Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Radiat Res ; 60(3): 308-317, 2019 May 01.
Article in English | MEDLINE | ID: mdl-31038675

ABSTRACT

There are no FDA-approved drugs that can be administered prior to ionizing radiation exposure to prevent hematopoietic-acute radiation syndrome (H-ARS). A suspension of synthetic genistein nanoparticles was previously shown to be an effective radioprotectant against H-ARS when administered prior to exposure to a lethal dose of total body radiation. Here we aimed to determine the time to protection and the duration of protection when the genistein nanosuspension was administered by intramuscular injection, and we also investigated the drug's mechanism of action. A single intramuscular injection of the genistein nanosuspension was an effective radioprotectant when given prophylactically 48 h to 12 h before irradiation, with maximum effectiveness occurring when administered 24 h before. No survival advantage was observed in animals administered only a single dose of drug after irradiation. The dose reduction factor of the genistein nanosuspension was determined by comparing the survival of treated and untreated animals following different doses of total body irradiation. As genistein is a selective estrogen receptor beta agonist, we also explored whether this was a central component of its radioprotective mechanism of action. Mice that received an intramuscular injection of an estrogen receptor antagonist (ICI 182,780) prior to administration of the genistein nanosuspension had significantly lower survival following total body irradiation compared with animals only receiving the nanosuspension (P < 0.01). These data define the time to and duration of radioprotection following a single intramuscular injection of the genistein nanosuspension and identify its likely mechanism of action.


Subject(s)
Acute Radiation Syndrome/drug therapy , Genistein/therapeutic use , Hematopoiesis , Nanoparticles/chemistry , Radiation-Protective Agents/therapeutic use , Animals , Dose-Response Relationship, Radiation , Genistein/administration & dosage , Hematopoiesis/drug effects , Injections, Intramuscular , Male , Mice , Radiation Exposure , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Suspensions
2.
Front Psychiatry ; 9: 612, 2018.
Article in English | MEDLINE | ID: mdl-30524321

ABSTRACT

Background: Early assessment and intervention are crucial to alleviate symptoms and prevent long-term negative outcomes in children suffering from Attention-deficit/hyperactivity disorder (ADHD). In Germany, at present, no standardized screening for ADHD is routinely administered. This study aims to evaluate a potential screening measure in a study population that is representative for a primary school entrance exam population in a German metropolitan region. Methods: Based on various socio-demographic variables, a sample of n = 500 5-year-old children (58% boys, 42% girls), representative of a primary school entrance exam population from a German metropolitan region, was selected. Their parents completed a written survey consisting of the CBCL and a brief screening tool for ADHD symptomatology based on the DISYPS-II questionnaire. Demographic data were also collected. Results: The subscale "Attention problems" of the CBCL/4-18 showed results in the clinical range for n = 10 (2%) participants. The ADHD screening identified n = 23 (4.6%) participants as suspect of having ADHD with a statistically significant gender difference (n = 17 boys vs. n = 6 girls, p = 0.03). In n = 5 (1%) participants, all boys, both CBCL/4-18 and the ADHD screening were indicative of ADHD. Conclusions: Results indicate that screening for ADHD in this population may be both feasible and reasonable given the high prevalence and chronic nature of this disorder and the benefit of an early initiation of treatment. Results match previously reported figures for prevalence of ADHD-related symptoms and gender differences in preschool and older pediatric populations and thus do not support the hypothesis that the prevalence of ADHD in a metropolitan region is significantly higher than in other regions.

3.
Horm Metab Res ; 49(6): 457-465, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28482370

ABSTRACT

17ß-Estradiol is known to regulate energy metabolism and body weight. Ovariectomy results in body weight gain while estradiol administration results in a reversal of weight gain. Isoflavones, found in rodent chow, can mimic estrogenic effects making it crucial to understand the role of these compounds on metabolic regulation. The goal of this study is to examine the effect of dietary isoflavones on body weight regulation in the ovariectomized rat. This study will examine how dietary isoflavones can interact with estradiol treatment to affect body weight. Consistent with previous findings, animals fed an isoflavone-rich diet had decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin levels (p<0.05) compared to animals fed an isoflavone-free diet. Estradiol replacement resulted in decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin (p<0.05). Current literature suggests the involvement of cytokines in the inflammatory response of body weight gain. We screened a host of cytokines and chemokines that may be altered by dietary isoflavones or estradiol replacement. Serum cytokine analysis revealed significant (p<0.05) diet-dependent increases in inflammatory cytokines (keratinocyte-derived chemokine). The isoflavone-free diet in OVX rats resulted in the regulation of the following cytokines and chemokines: interleukin-10, interleukin-18, serum regulated on activation, normal T cell expressed and secreted, and monocyte chemoattractant protein-1 (p<0.05). Overall, these results reveal that estradiol treatment can have differential effects on energy metabolism and body weight regulation depending on the presence of isoflavones in rodent chow.


