmic radiation (GCR) could enhance gastrointestinal (GI) cancer incidence among astronauts undertaking deep space exploration and GI-cancer mortality has been predicted to far exceed NASA's limit of < 3% REID (Radiation exposure-induced death) from cancer. Therefore, the development of countermeasure agents against high-LET radiation-induced GI cancer is needed to safeguard astronauts during and after an outer space mission. The cyclooxygenase-2/prostaglandin E2 (COX2/PGE2) mediated activation of pro-inflammatory and oncogenic signaling has been reported to play an important role in persistent inflammation and GI-tumorigenesis after high-LET radiation exposure. Therefore, aspirin, a well-known inhibitor of the COX/PGE2 pathway, was evaluated as a potential countermeasure against 28Si-induced PGE2 and tumorigenesis in Apc1638N/+, a murine model of human GI-cancer. Animals were fed either standard or aspirin supplemented diet (75, 150, or 300 mg/day of human equivalent dose) starting at the age of 4 weeks and continued till the end of the study, while mice were exposed to 28Si-ions (300 MeV/n; 69 keV/µm) at the age of 8 weeks. Serum PGE2 level, GI tumor size (>2mm2), number, and cluster (>5 adjoining tumors) were analyzed at 150 days post-exposure. Aspirin led to a significant reduction in PGE2 in a dose-dependent manner but did not reduce 28Si-induced GI tumorigenesis even at the highest (300 mg/day) dose. In summary, this study suggests that aspirin could reduce high-LET IR-induced pro-inflammatory PGE2 levels, however, lacks the ability to reduce high-LET IR-induced GI tumorigenesis in Apc1638N/+ mice.


Subject(s)
Cosmic Radiation , Dinoprostone , Animals , Aspirin , Carcinogenesis , Diet , Mice
10.
Int J Radiat Oncol Biol Phys ; 107(1): 202-211, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32036005

ABSTRACT

PURPOSE: Exposure to ionizing radiation increases risk of breast cancer. Although proton radiation is encountered in outer space and in medicine, we do not fully understand breast cancer risks from protons owing to limited in vivo data. The purpose of this study was to comparatively assess the effects of γ-rays and protons on mammary tumorigenesis in APCMin/+ mice. METHODS AND MATERIALS: Female APCMin/+ mice were exposed to 1 GeV protons (1.88 or 4.71 Gy) and 137Cs γ-rays (2 or 5 Gy). Mice were euthanized 100 to 110 days after irradiation, at which point mammary tumors were scored, tumor grades were assessed, and relative biological effectiveness was calculated. Molecular phenotypes were determined by assessing estrogen receptor α (ERα) and human epidermal growth factor receptor 2 (HER2) status. ERα downstream signaling was assessed by immunohistochemistry. RESULTS: Exposure to proton radiation led to increased mammary tumor frequency at both proton radiation doses compared with γ-rays. The calculated relative biological effectiveness for proton radiation-induced mammary tumorigenesis was 3.11 for all tumors and >5 for malignant tumors relative to γ-rays. Tumor frequency per unit of radiation was higher at the lower dose, suggesting a saturation effect at the higher dose. Protons induced more adenocarcinomas relative to γ-rays, and proton-induced tumors show greater ERα and HER2 positivity and higher activation of the ERα downstream PI3K/Akt and cyclin D1 pathways relative to γ-rays. CONCLUSIONS: Our data demonstrate that protons pose a higher risk of mammary tumorigenesis relative to γ-rays. We also show that proton radiation-induced tumors in APCMin/+ mice are ERα- and HER2-positive, which is consistent with our previous data on radiation-induced estrogenic response in wild-type mice. Although this study establishes APCMin/+ as a model with adequate signal-to-noise ratio for space radiation-induced mammary tumorigenesis, further studies will be required to address the uncertainties in space radiation-induced breast cancer risk estimation.


