Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 675
Filter
1.
Asian Pac J Cancer Prev ; 25(8): 2645-2654, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39205561

ABSTRACT

BACKGROUND: Radiotherapy (RT) is a crucial treatment for head and neck cancer however, it causes adverse reactions to the normal tissue and organs adjacent to target tumor. The present study was carried out to investigate possible association of single nucleotide polymorphism in DNA repair genes with toxicity effects of radiotherapy on normal tissue. METHODS: Three hundred and fifty head and neck cancer patients receiving radiotherapy treatment were enrolled in this study. The adverse after effects of radiotherapy on the normal tissue in the form of skin reactions were recorded. Single nucleotide polymorphisms of APE1 (rs1130409), hOGG1 (rs1052133) and Rad51 (rs1801320, rs1801321) genes were studied by polymerase chain reaction-Restriction fragment length polymorphism (PCR-RFLP) and direct DNA sequencing methods and their association with development of severe radio-toxicity effects was evaluated logistic regression analysis. RESULTS: The 172G/T polymorphism of Rad51 was 2.85 times higher and significantly associated with skin reactions (OR=2.85, 95% CI: 1.50-5.41; p=0.001) and severe oral mucositis (OR=4.96, 95% CI: 2.40-10.25; p<0.0001). These results suggested that the polymorphic nature of Rad51 is responsible for risk of radiotherapy adverse effects in HNC patients. The variant 326Cys and heterozygous 326Ser/Cys genotype of hOGG1 was significantly associated with high tumor grade (OR=3.16 95% CI: 1.66-5.99; p=0.0004, and OR=3.97 95% CI: 2.15-7.34; p=<0.0001 respectively). The homozygous variant 172TT genotype of Rad51 showed positive association with poor response of both tumor and nodes towards radiotherapy treatment (p=0.007 and p=0.022). CONCLUSIONS: Interpretation of our results revealed significant association of rs1801321 SNP of Rad51 with development of adverse toxicity reactions in normal tissue of head and neck cancer patients treated with radiotherapy.


Subject(s)
DNA Glycosylases , DNA-(Apurinic or Apyrimidinic Site) Lyase , Head and Neck Neoplasms , Polymorphism, Single Nucleotide , Rad51 Recombinase , Humans , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Male , Head and Neck Neoplasms/radiotherapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Female , Rad51 Recombinase/genetics , Middle Aged , DNA Glycosylases/genetics , Follow-Up Studies , Prognosis , Radiation Injuries/genetics , Radiation Injuries/etiology , Aged , Adult , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/pathology , Genotype , DNA Repair/genetics , Biomarkers, Tumor/genetics , Radiotherapy/adverse effects
2.
Radiat Res ; 202(3): 489-502, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39089691

ABSTRACT

Radiation exposure in a therapeutic setting or during a mass casualty event requires improved medical triaging, where the time to delivery and quantity of medical countermeasures are critical to survival. Radiation-induced liver injury (RILI) and fibrosis can lead to death, but clinical symptoms manifest late in disease pathogenesis and there is no simple diagnostic test to determine RILI. Because animal models do not completely recapitulate clinical symptoms, we used a human liver-on-a-chip model to identify biomarkers of RILI. The goals of this study were: 1. to establish a microfluidic liver-on-a-chip device as a physiologically relevant model for studying radiation-induced tissue damage; and 2. to determine acute changes in RNA expression and biological pathway regulation that identify potential biomarkers and mechanisms of RILI. To model functional human liver tissue, we used the Emulate organ-on-a-chip system to establish a co-culture of human liver sinusoidal endothelial cells (LSECs) and hepatocytes. The chips were subject to 0 Gy (sham), 1 Gy, 4 Gy, or 10 Gy irradiation and cells were collected at 6 h, 24 h, or 7 days postirradiation for RNA isolation. To identify significant expression changes in messenger RNA (mRNA) and long non-coding RNA (lncRNA), we performed RNA sequencing (RNASeq) to conduct whole transcriptome analysis. We found distinct differences in expression patterns by time, dose, and cell type, with higher doses of radiation resulting in the most pronounced expression changes, as anticipated. Ingenuity Pathway Analysis indicated significant inhibition of the cell viability pathway 24 h after 10 Gy exposure in LSECs but activation of this pathway in hepatocytes, highlighting differences between cell types despite receiving the same radiation dose. Overall, hepatocytes showed fewer gene expression changes in response to radiation, with only 3 statistically significant differentially expressed genes at 7 days: APOBEC3H, PTCHD4, and GDNF. We further highlight lncRNA of interest including DINO and PURPL in hepatocytes and TMPO-AS1 and PRC-AS1 in LSECs, identifying potential biomarkers of RILI. We demonstrated the potential utility of a human liver-on-a-chip model with primary cells to model organ-specific radiation injury, establishing a model for radiation medical countermeasure development and further biomarker validation. Furthermore, we identified biomarkers that differentiate radiation dose and defined cell-specific targets for potential radiation mitigation therapies.


Subject(s)
Lab-On-A-Chip Devices , Liver , Radiation Injuries , Humans , Liver/radiation effects , Liver/metabolism , Liver/pathology , Radiation Injuries/genetics , Radiation Injuries/pathology , Hepatocytes/radiation effects , Hepatocytes/metabolism , RNA/genetics , RNA/metabolism , Biomarkers/metabolism , Endothelial Cells/radiation effects , Endothelial Cells/metabolism
3.
Arh Hig Rada Toksikol ; 75(2): 91-101, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38963141

ABSTRACT

Even at low levels, exposure to ionising radiation can lead to eye damage. However, the underlying molecular mechanisms are not yet fully understood. We aimed to address this gap with a comprehensive in silico approach to the issue. For this purpose we relied on the Comparative Toxicogenomics Database (CTD), ToppGene Suite, Cytoscape, GeneMANIA, and Metascape to identify six key regulator genes associated with radiation-induced eye damage (ATM, CRYAB, SIRT1, TGFB1, TREX1, and YAP1), all of which have physical interactions. Some of the identified molecular functions revolve around DNA repair mechanisms, while others are involved in protein binding, enzymatic activities, metabolic processes, and post-translational protein modifications. The biological processes are mostly centred on response to DNA damage, the p53 signalling pathway in particular. We identified a significant role of several miRNAs, such as hsa-miR-183 and hsamiR-589, in the mechanisms behind ionising radiation-induced eye injuries. Our study offers a valuable method for gaining deeper insights into the adverse effects of radiation exposure.


Subject(s)
Data Mining , Radiation, Ionizing , Humans , Radiation Injuries/genetics , Radiation Injuries/etiology , Eye Injuries/etiology , Eye Injuries/genetics , Genomics , DNA Damage/radiation effects
4.
Toxicol Lett ; 399: 43-51, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39032790

ABSTRACT

In the event of a nuclear or radiation accident, rapid identification is required for those who exposed to potentially lethal dose irradiation. However, existing techniques are not adequate for the classification of lethal injury. Several studies have explored the potential of miRNAs as biomarkers for ionizing radiation injury, however, there are few miRNAs with specific expression for lethal radiation injury. Therefore, the aim of this study was to screen and validate the possibility of serum miRNAs as biomarkers of lethal radiation injury. We found the specific expression of mmu-miR-374c-5p / mmu-miR-194-5p on first day and mmu-miR-192-5p / mmu-miR-223-3p on third day in the mouse serum only under 10 Gy irradiation by miRNA sequencing and all significantly correlated with lymphocyte counts by Pearson's correlation analysis. In addition, it was found that among the 4 candidate serum miRNAs, only highly-expressed mmu-miR-192-5p in mouse serum irradiated at lethal doses was returned to sham-like expression levels at 3 days post-irradiation with amifostine pretreatment and closely correlated with survival rate. We demonstrated for the first time that mmu-miR-192-5p screened from lethally irradiated mice sera can be used as a potential biomarker for lethal irradiation injury, which will be helpful to improve efficiency of medical treatment to minimize casualties after a large-scale nuclear accident.


Subject(s)
Biomarkers , MicroRNAs , Animals , MicroRNAs/blood , MicroRNAs/genetics , Mice , Male , Biomarkers/blood , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/genetics , Radiation Injuries/blood , Radiation Injuries/genetics , Mice, Inbred C57BL
5.
Nat Commun ; 15(1): 4825, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862542

ABSTRACT

Our previous research revealed a key microRNA signature that is associated with spaceflight that can be used as a biomarker and to develop countermeasure treatments to mitigate the damage caused by space radiation. Here, we expand on this work to determine the biological factors rescued by the countermeasure treatment. We performed RNA-sequencing and transcriptomic analysis on 3D microvessel cell cultures exposed to simulated deep space radiation (0.5 Gy of Galactic Cosmic Radiation) with and without the antagonists to three microRNAs: miR-16-5p, miR-125b-5p, and let-7a-5p (i.e., antagomirs). Significant reduction of inflammation and DNA double strand breaks (DSBs) activity and rescue of mitochondria functions are observed after antagomir treatment. Using data from astronaut participants in the NASA Twin Study, Inspiration4, and JAXA missions, we reveal the genes and pathways implicated in the action of these antagomirs are altered in humans. Our findings indicate a countermeasure strategy that can potentially be utilized by astronauts in spaceflight missions to mitigate space radiation damage.


Subject(s)
Astronauts , Cosmic Radiation , MicroRNAs , Space Flight , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Cosmic Radiation/adverse effects , DNA Breaks, Double-Stranded/radiation effects , Radiation Injuries/genetics , Radiation Injuries/prevention & control , Male , Mitochondria/radiation effects , Mitochondria/metabolism , Mitochondria/genetics , Female , Adult
6.
Cell Commun Signal ; 22(1): 292, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802843

ABSTRACT

BACKGROUND: Hematopoietic stem cell (HSC) regeneration underlies hematopoietic recovery from myelosuppression, which is a life-threatening side effect of cytotoxicity. HSC niche is profoundly disrupted after myelosuppressive injury, while if and how the niche is reshaped and regulates HSC regeneration are poorly understood. METHODS: A mouse model of radiation injury-induced myelosuppression was built by exposing mice to a sublethal dose of ionizing radiation. The dynamic changes in the number, distribution and functionality of HSCs and megakaryocytes were determined by flow cytometry, immunofluorescence, colony assay and bone marrow transplantation, in combination with transcriptomic analysis. The communication between HSCs and megakaryocytes was determined using a coculture system and adoptive transfer. The signaling mechanism was investigated both in vivo and in vitro, and was consolidated using megakaryocyte-specific knockout mice and transgenic mice. RESULTS: Megakaryocytes become a predominant component of HSC niche and localize closer to HSCs after radiation injury. Meanwhile, transient insulin-like growth factor 1 (IGF1) hypersecretion is predominantly provoked in megakaryocytes after radiation injury, whereas HSCs regenerate paralleling megakaryocytic IGF1 hypersecretion. Mechanistically, HSCs are particularly susceptible to megakaryocytic IGF1 hypersecretion, and mTOR downstream of IGF1 signaling not only promotes activation including proliferation and mitochondrial oxidative metabolism of HSCs, but also inhibits ferritinophagy to restrict HSC ferroptosis. Consequently, the delicate coordination between proliferation, mitochondrial oxidative metabolism and ferroptosis ensures functional HSC expansion after radiation injury. Importantly, punctual IGF1 administration simultaneously promotes HSC regeneration and hematopoietic recovery after radiation injury, representing a superior therapeutic approach for myelosuppression. CONCLUSIONS: Our study identifies megakaryocytes as a last line of defense against myelosuppressive injury and megakaryocytic IGF1 as a novel niche signal safeguarding HSC regeneration.


Subject(s)
Ferroptosis , Hematopoietic Stem Cells , Insulin-Like Growth Factor I , Megakaryocytes , Regeneration , Animals , Hematopoietic Stem Cells/metabolism , Megakaryocytes/metabolism , Megakaryocytes/radiation effects , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/genetics , Ferroptosis/genetics , Mice , Mice, Inbred C57BL , Radiation Injuries/metabolism , Radiation Injuries/pathology , Radiation Injuries/genetics , Signal Transduction/radiation effects
7.
Nat Commun ; 15(1): 3018, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589357

ABSTRACT

Ionizing radiation induces cell death in the gastrointestinal (GI) epithelium by activating p53. However, p53 also prevents animal lethality caused by radiation-induced acute GI syndrome. Through single-cell RNA-sequencing of the irradiated mouse small intestine, we find that p53 target genes are specifically enriched in regenerating epithelial cells that undergo fetal-like reversion, including revival stem cells (revSCs) that promote animal survival after severe damage of the GI tract. Accordingly, in mice with p53 deleted specifically in the GI epithelium, ionizing radiation fails to induce fetal-like revSCs. Using intestinal organoids, we show that transient p53 expression is required for the induction of revival stem cells and is controlled by an Mdm2-mediated negative feedback loop. Together, our findings reveal that p53 suppresses severe radiation-induced GI injury by promoting fetal-like reprogramming of irradiated intestinal epithelial cells.


Subject(s)
Radiation Injuries , Tumor Suppressor Protein p53 , Mice , Animals , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Intestines , Gastrointestinal Tract/metabolism , Radiation Injuries/genetics , Radiation Injuries/metabolism , Stem Cells/metabolism , Apoptosis/genetics
8.
Int Immunopharmacol ; 133: 111987, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38652961

ABSTRACT

Radiation-induced kidney injury is a common side effect of radiotherapy, as the pelvic region is in close proximity to the kidneys, posing a risk of inducing radiation-induced kidney injury when treating any pelvic malignancies with radiotherapy. This type of injury typically manifests as chronic kidney disease a few months after radiotherapy, with the potential to progress to end-stage renal disease. Radiation-induced damage involves various components of the kidney, including glomeruli, tubules, interstitium, and extracellular matrix. Therefore, investigating its molecular mechanisms is crucial. In this study, we extensively searched literature databases, selecting recent transcriptomic studies related to acute kidney injury (AKI) published in the past decade. We downloaded the raw RNA sequencing datasets GSE30718 and GSE66494 related to AKI from the GEO database and identified that intestinal-type lectin ITLN1 plays a significant role in regulating radiation-induced kidney injury in rats. Differential gene analysis was performed using chip data from the GEO database, and further bioinformatics analysis identified 13 genes that may be involved in regulating kidney injury, with ITLN1 being the most relevant to kidney damage, thus selected as the target gene for this study. Subsequently, a rat model of radiation-induced kidney injury was established for experimental validation, assessing kidney tissue morphology and injury extent through staining observation and immunohistochemical staining. The protective effect of ITLN1 on kidney function was evaluated by measuring changes in rat body weight and blood pressure, serum kidney injury markers, and kidney structure. The experimental results indicate that overexpression of ITLN1 can improve kidney function in rats with radiation-induced kidney injury by activating the Akt/GSK-3ß/Nrf2 signaling pathway, suppressing oxidative stress, cell apoptosis, inflammation, cellular senescence, and fibrosis. This study highlights the significant role of ITLN1 in regulating kidney injury, providing a novel target for future treatments of radiation-induced kidney injury.