Subject(s)
Body Weight/drug effects , Diet , Estradiol/pharmacology , Hormone Replacement Therapy , Isoflavones/pharmacology , Ovariectomy , Abdominal Fat/pathology , Adipokines/blood , Animals , Cytokines/blood , Drinking/drug effects , Feeding Behavior/drug effects , Female , Organ Size , Rats, Sprague-Dawley , Uterus/drug effects , Uterus/pathology
4.
J Radiat Res ; 58(5): 636-646, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28340212

ABSTRACT

Accidental high-dose radiation exposures can lead to multi-organ injuries, including radiation dermatitis. The types of cellular damage leading to radiation dermatitis are not completely understood. To identify the cellular mechanisms that underlie radiation-induced skin injury in vivo, we evaluated the time-course of cellular effects of radiation (14, 16 or 17 Gy X-rays; 0.5 Gy/min) in the skin of C57BL/6 mice. Irradiation of 14 Gy induced mild inflammation, observed histologically, but no visible hair loss or erythema. However, 16 or 17 Gy radiation induced dry desquamation, erythema and mild ulceration, detectable within 14 days post-irradiation. Histological evaluation revealed inflammation with mast cell infiltration within 14 days. Fibrosis occurred 80 days following 17 Gy irradiation, with collagen deposition, admixed with neutrophilic dermatitis, and necrotic debris. We found that in cultures of normal human keratinocytes, exposure to 17.9 Gy irradiation caused the upregulation of p21/waf1, a marker of senescence. Using western blot analysis of 17.9 Gy-irradiated mice skin samples, we also detected a marker of accelerated senescence (p21/waf1) 7 days post-irradiation, and a marker of cellular apoptosis (activated caspase-3) at 30 days, both preceding histological evidence of inflammatory infiltrates. Immunohistochemistry revealed reduced epithelial stem cells from hair follicles 14-30 days post-irradiation. Furthermore, p21/waf1 expression was increased in the region of the hair follicle stem cells at 14 days post 17 Gy irradiation. These data indicate that radiation induces accelerated cellular senescence in the region of the stem cell population of the skin.


Subject(s)
Organ Specificity/radiation effects , Radiation Injuries/pathology , Skin Aging/radiation effects , Adult Stem Cells/radiation effects , Aging , Animals , Apoptosis/radiation effects , Cellular Senescence/radiation effects , Disease Models, Animal , Dose-Response Relationship, Radiation , Female , Fibrosis , Hair Follicle/pathology , Hair Follicle/radiation effects , Keratinocytes/pathology , Keratinocytes/radiation effects , Mice, Inbred C57BL , Skin/pathology , Skin/radiation effects , Ulcer/pathology
5.
Neurosci Lett ; 640: 53-59, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28077306

ABSTRACT

Phytoestrogens are plant derived, non-steroidal compounds naturally found in rodent chows that potentially have endocrine-disrupting effects. Isoflavones, the most common phytoestrogens, have a similar structure and molecular weight to 17ß-estradiol (E2) and have the ability to bind and activate both isoforms of the estrogen receptor (ER). Most isoflavones have a higher affinity for ERß, which is involved in sexually dimorphic behavioral regulation. The goal of this study was to examine the interaction of isoflavones and E2 presence in the OVX rat on anxiety- and depressive- like behavior and the related BDNF pathophysiology. E2 administration resulted in anxiogenic behaviors when isoflavones were present in the diet (p<0.05), but anxiolytic behaviors when isoflavones were not present (p<0.05). E2 resulted in antidepressive-like behaviors in animals fed an isoflavone-rich diet (p<0.05), with no effect when isoflavones were removed. Increased hippocampal BDNF expression was observed in animals fed an isoflavone-rich diet after E2 administration (p<0.05). BDNF expression in the amygdala and hypothalamus was increased after E2 treatment in animals fed an isoflavone-rich diet. Overall, these results demonstrate that the presence of dietary isoflavones can differentially regulate the effect of E2 replacement on behavior and BDNF expression.