Subject(s)
Carcinogenesis/radiation effects , Estrogen Receptor alpha/metabolism , Gamma Rays/adverse effects , Mammary Neoplasms, Experimental/pathology , Proton Therapy/adverse effects , Receptor, ErbB-2/metabolism , Relative Biological Effectiveness , Animals , Female , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/metabolism , Mice
11.
Life Sci Space Res (Amst) ; 27: 27-32, 2020 Nov.
Article in English | MEDLINE | ID: mdl-34756227

ABSTRACT

Long-duration space missions outside low earth orbit will expose astronauts to a cumulative dose of high-energy particle radiation especially to highly damaging heavy ion radiation, which poses considerable risk to astronauts' health. The purpose of the current study was to quantitatively identify oxidatively induced DNA base modifications and assess status of the repair pathways involved in removing the modified bases in mouse intestinal cells after exposure to γ-rays and iron radiation. Mice (C57BL/6J; 6 to 8 weeks; female) were exposed to 0.5 Gy of either γ-rays or iron radiation and control mice were sham-irradiated. Intestinal tissues were collected 2 months after radiation. DNA base lesions were measured using gas chromatography-tandem mass spectrometry with isotope­dilution. Base excision repair (BER) and nucleotide excision repair (NER) pathways were assessed using PCR and immunoblotting. Effects of iron radiation were compared to γ-rays and sham-irradiated controls. Exposure to iron radiation resulted in significantly higher levels of several DNA base lesions relative to control animals and those exposed to γ radiation. Assessment of BER and NER showed downregulation of pathway factors both at the RNA as well as at the protein levels. Our results not only provide important insight into DNA damage pattern in intestinal cells in response to iron radiation, but they also confirm our previous immunohistochemistry data on oxidatively induced DNA damage. We suggest that downregulation of the BER and NER pathways is contributing to ongoing DNA base damages long time after radiation exposure and has implications for chronic diseases including gastrointestinal diseases after heavy ion radiation exposure during space travel.


Subject(s)
Heavy Ions , Animals , DNA , DNA Damage , DNA Repair , Female , Heavy Ions/adverse effects , Mice , Mice, Inbred C57BL
12.
Int J Radiat Oncol Biol Phys ; 105(3): 525-536, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31271826

ABSTRACT

PURPOSE: Proton radiation is a major component of the radiation field in outer space and is used clinically in radiation therapy of resistant cancers. Although epidemiologic studies in atom bomb survivors and radiologic workers have established radiation as a risk factor for colorectal cancer (CRC), we have yet to determine the risk of CRC posed by proton radiation owing to a lack of sufficient human or animal data. The purpose of the current study was to quantitatively and qualitatively characterize differential effects of acute and fractionated high-energy protons on colorectal carcinogenesis. METHODS AND MATERIALS: We used ApcMin/+ mice, a well-studied CRC model, to examine acute versus fractionated proton radiation-induced differences in intestinal tumorigenesis and associated signaling pathways. Mice were exposed to 1.88 Gy of proton radiation delivered in a single fraction or in 4 equal daily fractions (0.47 Gy × 4). Intestinal tumor number and grade were scored 100 to 110 days after irradiation, and tumor and tumor-adjacent normal tissues were harvested to assess proliferative ß-catenin/Akt pathways and DNA damage response and repair pathways relevant to colorectal carcinogenesis. RESULTS: Significantly higher intestinal tumor number and grade, along with decreased differentiation, were observed after acute radiation relative to fractionated radiation. Acute protons induced upregulation of ß-catenin and Akt pathways with increased proliferative marker phospho-histone H3. Increased DNA damage along with decreased DNA repair factors involved in mismatch repair and nonhomologous end joining were also observed after exposure to acute protons. CONCLUSIONS: We show increased γH2AX, 53BP1, and 8-oxo-dG, suggesting that increased ongoing DNA damage along with decreased DNA repair factors and increased proliferative responses could be triggering a higher number of intestinal tumors after acute relative to fractionated proton exposures in ApcMin/+ mice. Taken together, our data suggest greater carcinogenic potential of acute relative to fractionated proton radiation.