Subject(s)
Kidney , Animals , Rats , Kidney/pathology , Kidney/metabolism , Kidney/radiation effects , Male , Acute Kidney Injury/metabolism , Acute Kidney Injury/etiology , Humans , Radiation Injuries/genetics , Rats, Sprague-Dawley , Signal Transduction , Radiation Injuries, Experimental/metabolism
9.
Biomol Biomed ; 24(5): 1331-1349, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-38552230

ABSTRACT

Radiation-induced lung injury (RILI) frequently occurs as a complication following radiotherapy for chest tumors like lung and breast cancers. However, the precise underlying mechanisms of RILI remain unclear. In this study, we generated RILI models in rats treated with a single dose of 20 Gy and examined lung tissues by single-cell RNA sequencing (scRNA-seq) 2 weeks post-radiation. Analysis of lung tissues revealed 18 major cell populations, indicating an increase in cell-cell communication following radiation exposure. Neutrophils, macrophages, and monocytes displayed distinct subpopulations and uncovered potential for pro-inflammatory effects. Additionally, endothelial cells exhibited a highly inflammatory profile and the potential for reactive oxygen species (ROS) production. Furthermore, smooth muscle cells (SMC) showed a high propensity for extracellular matrix (ECM) deposition. Our findings broaden the current understanding of RILI and highlight potential avenues for further investigation and clinical applications.


Subject(s)
Lung Injury , Single-Cell Analysis , Animals , Rats , Lung Injury/etiology , Lung Injury/genetics , Lung Injury/metabolism , Lung Injury/pathology , Single-Cell Analysis/methods , Transcriptome/radiation effects , Lung/pathology , Lung/radiation effects , Lung/metabolism , Reactive Oxygen Species/metabolism , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/genetics , Gene Expression Profiling/methods , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/radiation effects , Myocytes, Smooth Muscle/pathology , Male , Radiation Injuries/pathology , Radiation Injuries/genetics , Radiation Injuries/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/radiation effects , Rats, Sprague-Dawley
10.
J Stroke Cerebrovasc Dis ; 33(7): 107699, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38552890

ABSTRACT

BACKGROUND: Radiation treatment for diseases of the brain can result in hemorrhagic adverse radiation effects. The underlying pathologic substrate of brain bleeding after irradiation has not been elucidated, nor potential associations with induced somatic mutations. METHODS: We retrospectively reviewed our department's pathology database over 5 years and identified 5 biopsy specimens (4 patients) for hemorrhagic lesions after brain irradiation. Tissues with active malignancy were excluded. Samples were characterized using H&E, Perl's Prussian Blue, and Masson's Trichrome; immunostaining for B-cells (anti-CD20), T-cells (anti-CD3), endothelium (anti-CD31), macrophages (anti-CD163), α-smooth muscle actin, and TUNEL. DNA analysis was done by two panels of next-generation sequencing for somatic mutations associated with known cerebrovascular anomalies. RESULTS: One lesion involved hemorrhagic expansion among multifocal microbleeds that had developed after craniospinal irradiation for distant medulloblastoma treatment. Three bleeds arose in the bed of focally irradiated arteriovenous malformations (AVM) after confirmed obliteration. A fifth specimen involved the radiation field distinct from an irradiated AVM bed. From these, 2 patterns of hemorrhagic vascular pathology were identified: encapsulated hematomas and cavernous-like malformations. All lesions included telangiectasias with dysmorphic endothelium, consistent with primordial cavernous malformations with an associated inflammatory response. DNA analysis demonstrated genetic variants in PIK3CA and/or PTEN genes but excluded mutations in CCM genes. CONCLUSIONS: Despite pathologic heterogeneity, brain bleeding after irradiation is uniformly associated with primordial cavernous-like telangiectasias and disruption of genes implicated in dysangiogenesis but not genes implicated as causative of cerebral cavernous malformations. This may implicate a novel signaling axis as an area for future study.