Subject(s)
Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Brain/drug effects , Estradiol/pharmacology , Food-Drug Interactions , Isoflavones/administration & dosage , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Anxiety/psychology , Brain/metabolism , Depression/psychology , Diet , Estradiol/adverse effects , Female , Maze Learning/drug effects , Motor Activity/drug effects , Ovariectomy , Rats, Sprague-Dawley
6.
PLoS One ; 10(3): e0122258, 2015.
Article in English | MEDLINE | ID: mdl-25815474

ABSTRACT

We reported that microRNA-30c (miR-30c) plays a key role in radiation-induced human cell damage through an apoptotic pathway. Herein we further evaluated radiation-induced miR-30 expression and mechanisms of delta-tocotrienol (DT3), a radiation countermeasure candidate, for regulating miR-30 in a mouse model and human hematopoietic CD34+ cells. CD2F1 mice were exposed to 0 (control) or 7-12.5 Gy total-body gamma-radiation, and CD34+ cells were irradiated with 0, 2 or 4 Gy of radiation. Single doses of DT3 (75 mg/kg, subcutaneous injection for mice or 2 µM for CD34+ cell culture) were administrated 24 h before irradiation and animal survival was monitored for 30 days. Mouse bone marrow (BM), jejunum, kidney, liver and serum as well as CD34+ cells were collected at 1, 4, 8, 24, 48 or 72 h after irradiation to determine apoptotic markers, pro-inflammatory cytokines interleukin (IL)-1ß and IL-6, miR-30, and stress response protein expression. Our results showed that radiation-induced IL-1ß release and cell damage are pathological states that lead to an early expression and secretion of miR-30b and miR-30c in mouse tissues and serum and in human CD34+ cells. DT3 suppressed IL-1ß and miR-30 expression, protected against radiation-induced apoptosis in mouse and human cells, and increased survival of irradiated mice. Furthermore, an anti-IL-1ß antibody downregulated radiation-induced NFκBp65 phosphorylation, inhibited miR-30 expression and protected CD34+ cells from radiation exposure. Knockdown of NFκBp65 by small interfering RNA (siRNA) significantly suppressed radiation-induced miR-30 expression in CD34+ cells. Our data suggest that DT3 protects human and mouse cells from radiation damage may through suppression of IL-1ß-induced NFκB/miR-30 signaling.


Subject(s)
Lymphocytes/drug effects , MicroRNAs/genetics , Radiation Injuries/drug therapy , Radiation-Protective Agents/therapeutic use , Vitamin E/analogs & derivatives , Vitamins/therapeutic use , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Apoptosis/drug effects , Cytokines/genetics , Cytokines/metabolism , Gamma Rays , Humans , Lymphocytes/metabolism , Lymphocytes/radiation effects , Mice , MicroRNAs/radiation effects , Radiation Injuries/metabolism , Radiation Injuries/prevention & control , Radiation-Protective Agents/pharmacology , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Vitamin E/pharmacology , Vitamin E/therapeutic use , Vitamins/pharmacology
7.
Proteomes ; 3(3): 249-265, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-28248270

ABSTRACT

Damage to normal lung tissue is a limiting factor when ionizing radiation is used in clinical applications. In addition, radiation pneumonitis and fibrosis are a major cause of mortality following accidental radiation exposure in humans. Although clinical symptoms may not develop for months after radiation exposure, immediate events induced by radiation are believed to generate molecular and cellular cascades that proceed during a clinical latent period. Oxidative damage to DNA is considered a primary cause of radiation injury to cells. DNA can be repaired by highly efficient mechanisms while repair of oxidized proteins is limited. Oxidized proteins are often destined for degradation. We examined protein oxidation following 17 Gy (0.6 Gy/min) thoracic X-irradiation in C57BL/6J mice. Seventeen Gy thoracic irradiation resulted in 100% mortality of mice within 127-189 days postirradiation. Necropsy findings indicated that pneumonitis and pulmonary fibrosis were the leading cause of mortality. We investigated the oxidation of lung proteins at 24 h postirradiation following 17 Gy thoracic irradiation using 2-D gel electrophoresis and OxyBlot for the detection of protein carbonylation. Seven carbonylated proteins were identified using mass spectrometry: serum albumin, selenium binding protein-1, alpha antitrypsin, cytoplasmic actin-1, carbonic anhydrase-2, peroxiredoxin-6, and apolipoprotein A1. The carbonylation status of carbonic anhydrase-2, selenium binding protein, and peroxiredoxin-6 was higher in control lung tissue. Apolipoprotein A1 and serum albumin carbonylation were increased following X-irradiation, as confirmed by OxyBlot immunoprecipitation and Western blotting. Our findings indicate that the profile of specific protein oxidation in the lung is altered following radiation exposure.

8.
Radiat Res ; 180(6): 649-57, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24294967

ABSTRACT

We recently demonstrated that natural delta-tocotrienol (DT3) significantly enhanced survival in total-body irradiated (TBI) mice, and protected mouse bone marrow cells from radiation-induced damage through Erk activation-associated mTOR survival pathways. Here, we further evaluated the effects and mechanisms of DT3 on survival of radiation-induced mouse acute gastrointestinal syndrome. DT3 (75-100 mg/kg) or vehicle was administered as a single subcutaneous injection to CD2F1 mice 24 h before 10-12 Gy (60)Co total-body irradiation at a dose rate of 0.6 Gy/min and survival was monitored. In a separate group of mice, jejunum sections were stained with hematoxylin and eosin and the surviving crypts in irradiated mice were counted. Apoptosis in intestinal epithelial cells was measured by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) staining and bacterial translocation from gut to heart, spleen and liver in irradiated mice were evaluated. DT3 (75 mg/kg) significantly enhanced survival in mice that received 10, 10.5, 11 or 12 Gy TBI. Administration of DT3 protected intestinal tissue, decreased apoptotic cells in jejunum and inhibited gut bacterial translocation in irradiated mice. Furthermore, DT3 significantly inhibited radiation-induced production of pro-inflammatory factors interleukin-1ß and -6 and suppressed expression of protein tyrosine kinase 6 (PTK6), a stress-induced kinase that promotes apoptosis in mouse intestinal cells. Our data demonstrate that administration of DT3 protected mice from radiation-induced gastrointestinal system damage.


Subject(s)
Gastrointestinal Tract/injuries , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/pharmacology , Vitamin E/analogs & derivatives , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bacterial Translocation/drug effects , Bacterial Translocation/radiation effects , Carrier Proteins/metabolism , Cell Survival/drug effects , Cell Survival/radiation effects , Cobalt Radioisotopes/adverse effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Epithelial Cells/drug effects , Epithelial Cells/radiation effects , Gamma Rays/adverse effects , Gastrointestinal Tract/cytology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/radiation effects , Jejunum/cytology , Jejunum/drug effects , Jejunum/radiation effects , Male , Mice , Microfilament Proteins , Photons/adverse effects , Protein-Tyrosine Kinases/metabolism , Survival Analysis , Vitamin E/pharmacology
9.
Radiat Res ; 180(3): 316-25, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23952576

ABSTRACT

Previous studies demonstrated that genistein protects mice from radiation-induced bone marrow failure. To overcome genistein's extremely low water solubility, a nanoparticle suspension of genistein has been formulated for more rapid dissolution. In the current study, we evaluated the radioprotective effects of a nanoparticle formulation of genistein on survival and hematopoietic recovery in mice exposed to total-body gamma irradiation. A single intramuscular injection of a saline-based genistein nanosuspension (150 mg/kg) administered to CD2F1 mice 24 h before 9.25 Gy (60)Co radiation exposure resulted in a 30-day survival rate of 95% compared to 25% in vehicle-treated animals. In mice irradiated at 7 Gy, the genistein nanosuspension increased mouse bone marrow cellularity from approximately 2.9% (vehicle treated) to 28.3% on day 7 postirradiation. Flow cytometry analysis demonstrated decreased radiation-induced hematopoietic stem and progenitor cell (HSPC, Lineage(-)/cKit(+)) death from 77.0% (vehicle) to 43.9% (genistein nanosuspension) with a significant recovery of clonogenicity 7 days after irradiation. The genistein nanosuspension also attenuated the radiation-induced elevation of proinflammatory factors interleukin 1 beta (IL-1ß), IL-6 and cyclooxygenase-2 (COX-2) in mouse bone marrow and spleen, which may contribute to protecting HSPCs.


Subject(s)
Bone Marrow/radiation effects , Genistein/administration & dosage , Nanoparticles , Spleen/radiation effects , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Gamma Rays , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , Spleen/metabolism , Spleen/pathology , Whole-Body Irradiation
10.
Exp Hematol ; 39(3): 293-304, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21146580

ABSTRACT

OBJECTIVE: Our laboratory reported that the angiotensin converting enzyme inhibitor captopril improves erythroid recovery from total body irradiation (TBI) in mice when administered after irradiation. However, captopril administered before TBI attenuates erythroid recovery. Here we investigate captopril and radiation regulation of erythropoietin (EPO) and thrombopoietin (TPO), key effectors of erythroid progenitor proliferation and differentiation. MATERIALS AND METHODS: C57BL/6 mice, nonirradiated or exposed to 7.5 Gy TBI ((60)Co, 0.6 Gy/min) were untreated or administered captopril. Plasma EPO and TPO levels were measured by enzyme-linked immunosorbent assay. Gene expression of EPO was determined by quantitative reverse transcription polymerase chain reaction. The hypoxia-inducible factors (HIF)-1α and -2α were measured by immunoblotting. RESULTS: In nonirradiated mice, continuous captopril administration in the water transiently reduced reticulocytes and red blood cells after 7 and 10 days, respectively. EPO plasma levels and gene expression were reduced below detectable limits after 2 days of captopril treatment, but recovered within 7 days. HIF-1α and HIF-2α were activated preceding reticulocyte and red blood cell recovery. TBI, which ablates early and late-stage erythroid progenitors, activated both HIFs and increased EPO and TPO. Captopril treatment postirradiation suppressed radiation-induced HIF activation and EPO expression. In contrast, captopril administration for 7 days before TBI resulted in earlier EPO induction and activation. Captopril treatment lowered TPO levels in nonirradiated mice, but had minimal effects on radiation-induced TPO. CONCLUSIONS: In nonirradiated mice, captopril biphasically regulates EPO via HIF activation. TBI ablates erythroid progenitors, resulting in hypoxia, HIF activation, and increased EPO expression that are modulated by captopril treatment. These data suggest that short-term suppression of radiation-induced EPO immediately after TBI is favorable for erythroid recovery.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Captopril/pharmacology , Erythroid Precursor Cells/metabolism , Erythropoietin/biosynthesis , Gamma Rays/adverse effects , Gene Expression Regulation , Whole-Body Irradiation , Animals , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Mice , Thrombopoietin/biosynthesis , Time Factors
11.
J Am Assoc Lab Anim Sci ; 49(3): 323-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20587164

ABSTRACT

Concern regarding the potential for radiation exposure from accidents or nuclear and radiologic terrorism is increasing. The purpose of this study was to determine whether the addition of minimal supportive care consisting of hydration or nutritional gels could be used to reduce mortality in mice exposed to (60)Co gamma-radiation. Male CD2F1 mice received 0, 8.50, or 9.25 Gy (60)Co at a dose rate of 0.6 Gy/min. These groups were further divided into 3 treatment groups that-in addition to pelleted food and water-received no supportive care, hydration gel, or nutritional gel. Overall survival, mean survival time, consumption of pelleted food and gel, and body weight were recorded for 30 d. Radiation caused dose-dependent decreases in overall survival, consumption of pelleted food and supplemental gel, and body weight. However, at each radiation dose (0, 8.50, 9.25 Gy), the type of supportive care did not modify overall survival, mean survival time, or changes in body weight. These results demonstrate that hydration and nutritional gels were not effective methods of supportive care after high-dose total body irradiation in mice.


Subject(s)
Fluid Therapy , Nutritional Support , Radiation Injuries, Experimental/therapy , Animals , Body Weight , Cobalt Radioisotopes , Gamma Rays , Male , Mice , Mice, Inbred Strains , Radiation Injuries, Experimental/mortality , Whole-Body Irradiation
12.
Exp Hematol ; 38(4): 270-81, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20116413

ABSTRACT

OBJECTIVE: Angiotensin II (Ang II), a potent vasoconstrictor, affects the growth and development of hematopoietic cells. Mixed findings have been reported for the effects of angiotensin-converting enzyme (ACE) inhibitors on radiation-induced injury to the hematopoietic system. We investigated the consequences of different regimens of the ACE inhibitor captopril on radiation-induced hematopoietic injury. MATERIALS AND METHODS: C57BL/6 mice were either sham-irradiated or exposed to (60)Co total body irradiation (0.6 Gy/min). Captopril was provided in the water for different time periods relative to irradiation. RESULTS: In untreated mice, the survival rate from 7.5 Gy was 50% at 30 days postirradiation. Captopril treatment for 7 days prior to irradiation resulted in radiosensitization with 100% lethality and a rapid decline in mature blood cells. In contrast, captopril treatment beginning 1 hour postirradiation and continuing for 30 days resulted in 100% survival, with improved recovery of mature blood cells and multilineage hematopoietic progenitors. In nonirradiated control mice, captopril biphasically modulated Lin(-) marrow progenitor cell cycling. After 2 days, captopril suppressed G(0)-G(1) transition and a greater number of cells entered a quiescent state. However, after 7 days of captopril treatment Lin(-) progenitor cell cycling increased compared to untreated control mice. CONCLUSION: These findings suggest that ACE inhibition affects hematopoietic recovery following radiation by modulating the hematopoietic progenitor cell cycle. The timing of captopril treatment relative to radiation exposure differentially affects the viability and repopulation capacity of spared hematopoietic stem cells and, therefore, can result in either radiation protection or radiation sensitization.


Subject(s)
Captopril/pharmacology , Hematopoietic Stem Cells/drug effects , Radiation Protection/methods , Whole-Body Irradiation , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Captopril/administration & dosage , Female , Mice , Mice, Inbred C57BL , Models, Animal , Time Factors
13.
Int Immunopharmacol ; 9(12): 1401-10, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19716438

ABSTRACT

The development of an effective pharmacological countermeasure is needed to reduce the morbidity and mortality in military and civilian populations associated with possible exposure to ionizing radiation. We previously demonstrated that a single subcutaneous (sc) administration of genistein at a non-toxic dose provided protection against acute radiation injury and that the radioprotective effects were associated with multilineage, hematopoietic progenitor cell recovery. The purpose of this study was to determine whether hematopoietic recovery was preceded by cytokine induction. In mice treated with sc genistein 24 h before irradiation (7 Gy 60Co), we quantified serum cytokine levels by multiplex Luminex and also investigated a larger number of cytokines using cytokine arrays. Genistein administration stimulated serum granulocyte-colony stimulating factor (G-CSF) 4h and 24h after sham irradiation or gamma-irradiation. Interleukin-6 (IL-6) was significantly increased in genistein-treated animals 4h after irradiation. Because G-CSF and IL-6 are important hematopoietic factors, these results support our hypothesis that the previously observed radioprotective efficacy by genistein may be a result of early recovery of hematopoietic cells due to enhanced production of G-CSF and IL-6.


Subject(s)
Genistein/administration & dosage , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-6/metabolism , Radiation Injuries/immunology , Radiation Injuries/prevention & control , Radiation-Protective Agents/administration & dosage , Whole-Body Irradiation , Animals , Disease Models, Animal , Gene Expression Profiling , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/immunology , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Inbred Strains , Oligonucleotide Array Sequence Analysis , Radiation Injuries/blood
14.
Int J Radiat Biol ; 85(7): 539-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19557599

ABSTRACT

PURPOSE: The search for ideal protective agents for use in a variety of radiation scenarios has continued for more than six decades. This review evaluates agents and procedures that have the potential to protect against acute and late effects of ionising radiation when administered either before or after radiation exposure. CONCLUSION: Over the years, extensive experimental studies of radiation-protective agents have enhanced our knowledge of radiation physics, chemistry, and biology. However, translation of agents from animal testing to use in various scenarios, such as prophylactic adjuncts in radiotherapy or post-exposure treatments for potential victims of radiation accidents/incidents, has been slow. Nevertheless, a number of compounds are now available for use in a variety of radiation situations. These include agents approved by the U.S. Food and Drug Administration for use in reducing exposure to internal radionuclides (Prussian blue, calcium diethylenetriamene pentaacetate (DTPA) and zinc DTPA, potassium iodide) and amifostine for alleviating xerostomia associated with radiotherapy. Consensus groups have also recommended other therapies such as granulocyte colony-stimulating factor for radiation-induced neutropenia. The variety of prophylactic and therapeutic agents in the research pipeline includes those that are naturally-occurring with low toxicity, provide a long window of protection, protect normal tissue while sensitising tumours, or act via receptors and modulate biological processes such as induction of genes responsible for radioresistance. The search for agents that protect against acute and late effects of ionising radiation injury will undoubtedly continue into the future and influence other areas of radiation research.


Subject(s)
Radiation-Protective Agents/pharmacology , Animals , Antioxidants/pharmacology , Drug Discovery , Glucans/pharmacology , Humans , Plant Extracts/pharmacology , Radioactive Hazard Release
15.
Int J Radiat Biol ; 84(9): 713-26, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18821385

ABSTRACT

PURPOSE: In this study we addressed whether genistein-induced radioprotection in mice is associated with alterations of the cell cycle of hematopoietic stem and progenitor cells. MATERIALS AND METHODS: C57BL/6J female mice received a single subcutaneous injection of genistein (200 mg/kg) 24 h prior to a lethal dose (7.75 Gy, (60)Co) of total body irradiation. Proliferation-associated Ki-67 protein/7-aminoactinomycin-D (Ki67/7AAD) cell cycle staining was used to differentiate between G(0), G(1), and S/G(2)/M in bone marrow cell populations negative for expression of mature hematopoietic lineage marker cells but positive for expression of stem cell antigen-1 and tyrosine kinase receptor for stem cell factor (Lin(-)Sca-1(+)cKit(+), LSK(+)). Quantitative real-time polymerase chain reaction (qRT-PCR) microarrays were utilized to examine cell cycle specific genes. RESULTS: At 24 h following radiation exposure, a greater percentage of LSK(+) in genistein-treated mice accumulated in the G(0) phase of the cell cycle, whereas a large percentage of LSK(+) bone marrow cells from untreated and vehicle (PEG-400)-treated mice progressed into the G(1) and S/G(2)/M phases. Moreover, the absolute number of marrow total LSK(+), long-term LSK(+), and short-term LSK(+) increased 2.8, 12.1, and 4.2-fold, respectively, at 7 days post-irradiation in genistein-treated vs. untreated irradiated mice. Lin(-) cells from genistein-treated mice expressed fewer DNA damage responsive and cell cycle checkpoint genes than LSK(+) from untreated or vehicle-treated mice. CONCLUSION: Pretreatment with genistein provides in vivo protection from acute myelotoxicity through extended quiescence followed by reduced senescence of marrow repopulating LSK(+).