Subject(s)
DNA Breaks, Double-Stranded , DNA Mismatch Repair , Intestinal Neoplasms/genetics , Neoplasms, Radiation-Induced/genetics , Protons/adverse effects , 8-Hydroxy-2'-Deoxyguanosine/metabolism , Animals , Carcinogenesis/genetics , Cell Differentiation , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclin D1/metabolism , DNA End-Joining Repair , Disease Models, Animal , Dose Fractionation, Radiation , Female , Gene Expression , Genes, APC , Histones/metabolism , Immunoblotting/methods , Intestinal Neoplasms/pathology , Intestine, Small/metabolism , Intestine, Small/radiation effects , Mice , Mice, Inbred C57BL , Neoplasms, Radiation-Induced/pathology , Proto-Oncogene Proteins c-akt/metabolism , Radiation Dosage , Radiation Exposure/adverse effects , Space Flight , Tumor Suppressor p53-Binding Protein 1/metabolism , Up-Regulation/radiation effects , beta Catenin/metabolism
13.
Aging (Albany NY) ; 11(12): 4145-4158, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31239406

ABSTRACT

Heavy ion radiation, prevalent in outer space and relevant for radiotherapy, is densely ionizing and poses risk to stem cells that are key to intestinal homeostasis. Currently, the molecular spectrum of heavy ion radiation-induced perturbations in intestinal stem cells (ISCs), that could trigger intestinal pathologies, remains largely unexplored. The Lgr5-EGFP-IRES-creERT mice were exposed to 50 cGy of iron radiation. Mice were euthanized 60 d after exposure and ISCs were sorted using fluorescence activated cell sorting. Reactive oxygen species (ROS) and mitochondrial superoxide were measured using fluorescent probes. Since DNA damage is linked to senescence and senescent cells acquire senescence-associated secretory phenotype (SASP), we stained ISCs for both senescence markers p16, p21, and p19 as well as SASP markers IL6, IL8, and VEGF. Due to potential positive effects of SASP on proliferation, we also stained for PCNA. Data show increased ROS and ongoing DNA damage, by staining for γH2AX, and 53BP1, along with accumulation of senescence markers. Results also showed increased SASP markers in senescent cells. Collectively, our data suggest that heavy-ion-induced chronic stress and ongoing DNA damage is promoting SASP in a fraction of the ISCs, which has implications for gastrointestinal function, inflammation, and carcinogenesis in astronauts and patients.


Subject(s)
Cellular Senescence/radiation effects , DNA Damage/radiation effects , Epithelial Cells/radiation effects , Heavy Ions , Stem Cells/radiation effects , Animals , Flow Cytometry , Green Fluorescent Proteins , Humans , Intestinal Mucosa/cytology , Iron , Male , Mice , Reactive Oxygen Species , Receptors, G-Protein-Coupled
14.
Proc Natl Acad Sci U S A ; 115(42): E9832-E9841, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30275302

ABSTRACT

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/ß-catenin and its downstream EphrinB/EphB signaling are key to intestinal epithelial cell (IEC) proliferation and positioning during migration, and both are up-regulated 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.


