Low intensity microwave radiation induced oxidative stress, inflammatory response and DNA damage in rat brain

2015 ◽  
Vol 51 ◽  
pp. 158-165 ◽  
Author(s):  
Kanu Megha ◽  
Pravin Suryakantrao Deshmukh ◽  
Basu Dev Banerjee ◽  
Ashok Kumar Tripathi ◽  
Rafat Ahmed ◽  
...  
2015 ◽  
Vol 73 (1) ◽  
pp. 93-100 ◽  
Author(s):  
Kanu Megha ◽  
Pravin S. Deshmukh ◽  
Alok K. Ravi ◽  
Ashok K. Tripathi ◽  
Mahesh P. Abegaonkar ◽  
...  

Antioxidants ◽  
2021 ◽  
Vol 10 (11) ◽  
pp. 1850
Author(s):  
Jinlong Wei ◽  
Qin Zhao ◽  
Yuyu Zhang ◽  
Weiyan Shi ◽  
Huanhuan Wang ◽  
...  

This article mainly observed the protective effect of sulforaphane (SFN) on radiation-induced skin injury (RISI). In addition, we will discuss the mechanism of SFN’s protection on RISI. The RISI model was established by the irradiation of the left thigh under intravenous anesthesia. Thirty-two C57/BL6 mice were randomly divided into control group (CON), SFN group, irradiation (IR) group, and IR plus SFN (IR/SFN) group. At eight weeks after irradiation, the morphological changes of mouse skin tissues were detected by H&E staining. Then, the oxidative stress and inflammatory response indexes in mouse skin tissues, as well as the expression of Nrf2 and its downstream antioxidant genes, were evaluated by ELISA, real-time PCR, and Western blotting. The H&E staining showed the hyperplasia of fibrous tissue in the mouse dermis and hypodermis of the IR group. Western blotting and ELISA results showed that the inflammasome of NLRP3, caspase-1, and IL-1β, as well as oxidative stress damage indicators ROS, 4-HNE, and 3-NT, in the skin tissues of mice in the IR group were significantly higher than those in the control group (p < 0.05). However, the above pathological changes declined sharply after SFN treatment (p < 0.05). In addition, the expressions of Nrf2 and its regulated antioxidant enzymes, including CAT and HO-1, were higher in the skin tissues of SFN and IR/SFN groups, but lower in the control and IR groups (p < 0.05). SFN may be able to suppress the oxidative stress by upregulating the expression and function of Nrf2, and subsequently inhibiting the activation of NLRP3 inflammasome and DNA damage, so as to prevent and alleviate the RISI.


2021 ◽  
Vol 69 (2) ◽  
pp. 113
Author(s):  
Aymen Mabrouk ◽  
Amina Sakly ◽  
Feiza Mnasria ◽  
Lobna Ezzi

2021 ◽  
Vol 2021 ◽  
pp. 1-18
Author(s):  
Clarissa S. Schütz ◽  
Matthias B. Stope ◽  
Sander Bekeschus

At serine139-phosphorylated gamma histone H2A.X (γH2A.X) has been established over the decades as sensitive evidence of radiation-induced DNA damage, especially DNA double-strand breaks (DSBs) in radiation biology. Therefore, γH2A.X has been considered a suitable marker for biomedical applications and a general indicator of direct DNA damage with other therapeutic agents, such as cold physical plasma. Medical plasma technology generates a partially ionized gas releasing a plethora of reactive oxygen and nitrogen species (ROS) simultaneously that have been used for therapeutic purposes such as wound healing and cancer treatment. The quantification of γH2A.X as a surrogate parameter of direct DNA damage has often been used to assess genotoxicity in plasma-treated cells, whereas no sustainable mutagenic potential of the medical plasma treatment could be identified despite H2A.X phosphorylation. However, phosphorylated H2A.X occurs during apoptosis, which is associated with exposure to cold plasma and ROS. This review summarizes the current understanding of γH2A.X induction and function in oxidative stress in general and plasma medicine in particular. Due to the progress towards understanding the mechanisms of H2A.X phosphorylation in the absence of DSB and ROS, observations of γH2A.X in medical fields should be carefully interpreted.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2359-2359
Author(s):  
Larisa Pereboeva ◽  
Erik Westin ◽  
Toral Patel ◽  
Ian Flaniken ◽  
Lawrence S. Lamb ◽  
...  