Subject(s)
Cell Cycle/drug effects , Genistein/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Radiation-Protective Agents/pharmacology , Animals , Biomarkers/metabolism , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/physiology , Bone Marrow/radiation effects , Cell Cycle/radiation effects , Cell Lineage/drug effects , Cell Lineage/radiation effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cellular Senescence/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , Genistein/administration & dosage , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Injections, Subcutaneous , Mice , Radiation Dosage , Radiation-Protective Agents/administration & dosage
16.
J Radiat Res ; 49(4): 361-72, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18434686

ABSTRACT

The effects of genistein on 30-day survival and delayed lung injury were examined in C57BL/6J female mice. A single subcutaneous injection of vehicle (PEG-400) or genistein (200 mg/kg) was administered 24 h before total body irradiation (7.75 Gy (60)Co, 0.6 Gy/min). Experimental groups were: No treatment + Sham (NC), Vehicle + Sham (VC), Genistein + Sham (GC), Radiation only (NR), Vehicle + Radiation (VR), Genistein + Radiation (GR). Thirty-day survivals after 7.75 Gy were: NR 23%, VR 53%, and GR 92%, indicating significant protection from acute radiation injury by genistein. Genistein also mitigated radiation-induced weight loss on days 13-28 postirradiation. First generation lung fibroblasts were analyzed for micronuclei 24 h postirradiation. Fibroblasts from the lungs of GR-treated mice had significantly reduced micronuclei compared with NR mice. Collagen deposition was examined by histochemical staining. At 90 days postirradiation one half of the untreated and vehicle irradiated mice had focal distributions of small collagen-rich plaques in the lungs, whereas all of the genistein-treated animals had morphologically normal lungs. Radiation reduced the expression of COX-2, transforming growth factor-beta receptor (TGFbetaR) I and II at 90 days after irradiation. Genistein prevented the reduction in TGFbetaRI. However, by 180 days postirradiation, these proteins normalized in all groups. These results demonstrate that genistein protects against acute radiation-induced mortality in female mice and that GR-treated mice have reduced lung damage compared to NR or VR. These data suggest that genistein is protective against a range of radiation injuries.


Subject(s)
Cytokines/analysis , Genistein/administration & dosage , Radiation Injuries/metabolism , Radiation Injuries/prevention & control , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/prevention & control , Whole-Body Irradiation , Animals , Biomarkers/metabolism , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Lung/drug effects , Lung/metabolism , Lung/radiation effects , Mice , Mice, Inbred C57BL , Radiation Dosage , Radiation-Protective Agents/administration & dosage , Survival Analysis , Survival Rate
17.
Int J Radiat Biol ; 83(3): 141-51, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17378522

ABSTRACT

PURPOSE: Genistein, a non-toxic isoflavone from soybeans, has immunomodulating and radioprotective properties. In this study we investigated the mechanism for genistein-induced radioprotection by evaluating the recovery of bone marrow cells and peripheral blood hematology in lethally irradiated mice. MATERIALS AND METHODS: CD2F1 male mice received a single subcutaneous injection of genistein (200 mg/kg) 24 h prior to a lethal, total body irradiation dose (8.75 Gy) of cobalt-60 gamma radiation. Survival and hematopoietic reconstitution were evaluated over nine weeks post-irradiation. Hematopoietic progenitor colony-forming cell assays were used to assess the reconstitution of bone marrow after radiation-induced myelosuppression. RESULTS: A total of 97% of genistein-treated mice survived after 30 days while 31% of vehicle-treated and 0% of untreated mice survived. The improvement in survival was related to accelerated neutrophil and platelet recovery, resulting from earlier and more pronounced multilineage, hematopoietic progenitor cell reconstitution in the femoral marrow compartment. Myeloid and erythroid progenitor cell numbers at day 15 post-irradiation were 6-fold to 20-fold higher in genistein-treated animals than in control animals. CONCLUSIONS: These results demonstrate that a single subcutaneous administration of genistein 24 h before irradiation provides significant radioprotection to the hematopoietic progenitor cell compartment.


Subject(s)
Cell Survival/radiation effects , Genistein/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Cell Survival/drug effects , Colony-Forming Units Assay , Gamma Rays , Genistein/administration & dosage , Hematopoietic Stem Cells/cytology , Injections, Subcutaneous , Kinetics , Male , Mice , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/pharmacology , Time Factors , Whole-Body Irradiation
18.
J Basic Microbiol ; 46(4): 329-35, 2006.
Article in English | MEDLINE | ID: mdl-16847837