Subject(s)
Mutation , Radiation Injuries , Adult , Female , Humans , Male , Middle Aged , Young Adult , Biopsy , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Cranial Irradiation/adverse effects , Databases, Factual , DNA Mutational Analysis , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Intracranial Arteriovenous Malformations/genetics , Intracranial Arteriovenous Malformations/radiotherapy , Intracranial Arteriovenous Malformations/pathology , Intracranial Hemorrhages/genetics , Intracranial Hemorrhages/etiology , Intracranial Hemorrhages/pathology , Phenotype , PTEN Phosphohydrolase/genetics , Radiation Injuries/genetics , Radiation Injuries/pathology , Radiation Injuries/etiology , Retrospective Studies , Risk Factors
11.
Front Immunol ; 15: 1338922, 2024.
Article in English | MEDLINE | ID: mdl-38426100

ABSTRACT

This review explores the mechanisms of chronic radiation-induced skin injury fibrosis, focusing on the transition from acute radiation damage to a chronic fibrotic state. It reviewed the cellular and molecular responses of the skin to radiation, highlighting the role of myofibroblasts and the significant impact of Transforming Growth Factor-beta (TGF-ß) in promoting fibroblast-to-myofibroblast transformation. The review delves into the epigenetic regulation of fibrotic gene expression, the contribution of extracellular matrix proteins to the fibrotic microenvironment, and the regulation of the immune system in the context of fibrosis. Additionally, it discusses the potential of biomaterials and artificial intelligence in medical research to advance the understanding and treatment of radiation-induced skin fibrosis, suggesting future directions involving bioinformatics and personalized therapeutic strategies to enhance patient quality of life.


Subject(s)
Artificial Intelligence , Radiation Injuries , Humans , Epigenesis, Genetic , Quality of Life , Fibrosis , Transforming Growth Factor beta/metabolism , Radiation Injuries/genetics
12.
Sci Rep ; 14(1): 2681, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38302506

ABSTRACT

A radiological accident, whether from industrial, medical, or malicious origin, may result in localized exposure to high doses of ionizing radiations, leading to the development of local radiation injury (LRI), that may evolve toward deep ulceration and necrosis of the skin and underlying tissues. Early diagnosis is therefore crucial to facilitate identification and management of LRI victims. Circulating microRNAs (miRNA) have been studied as potential diagnostic biomarkers of several diseases including hematological defects following whole-body irradiation (WBI). This study aims to identify a blood miRNA signature associated with LRI in a preclinical C57BL/6J mouse model of hindlimb irradiation using different 10-MV X-ray doses that lead to injuries of different severities. To this end, we first performed broad-spectrum plasma miRNA profiling, followed by a targeted validation step, on two independent animal cohorts. Using a multivariate sparse partial least square discriminant analysis, we identified a panel of eight circulating miRNAs able to segregate mice according to LRI severity. Interestingly, these miRNAs were previously associated with WBI (miR-150-5p, miR-342-3p, miR-146a-5p), inflammation (miR-18a-5p, miR-148b-3p, miR-532-5p) and skin diseases (miR-139-5p, miR-195-5p). Our results suggest the use of circulating miRNAs as suitable molecular biomarkers for LRI prognosis and diagnosis.


Subject(s)
Circulating MicroRNA , MicroRNAs , Radiation Injuries , Humans , Animals , Mice , MicroRNAs/genetics , Mice, Inbred C57BL , Biomarkers , Circulating MicroRNA/genetics , Radiation Injuries/diagnosis , Radiation Injuries/genetics , Gene Expression Profiling
13.
Int J Radiat Biol ; 100(9): 1240-1252, 2024.
Article in English | MEDLINE | ID: mdl-38190433

ABSTRACT

PURPOSE: The purpose of this paper is to provide an overview of the methodology used to estimate radiation genetic risks and quantify the risk of hereditary effects as outlined in the ICRP Publication 103. It aims to highlight the historical background and development of the doubling dose method for estimating radiation-related genetic risks and its continued use in radiological protection frameworks. RESULTS: This article emphasizes the complexity associated with quantifying the risk of hereditary effects caused by radiation exposure and highlights the need for further clarification and explanation of the calculation method. As scientific knowledge in radiation sciences and human genetics continues to advance in relation to a number of factors including stability of disease frequency, selection pressures, and epigenetic changes, the characterization and quantification of genetic effects still remains a major issue for the radiological protection system of the International Commission on Radiological Protection. CONCLUSION: Further research and advancements in this field are crucial for enhancing our understanding and addressing the complexities involved in assessing and managing the risks associated with hereditary effects of radiation.