Subject(s)
Cell Movement/radiation effects , Cellular Senescence/radiation effects , Epithelial Cells/radiation effects , Gamma Rays/adverse effects , Intestines/pathology , Stress, Physiological/radiation effects , Whole-Body Irradiation/adverse effects , Animals , Intestines/radiation effects , Iron Radioisotopes/adverse effects , Male , Mice , Mice, Inbred C57BL , Signal Transduction/radiation effects
15.
Oncotarget ; 9(33): 23078-23090, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29796173

ABSTRACT

The availability of robust classification algorithms for the identification of high risk individuals with resectable disease is critical to improving early detection strategies and ultimately increasing survival rates in PC. We leveraged high quality biospecimens with extensive clinical annotations from patients that received treatment at the Medstar-Georgetown University hospital. We used a high resolution mass spectrometry based global tissue profiling approach in conjunction with multivariate analysis for developing a classification algorithm that would predict early stage PC with high accuracy. The candidate biomarkers were annotated using tandem mass spectrometry. We delineated a six metabolite panel that could discriminate early stage PDAC from benign pancreatic disease with >95% accuracy of classification (Specificity = 0.85, Sensitivity = 0.9). Subsequently, we used multiple reaction monitoring mass spectrometry for evaluation of this panel in plasma samples obtained from the same patients. The pattern of expression of these metabolites in plasma was found to be discordant as compared to that in tissue. Taken together, our results show the value of using a metabolomics approach for developing highly predictive panels for classification of early stage PDAC. Future investigations will likely lead to the development of validated biomarker panels with potential for clinical translation in conjunction with CA-19-9 and/or other biomarkers.

16.
Free Radic Res ; 52(5): 556-567, 2018 May.
Article in English | MEDLINE | ID: mdl-29544379

ABSTRACT

Carbon irradiation due to its higher biological effectiveness relative to photon radiation is a concern for toxicity to proliferative normal gastrointestinal (GI) tissue after radiotherapy and long-duration space missions such as mission to Mars. Although radiation-induced oxidative stress is linked to chronic diseases such as cancer, effects of carbon irradiation on normal GI tissue have not been fully understood. This study assessed and compared chronic oxidative stress in mouse intestine and colon after different doses of carbon and γ radiation, which are qualitatively different. Mice (C57BL/6J) were exposed to 0.5 or 1.3 Gy of γ or carbon irradiation, and intestinal and colonic tissues were collected 2 months after irradiation. While part of the tissues was used for isolating epithelial cells, tissue samples were also fixed and paraffin embedded for 4 µm thick sections as well as frozen for biochemical assays. In isolated epithelial cells, reactive oxygen species and mitochondrial status were studied using fluorescent probes and flow cytometry. We assessed antioxidant enzymes and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity in tissues and formalin-fixed tissue sections were stained for 4-hydroxynonenal, a lipid peroxidation marker. Data show that mitochondrial deregulation, increased NADPH oxidase activity, and decreased antioxidant activity were major contributors to carbon radiation-induced oxidative stress in mouse intestinal and colonic cells. When considered along with higher lipid peroxidation after carbon irradiation relative to γ-rays, our data have implications for functional changes in intestine and carcinogenesis in colon after carbon radiotherapy as well as space travel.


Subject(s)
Carbon/chemistry , Colon/radiation effects , Gamma Rays , Intestine, Small/radiation effects , Oxidative Stress/radiation effects , Aldehydes/metabolism , Animals , Catalase/metabolism , Colon/cytology , Colon/metabolism , Dose-Response Relationship, Radiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Intestine, Small/cytology , Intestine, Small/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/radiation effects , NADPH Oxidases/metabolism , Radiation, Ionizing , Radiometry , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Whole-Body Irradiation
17.
Alcohol ; 66: 21-26, 2018 02.
Article in English | MEDLINE | ID: mdl-29277284

