RESUMO
BACKGROUND/AIMS: One of the treatments for breast cancer is surgical resection of the tumour and prevention of recurrence with postoperative radiotherapy. Unfortunately, radiotherapy is not always effective enough due to the low sensitivity of cancer cells to ionising radiation. This study aimed to evaluate the radiosensitising properties of resveratrol, piceatannol and polydatin on breast cancer cells, which differ in the presence of hormonal receptors on their surface. METHODS: The experimental part was carried out on triple-negative breast cancer cells (HCC38) and hormone-dependent cells (MCF7). The study assessed the level of cell death, changes in the expression of genes (BAX, BCL-2) and proteins related to the apoptosis process (CASPASE 3, 8 and P53), changes in the expression of antioxidant enzymes (CATALASE, SOD, GPx1/2) and NRF-2. Additionally, the expression level of RAD51 protein and histone H2AX, which are involved in DNA repair processes, was assessed. Statistical significance was evaluated by a two-way analysis of variance (ANOVA) followed by Tukey's post hoc test (p < 0.05). RESULTS: Ionising radiation in combination with resveratrol or piceatannol activates the apoptosis process by internal and external pathways. Greater sensitivity of MCF7 cells compared to HCC38 cells to ionising radiation in combination with resveratrol is associated with a weaker antioxidant response of cells and reduced intensity of DNA damage repair. DNA repair induced by ionising radiation occurs more effectively in HCC38 cells than in MCF7 cells. CONCLUSION: Resveratrol has the highest radiosensitising potential among the tested stilbene for cells of both lines. The effectiveness of ionizing radiation in combination with resveratrol (to a lesser extent with piceatannol) is more significant in MCF7 than in HCC38 cells.
Assuntos
Apoptose , Radiação Ionizante , Radiossensibilizantes , Resveratrol , Estilbenos , Humanos , Estilbenos/farmacologia , Resveratrol/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Feminino , Radiossensibilizantes/farmacologia , Linhagem Celular Tumoral , Células MCF-7 , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/radioterapia , Neoplasias da Mama/tratamento farmacológico , Histonas/metabolismo , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Rad51 Recombinase/metabolismo , Caspase 3/metabolismo , GlucosídeosRESUMO
Glutathione S-transferase-Pi 1 (GSTP1) is an isozyme that plays a key role in detoxification and antioxidative damage. It also confers resistance to tumor therapy. However, the specific role of GSTP1 in radiotherapy resistance in pancreatic cancer (PC) is not known. In this study, we investigated how GSTP1 imparts radioresistance in PC. The findings of previous studies and this study revealed that ionizing radiation (IR) induces ferroptosis in pancreatic cancer cells, primarily by upregulating the expression of ACSL4. Our results showed that after IR, GSTP1 prolonged the survival of pancreatic cancer cells by inhibiting ferroptosis but did not affect apoptosis. The expression of GSTP1 reduced cellular ferroptosis by decreasing the levels of ACSL4 and increasing the GSH content. These changes increase the resistance of pancreatic cancer cells and xenograft tumors to IR. Our findings indicate that ferroptosis participates in irradiation-induced cell death and that GSTP1 prevents IR-induced death of pancreatic cancer cells by inhibiting ferroptosis.