Subject(s)
Radiation Protection , Humans , Radiation Protection/methods , Risk Assessment , Radiation Exposure/adverse effects , Radiation Dosage , Radiation Injuries/prevention & control , Radiation Injuries/genetics
14.
Pract Radiat Oncol ; 14(1): e29-e39, 2024.
Article in English | MEDLINE | ID: mdl-37742832

ABSTRACT

PURPOSE: There are mixed and limited data regarding radiation therapy (RT) tolerance in carriers of a germline pathogenic or likely pathogenic (P/LP) ATM variant. We investigated RT-related toxic effects in carriers of an ATM variant who received treatment for breast cancer. METHODS AND MATERIALS: We identified 71 patients treated with adjuvant RT for breast cancer who were carriers of a variant in ATM: 15 were classified as P/LP and 56 classified as variants of unknown significance (VUS). We additionally identified 205 consecutively treated patients during a similar timeframe who were either confirmed ATM wild type or had no prior genetic testing. RT plans were reviewed. Acute and chronic toxic effects were evaluated using Common Terminology Criteria for Adverse Events version 4.0 criteria. Fisher's exact tests for count data were performed to compare toxic effects between the cohorts (P/LP vs VUS vs control). Wilcoxon rank-sum testing was performed to assess for differences in patient characteristics. RESULTS: The median toxicity follow-up was 19.4 months; median follow-up for the subcohorts was 13.3 months (P/LP), 12.6 months (VUS), and 23.3 months (control). There were no significant differences in radiation plan heterogeneity, receipt of a boost, or size of breast/chest wall planning target volume. There was greater use of hypofractionated RT in the control cohort (P = .023). After accounting for patient- and treatment-related factors that may affect toxic effects, we found no significant differences with respect to acute dermatitis, hyperpigmentation, moist desquamation, breast/chest wall pain, or breast edema. Additionally, we found no significant differences with respect to chronic breast/chest wall pain, induration, telangiectasia, or cosmetic outcome. CONCLUSIONS: RT as part of the management of breast cancer was well tolerated in carriers of a P/LP ATM variant, with toxic effect profiles that were similar to those seen in patients without known ATM mutations. High rates of excellent or good cosmesis were observed in carriers of a P/LP ATM variant who underwent breast conservation.


Subject(s)
Breast Neoplasms , Radiation Injuries , Humans , Female , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Breast Neoplasms/pathology , Radiation Injuries/genetics , Radiation Injuries/pathology , Pain , Ataxia Telangiectasia Mutated Proteins/genetics
15.
Oncology ; 102(7): 1, 2024.
Article in English | MEDLINE | ID: mdl-38160665

ABSTRACT

INTRODUCTION: Bladder cancer (BC) is sensitive to radiation treatment and a subset of patients experience radiation-induced injuries including shrinkage of bladder due to bladder fibrosis. METHODS: This study is a retrospective cohort study. Three Japanese BC patients were randomly selected. Using a microRNA (miRNA) array, comparing their samples with or without radiation-induced injuries, we have checked the clustering of miRNA expression. RESULTS: Hsa-miR-130a, hsa-miR-200c, hsa-miR-141, and hsa-miR-96 were found to be highly expressed (>50 times) in patients with fibrotic bladder shrinkage (FBS) compared to those with intact bladder (IB) function. In patients with FBS, hsa-miR-6835, hsa-miR-4675, hsa-miR-371a, and hsa-miR-6885 were detected to have lesser than half expression to IB patients. We have analyzed the significance of these genes in relation to overall survival of 409 BC patients retrieved from the Cancer Genome Atlas data set. All available cutoff values between the lower and upper quartiles of expression are used for the selected genes, and false discovery rate using the Benjamini-Hochberg method is computed to correct for multiple hypothesis testing. We have run combined survival analysis of the mean expression of these four miRNAs highly expressed in FBS patients. 175 patients with high expression had a longer median survival of 98.47 months than 23.73 months in 233 patients with low expression (hazard ratio [HR]: 0.53; 0.39-0.72, log-rank p value: 7.3e-0.5). Combination analysis of all 8 genes including hsa-miR-6835, hsa-miR-4675, hsa-miR-371a, and hsa-miR-6885 showed the same HR for OS. Target scanning for these miRNAs matched specific cytokines known as an early biomarker to develop radiation-induced fibrosis. CONCLUSIONS: BC patients with fibrotic radiation injury have specific miRNA expression profile targeting profibrotic cytokines and these miRNAs possibly render to favorable survival.


Subject(s)
MicroRNAs , Radiation Injuries , Urinary Bladder Neoplasms , Urinary Bladder , Humans , MicroRNAs/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/radiotherapy , Urinary Bladder Neoplasms/pathology , Male , Retrospective Studies , Female , Radiation Injuries/genetics , Radiation Injuries/pathology , Aged , Urinary Bladder/pathology , Urinary Bladder/radiation effects , Urinary Bladder/metabolism , Middle Aged , Aged, 80 and over , Fibrosis/genetics
16.
Int J Mol Sci ; 24(23)2023 Dec 02.
Article in English | MEDLINE | ID: mdl-38069378