ABSTRACT

We previously reported increased current density through P-type voltage-gated Ca2+ channels in inferior colliculus (IC) neurons during alcohol withdrawal. However, the molecular correlate of this increased P-type channel current is currently unknown. Here, we probe changes in mRNA and protein expression of the pore-forming CaV2.1-α1 (P/Q-type) subunits in IC neurons during the course of alcohol withdrawal-induced seizures (AWSs). Rats received three daily doses of ethanol or the vehicle every 8 h for 4 consecutive days. The IC was dissected at various time intervals following alcohol withdrawal, and the mRNA and protein levels of the CaV2.1-α1 subunits were measured. In separate experiments, rats were tested for acoustically evoked seizure susceptibility 3, 24, and 48 h after alcohol withdrawal. AWSs were observed 24 h after withdrawal; no seizures were observed at 3 or 48 h or in the control-treated rats. Compared to control-treated rats, the mRNA levels of the CaV2.1-α1 subunit were increased 1.9-fold and 2.1-fold at 3 and 24 h, respectively; change in mRNA expression was nonsignificant at 48 h following alcohol withdrawal. Western blot analyses revealed that protein levels of the CaV2.1-α1 subunits were not altered in IC neurons following alcohol withdrawal. We conclude that expression of the Cacna1a mRNA increased before the onset of AWS susceptibility, suggesting that altered CaV2.1 channel expression may play a role in AWS pathogenesis.


Subject(s)
Alcohol Withdrawal Seizures/metabolism , Calcium Channels, N-Type/metabolism , Ethanol , Inferior Colliculi/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Alcohol Withdrawal Seizures/genetics , Alcohol Withdrawal Seizures/physiopathology , Animals , Calcium Channels, N-Type/genetics , Disease Models, Animal , Inferior Colliculi/physiopathology , Male , Membrane Potentials , RNA, Messenger/genetics , Rats, Sprague-Dawley , Time Factors , Up-Regulation
18.
J Cancer ; 8(10): 1769-1773, 2017.
Article in English | MEDLINE | ID: mdl-28819373

ABSTRACT

The purpose of the study was to assess transgenerational intestinal tumorigenic effects of low dose ionizing radiation employing a well-characterized mouse model of human colorectal cancer. Mice (6 to 8 weeks old APC1638N/+ mice; n=20 per study group) were exposed to whole-body 25 cGy x-rays and mated 2 days post-irradiation. Intestinal tumorigenesis in male and female F1 mice from No Parents Irradiated (NPI), Both Parents Irradiated (BPI), and Male Parent Irradiated (MPI) groups were compared 210 days after birth. Male and female Direct Parent Irradiated (DPI) groups were additional controls for male and female F1 groups respectively. Data showed higher intestinal tumor frequency (± standard error of the mean) in male and female F1 from BPI (male: 7.81 ± 0.91; female: 5.45 ± 0.36) as well as from MPI (male: 6.30 ± 0.33; female: 4.45 ± 0.33) mice relative to F1 from NPI mice (male: 4.2 ± 0.48; female: 3.35 ± 0.37). Compared to male and female DPI (male: 5.55 ± 0.40; female: 3.60 ± 0.22), tumor frequency in F1 mice of BPI and MPI, though higher, was not statistically significant except for DPI vs. BPI in male mice. Additionally, both BPI and MPI showed increased frequency of larger tumors relative to NPI. In summary, our observations demonstrated that the APC1638N/+ mice due to its low spontaneous tumor frequency could serve as an effective model to study risk of transgenerational carcinogenesis in gastrointestinal tissues after exposure to clinically relevant low doses of ionizing radiation.