Assuntos
Ferroptose , Glutationa S-Transferase pi , Neoplasias Pancreáticas , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/radioterapia , Glutationa S-Transferase pi/metabolismo , Glutationa S-Transferase pi/genética , Humanos , Animais , Linhagem Celular Tumoral , Camundongos , Camundongos Nus , Coenzima A Ligases/metabolismo , Coenzima A Ligases/genética , Apoptose/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto , Radiação Ionizante , Tolerância a Radiação , Camundongos Endogâmicos BALB C , Glutationa/metabolismoRESUMO
BACKGROUND: Ionizing radiation (IR), including radiotherapy, can exert lasting harm on living organisms. While liposaccharide (LPS) offers resistance to radiation damage, it also induces toxic responses. Thankfully, an LPS analogue called N-formylmethionine-leucyl-phenylalanine (fMLP) holds the potential to mitigate this toxicity, offering hope for radiation protection. METHODS: Survival of C57BL/6 mice exposed to IR after administration with fMLP/LPS/WR-2721 or saline was recorded. Cell viability and apoptosis assay of bone marrow (BMC), spleen and small intestinal epithelial (HIECs) cells were tested by Cell Counting Kit-8 (CCK-8) and flow cytometry assay. Tissue damage was evaluated by Hematoxilin and Eosin (H&E), Ki-67, and TUNEL staining. RNA sequencing was performed to reveal potential mechanisms of fMLP-mediated radiation protection. Flow cytometry and western blot were performed to verify the radiation protection mechanism of fMLP on the cell cycle. RESULTS: The survival rates of C57BL/6 mice exposed to ionizing radiation after administering fMLP increased. fMLP demonstrated low toxicity in vitro and in vivo, maintaining cell viability and mitigating radiation-induced apoptosis. Moreover, it protected against tissue damage in the hematopoietic and intestinal system. RNA sequencing shed light on fMLP's potential mechanism, suggesting its role in modulating innate immunity and cell cycling. This was evidenced by its ability to reverse radiation-induced G2/M phase arrests in HIECs. CONCLUSION: fMLP serves as a promising radioprotective agent, preserving cells and radiosensitive tissues from IR. Through its influence on the cell cycle, particularly reversing radiation-induced arrest in G2/M phases, fMLP offers protection against IR's detrimental effects.
Assuntos
Apoptose , Hematopoese , Protetores contra Radiação , Animais , Camundongos , Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Protetores contra Radiação/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Camundongos Endogâmicos C57BL , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Radiação Ionizante , Intestinos/efeitos dos fármacos , Intestinos/efeitos da radiação , Intestinos/patologia , MasculinoRESUMO
Ionizing radiation is a significant risk factor for cataracts, but the pathogenesis of radiation-induced cataracts remains incompletely understood. Ferroptosis, an iron-dependent form of programmed cell death discovered in recent years, has gained increasing attention for its role in various diseases. This article systematically reviews research progress on ionizing radiation, ferroptosis, age-related cataracts, and radiation-induced cataracts. It proposes the "ferroptosis hypothesis" for the pathogenesis of radiation-induced cataracts. Through ionization and oxidative stress effects, ionizing radiation leads to elevated free iron levels and exacerbated lipid peroxidation in lens cells, activating the ferroptosis pathway and resulting in lens opacity. The involvement of ferroptosis in the development of age-related cataracts suggests that it may also be an important pathogenic mechanism of radiation-induced cataracts. Targeting the ferroptosis pathway may be a novel strategy for preventing and treating radiation-induced cataracts. Furthermore, developing new ferroptosis-specific inhibitors with improved targeting and pharmacokinetic properties is also an essential direction for research on preventing and treating radiation-induced cataracts. The study of ferroptosis provides new insights into the mechanism and management of radiation-induced cataracts, potentially transforming radiation-induced cataracts from "inevitable" to "preventable and treatable."