Abstract Abstract 2359 Introduction: Dyskeratosis congenita (DC) is an inherited multisystem disorder consisting of premature aging, cancer predisposition, bone marrow failure and the characteristic triad of mucosal leukoplakia, skin dyspigmentation and nail dystrophy. Symptomology associated with DC arises as a consequence of mutations within genes associated with telomeres and telomerase activity manifested by critically shortened telomeres in affected cells. We have previously reported a growth disadvantage and increased intracellular oxidative stress in cultured somatic cells obtained from patients with DC. We hypothesize that telomere maintenance is closely linked to dysregulation in oxidative pathways and consequent DNA damage. Our objective was to discern whether pharmacologic intervention to alleviate oxidative stress imparts a protective effect in DC cells. Methods: T lymphocytes from both DC subjects with hTERC mutations and age-matched controls were cultured and expanded in vitro using CD3/CD28 beads. DNA damage to cells was induced using paclitaxel, etoposide, or ionizing radiation during log-phase of cell growth. Cellular proliferation and apoptosis were monitored by cell counting and flow cytometry (FACS) using Annexin V antibody and propidium iodide. Western blotting was used to measure basal and radiation-induced expression of DNA damage response (DDR) proteins, including total p53 and its activated form (serine 15 phosphorylated; p53S15), p21WAF, and phosphorylated H2AX (gH2AX). Level of oxidative stress was determined by FACS using the cell-permeable fluorogenic probe DCFH and dihydroethedium (DHE) detecting reactive oxygen species (ROS). Anti-oxidants, including vitamin E and N acetyl cysteine (NAC), were used in vitro to modulate levels of oxidative stress in control and radiated cells. Results: Comparison of growth curves demonstrated a significant decrease in proliferation of T cells obtained from DC patients versus control T cells. This growth disadvantage was more pronounced following cell exposure to radiation, paclitaxel, and etoposide. To explain these differences we investigated several parameters indicative of DNA damage. DC lymphocytes had higher basal levels of apoptosis, while radiation resulted in comparable levels of apoptosis in both DC and control cultures. Similarly, DDR markers p53 and p53S15, but not p21 and g-H2AX, were basally expressed at higher levels in DC lymphocytes while radiation, in a dose-dependent manner, upregulated expression of p53, p53S15, p21 and g-H2AX in both DC and control lymphocytes. Consistent with DDR data, elevated basal levels of ROS were found in short term DC cultures. Additionally, in a dose dependent manner, the anti-oxidant NAC partially ameliorated the growth disadvantage of DC cells. Importantly, NAC also decreased radiation-induced apoptosis and oxidative stress in DC cells. Studies are ongoing to characterize the modulation of DDR markers in NAC-treated cells. Conclusions: DC is an important disease model for studying the effects of telomere shortening on cellular proliferation and other molecular pathways involved in cell senescence and aging. Our findings of elevated basal levels of apoptosis, DDR proteins and oxidative stress in DC lymphocytes, as well as increased sensitivity of DC cells to cytotoxic agents suggests a role of telomerase and/or telomere length in regulating oxidative and DNA damage response pathways. This data also validates the clinical finding of DC patients' intolerance to myeloablative therapy. Finally a pharmacologic approach to reduce oxidative stress may alleviate some of the untoward toxicities associated with current cytotoxic treatments in DC. Clinical trials testing various anti-oxidant therapies are currently under design. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document