19.
Life Sci Space Res (Amst) ; 13: 45-50, 2017 May.
Article in English | MEDLINE | ID: mdl-28554509

ABSTRACT

Ionizing radiation (IR) is a recognized risk factor for colorectal cancer (CRC) and astronauts undertaking long duration space missions are expected to receive IR doses in excess of permissible limits with implications for colorectal carcinogenesis. Exposure to IR in outer space occurs at low doses and dose rates, and energetic heavy ions due to their high linear energy transfer (high-LET) characteristics remain a major concern for CRC risk in astronauts. Previously, we have demonstrated that intestinal tumorigenesis in a mouse model (APC1638N/+) of human colorectal cancer was significantly higher after exposure to high dose rate energetic heavy ions relative to low-LET γ radiation. The purpose of the current study was to compare intestinal tumorigenesis in APC1638N/+ mice after exposure to energetic heavy ions at high (50cGy/min) and relatively low (0.33cGy/min) dose rate. Male and female mice (6-8 weeks old) were exposed to either 10 or 50cGy of 28Si (energy: 300MeV/n; LET: 70keV/µm) or 56Fe (energy: 1000MeV/n; LET: 148keV/µm) ions at NASA Space Radiation Laboratory in Brookhaven National Laboratory. Mice (n=20 mice/group) were euthanized and intestinal and colon tumor frequency and size were counted 150days after radiation exposure. Intestinal tumorigenesis in male mice exposed to 56Fe was similar for high and low dose rate exposures. Although male mice showed a decreasing trend at low dose rate relative to high dose rate exposures, the differences in tumor frequency between the two types of exposures were not statistically significant after 28Si radiation. In female mice, intestinal tumor frequency was similar for both radiation type and dose rates tested. In both male and female mice intestinal tumor size was not different after high and low dose rate radiation exposures. Colon tumor frequency in male and female mice after high and low dose rate energetic heavy ions was also not significantly different. In conclusion, intestinal and colonic tumor frequency and size was similar irrespective of energetic heavy ion radiation dose rate suggesting that carcinogenic potential of energetic heavy ions is independent of dose rate.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Carcinogenesis/pathology , Colonic Neoplasms/pathology , Disease Models, Animal , Heavy Ion Radiotherapy/adverse effects , Intestinal Neoplasms/pathology , Neoplasms, Radiation-Induced/pathology , Animals , Colonic Neoplasms/etiology , Dose-Response Relationship, Radiation , Female , Intestinal Neoplasms/etiology , Male , Mice , Mice, Inbred C57BL , Neoplasms, Radiation-Induced/etiology , Radiation Exposure/adverse effects
20.
Radiat Res ; 187(4): 476-482, 2017 04.
Article in English | MEDLINE | ID: mdl-28218889

ABSTRACT

Health risks from space radiations, particularly from densely ionizing radiations, represent an important challenge for long-ranged manned space missions. Reliable methods are needed for scaling low-LET to high-LET radiation risks for humans, based on animal or in vitro studies comparing these radiations. The current standard metric, relative biological effectiveness (RBE) compares iso-effect doses of two radiations. By contrast, a proposed new metric, radiation effects ratio (RER), compares effects of two radiations at the same dose. This definition of RER allows direct scaling of low-LET to high-LET radiation risks in humans at the dose or doses of interest. By contrast to RBE, RER can be used without need for detailed information about dose response shapes for compared radiations. This property of RER allows animal carcinogenesis experiments to be simplified by reducing the number of tested radiation doses. For simple linear dose-effect relationships, RBE = RER. However, for more complex dose-effect relationships, such as those with nontargeted effects at low doses, RER can be lower than RBE. We estimated RBE and RER values and uncertainties using heavy ion (12C, 28Si, 56Fe) and gamma-ray-induced tumors in a mouse model for intestinal cancer (APC1638N/+), and used both RBE and RER to estimate low-LET to high-LET risk scaling factors. The data showed clear evidence of nontargeted effects at low doses. In situations, such as the ones discussed here where nontargeted effects dominate at low doses, RER was lower than RBE by factors around 2.8-3.5 at 0.03 Gy and 1.3-1.4 at 0.3 Gy. It follows that low-dose high-LET human cancer risks scaled from low-LET human risks using RBE may be correspondingly overestimated.


Subject(s)
Carcinogenesis/radiation effects , Linear Energy Transfer , Models, Biological , Neoplasms, Radiation-Induced/etiology , Animals , Disease Models, Animal , Dose-Response Relationship, Radiation , Female , Humans , Intestinal Neoplasms/etiology , Male , Mice , Neoplasms, Experimental/etiology , Radiation Exposure/adverse effects , Radiation Exposure/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...