Assuntos
Catarata , Ferroptose , Catarata/etiologia , Humanos , Peroxidação de Lipídeos , Estresse Oxidativo , Lesões por Radiação/etiologia , Animais , Radiação Ionizante , Cristalino/efeitos da radiação , Ferro/metabolismoRESUMO
Background: Radiation therapy is an effective local therapy for lung cancer. However, the interaction between genes and radiotherapy is multifaceted and intricate. Therefore, we explored the role of miR-93-5p in the proliferation, apoptosis, and migration abilities of A549 cells. Simultaneously, we also investigated the interactions between miR-93-5p and ionizing radiation (IR). Methods: Cell Counting Kit-8, transwell, and apoptotic assay were performed to measure the proliferation, migration, and apoptosis abilities. The expression levels of miR-93-5p and its target gene in lung cancer were predicted using starBase v3.0. Then, data were validated using qPCR and western blot. Results: miR-93-5p significantly promoted the proliferation (P < 0.01) and migration abilities (P < 0.001) of A549 cells. Gasdermin E (GSDME) was identified to be a putative target of miR-93-5p and had a negative correlation with miR-93-5p (P < 0.001). Overexpression of miR-93-5p significantly decreased GSDME in A549 (P < 0.001). Interestingly, miR-93-5p decreased cell proliferation (P < 0.01) and cell migration (P < 0.01) and increased apoptosis (P < 0.01) in A549 cells after exposure to IR. Conclusions: miR-93-5p is presumed to play an oncogenic role in lung cancer by enhancing A549 cell proliferation and migration. It can enhance the sensitivity of radiotherapy under IR conditions. We speculate that the miR-93-5p/GSDME pathway was inhibited, activating the GSDME-related pyroptosis pathway when the cells were exposed to IR. Therefore, miR-93-5p can overcome resistance to radiotherapy and improve the efficacy of radiotherapy.
Assuntos
Apoptose , Carcinoma Pulmonar de Células não Pequenas , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares , MicroRNAs , Radiação Ionizante , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Células A549 , Movimento Celular/efeitos da radiação , Movimento Celular/genética , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proliferação de Células/efeitos da radiação , Proliferação de Células/genética , Apoptose/efeitos da radiação , Apoptose/genética , Regulação Neoplásica da Expressão Gênica/efeitos da radiaçãoRESUMO
BACKGROUND: Exposure to ionizing radiation has been linked to cardiovascular diseases. However, the impact of moderate doses of radiation on abdominal aortic aneurysm (AAA) remains unknown. METHODS: Angiotensin II-infused Apoe-/- mice were irradiated (acute, 1 Gray) either 3 days before (Day-3) or 1 day after (Day+1) pomp implantation. Isolated primary aortic vascular smooth muscle cells (VSMCs) were irradiated (acute 1 Gray) for mechanistic studies and functional testing in vitro. RESULTS: Day-3 and Day+1 irradiation resulted in a significant reduction in aorta dilation (Control: 1.39+/-0.12; Day-3: 1.12+/-0.11; Day+1: 1.15+/-0.08 mm, P<0.001) and AAA incidence (Control: 81.0%; Day-3: 33.3%, Day+1: 53.3%) compared to the non-irradiated group. Day-3 and Day+1 irradiation led to an increase in collagen content in the adventitia (Thickness control: 23.64+/-2.9; Day-3: 54.39+/-15.5; Day+1 37.55+/-10.8 mm, P = 0.006). However, the underlying protective mechanisms were different between Day-3 and Day+1 groups. Irradiation before Angiotensin II (AngII) infusion mainly modulated vascular smooth muscle cell (VSMC) phenotype with a decrease in contractile profile and enhanced proliferative and migratory activity. Irradiation after AngII infusion led to an increase in macrophage content with a local anti-inflammatory phenotype characterized by the upregulation of M2-like gene and IL-10 expression. CONCLUSION: Moderate doses of ionizing radiation mitigate AAA either through VSCM phenotype or inflammation modulation, depending on the time of irradiation.
Assuntos
Angiotensina II , Aneurisma da Aorta Abdominal , Músculo Liso Vascular , Miócitos de Músculo Liso , Radiação Ionizante , Animais , Aneurisma da Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/etiologia , Camundongos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efeitos da radiação , Músculo Liso Vascular/patologia , Angiotensina II/farmacologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/efeitos da radiação , Miócitos de Músculo Liso/patologia , Masculino , Modelos Animais de Doenças , Interleucina-10/metabolismo , Interleucina-10/genética , Colágeno/metabolismo , Proliferação de Células/efeitos da radiaçãoRESUMO
The genome is constantly exposed to DNA damage from endogenous and exogenous sources. Fine modulation of DNA repair, chromatin remodeling, and transcription factors is necessary for protecting genome integrity, but the precise mechanisms are still largely unclear. We found that after ionizing radiation (IR), global trimethylation of histone H3 at lysine 4 (H3K4me3) was decreased at an early (5 min) post-IR phase but increased at an intermediate (180 min) post-IR phase in both human and mouse hematopoietic cells. We demonstrated that PTIP, a component of the MLL histone methyltransferase complex, is required for H3K4me3 upregulation in the intermediate post-IR phase and promotes cell cycle arrest by epigenetically inducing a cell cycle inhibitor, PRDM1. In addition, we found that PTIP expression is specifically downregulated in acute myeloid leukemia patients. These findings collectively suggest that the PTIP-PRDM1 axis plays an essential role in proper DNA damage response and its deregulation contributes to leukemogenesis.
Assuntos
Pontos de Checagem do Ciclo Celular , Dano ao DNA , Proteínas de Ligação a DNA , Fator 1 de Ligação ao Domínio I Regulador Positivo , Animais , Humanos , Camundongos , Proteínas de Transporte/metabolismo , Proteínas de Transporte/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Epigênese Genética , Histonas/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Radiação Ionizante , Regulação para CimaRESUMO
In major radiological events, rapid assays to detect ionizing radiation exposure are crucial for effective medical interventions. The purpose of these assays is twofold: to categorize affected individuals into groups for initial treatments, and to provide definitive dose estimates for continued care and epidemiology. However, existing high-throughput cytogenetic biodosimetry assays take about 3 days to yield results, which delays critical interventions. We have developed a multiwell-based variant of the chemical-induced G0-phase Premature Chromosome Condensation Assay that delivers same-day results. Our findings revealed that using a concentration of phosphatase inhibitor lower than recommended significantly increases the yield of cells with highly condensed chromosomes. These chromosomes exhibited increased fragmentation in a dose-dependent manner, enabling to quantify radiation damage using a custom Deep Learning algorithm. This algorithm demonstrated reasonable performance in categorizing doses into distinct treatment groups (84% and 80% accuracy for three and four iso-treatment dose bins, respectively) and showed reliability in determining the actual doses received (correlation coefficient of 0.879). This method is amendable to full automation and has the potential to address the need for same-day, high-throughput cytogenetic test for both dose categorization and dose reconstruction in large-scale radiation emergencies.
Assuntos
Radiometria , Humanos , Radiometria/métodos , Radiação Ionizante , Bioensaio/métodos , Relação Dose-Resposta à Radiação , Ensaios de Triagem em Larga Escala/métodosRESUMO
Ubiquitination is a crucial posttranslational modification required for the proper repair of DNA double-strand breaks (DSBs) induced by ionizing radiation (IR). DSBs are mainly repaired through homologous recombination (HR) when template DNA is present and nonhomologous end joining (NHEJ) in its absence. In addition, microhomology-mediated end joining (MMEJ) and single-strand annealing (SSA) provide backup DSBs repair pathways. However, the mechanisms controlling their use remain poorly understood. By using a high-resolution CRISPR screen of the ubiquitin system after IR, we systematically uncover genes required for cell survival and elucidate a critical role of the E3 ubiquitin ligase SCFcyclin F in cell cycle-dependent DSB repair. We show that SCFcyclin F-mediated EXO1 degradation prevents DNA end resection in mitosis, allowing MMEJ to take place. Moreover, we identify a conserved cyclin F recognition motif, distinct from the one used by other cyclins, with broad implications in cyclin specificity for cell cycle control.