ABSTRACT

Genistein, a radioprotective soy isoflavone and protein kinase inhibitor, blocks the invasion of pathogenic bacteria in mammalian epithelial cells. The purpose of this study was to evaluate the direct effect of genistein on the survival and growth of the probiotic Lactobacillus reuteri and selected opportunistic bacteria in vitro as a prelude to in vivo use for managing postirradiation sepsis. We evaluated the opportunistic bacterial enteropathogens Escherichia coli, Shigella sonnei, and Staphylococcus aureus as well as Klebsiella pneumoniae and the non-pathogenic organism, Bacillus anthracis (Sterne). The latter two bacteria are found in the environment and may be of concern in irradiated individuals. A standard in vitro test was employed to evaluate the direct effect of genistein on the bacteria. This test involved determining bacterial colony forming unit (CFU) counts at a single concentration of genistein. In the CFU assays, significant reductions in CFUs were found for S. aureus and B. anthracis when cultured in the presence of 100 muM genistein. However, L. reuteri, E. coli, S. sonnei, and K. pneumoniae were not altered by in vitro culturing in the presence of 100 muM genistein. These results demonstrate the in vitro antimicrobial activity of genistein. Furthermore, the use of genistein in combination with probiotics may augment the effectiveness of antimicrobial therapies currently used in the management of infections, including those induced by ionizing irradiation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Limosilactobacillus reuteri/drug effects , Bacillus anthracis/drug effects , Colony Count, Microbial , Limosilactobacillus reuteri/growth & development , Probiotics , Staphylococcus aureus/drug effects
19.
J Appl Toxicol ; 23(6): 379-85, 2003.
Article in English | MEDLINE | ID: mdl-14635262

ABSTRACT

The radioprotective and behavioral effects of an acute administration of the isoflavone genistein (4',5,7-trihydroxyflavone) were investigated in adult CD2F1 male mice. Mice were administered a single subcutaneous (s.c.) dose of genistein either 24 h or 1 h before a lethal dose of gamma radiation (9.5-Gy of cobalt-60 at 0.6 Gy min(-1)). Mice received saline, PEG-400 vehicle or genistein at 3.125, 6.25, 12.5, 25, 50, 100, 200, or 400 mg kg(-1) body weight. For mice treated 24 h before irradiation there was a significant increase in 30-day survival for animals receiving genistein doses of 25 to 400 mg kg(-1) (p<0.001). In contrast, the 30-day survival rates of mice treated with genistein 1 h before irradiation were not significantly different from those of the vehicle control group. Additionally, the acute toxicity of genistein was evaluated in non-irradiated male mice administered a single s.c. injection of saline, vehicle, or genistein at 100, 200 or 400 mg kg(-1). At these genistein doses there were no adverse effects, compared with controls, on locomotor activity, grip strength, motor coordination, body weight, testes weight, or histopathology. These results demonstrate that a single s.c. administration of the flavonoid genistein at non-toxic doses provides protection against acute radiation injury.


Subject(s)
Genistein/pharmacology , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/pharmacology , Animals , Body Weight/drug effects , Body Weight/radiation effects , Dose-Response Relationship, Drug , Genistein/administration & dosage , Genistein/toxicity , Injections, Subcutaneous , Male , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Motor Activity/radiation effects , Organ Size/drug effects , Organ Size/radiation effects , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/toxicity , Survival Rate , Testis/drug effects , Testis/radiation effects , Time Factors
20.
Toxicology ; 189(1-2): 1-20, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12821279

ABSTRACT

The potential of antioxidants to reduce the cellular damage induced by ionizing radiation has been studied in animal models for more than 50 years. The application of antioxidant radioprotectors to various human exposure situations has not been extensive although it is generally accepted that endogenous antioxidants, such as cellular non-protein thiols and antioxidant enzymes, provide some degree of protection. This review focuses on the radioprotective efficacy of naturally occurring antioxidants, specifically antioxidant nutrients and phytochemicals, and how they might influence various endpoints of radiation damage. Results from animal experiments indicate that antioxidant nutrients, such as vitamin E and selenium compounds, are protective against lethality and other radiation effects but to a lesser degree than most synthetic protectors. Some antioxidant nutrients and phytochemicals have the advantage of low toxicity although they are generally protective when administered at pharmacological doses. Naturally occurring antioxidants also may provide an extended window of protection against low-dose, low-dose-rate irradiation, including therapeutic potential when administered after irradiation. A number of phytochemicals, including caffeine, genistein, and melatonin, have multiple physiological effects, as well as antioxidant activity, which result in radioprotection in vivo. Many antioxidant nutrients and phytochemicals have antimutagenic properties, and their modulation of long-term radiation effects, such as cancer, needs further examination. In addition, further studies are required to determine the potential value of specific antioxidant nutrients and phytochemicals during radiotherapy for cancer.


Subject(s)
Antioxidants/pharmacology , Radiation Injuries/prevention & control , Radiation-Protective Agents/pharmacology , Animals , Ascorbic Acid/pharmacology , Humans , Mice , Radiation, Ionizing , Selenium/pharmacology , Vitamin E/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...