ABSTRACT

Patients receiving cranial radiotherapy for primary and metastatic brain tumors may experience radiation-induced brain injury (RIBI). Thus far, there has been a lack of effective preventive and therapeutic strategies for RIBI. Due to its complicated underlying pathogenic mechanisms, it is rather difficult to develop a single approach to target them simultaneously. We have recently reported that Reprimo (RPRM), a tumor suppressor gene, is a critical player in DNA damage repair, and RPRM deletion significantly confers radioresistance to mice. Herein, by using an RPRM knockout (KO) mouse model established in our laboratory, we found that RPRM deletion alleviated RIBI in mice via targeting its multiple underlying mechanisms. Specifically, RPRM knockout significantly reduced hippocampal DNA damage and apoptosis shortly after mice were exposed to whole-brain irradiation (WBI). For the late-delayed effect of WBI, RPRM knockout obviously ameliorated a radiation-induced decline in neurocognitive function and dramatically diminished WBI-induced neurogenesis inhibition. Moreover, RPRM KO mice exhibited a significantly lower level of acute and chronic inflammation response and microglial activation than wild-type (WT) mice post-WBI. Finally, we uncovered that RPRM knockout not only protected microglia against radiation-induced damage, thus preventing microglial activation, but also protected neurons and decreased the induction of CCL2 in neurons after irradiation, in turn attenuating the activation of microglial cells nearby through paracrine CCL2. Taken together, our results indicate that RPRM plays a crucial role in the occurrence of RIBI, suggesting that RPRM may serve as a novel potential target for the prevention and treatment of RIBI.


Subject(s)
Brain Injuries , Radiation Injuries , Animals , Humans , Mice , Apoptosis , Brain/pathology , Brain Injuries/genetics , Brain Injuries/prevention & control , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Glycoproteins/antagonists & inhibitors , Glycoproteins/metabolism , Inflammation/pathology , Microglia , Radiation Injuries/genetics , Radiation Injuries/prevention & control , Radiation Injuries/pathology
17.
Radiat Res ; 200(6): 556-568, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37874034

ABSTRACT

Ionizing radiation in space, radiation devices or nuclear disasters are major threats to human health and public security. Expanding countermeasures for dealing with accidental or occupational radiation exposure is crucial for the protection of radiation injuries. Circulating microRNAs (miRNAs) have emerged as promising radiation biomarkers in recent years. However, the origin, distribution and functions of radiosensitive circulating miRNAs remain unclear, which obstructs their clinical applications in the future. In this study, we found that mmu-miR-342-3p (miR-342) in mouse serum presents a stable and significant decrease after X-ray total-body irradiation (TBI). Focusing on this miRNA, we investigated the influences of circulating miR-342 on the radiation-induced injury. Through tail vein injection of Cy5-labeled synthetic miR-342, we found the exogenous miR-342-Cy5 was mainly enriched in metabolic and immune organs. Besides, the bioinformatic analysis predicted that miR-342 might involve in immune-related processes or pathways. Further, mice were tail vein injected with synthetic miR-342 mimetics (Ago-miR-342) after irradiation to upregulate the level of miR-342 in circulating blood. The results showed that the upregulation of circulating miR-342 alleviated the radiation-induced depletion of CD3+CD4+ T lymphocytes and influenced the levels of IL-2 and IL-6 in irradiated mice. Moreover, the injection of Ago-miR-342 improved the survival rates of mice with acute radiation injury. Our findings demonstrate that upregulation of circulating miR-342 alleviates the radiation-induced immune system injury, which provides us new insights into the functions of circulating miRNAs and the prospect as the targets for mitigation of radiation injuries.


Subject(s)
Circulating MicroRNA , MicroRNAs , Radiation Injuries , Animals , Mice , Biomarkers , Circulating MicroRNA/genetics , Circulating MicroRNA/metabolism , Immune System/radiation effects , MicroRNAs/genetics , Radiation Injuries/genetics
18.
Radiat Prot Dosimetry ; 199(14): 1533-1538, 2023 Sep 18.
Article in English | MEDLINE | ID: mdl-37721085

ABSTRACT

Radiosensitivity in humans can influence radiation-induced normal tissue toxicity. As radiosensitivity has a genetic predisposition, we aimed to investigate the possible association between four single nucleotide polymorphism (SNP) sites and the radiosensitivity in healthy people. We genotyped four selected SNPs: TRIP12 (rs13018957), UIMC1 (rs1700490) and POLN (rs2022302), and analyzed the association between SNP and the radiosensitivity in healthy people. We distinguished radiosensitivity by chromosome aberration analysis in healthy individuals. Healthy donors were classified into three groups based on chromosomal aberrations: resistant, normal and sensitive. Using the normal group as a reference, the genotypes CT and CC of rs13018957 (CT: OR = 26.13; CC: OR = 15.97), AA of rs1700490 (OR = 32.22) and AG of rs2022302 (OR = 13.98) were risk factors for radiosensitivity. The outcomes of the present study suggest that four SNPs are associated with radiosensitivity. This study lends insights to the underlying mechanisms of radiosensitivity and improves our ability to identify radiosensitive individuals.


