Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 300
Filter
1.
Exp Neurol ; 341: 113705, 2021 07.
Article in English | MEDLINE | ID: mdl-33753139

ABSTRACT

Anticancer therapeutics can provoke severe side effects that impair the patient's quality of life. A frequent dose-limiting side effect of platinum-based anticancer therapy is neurotoxicity. Its pathophysiology is poorly understood, and effective preventive or therapeutic measures are missing. Therefore, elucidation of the molecular mechanism of platinating drug-induced neurotoxicity and the development of preventive strategies is urgently needed. To this end, we aim to use C. elegans as a 3R-compliant in vivo model. The 3R principles were conceived for animal welfare in science concerning animal experiments, which should be replaced, reduced or refined. We can analytically demonstrate dose-dependent uptake of cisplatin (CisPt) in C. elegans, as well as genotoxic and cytotoxic effects based on DNA adduct formation (i.e., 1,2-GpG intrastrand crosslinks), induction of apoptosis, and developmental toxicity. Measuring the impairment of pharyngeal pumping as a marker of neurotoxicity, we found that especially CisPt reduces the pumping frequency at concentrations where basal and touch-provoked movement were not yet affected. CisPt causes glutathione (GSH) depletion and RNAi-mediated knockdown of the glutamate-cysteine ligase GCS-1 aggravates the CisPt-induced inhibition of pharyngeal pumping. Moreover, N-acetylcysteine (NAC) mitigated CisPt-triggered toxicity, indicating that GSH depletion contributes to the CisPt-induced pharyngeal damage. In addition to NAC, amifostine (WR1065) also protected the pharynx of C. elegans from the toxic effects of CisPt. Measuring pharyngeal activity by the electrophysiological recording of neurotransmission in the pharynx, we confirmed that CisPt is neurotoxic in C. elegans and that NAC is neuroprotective in the nematode. The data support the hypothesis that monitoring the pharyngeal activity of C. elegans is a useful surrogate marker of CisPt-induced neurotoxicity. In addition, a low GSH pool reduces the resistance of neurons to CisPt treatment, and both NAC and WR1065 are capable of attenuating platinum-induced neurotoxicity during post-incubation in C. elegans. Overall, we propose C. elegans as a 3R-compliant in vivo model to study the molecular mechanisms of platinum-induced neurotoxicity and to explore novel neuroprotective therapeutic strategies to alleviate respective side effects of platinum-based cancer therapy.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Disease Models, Animal , Neurotoxicity Syndromes/prevention & control , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Chemoprevention/methods , Dose-Response Relationship, Drug , Mercaptoethylamines/pharmacology , Mercaptoethylamines/therapeutic use , Neurotoxicity Syndromes/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Platinum Compounds/toxicity
2.
Cells ; 11(1)2021 12 29.
Article in English | MEDLINE | ID: mdl-35011655

ABSTRACT

The search for effective neuroprotective agents for the treatment of neurotrauma has always been of great interest to researchers around the world. Extracellular heat shock protein 70 (eHsp70) is considered a promising agent to study, as it has been demonstrated to exert a significant neuroprotective activity against various neurodegenerative diseases. We showed that eHsp70 can penetrate neurons and glial cells when added to the incubation medium, and can accumulate in the nuclei of neurons and satellite glial cells after axotomy. eHsp70 reduces apoptosis and necrosis of the glial cells, but not the neurons. At the same time, co-localization of eHsp70 with p53 protein, one of the key regulators of apoptosis, was noted. eHsp70 reduces the level of the p53 protein apoptosis promoter both in glial cells and in the nuclei and cytoplasm of neurons, which indicates its neuroprotective effect. The ability of eHsp70 to reverse the proapoptotic effect of the p53 activator WR1065 may indicate its ability to regulate p53 activity or its proteosome-dependent degradation.


Subject(s)
Apoptosis , Astacoidea/metabolism , Axotomy , HSP70 Heat-Shock Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Down-Regulation , E2F1 Transcription Factor/metabolism , HSP70 Heat-Shock Proteins/isolation & purification , Humans , Mechanoreceptors/metabolism , Mercaptoethylamines/pharmacology , Necrosis , Neuroglia/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
3.
Cell Mol Neurobiol ; 39(5): 619-636, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30874981

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a side effect of platinum-based chemotherapy and decreases the quality of life of cancer patients. We compared neuroprotective properties of several agents using an in vitro model of terminally differentiated human cells NT2-N derived from cell line NT2/D1. Sodium azide and an active metabolite of amifostine (WR1065) increase cell viability in simultaneous treatment with cisplatin. In addition, WR1065 protects the non-dividing neurons by decreasing cisplatin caused oxidative stress and apoptosis. Accumulation of Pt in cisplatin-treated cells was heterogeneous, but the frequency and concentration of Pt in cells were lowered in the presence of WR1065 as shown by X-ray fluorescence microscopy (XFM). Transition metals accumulation accompanied Pt increase in cells; this effect was equally diminished in the presence of WR1065. To analyze possible chemical modulation of Pt-DNA bonds, we examined the platinum LIII near edge spectrum by X-ray absorption spectroscopy. The spectrum found in cisplatin-DNA samples is altered differently by the addition of either WR1065 or sodium azide. Importantly, a similar change in Pt edge spectra was noted in cells treated with cisplatin and WR1065. Therefore, amifostine should be reconsidered as a candidate for treatments that reduce or prevent CIPN.


Subject(s)
Antioxidants/pharmacology , Cisplatin/adverse effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitory Concentration 50 , Mercaptoethylamines/pharmacology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sodium Azide/pharmacology
4.
Sci Rep ; 9(1): 1949, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760738

ABSTRACT

Unresectable pancreatic cancer is almost universally lethal because chemotherapy and radiation cannot completely stop the growth of the cancer. The major problem with using radiation to approximate surgery in unresectable disease is that the radiation dose required to ablate pancreatic cancer exceeds the tolerance of the nearby duodenum. WR-2721, also known as amifostine, is a well-known radioprotector, but has significant clinical toxicities when given systemically. WR-2721 is a prodrug and is converted to its active metabolite, WR-1065, by alkaline phosphatases in normal tissues. The small intestine is highly enriched in these activating enzymes, and thus we reasoned that oral administration of WR-2721 just before radiation would result in localized production of the radioprotective WR-1065 in the small intestine, providing protective benefits without the significant systemic side effects. Here, we show that oral WR-2721 is as effective as intraperitoneal WR-2721 in promoting survival of intestinal crypt clonogens after morbid irradiation. Furthermore, oral WR-2721 confers full radioprotection and survival after lethal upper abdominal irradiation of 12.5 Gy × 5 fractions (total of 62.5 Gy, EQD2 = 140.6 Gy). This radioprotection enables ablative radiation therapy in a mouse model of pancreatic cancer and nearly triples the median survival compared to controls. We find that the efficacy of oral WR-2721 stems from its selective accumulation in the intestine, but not in tumors or other normal tissues, as determined by in vivo mass spectrometry analysis. Thus, we demonstrate that oral WR-2721 is a well-tolerated, and quantitatively selective, radioprotector of the intestinal tract that is capable of enabling clinically relevant ablative doses of radiation to the upper abdomen without unacceptable gastrointestinal toxicity.


Subject(s)
Amifostine/pharmacology , Mercaptoethylamines/pharmacology , Radiation-Protective Agents/therapeutic use , Administration, Oral , Amifostine/metabolism , Animals , Female , Intestine, Small/drug effects , Male , Mercaptoethylamines/metabolism , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/drug therapy , Radiation Dosage , Radiation Protection/methods , Pancreatic Neoplasms
5.
Med Chem ; 14(6): 585-594, 2018.
Article in English | MEDLINE | ID: mdl-29663895

ABSTRACT

BACKGROUND: Methicillin resistant Staphylococcus aureus (MRSA) usually invalidate powerful antibiotics in the clinic. Pleuromutilin derivatives have been reported to possess antibacterial activity against MRSA. OBJECTIVE: The antibacterial activities against MRSA of a series of thirteen synthetic pleuromutilin derivatives were investigated through in vitro models. METHODS: A series of novel thioehter pleuromutilin derivatives incorporating various aromatic substituents into the C14 side chain have been reported. The in vitro antibacterial activities of these derivatives against MRSA and Escherichia coli were tested by the broth dilution method. RESULTS: Twelve pleuromutilin derivatives were designed, synthesized and evaluated for in vitro antibacterial activities against four Staphylococcus aureus strains. From structure-activity relationship studies, compound 11c was identified as promising compounds with the most potent in vitro antibacterial activity among the series (MIC = 0.0625-0.125 µg/ml) against Staphylococcus aureus strains. The binding of compound 11c to the 50s ribosome was investigated by molecular modeling. CONCLUSION: It was found that there is a reasonable correlation between the binding free energy and the antibacterial activity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Mercaptoethylamines/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Diterpenes/chemical synthesis , Diterpenes/chemistry , Diterpenes/pharmacology , Escherichia coli/drug effects , Mercaptoethylamines/chemical synthesis , Mercaptoethylamines/chemistry , Microbial Sensitivity Tests , Molecular Docking Simulation , Polycyclic Compounds , Ribosome Subunits, Large, Bacterial/chemistry , Staphylococcus aureus/drug effects , Structure-Activity Relationship , Pleuromutilins
6.
Redox Biol ; 6: 73-79, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26188467

ABSTRACT

Exposure to radiation can damage endothelial cells in the irradiated area via the production of reactive oxygen species. We synthesized phosphine-borane complexes that reduce disulfide bonds and had previously been shown to interfere with redox-mediated signaling of cell death. We hypothesized that this class of drugs could interfere with the downstream effects of oxidative stress after irradiation and rescue endothelial cells from radiation damage. Cultured bovine aortic endothelial cells were plated for clonogenic assay prior to exposure to varying doses of irradiation from a (137)Cs irradiator and treated with various concentrations of bis(3-propionic acid methyl ester)phenylphosphine borane complex (PB1) at different time points. The clone-forming ability of the irradiated cells was assessed seven days after irradiation. We compared the radioprotective effects of PB1 with the aminothiol radioprotectant WR1065 and known superoxide scavengers. PB1 significantly protected bovine aortic endothelial cells from radiation damage, particularly when treated both before and after radiation. The radioprotection with 1 µM PB1 corresponded to a dose-reduction factor of 1.24. Radioprotection by PB1 was comparable to the aminothiol WR1065, but was significantly less toxic and required much lower concentrations of drug (1 µM vs. 4 mM, respectively). Superoxide scavengers were not radioprotective in this paradigm, indicating the mechanisms for both loss of clonogenicity and PB1 radioprotection are independent of superoxide signaling. These data demonstrate that PB1 is an effective redox-active radioprotectant for endothelial cells in vitro, and is radioprotective at a concentration approximately 4 orders of magnitude lower than the aminothiol WR1065 with less toxicity.


Subject(s)
Boranes/pharmacology , Endothelial Cells/drug effects , Gamma Rays/adverse effects , Phosphines/pharmacology , Radiation-Protective Agents/pharmacology , Superoxides/antagonists & inhibitors , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Aorta/radiation effects , Cattle , Cells, Cultured , Clone Cells , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Humans , Mercaptoethylamines/pharmacology , Metalloporphyrins/pharmacology , Oxidation-Reduction , Polyethylene Glycols/pharmacology , Signal Transduction , Superoxide Dismutase/pharmacology , Superoxides/metabolism
7.
Redox Biol ; 2: 590-8, 2014.
Article in English | MEDLINE | ID: mdl-24688895

ABSTRACT

Many carbonyl species from either lipid peroxidation or glycoxidation are extremely reactive and can disrupt the function of proteins and enzymes. 4-hydroxynonenal and methylglyoxal are the most abundant and toxic lipid-derived reactive carbonyl species. The presence of these toxics leads to carbonyl stress and cause a significant amount of macromolecular damages in several diseases. Much evidence indicates trapping of reactive carbonyl intermediates may be a useful strategy for inhibiting or decreasing carbonyl stress-associated pathologies. There is no rapid and convenient analytical method available for the assessment of direct carbonyl scavenging capacity, and a very limited number of carbonyl scavengers have been identified to date, their therapeutic potential being highlighted only recently. In this context, we have developed a new and rapid sensitive fluorimetric method for the assessment of reactive carbonyl scavengers without involvement glycoxidation systems. Efficacy of various thiol- and non-thiol-carbonyl scavenger pharmacophores was tested both using this screening assay adapted to 96-well microplates and in cultured cells. The scavenging effects on the formation of Advanced Glycation End-product of Bovine Serum Albumin formed with methylglyoxal, 4-hydroxynonenal and glucose-glycated as molecular models were also examined. Low molecular mass thiols with an α-amino-ß-mercaptoethane structure showed the highest degree of inhibitory activity toward both α,ß-unsaturated aldehydes and dicarbonyls. Cysteine and cysteamine have the best scavenging ability toward methylglyoxal. WR-1065 which is currently approved for clinical use as a protective agent against radiation and renal toxicity was identified as the best inhibitor of 4-hydroxynonenal.


Subject(s)
Aldehydes/pharmacology , Cysteamine/pharmacology , Cysteine/pharmacology , High-Throughput Screening Assays/methods , Pyruvaldehyde/pharmacology , Aldehydes/antagonists & inhibitors , Animals , Caco-2 Cells , Cell Line, Tumor , Glycation End Products, Advanced/metabolism , Humans , Mercaptoethylamines/pharmacology , Mice , Pyruvaldehyde/antagonists & inhibitors , Sensitivity and Specificity , Serum Albumin, Bovine/metabolism
8.
Radiat Res ; 179(2): 115-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23237540

ABSTRACT

Very low doses of ionizing radiation, 5 to 100 mGy, can induce adaptive responses characterized by elevation in cell survival and reduction in micronuclei formation. Utilizing these end points, RKO human colon carcinoma and transformed mouse embryo fibroblasts (MEF), wild-type or knockout cells missing TNF receptors 1 and 2 (TNFR1(-)R2(-)), and C57BL/6 and TNFR1(-)R2(-) knockout mice, we demonstrate that intact TNF signaling is required for induction of elevated manganese superoxide dismutase (SOD2) activity (P < 0.001) and the subsequent expression of these SOD2-mediated adaptive responses when cells are challenged at a later time with 2 Gy. In contrast, amifostine's free thiol form WR1065 can directly activate NF-κB giving rise to elevated SOD2 activity 24 h later and induce an adaptive response in both MEF wild-type and TNF signaling defective TNFR1(-)R2(-) cells. Transfection of cells with SOD2 siRNA completely abolishes both the elevation in SOD2 activity and expression of the adaptive responses. These results were confirmed in vivo using a micronucleus assay in splenocytes derived from C57BL/6 and TNFR1(-)R2(-) knockout mice that were exposed to 100 mGy or 400 mg/kg amifostine 24 h prior to exposure to a 2 Gy whole-body dose. A dose of 100 mGy also conferred enhanced protection to C57BL/6 mice exposed 24 h later to 100 mg/kg of N-Ethyl-N-nitrosourea (ENU). While very low radiation doses require an intact TNF signaling process to induce a SOD2-mediated adaptive response, amifostine can induce a similar adaptive response in both TNF receptor competent and knockout cells, respectively.


Subject(s)
Adaptation, Physiological/drug effects , Adaptation, Physiological/radiation effects , Superoxide Dismutase/metabolism , Alkylating Agents/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Ethylnitrosourea/pharmacology , Female , Genomic Instability/drug effects , Genomic Instability/radiation effects , Humans , Mercaptoethylamines/pharmacology , Mice , Mice, Inbred C57BL , Pregnancy , Signal Transduction/drug effects , Signal Transduction/radiation effects
9.
Free Radic Biol Med ; 51(10): 1918-25, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21945096

ABSTRACT

Manganese superoxide dismutase (SOD2)-mediated adaptive processes that protect against radiation-induced micronucleus formation can be induced in cells after a 2-Gy exposure by previously exposing them to either low-dose ionizing radiation (10cGy) or WR1065 (40µM), the active thiol form of amifostine. Although both adaptive processes culminate in elevated levels of SOD2 enzymatic activity, the underlying pathways differ in complexity, with the tumor necrosis factor α (TNFα) signaling pathway implicated in the low-dose radiation-induced response, but not in the thiol-induced pathway. The goal of this study was the characterization of the effects of TNFα receptors 1 and 2 (TNFR1, TNFR2) on the adaptive responses induced by low-dose irradiation or thiol exposure using micronucleus formation as an endpoint. BFS-1 wild-type cells with functional TNFR1 and 2 were exposed 24h before a 2-Gy dose of ionizing radiation to either 10cGy or a 40µM dose of WR1065. BFS2C-SH02 cells, defective in TNFR1, and BFS2C-SH22 cells, defective in both TNFR1 and TNFR2 and generated from BFS2C-SH02 cells by transfection with a murine TNFR2-targeting vector and confirmed to be TNFR2 defective by quantitative PCR, were also exposed under similar conditions for comparison. A 10-cGy dose of radiation induced a significant elevation in SOD2 activity in BFS-1 (P<0.001) and BFS2C-SH02 (P=0.005) but not BFS2C-SH22 cells (P=0.433), compared to their respective untreated controls. In contrast, WR1065 significantly induced elevations in SOD2 activity in all three cell lines (P=0.001, P=0.007, P=0.020, respectively). A significant reduction in the frequency of radiation-induced micronuclei was observed in each cell line when exposure to a 2-Gy challenge dose of radiation occurred during the period of maximal elevation in SOD2 activity. However, this adaptive effect was completely inhibited if the cells were transfected 24h before low-dose radiation or thiol exposure with SOD2 siRNA. Under the conditions tested, TNFR1 and 2 inhibition negatively affected the low-dose radiation-induced but not the thiol-induced adaptive responses observed to be mediated by elevations in SOD2 activity.


Subject(s)
Mercaptoethylamines/pharmacology , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Amifostine/analogs & derivatives , Amifostine/chemistry , Animals , Cell Line, Tumor , Enzyme Activation/genetics , Enzyme Activation/radiation effects , Mercaptoethylamines/chemistry , Mice , Micronuclei, Chromosome-Defective/drug effects , Micronuclei, Chromosome-Defective/radiation effects , Micronucleus Tests , RNA, Small Interfering/genetics , Radiation, Ionizing , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Superoxide Dismutase/genetics
10.
Radiat Res ; 175(1): 57-65, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21175348

ABSTRACT

RKO36 cells exposed to either WR1065 or 10 cGy X rays show elevated SOD2 gene expression and SOD2 enzymatic activity. Cells challenged at this time with 2 Gy exhibit enhanced radiation resistance. This phenomenon has been identified as a delayed radioprotective effect or an adaptive response when induced by thiols or low-dose radiation, respectively. In this study we investigated the relative effectiveness of both WR1065 and low-dose radiation in reducing the incidence of radiation-induced micronucleus formation in binucleated RKO36 human colon carcinoma cells. The role of SOD2 in this process was assessed by measuring changes in enzymatic activity as a function of the inducing agent used, the level of protection afforded, and the inhibitory effects of short interfering RNA (SOD2 siRNA). Both WR1065 and 10 cGy X rays effectively induced a greater than threefold elevation in SOD2 activity 24 h after exposure. Cells irradiated at this time with 2 Gy exhibited a significant resistance to micronucleus formation (P < 0.05; Student's two-tailed t test). This protective effect was significantly inhibited in cells transfected with SOD2 siRNA. SOD2 played an important role in the adaptive/delayed radioprotective response by inhibiting the initiation of a superoxide anion-induced ROS cascade leading to enhanced mitochondrial and nuclear damages.


Subject(s)
Colorectal Neoplasms/genetics , Mercaptoethylamines/pharmacology , Micronuclei, Chromosome-Defective , Radiation-Protective Agents/pharmacology , Superoxide Dismutase/physiology , Adaptation, Physiological , Cell Line, Tumor , Humans , Reactive Oxygen Species/metabolism , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/radiation effects , Superoxides/metabolism
11.
Radiat Res ; 174(6): 719-26, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21128795

ABSTRACT

The DEL assay in yeast detects DNA deletions that are inducible by many carcinogens. Here we use the colorimetric agent MTS to adapt the yeast DEL assay for microwell plate measurement of ionizing radiation-induced cell killing and DNA deletions. Using the microwell-based DEL assay, cell killing and genotoxic DNA deletions both increased with radiation dose between 0 and 2000 Gy. We used the microwell-based DEL assay to assess the effectiveness of varying concentrations of five different radioprotectors, N-acetyl-l-cysteine, l-ascorbic acid, DMSO, Tempol and Amifostine, and one radiosensitizer, 5-bromo-2-deoxyuridine. The microwell format of the DEL assay was able to successfully detect protection against and sensitization to both radiation-induced cytotoxicity and genotoxicity. Such radioprotection and sensitization detected by the microwell-based DEL assay was validated and compared with similar measurements made using the traditional agar-based assay format. The yeast DEL assay in microwell format is an effective tool for rapidly detecting chemical protectors and sensitizers to ionizing radiation and is automatable for chemical high-throughput screening purposes.


Subject(s)
Gene Deletion , Radiation-Protective Agents/pharmacology , Saccharomyces cerevisiae/radiation effects , Cell Survival/radiation effects , Mercaptoethylamines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/genetics
12.
J Altern Complement Med ; 16(5): 561-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20491513

ABSTRACT

BACKGROUND: Ionizing radiation (IR) initiates intracellular oxidative stress through enhanced formation of reactive oxygen species (ROS) that attack DNA leading to cell death. Because of the diversity of IR applied in medicine, agriculture, industry, and the growing threats of global terrorism, the acquisition of radioprotectors is an urgent need for the nation. However, the applicability of radioprotectors currently under investigation is limited due to their inherent toxicity. OBJECTIVE: This study investigated the effect of a standardized North American ginseng extract (NAGE, total ginsenoside content: 11.7%) on DNA damage in human lymphocytes at 90 minutes postirradiation. DESIGN: With the application of NAGE (250-1000 microg mL(-1)) at 90 minutes postirradiation (1 and 2 Gy), DNA damage in lymphocytes obtained from 40 healthy individuals was evaluated by cytokinesis-block micronucleus assay. Similar experiments were also performed in lymphocytes treated with WR-1065 (1 mmol/L or 3 mmol/L). In addition, before and after irradiation, lymphocytes obtained from 10 individuals were measured for their total antioxidant capacity (TAC) and the reactive oxygen species (ROS). RESULTS: The significant effect of NAGE against (137)Cs-induced micronuclei (MN) in lymphocytes is concentration dependent. NAGE (750 microg mL(-1)) reduced MN yield by 50.7% after 1 Gy and 35.9% after 2 Gy exposures, respectively; these results were comparable to that of WR-1065. Furthermore, we also found that NAGE reduces MN yield and ROS but increases TAC in lymphocytes. CONCLUSIONS: Our results suggest that NAGE is a relatively nontoxic natural compound that holds radioprotective potential in human lymphocytes even when applied at 90 minutes postirradiation. One of the radioprotective mechanisms may be mediated through the scavenging of free radicals and enhancement of the intracellular TAC.


Subject(s)
Lymphocytes/drug effects , Oxidative Stress/drug effects , Panax/chemistry , Phytotherapy , Plant Extracts/therapeutic use , Radiation Injuries/prevention & control , Radiation-Protective Agents/therapeutic use , Adult , Antioxidants/pharmacology , Antioxidants/therapeutic use , DNA Damage , Female , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Humans , Lymphocytes/radiation effects , Male , Mercaptoethylamines/pharmacology , Micronuclei, Chromosome-Defective , Micronucleus Tests , Middle Aged , Plant Extracts/pharmacology , Radiation Injuries/genetics , Radiation, Ionizing , Radiation-Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Single-Blind Method
13.
Radiat Res ; 173(2): 175-83, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20095849

ABSTRACT

Repair of DNA damage through homologous recombination (HR) pathways plays a crucial role in maintaining genome stability. However, overstimulation of HR pathways in response to genotoxic stress may abnormally elevate recombination frequencies, leading to increased mutation rates and delayed genomic instability. Radiation-induced genomic instability has been detected after exposure to both low- and high-linear energy transfer (LET) radiations, but the mechanisms responsible for initiating or propagating genomic instability are not known. We have demonstrated that WR-1065, the active metabolite of amifostine, protects against radiation-induced cell killing and delayed genomic instability. We hypothesize that hyperstimulation of HR pathways plays a mechanistic role in radiation-induced genomic instability and that, in part, WR-1065 exerts it radioprotective effect through suppression of the HR pathway. Results of this study demonstrate that WR-1065 treatment selectively protected against radiation-induced cell killing in HR-proficient cell lines compared to an HR-deficient cell line. Further, WR-1065 treatment decreases HR in response to DNA damage using two different mammalian cell systems. This suppression of hyper-recombination is a previously unrecognized mechanism by which WR-1065 effects radioprotection in mammalian cells.


Subject(s)
Amifostine/pharmacology , Mercaptoethylamines/pharmacology , Radiation-Protective Agents/pharmacology , Recombination, Genetic , Animals , CHO Cells , Camptothecin/pharmacology , Cell Line, Tumor , Cricetinae , Cricetulus , DNA-Binding Proteins/metabolism , Genomic Instability , Humans , Hydroxyurea/pharmacology , RNA Interference , Rad51 Recombinase/metabolism , Reactive Oxygen Species/metabolism
14.
Environ Mol Mutagen ; 50(6): 460-72, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19334055

ABSTRACT

The success of nucleoside reverse transcriptase inhibitors (NRTIs) in treating HIV-1 infection and reducing mother-to-child transmission of the virus during pregnancy is accompanied by evidence that NRTIs cause long-term health risks for cancer and mitochondrial disease. Thus, agents that mitigate toxicities of the current combination drug therapies are needed. Previous work had shown that the NRTI-drug pair zidovudine (AZT)-didanosine (ddI) was highly cytotoxic and mutagenic; thus, we conducted preliminary studies to investigate the ability of the active moiety of amifostine, WR1065, to protect against the deleterious effects of this NRTI-drug pair. In TK6 cells exposed to 100 muM AZT-ddI (equimolar) for 3 days with or without 150 muM WR1065, WR1065 enhanced long-term cell survival and significantly reduced AZT-ddI-induced mutations. Follow-up studies were conducted to determine if coexposure to AZT and WR1065 abrogated the antiretroviral efficacy of AZT. In human T-cell blasts infected with HIV-1 in culture, inhibition of p24 protein production was observed in cells treated with 10 muM AZT in the absence or presence of 5-1,000 muM WR1065. Surprisingly, WR1065 alone exhibited dose-related inhibition of HIV-1 p24 protein production. WR1065 also had antiviral efficacy against three species of adenovirus and influenza A and B. Intracellular levels of unbound WR1065 were measured following in vitro/in vivo drug exposure. These pilot study results indicate that WR1065, at low intracellular levels, has cytoprotective and antimutagenic activities against the most mutagenic pair of NRTIs and has broad spectrum antiviral effects. These findings suggest that the activities have a possible common mode of action that merits further investigation.


Subject(s)
Didanosine/analogs & derivatives , Dideoxynucleotides/toxicity , Mercaptoethylamines/pharmacology , Mutagenesis/drug effects , Virus Replication/drug effects , Zidovudine/analogs & derivatives , Adenoviridae/drug effects , Adenoviridae/physiology , Cell Line , Cytoplasm/drug effects , Cytoplasm/metabolism , Didanosine/toxicity , Dose-Response Relationship, Drug , HIV Core Protein p24/metabolism , HIV-1/drug effects , HIV-1/physiology , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Influenza A virus/drug effects , Influenza A virus/physiology , Influenza B virus/drug effects , Influenza B virus/physiology , Intracellular Space/drug effects , Intracellular Space/metabolism , Lymphocytes/drug effects , Lymphocytes/virology , Mutation/genetics , Phytohemagglutinins/pharmacology , Serotyping , Time Factors , Zidovudine/toxicity
15.
J Nutr Sci Vitaminol (Tokyo) ; 55(1): 66-74, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19352065

ABSTRACT

Rats were fed diets with and without 0.5% L-cysteine supplement for 14 d or shorter periods to clarify the mechanism by which dietary cysteine elicits its hypohomocysteinemic effect. Cysteine supplementation significantly decreased plasma homocysteine concentration with an increase in plasma cysteine concentration in rats fed 10% casein diet (10C) or 15% soybean protein diet (15S) but not in rats fed 25% casein diet (25C) or 25% soybean protein diet. Cysteine supplementation also significantly suppressed hyperhomocysteinemia induced by choline-deprived 10C with an increase in plasma cysteine concentration but not that induced by 25C+0.65% methionine or 25C+0.4% guanidinoacetic acid. Hepatic S-adenosylmethionine (SAM) and homocysteine concentrations were significantly decreased by cysteine supplementation of 15S. These decreases in plasma homocysteine concentration and hepatic SAM and homocysteine concentrations due to cysteine supplementation disappeared when 15S was fortified with 0.3% methionine. The plasma homocysteine concentration significantly decreased with an increase in plasma cysteine concentration only 1 d after diet change from 15S to cysteine-supplemented 15S, while hepatic cystathionine beta-synthase and betaine-homocysteine S-methyltransferase activities were not altered. Unlike cysteine, cysteic acid and 2-mercaptoethylamine did not decrease plasma homocysteine concentration. These results indicate that cysteine markedly decreases plasma homocysteine concentration only when added to diets low in both protein and methionine levels and suggest that increased plasma cysteine concentration and decreased flow of methionine toward homocysteine formation, but not alteration of homocysteine-metabolizing enzyme activities, are associated with the hypohomocysteinemic effect of cysteine.


Subject(s)
Cysteine/blood , Cysteine/pharmacology , Diet, Protein-Restricted , Homocysteine/metabolism , Hyperhomocysteinemia/prevention & control , Methionine/administration & dosage , Animals , Betaine-Homocysteine S-Methyltransferase/metabolism , Caseins/pharmacology , Choline/pharmacology , Cystathionine beta-Synthase/metabolism , Cysteic Acid/pharmacology , Cysteine/therapeutic use , Dietary Supplements , Glycine/analogs & derivatives , Glycine/pharmacology , Male , Mercaptoethylamines/pharmacology , Methionine/pharmacology , Rats , Rats, Wistar , S-Adenosylhomocysteine/metabolism , Soybean Proteins/pharmacology
16.
Int J Radiat Oncol Biol Phys ; 73(3): 886-96, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19215822

ABSTRACT

PURPOSE: To determine whether amifostine can induce elevated manganese superoxide dismutase (SOD2) in murine tissues and a transplantable SA-NH tumor, resulting in a delayed tumor cell radioprotective effect. METHODS AND MATERIALS: SA-NH tumor-bearing C3H mice were treated with a single 400 mg/kg or three daily 50 mg/kg doses of amifostine administered intraperitoneally. At selected time intervals after the last injection, the heart, liver, lung, pancreas, small intestine, spleen, and SA-NH tumor were removed and analyzed for SOD2, catalase, and glutathione peroxidase (GPx) enzymatic activity. The effect of elevated SOD2 enzymatic activity on the radiation response of SA-NH cells was determined. RESULTS: SOD2 activity was significantly elevated in selected tissues and a tumor 24 h after amifostine treatment. Catalase and GPx activities remained unchanged except for significant elevations in the spleen. GPx was also elevated in the pancreas. SA-NH tumor cells exhibited a twofold elevation in SOD2 activity and a 27% elevation in radiation resistance. Amifostine administered in three daily fractions of 50 mg/kg each also resulted in significant elevations of these antioxidant enzymes. CONCLUSIONS: Amifostine can induce a delayed radioprotective effect that correlates with elevated levels of SOD2 activity in SA-NH tumor. If limited to normal tissues, this delayed radioprotective effect offers an additional potential for overall radiation protection. However, amifostine-induced elevation of SOD2 activity in tumors could have an unanticipated deleterious effect on tumor responses to fractionated radiation therapy, given that the radioprotector is administered daily just before each 2-Gy fractionated dose.


Subject(s)
Amifostine/pharmacology , Catalase/metabolism , Glutathione Peroxidase/metabolism , Radiation-Protective Agents/pharmacology , Superoxide Dismutase/metabolism , Animals , Female , Liver/enzymology , Lung/enzymology , Mercaptoethylamines/pharmacology , Mice , Mice, Inbred C3H , Myocardium/enzymology , Pancreas/enzymology , Radiation Tolerance/drug effects , Sarcoma, Experimental/enzymology
17.
Fertil Steril ; 91(1): 173-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18206143

ABSTRACT

OBJECTIVE: To determine the effect of 4-hydroperoxycyclophosphamide (4OOH-CP) on the respiration of human sperm, and investigate the protective properties of mesna and WR-1065. SETTING: SUNY Upstate Medical University, Syracuse, NY. PATIENT(S): Men (n = 12) visited the Andrology Department for fertility evaluation. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Sperm respiration. RESULT(S): Immediate decline in the rate of respiration was observed when 4OOH-CP was added to washed sperm or semen. The inhibition was concentration dependent. The respiration was less affected when 4OOH-CP was added to semen, suggesting the presence of protective factors in the seminal plasma. Excess of mesna or WR-1065 ameliorated the effect of 4OOH-CP. Mesna was the more potent of the two compounds. 4OOH-CP also inhibited the respiration of mitochondria from beef heart. CONCLUSION(S): These findings emphasize the adverse effects of alkylating agents on sperm function. The results also provide a framework for thiol drug administration with high-dose alkylating agents to protect male fertility. The protective capacity of seminal plasma deserves further testing.


Subject(s)
Cyclophosphamide/analogs & derivatives , Mercaptoethylamines/pharmacology , Mesna/pharmacology , Mitochondria/metabolism , Oxygen Consumption/drug effects , Protective Agents/pharmacology , Semen/physiology , Spermatozoa/physiology , Alkylating Agents/pharmacology , Cyclophosphamide/pharmacology , Humans , Male , Mitochondria/drug effects , Semen/drug effects , Spermatozoa/drug effects
18.
Int J Radiat Biol ; 84(11): 900-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19016138

ABSTRACT

PURPOSE: N-(2-mercaptoethyl)1,3-diaminopropane (WR-1065), is the active metabolite of amifostine, a broad spectrum cytoprotective agent used in conjunction with both chemo- and radiotherapy of certain cancers. This report describes for the first time an oral formulation of WR-1065 and follows on from our earlier report of a similar oral formulation of amifostine. MATERIALS AND METHODS: The nanoparticles of WR-1065 were prepared by spray drying technique using poly lactide-co-glycolide (PLGA) as the polymer matrix. Radioprotection was determined by measuring reductions in radiation-induced: (i) 30-day survival; (ii) bone marrow suppression; and (iii) intestinal injury following 9 Gray (Gy) whole body gamma irradiation in mice. All treatments were given 1 hour pre-irradiation and WR-1065 was tested at the dose of 500 mg/kg. RESULTS: The WR-1065/PLGA nanoparticles were smooth and spherical with the average diameter of 206 nm and contained 21.7% (w/w) WR-1065. While irradiation markedly reduced 30-day survival in non-treated control mice, and caused significant bone marrow suppression and intestinal injury in surviving mice, oral administration of WR-1065/PLGA nanoparticles resulted in significant radioprotection as evidenced by a marked reduction in all three of the above mentioned parameters of radiation injury. CONCLUSIONS: These findings clearly demonstrate the feasibility of developing an effective oral formulation of WR-1065 as a radioprotective agent.


Subject(s)
Mercaptoethylamines/administration & dosage , Mercaptoethylamines/pharmacology , Nanoparticles/chemistry , Polyglactin 910/chemistry , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/pharmacology , Administration, Oral , Animals , Bone Marrow/drug effects , Bone Marrow/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Jejunum/cytology , Jejunum/drug effects , Jejunum/radiation effects , Male , Mercaptoethylamines/chemistry , Mice , Radiation-Protective Agents/chemistry , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/radiation effects , Survival Rate
19.
Free Radic Biol Med ; 45(12): 1674-81, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18845240

ABSTRACT

Compounds that can protect cells from the effects of radiation are important for clinical use, in the event of an accidental or terrorist-generated radiation event, and for astronauts traveling in space. One of the major concerns regarding the use of radio-protective agents is that they may protect cells initially, but predispose surviving cells to increased genomic instability later. In this study we used WR-1065, the active metabolite of amifostine, to determine how protection from direct effects of high- and low-LET radiation exposure influences genomic stability. When added 30 min before irradiation and in high concentrations, WR-1065 protected cells from immediate radiation-induced effects as well as from delayed genomic instability. Lower, nontoxic concentrations of WR-1065 did not protect cells from death; however, it was effective in significantly decreasing delayed genomic instability in the progeny of irradiated cells. The observed increase in manganese superoxide dismutase protein levels and activity may provide an explanation for this effect. These results confirm that WR-1065 is protective against both low- and high-LET radiation-induced genomic instability in surviving cells.


Subject(s)
Amifostine/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Genomic Instability/drug effects , Mercaptoethylamines/pharmacology , Radiation-Protective Agents/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Dose-Response Relationship, Radiation , Green Fluorescent Proteins/metabolism , Humans , Micronucleus Tests , Radiation Tolerance , Superoxide Dismutase/metabolism , X-Rays
20.
Phytother Res ; 22(12): 1614-22, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18803249

ABSTRACT

To explore the radioprotective effect of a standardized North American ginseng extract (NAGE) on human peripheral blood lymphocytes (PBL), a micronuclei (MN) assay was conducted in PBL obtained from 12 volunteers. NAGE (50-1000 microg/mL) and WR-1065 (1 mM and 3 mM) were applied to PBL cultures at 0 h and 90 min post-irradiation. It was found that (1) the baseline MN yield of PBL ranged from 14.4 +/- 1.5 to 15.9 +/- 1.5 per 1000 binucleated cells (p > 0.05); after irradiation (1 Gy and 2 Gy), the MN yield increased sharply; (2) MN yields declined with increasing concentrations of NAGE and WR-1065. Even at 90 min post-irradiation of 1 Gy, the maximum level of MN reduction rate caused by NAGE and WR-1065 was 53.8% and 59.2%, respectively; after 2 Gy irradiation, it was 37.3% and 42%, respectively; (3) the MN distribution in PBL followed a non-Poisson distribution in all cases; and (4) both NAGE and WR-1065 showed no significant effect on the proliferation index of lymphocytes. The results indicate that NAGE is a relatively non-toxic natural product, which can be administered as a dietary supplement and has the potential to be a radiation countermeasure.


Subject(s)
Lymphocytes/drug effects , Mercaptoethylamines/pharmacology , Panax/chemistry , Plant Extracts/pharmacology , Radiation-Protective Agents/pharmacology , Adult , Cells, Cultured , Cesium/toxicity , Chlorides/toxicity , Dose-Response Relationship, Drug , Ginsenosides/pharmacology , Humans , Lymphocytes/radiation effects , Micronuclei, Chromosome-Defective/drug effects , Micronuclei, Chromosome-Defective/radiation effects , Micronucleus Tests , Middle Aged , Radiation, Ionizing
SELECTION OF CITATIONS
SEARCH DETAIL
...