Assuntos
Ciclo Celular , Ciclinas , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Exodesoxirribonucleases , Humanos , Ciclo Celular/genética , Exodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/genética , Ciclinas/metabolismo , Ciclinas/genética , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/genética , Reparo do DNA por Junção de Extremidades , Ubiquitinação , Radiação IonizanteRESUMO
Resistance to radiotherapy is a major barrier during cancer treatment. Here using genome-scale CRISPR/Cas9 screening, we identify CD274 gene, which encodes PD-L1, to confer lung cancer cell resistance to ionizing radiation (IR). Depletion of endogenous PD-L1 delays the repair of IR-induced DNA double-strand breaks (DSBs) and PD-L1 loss downregulates non-homologous end joining (NHEJ) while overexpression of PD-L1 upregulates NHEJ. IR induces translocation of PD-L1 from the membrane into nucleus dependent on deglycosylation of PD-L1 at N219 and CMTM6 and leads to PD-L1 recruitment to DSBs foci. PD-L1 interacts with Ku in the nucleus and enhances Ku binding to DSB DNA. The interaction between the IgC domain of PD-L1 and the core domain of Ku is required for PD-L1 to accelerate NHEJ-mediated DSB repair and produce radioresistance. Thus, PD-L1, in addition to its immune inhibitory activity, acts as mechanistic driver for NHEJ-mediated DSB repair in cancer.
Assuntos
Antígeno B7-H1 , Núcleo Celular , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Autoantígeno Ku , Humanos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Autoantígeno Ku/metabolismo , Autoantígeno Ku/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Animais , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/patologia , Camundongos , Glicosilação , Radiação Ionizante , Sistemas CRISPR-CasRESUMO
A model for accelerated aging in mice was developed: CB6F2 mice aged 39-45 days were exposed to fractionated 4-fold relatively uniform γ-radiation (137Cs, 0.98 Gy/min) at a total dose of 6.8 Gy. Radiation exposure led to delayed active growth, leukopenia, and lymphopenia for over 1 year during the post-radiation period. The death of irradiated males and females occurred significantly earlier than in control group animals. Median lifespans in the experimental group were 35-38% lower than in the control group (p<0.001). Ionizing radiation exposure led to the early development of hair depigmentation, cachexia, and the development of aging-associated diseases. In irradiated mice, oncological pathology constituted 30-35% in the mortality structure, which is twice as often as in the control group. The developed model can be used to study the pathogenesis of accelerated aging under radiation exposure and the search for means of its prevention and treatment.
Assuntos
Senilidade Prematura , Raios gama , Animais , Camundongos , Masculino , Feminino , Raios gama/efeitos adversos , Senilidade Prematura/patologia , Senilidade Prematura/genética , Senilidade Prematura/etiologia , Longevidade/efeitos da radiação , Radiação Ionizante , Envelhecimento/efeitos da radiação , Caquexia/patologia , Caquexia/etiologia , Radioisótopos de CésioRESUMO
Breast cancer is the most diagnosed cancer in the world, and it is the primary cause of cancer death for women. The risk of breast cancer is increased by endogenous factors like hormones and exogenous factors like radiation exposure that causes damage to the mammary epithelial cells leading to an inflammatory response. Chronic inflammation creates a microenvironment composed of, among other factors, chemokines, and interleukins, which promote cancer. The gene expression of the interleukin 1 receptor type 1, the interleukin 1 receptor antagonist, the Interleukin 1 Receptor Accessory Protein, the interleukin 6 cytokine family signal transducer, the C-X-C motif chemokine ligand 3, the C-X-C motif chemokine ligand 5, and the C-X-C motif chemokine ligand 6 were analyzed in an estrogen and radiation experimental breast cancer model. Furthermore, the expression of these genes was correlated with immune cell infiltration, estrogen receptor expression, and their clinical relevance in breast cancer patients based on data provided by The Cancer Genome Atlas database online. Results given by the experimental breast cancer model showed that all genes related to inflammation respond to ionizing radiation alone or in combination with estrogen. On the other hand, the immune response depended on the breast cancer type and on the expression of the gene that encoded the estrogen receptor. Finally, the importance of the expression of these genes in breast cancer is such that high IL1R1 or IL1RAP is strongly related to patient survival. These findings may help to improve the understanding of the role of immune molecules in carcinogenesis and enhance therapeutic approaches.
Assuntos
Neoplasias da Mama , Receptor alfa de Estrogênio , Estrogênios , Inflamação , Radiação Ionizante , Feminino , Receptor alfa de Estrogênio/metabolismo , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/genética , Inflamação/metabolismo , Humanos , Camundongos , Regulação Neoplásica da Expressão Gênica , Microambiente TumoralRESUMO
BACKGROUND: Gastrointestinal-acute radiation syndrome (GI-ARS) caused by moderate to high doses of ionizing radiation exposure contribute to early death in humans. GI injury is also a common adverse effect seen in cancer patients undergoing abdominal/pelvic radiotherapy. Currently, no countermeasure agents have been approved for medical management of GI-ARS. The present study aims to evaluate the mechanism of action of Trichostatin A(TSA), a pan histone deacetylase inhibitor, against radiation-induced GI injury. METHODS: TSA (150 ng/kg bw) was administered to mice 1 h and 24 h after 15 Gy abdominal irradiation. Expression of various markers of oxidative stress, mitochondrial dysfunction, and apoptosis were checked in the jejunum, and their possible regulation through the Nrf2 signaling pathway was evaluated. RESULTS: TSA administered post-irradiation (15 Gy + TSA) elevated intestinal total antioxidant and glutathione levels by regulating the expression of Slc7A11 and antioxidant proteins, GCLC, GPX4, and TXNRD1. Improved mitochondrial membrane potential, ATP levels, downregulation of mitochondrial quality control proteins, (PINK1 and PARKIN), and differential regulation of the apoptotic proteins, (BAX, PUMA and BCL2) with reduced intestinal epithelial cell apoptosis in the TSA-adminstered group were observed. TSA also upregulated Nrf2 in the presence of its specific inhibitor, ML385, suggesting its involvement in regulating Nrf2 signaling during oxidative stress induced by radiation in intestine. H & E stained jejunum cross-sections revealed that TSA mitigated radiation-mediated intestinal injury in mice. CONCLUSIONS: Present findings indicate that TSA is beneficial in mitigating the damaging effects of ionizing radiation in the intestine.
Assuntos
Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos , Fator 2 Relacionado a NF-E2 , Estresse Oxidativo , Proteínas Quinases , Radiação Ionizante , Transdução de Sinais , Ubiquitina-Proteína Ligases , Animais , Fator 2 Relacionado a NF-E2/metabolismo , Camundongos , Ácidos Hidroxâmicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Proteínas Quinases/metabolismo , Masculino , Ubiquitina-Proteína Ligases/metabolismo , Oxirredução/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Glutationa Peroxidase/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/efeitos da radiação , Intestinos/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos da radiação , Síndrome Aguda da Radiação/tratamento farmacológico , Síndrome Aguda da Radiação/metabolismoRESUMO
DNA damage has been implicated in the stimulation of the type 1 interferon (T1IFN) response. Here, we show that downregulation of the DNA repair protein, polynucleotide kinase/phosphatase (PNKP), in a variety of cell lines causes robust phosphorylation of STAT1, upregulation of interferon-stimulated genes and persistent accumulation of cytosolic DNA, all of which are indicators for the activation of the T1IFN response. Furthermore, this did not require damage induction by ionizing radiation. Instead, our data revealed that production of reactive oxygen species (ROS) synergises with PNKP loss to potentiate the T1IFN response, and that loss of PNKP significantly compromises mitochondrial DNA (mtDNA) integrity. Depletion of mtDNA or treatment of PNKP-depleted cells with ROS scavengers abrogated the T1IFN response, implicating mtDNA as a significant source of the cytosolic DNA required to potentiate the T1IFN response. The STING signalling pathway is responsible for the observed increase in the pro-inflammatory gene signature in PNKP-depleted cells. While the response was dependent on ZBP1, cGAS only contributed to the response in some cell lines. Our data have implications for cancer therapy, since PNKP inhibitors would have the potential to stimulate the immune response, and also to the neurological disorders associated with PNKP mutation.
Assuntos
Enzimas Reparadoras do DNA , DNA Mitocondrial , Interferon Tipo I , Fosfotransferases (Aceptor do Grupo Álcool) , Radiação Ionizante , Espécies Reativas de Oxigênio , Humanos , Interferon Tipo I/metabolismo , Interferon Tipo I/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/genética , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Reparo do DNA , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/genética , Dano ao DNA , Linhagem Celular , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Transdução de Sinais , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Fosforilação , Citosol/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genéticaRESUMO
The concept of 'hormesis' is defined as a dose-response relationship whereby low doses of various toxic substances or physical stressors trigger bio-positive effects in diverse biological systems, whereas high doses cause inhibition of cellular performance (e.g. growth, viability). The two-sided phenomenon of specific low-dose stimulation and high-dose inhibition imposed by a 'hormetic-factor' has been well documented in toxicology and pharmacology. Multitudinous factors have been identified that correspondingly cause hormetic effects in diverse taxa of animals, fungi, and plants. This study particularly aims to elucidate the molecular basis for stimulatory implications of ionizing radiation (IR) on plant male gametophytes (pollen). Beyond that, this analysis impacts general research on cell growth, plant breeding, radiation protection, and, in a wider sense, medical treatment. For this purpose, IR-related data were surveyed and discussed in connection with the present knowledge about pollen physiology. It is concluded that IR-induced reactive oxygen species (ROS) have a key role here. Moreover, it is hypothesized that IR-exposure shifts the ratio between diverse types of ROS in the cell. The interrelation between ROS, intracellular Ca2+-gradient, NADPH oxidases, ROS-scavengers, actin dynamics, and cell wall properties are most probably involved in IR-hormesis of pollen germination and tube growth. Modulation of gene expression, phytohormone signalling, and cellular antioxidant capacity are also implicated in IR-hormesis.
Assuntos
Pólen , Radiação Ionizante , Espécies Reativas de Oxigênio , Pólen/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Hormese/efeitos da radiação , Germinação/efeitos da radiaçãoRESUMO
In the United States, the Federal Aviation Administration has officially classified flight crews (FC) consisting of commercial pilots, cabin crew, or flight attendants as "radiation workers" since 1994 due to the potential for cosmic ionizing radiation (CIR) exposure at cruising altitudes originating from solar activity and galactic sources. Several epidemiological studies have documented elevated incidence and mortality for several cancers in FC, but it has not yet been possible to establish whether this is attributable to CIR. CIR and its constituents are known to cause a myriad of DNA lesions, which can lead to carcinogenesis unless DNA repair mechanisms remove them. But critical knowledge gaps exist with regard to the dosimetry of CIR, the role of other genotoxic exposures among FC, and whether possible biological mechanisms underlying higher cancer rates observed in FC exist. This review summarizes our understanding of the role of DNA damage and repair responses relevant to exposure to CIR in FC. We aimed to stimulate new research directions and provide information that will be useful for guiding regulatory, public health, and medical decision-making to protect and mitigate the risks for those who travel by air.
Assuntos
Radiação Cósmica , Dano ao DNA , Exposição Ocupacional , Humanos , Radiação Cósmica/efeitos adversos , Exposição Ocupacional/efeitos adversos , Reparo do DNA , Radiação Ionizante , Neoplasias Induzidas por Radiação/etiologia , Neoplasias Induzidas por Radiação/genética , Neoplasias/etiologia , Neoplasias/genéticaRESUMO
Radiation cytogenetics has a rich history seldom appreciated by those outside the field. Early radiobiology was dominated by physics and biophysical concepts that borrowed heavily from the study of radiation-induced chromosome aberrations. From such studies, quantitative relationships between biological effect and changes in absorbed dose, dose rate and ionization density were codified into key concepts of radiobiological theory that have persisted for nearly a century. This review aims to provide a historical perspective of some of these concepts, including evidence supporting the contention that chromosome aberrations underlie development of many, if not most, of the biological effects of concern for humans exposed to ionizing radiations including cancer induction, on the one hand, and tumor eradication on the other. The significance of discoveries originating from these studies has widened and extended far beyond their original scope. Chromosome structural rearrangements viewed in mitotic cells were first attributed to the production of breaks by the radiations during interphase, followed by the rejoining or mis-rejoining among ends of other nearby breaks. These relatively modest beginnings eventually led to the discovery and characterization of DNA repair of double-strand breaks by non-homologous end joining, whose importance to various biological processes is now widely appreciated. Two examples, among many, are V(D)J recombination and speciation. Rapid technological advancements in cytogenetics, the burgeoning fields of molecular radiobiology and third-generation sequencing served as a point of confluence between the old and new. As a result, the emergent field of "cytogenomics" now becomes uniquely positioned for the purpose of more fully understanding mechanisms underlying the biological effects of ionizing radiation exposure.
Assuntos
Aberrações Cromossômicas , Citogenética , Radiobiologia , Humanos , Aberrações Cromossômicas/efeitos da radiação , Animais , Reparo do DNA/efeitos da radiação , Radiação Ionizante , História do Século XX , Quebras de DNA de Cadeia Dupla/efeitos da radiaçãoRESUMO
This historical review of extracellular vesicles in the setting of exposure to ionizing radiation (IR) traces our understanding of how vesicles were initially examined and reported in the literature in the late 1970s (for secreted exosomes) and early 1980s (for plasma membrane-derived, exfoliated vesicles) to where we are now and where we may be headed in the next decade. An emphasis is placed on biophysical properties of extracellular vesicles, energy consumption and the role of vesiculation as an essential component of membrane turnover. The impact of intercellular signal trafficking by vesicle surface and intra-vesicular lipids, proteins, nucleic acids and metabolites is reviewed in the context of biomarkers for estimating individual radiation dose after exposure to radiation, pathogenesis of disease and development of cell-free therapeutics. Since vesicles express both growth stimulatory and inhibitory molecules, a hypothesis is proposed to consider superposition in a shared space and entanglement of molecules by energy sources that are external to human cells. Implications of this approach for travel in deep space are briefly discussed in the context of clinical disorders that have been observed after space travel.
Assuntos
Membrana Celular , Humanos , Membrana Celular/metabolismo , Membrana Celular/efeitos da radiação , Vesículas Extracelulares/efeitos da radiação , Vesículas Extracelulares/metabolismo , Radiometria , Voo Espacial , Animais , Sistema Livre de Células , História do Século XX , Radiação IonizanteRESUMO
The increased risk of liver malignancies was found in workers of the first Russian nuclear production facility, Mayak Production Association, who had been chronically exposed to gamma rays externally and to alpha particles internally due to plutonium inhalation. In the present study, we updated the radiogenic risk estimates of the hepatobiliary malignancies using the extended follow-up period (1948-2018) of the Mayak worker cohort and the improved «Mayak worker dosimetry system-2013¼. The cohort comprised 22,377 workers hired at the Mayak PA between 1948 and 1982. The analysis considered 62 liver malignancies (32 hepatocellular carcinomas, 13 intrahepatic cholangiocarcinomas, 16 angiosarcomas, and 1 anaplastic cancer) and 33 gallbladder adenocarcinomas. The analysis proved the positive significant association of the liver malignancy risk (the total of histological types, hepatocellular carcinoma) with the liver absorbed alpha dose from internal exposure. The excess relative risk per Gy (95% confidence interval) of alpha dose (the linear model) was 7.56 (3.44; 17.63) for the total of histological types and 3.85 (0.95; 13.30) for hepatocellular carcinoma. Indications of non-linearity were observed in the dose-response for internal exposure to alpha radiation. No impact of external gamma-ray exposure on the liver malignancy incidence was found. In the study cohort, the number of angiosarcomas among various types of liver malignancies was very high (25.8%), and most of these tumors (73.3%) were registered in individuals internally exposed to alpha radiation at doses ranging between 6.0 and 21.0 Gy. No association with chronic occupational radiation exposure was observed for the incidence of gallbladder malignancies.