Subject(s)
Polymorphism, Single Nucleotide , Radiation Injuries , Humans , Chromosome Aberrations , Health Status , Radiation Injuries/genetics , Radiation Tolerance/genetics , DNA Damage/genetics , Carrier Proteins , Ubiquitin-Protein Ligases
19.
Cell Biochem Funct ; 41(8): 1115-1132, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37653677

ABSTRACT

The biogenic synthesis of nanoparticles has drawn significant attention. The spleen is the largest lymphatic organ that is adversely impacted during irradiation. The current study was designated to evaluate the possible anti-inflammatory effect of matcha-silver nanoparticles (M-AgNPs) to reduce inflammation associated with γ-radiation induced-oxidative stress and inflammation in rats' spleen. Silver nanoparticles (AgNPs) were synthesized by biogenic synthesis using a green sonochemical method from matcha (M) green tea. The obtained M-AgNPs were extensively characterized by dynamic light scattering, transmission electron microscopy, thermogravimetric analysis, and Fourier-transform infrared spectroscopy. Using zetasizer analysis, the surface charge, particle size, and radical scavenging DPPH assay of M-AgNPs were also examined. Biocompatibility and cytotoxicity were analyzed by MTT assay, and the IC50 was calculated. Four groups of 24 Wistar rats each had an equal number of animals. The next step involved measuring the levels of oxidative stress markers in the rat splenic tissue. Additionally, the amounts of inflammatory protein expression were evaluated using the ELISA analysis. The results indicated the formation of spherical nanoparticles of pure Ag° coated with matcha polyphenols at the nanoscale, as well as uniform monodisperse particles suited for cellular absorption. Results revealed that M-AgNPs improved all biochemical parameters. Furthermore, M-AgNPs relieve inflammation by reducing the expression of NOD-like receptor family pyrin domain-containing 3 (NLRP3), interleukin-1ß (IL-1ß), and enhancing the levels of ileSnt information regulator 1 (SIRT1). Histopathological examinations demonstrated the ability of M-AgNPs to overcome the damage consequent to irradiation and recover the spleen's cellular structure. These results confirmed that matcha is a potential biomaterial for synthesizing AgNPs, which can be exploited for their anti-inflammatory activity.


Subject(s)
Metal Nanoparticles , Silver , Animals , Rats , Anti-Inflammatory Agents , Gamma Rays , Inflammation/drug therapy , Inflammation/pathology , Metal Nanoparticles/chemistry , Oxidative Stress , Rats, Wistar , Signal Transduction , Silver/pharmacology , Silver/chemistry , Silver/therapeutic use , Sirtuin 1 , Spleen , Radiation Injuries/genetics , Radiation Injuries/metabolism , Radiation Injuries/prevention & control , Tea
20.
Asian Pac J Cancer Prev ; 24(9): 3049-3057, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37774056

ABSTRACT

BACKGROUND: The genetic polymorphisms in DNA repair genes and their correlation with normal tissue toxicity in response to radiation therapy has not been consistently proven in many of the studies done in head and neck cancers (HNC). This study was intended to investigate the association of most common single nucleotide polymorphisms of DNA repair genes with acute radiation induced toxicities such as skin reactions and oral mucositis in normal tissue from HNC patients receiving radiotherapy from South-Western Maharashtra. METHODS: Two hundred HNC patients receiving radiotherapy were enrolled in this study and the radiation injuries in the form of skin reactions and oral mucositis were recorded. Three single nucleotide polymorphisms (SNPs) rs1799782, rs25489) rs25487 of XRCC1 gene, rs3218536in XRCC2 gene and rs861539 SNP of XRCC3 gene were studied by PCR-RFLP and direct DNA sequencing.  Results: The univariate analysis of SNPs of XRCC1, XRCC2 and XRCC3, the obtained results verified that XRCC1 polymorphism at 194Trp of exon 6 (OR=0.69, 95% CI: 0.28-1.71; p=0.433), codon 280 at exon 9 ((OR=1.05, 95% CI: 0.42-2.63; p=0.911) and codon 399 of at exon 10(OR=1.06, 95% CI: 0.52-2.15; p=0.867) and XRCC2 polymorphism at codon 188 at exon 3 (OR=1.07, 95% CI: 0.46-2.47; p=0.866) and 241Met variant genotype of XRCC3 (OR=2.63 95% CI: 0.42-16.30; p=0.298) showed no association with degree of radiotherapy associated dermatitis or mucositis in HNC patients. CONCLUSION: The findings from this study postulated that none of rs1799782, rs25489, rs25487 SNPs of XRCC1, rs3218536 SNP of XRCC2 nor rs861539 SNP of XRCC3 were associated with increased toxicity of radiotherapy in HNC patients of south-western Maharashtra. 
.


Subject(s)
Head and Neck Neoplasms , Radiation Injuries , Stomatitis , Humans , Polymorphism, Single Nucleotide/genetics , Genetic Predisposition to Disease , DNA Repair/genetics , India , X-ray Repair Cross Complementing Protein 1/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/radiotherapy , Genotype , Radiation Injuries/etiology , Radiation Injuries/genetics , Case-Control Studies , DNA-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL