Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Environ Toxicol ; 33(9): 931-945, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29968967

ABSTRACT

Deleterious effects of MW radiation on the male reproduction are well studied. Previous reports although suggest that 2.45 GHz MW irradiation induced oxidative and nitrosative stress adversely affects the male reproductive function but the detailed molecular mechanism occurring behind it has yet to be elucidated. The aim of present study was to investigate the underlying detailed pathway of the testicular apoptosis induced by free radical load and redox imbalance due to 2.45 GHz MW radiation exposure and the degree of severity along with the increased exposure duration. Twelve-week old male mice were exposed to 2.45 GHz MW radiation [continuous-wave (CW) with overall average Power density of 0.0248 mW/cm2 and overall average whole body SAR value of 0.0146 W/kg] for 2 hr/day over a period of 15, 30, and 60 days. Testicular histology, serum testosterone, ROS, NO, MDA level, activity of antioxidant enzymes, expression of pro-apoptotic proteins (p53 and Bax), anti-apoptotic proteins (Bcl-2 and Bcl-xL ), cytochrome-c, inactive/active caspase-3, and uncleaved PARP-1 were evaluated. Findings suggest that 2.45 GHz MW radiation exposure induced testicular redox imbalance not only leads to enhanced testicular apoptosis via p53 dependent Bax-caspase-3 mediated pathway, but also increases the degree of apoptotic severity in a duration dependent manner.


Subject(s)
Apoptosis/radiation effects , Caspase 3/metabolism , Microwaves , Nitrosative Stress/radiation effects , Oxidative Stress/radiation effects , Testis/radiation effects , Animals , Antioxidants/metabolism , Male , Mice , Oxidation-Reduction , Poly (ADP-Ribose) Polymerase-1/metabolism , Testis/metabolism , Testis/pathology , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
2.
Toxicol Sci ; 161(2): 349-374, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29069439

ABSTRACT

Microwave (MW) radiation induced oxidative stress reduces dendritic arborization, spine density and number of hippocampal pyramidal neurons and hence, impair learning and spatial memory through p53-dependent/independent apoptosis of hippocampal neuronal and nonneuronal cells. However, the mechanisms responsible for MW radiation induced impairment in memory formation remains still unknown. This study elucidates the effect of short (15 days) and long-term (30 and 60 days) low level 2.45 GHz MW radiation-induced local stress on the hippocampal spatial memory formation pathway in adult male mice. Twelve-weeks old mice were exposed to 2.45 GHz MW radiation (continuous-wave with overall average Power density of 0.0248 mW/cm2 and overall average whole body SAR value of 0.0146 W/Kg) @ 2 h/d for 15, 30, and 60 days. Learning and spatial memory was assessed by 8-arm radial maze. We have investigated the alterations in serum corticosterone level and the expression of glucocorticoid receptor, corticotropin-releasing hormone (CRH), inducible nitric oxide synthase (i-NOS), iGluRs, PSD-95-neuronal NOS (n-NOS) system, protein kinase A, protein kinase Cε-ERK1/2-pERK1/2 in all the hippocampal subregions, viz. CA1, CA2, CA3, and DG through immunohistochemistry/immunofluorescence and alterations in the expression of hippocampal glucocorticoid receptor, CRH-receptor 1 (CRH-R1), cAMP-response element-binding (CREB), and phosphorylated-CREB (p-CREB) through western blot analysis. We observed that 2.45 GHz MW irradiated mice showed slow learning and significantly increased number of working and reference memory errors in radial maze task. Further, 2.45 GHz MW radiation exposure increases serum corticosterone level and the expression of CRH, CRH-R1, and i-NOS, while the expression of iGluRs, n-NOS, PSD-95, protein kinase Cε, protein kinase A, ERK-p-ERK, CREB, and p-CREB decreases in above mentioned hippocampal subregions in a duration dependent manner. Our findings led us to conclude that 2.45 GHz MW radiation exposure induced local stress suppresses signaling mechanism(s) of hippocampal memory formation.


Subject(s)
Hippocampus/radiation effects , Learning/radiation effects , Microwaves/adverse effects , Oxidative Stress/radiation effects , Signal Transduction/radiation effects , Spatial Memory/radiation effects , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Radiation , Extracellular Signal-Regulated MAP Kinases/metabolism , Hippocampus/metabolism , Male , Mice , Receptors, Ionotropic Glutamate/metabolism
3.
Reprod Toxicol ; 73: 41-60, 2017 10.
Article in English | MEDLINE | ID: mdl-28780396

ABSTRACT

Present study investigated the long-term effects of mobile phone (1800MHz) radiation in stand-by, dialing and receiving modes on the female reproductive function (ovarian and uterine histo-architecture, and steroidogenesis) and stress responses (oxidative and nitrosative stress). We observed that mobile phone radiation induces significant elevation in ROS, NO, lipid peroxidation, total carbonyl content and serum corticosterone coupled with significant decrease in antioxidant enzymes in hypothalamus, ovary and uterus of mice. Compared to control group, exposed mice exhibited reduced number of developing and mature follicles as well as corpus lutea. Significantly decreased serum levels of pituitary gonadotrophins (LH, FSH), sex steroids (E2 and P4) and expression of SF-1, StAR, P-450scc, 3ß-HSD, 17ß-HSD, cytochrome P-450 aromatase, ER-α and ER-ß were observed in all the exposed groups of mice, compared to control. These findings suggest that mobile phone radiation induces oxidative and nitrosative stress, which affects the reproductive performance of female mice.


Subject(s)
Cell Phone , Electromagnetic Radiation , Ovary , Reproduction , Uterus , Animals , Corticosterone/blood , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Hydroxysteroid Dehydrogenases/metabolism , Hypothalamus/metabolism , Luteinizing Hormone/blood , Mice , Nitrates/metabolism , Nitrites/metabolism , Ovary/metabolism , Ovary/pathology , Oxidative Stress , Progesterone/blood , Reactive Oxygen Species , Uterus/metabolism , Uterus/pathology
4.
Toxicol Sci ; 148(2): 380-99, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26396154

ABSTRACT

A close association between microwave (MW) radiation exposure and neurobehavioral disorders has been postulated but the direct effects of MW radiation on central nervous system still remains contradictory. This study was performed to understand the effect of short (15 days) and long-term (30 and 60 days) low-level MW radiation exposure on hippocampus with special reference to spatial learning and memory and its underlying mechanism in Swiss strain male mice, Mus musculus. Twelve-weeks old mice were exposed to 2.45 GHz MW radiation (continuous-wave [CW] with overall average power density of 0.0248 mW/cm(2) and overall average whole body specific absorption rate value of 0.0146 W/Kg) for 2 h/day over a period of 15, 30, and 60 days). Spatial learning and memory was monitored by Morris Water Maze. We have checked the alterations in hippocampal oxidative/nitrosative stress, neuronal morphology, and expression of pro-apoptotic proteins (p53 and Bax), inactive executioner Caspase- (pro-Caspase-3), and uncleaved Poly (ADP-ribose) polymerase-1 in the hippocampal subfield neuronal and nonneuronal cells (DG, CA1, CA2, and CA3). We observed that, short-term as well as long-term 2.45 GHz MW radiation exposure increases the oxidative/nitrosative stress leading to enhanced apoptosis in hippocampal subfield neuronal and nonneuronal cells. Present findings also suggest that learning and spatial memory deficit which increases with the increased duration of MW exposure (15 < 30 < 60 days) is correlated with a decrease in hippocampal subfield neuronal arborization and dendritic spines. These findings led us to conclude that exposure to CW MW radiation leads to oxidative/nitrosative stress induced p53-dependent/independent activation of hippocampal neuronal and nonneuronal apoptosis associated with spatial memory loss.


Subject(s)
Apoptosis/radiation effects , Behavior, Animal/radiation effects , Hippocampus/drug effects , Maze Learning/radiation effects , Microwaves/adverse effects , Oxidative Stress/radiation effects , Reactive Nitrogen Species/metabolism , Spatial Memory/drug effects , Tumor Suppressor Protein p53/metabolism , Animals , Caspase 3/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Lipid Peroxidation/radiation effects , Male , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Carbonylation/radiation effects , Reactive Oxygen Species/metabolism , Signal Transduction/radiation effects , Time Factors , bcl-2-Associated X Protein/metabolism
5.
Appl Biochem Biotechnol ; 169(5): 1727-51, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23334843

ABSTRACT

The present experiment was designed to study the 2.45 GHz low-level microwave (MW) irradiation-induced stress response and its effect on implantation or pregnancy in female mice. Twelve-week-old mice were exposed to MW radiation (continuous wave for 2 h/day for 45 days, frequency 2.45 GHz, power density=0.033549 mW/cm(2), and specific absorption rate=0.023023 W/kg). At the end of a total of 45 days of exposure, mice were sacrificed, implantation sites were monitored, blood was processed to study stress parameters (hemoglobin, RBC and WBC count, and neutrophil/lymphocyte (N/L) ratio), the brain was processed for comet assay, and plasma was used for nitric oxide (NO), progesterone and estradiol estimation. Reactive oxygen species (ROS) and the activities of ROS-scavenging enzymes- superoxide dismutase, catalase, and glutathione peroxidase-were determined in the liver, kidney and ovary. We observed that implantation sites were affected significantly in MW-irradiated mice as compared to control. Further, in addition to a significant increase in ROS, hemoglobin (p<0.001), RBC and WBC counts (p<0.001), N/L ratio (p<0.01), DNA damage (p<0.001) in brain cells, and plasma estradiol concentration (p<0.05), a significant decrease was observed in NO level (p<0.05) and antioxidant enzyme activities of MW-exposed mice. Our findings led us to conclude that a low level of MW irradiation-induced oxidative stress not only suppresses implantation, but it may also lead to deformity of the embryo in case pregnancy continues. We also suggest that MW radiation-induced oxidative stress by increasing ROS production in the body may lead to DNA strand breakage in the brain cells and implantation failure/resorption or abnormal pregnancy in mice.


Subject(s)
Brain/radiation effects , Embryo Implantation/radiation effects , Ovary/radiation effects , Pregnancy, Animal/radiation effects , Animals , Brain/enzymology , Catalase/metabolism , Comet Assay , DNA Damage , Estradiol/blood , Female , Glutathione Peroxidase/metabolism , Kidney/enzymology , Kidney/radiation effects , Lipid Peroxidation/radiation effects , Liver/enzymology , Liver/radiation effects , Mice , Microwaves , Ovary/enzymology , Oxidative Stress/radiation effects , Pregnancy , Pregnancy, Animal/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL