Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Toxicol ; 42(3): 278-286, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36941229

RESUMO

The nitramine explosive, hexahydro-1,3,5-trinitro-1,3,5-triazine (RDX) is associated with acute and chronic toxicity in mammals and targets both the central nervous system and liver. After a single oral dose of RDX in male rats, the systemic distribution of RDX and the toxicodynamic response was measured using clinical chemistry and Affymetrix Rat Genome® 230 2.0 gene expression arrays, respectively. Nominal doses of 0, 9 and 36 mg/kg pure RDX were administered to animals followed by liver, cerebral cortex, and hippocampus gene expression analysis at 0, 3.5, 24, and 48 hours. RDX quickly entered the liver and brain, increasing up to 24 hours. For the 36 mg/kg dose, RDX was still measurable in liver and brain at 48 hours, but was non-detectible for the 9 mg/kg dose. At 3.5 hours, the time within which most convulsions reportedly occur after RDX ingestion, the hippocampus displayed the highest response for both gene expression and pathways, while the cortex was relatively non-responsive. The top 2 impacted pathways, primarily involved in neurotransmission, were the GABAergic and glutamatergic pathways. High numbers of genes also responded to RDX in the liver with P450 metabolism pathways significantly involved. Compared to the liver, the hippocampus displayed more consistent biological effects across dose and time with neurotransmission pathways predominating. Overall, based on gene expression data, RDX responses were high in both the hippocampus and liver, but were minimal in the cerebral cortex. These results identify the hippocampus as an important target for RDX based on gene expression.


Assuntos
Substâncias Explosivas , Ratos , Masculino , Animais , Substâncias Explosivas/toxicidade , Fígado , Triazinas/toxicidade , Encéfalo/metabolismo , Expressão Gênica , Mamíferos/metabolismo
2.
Chem Res Toxicol ; 29(10): 1602-1610, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27617619

RESUMO

Sulfur mustard (HD) is a vesicating and alkylating agent widely used on the battlefield during World War I and more recently in the Iran-Iraq War. It targets the eyes, skin, and lungs, producing skin burns, conjunctivitis, and compromised respiratory function; early acute effects lead to long-term consequences. However, it is the effects on the lungs that drive morbidity and eventual mortality. The temporal postexposure response to HD within lung tissue raises the question of whether toxicity is driven by the alkylating properties of HD on critical homeostatic pathways. We have established an anesthetized swine model of inhaled HD vapor exposure to investigate the toxic effects of HD 12 h postexposure. Large white female swine were anesthetized and instrumented prior to exposure to air, 60 (sublethal) or 100 µg·kg-1 (∼LD40) doses of HD (10 min). Physiological parameters were continuously assessed. Data indicate that exposure to 100 µg·kg-1 HD lowered arterial blood oxygenation and increased shunt fraction and lavage protein compared with those of air-exposed controls and the 60 µg·kg-1 dose of HD. Histopathology showed an increased total pathology score between the 100 µg·kg-1 HD group and air-exposed controls. Principal component analysis of differentially expressed genes demonstrated a distinct and separable response of inhaled HD between air-exposed controls and the 60 and 100 µg·kg-1 doses of HD. Canonical pathway analysis demonstrated changes in acute phase response signaling, aryl hydrocarbon receptor signaling, NRF-2 mediated oxidative stress, and zymosterol biosynthesis in the 60 and 100 µg·kg-1 HD dose group. Transcriptional changes also indicated alterations in immune response, cancer, and cell signaling and metabolism canonical pathways. The 100 µg·kg-1 dose group also showed significant changes in cholesterol biosynthesis. Taken together, exposure to inhaled HD had a significant effect on physiological responses coinciding with acute changes in gene expression and lung histopathology. In addition, transcriptomics support the observed beneficial effects of N-acetyl-l-cysteine for treatment of acute inhalation HD exposure.


Assuntos
Anestesia , Perfilação da Expressão Gênica , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Gás de Mostarda/administração & dosagem , Gás de Mostarda/toxicidade , Acetilcisteína/uso terapêutico , Administração por Inalação , Animais , Relação Dose-Resposta a Droga , Feminino , Exposição por Inalação , Modelos Animais , Suínos , Testes de Toxicidade
3.
Toxicol Lett ; 244: 8-20, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26562770

RESUMO

Toxic industrial chemicals are used throughout the world to produce everyday products such as household and commercial cleaners, disinfectants, pesticides, pharmaceuticals, plastics, paper, and fertilizers. These chemicals are produced, stored, and transported in large quantities, which poses a threat to the local civilian population in cases of accidental or intentional release. Several of these chemicals have no known medical countermeasures for their toxic effects. Phosgene is a highly toxic industrial chemical which was used as a chemical warfare agent in WWI. Exposure to phosgene causes latent, non-cardiogenic pulmonary edema which can result in respiratory failure and death. The mechanisms of phosgene-induced pulmonary injury are not fully identified, and currently there is no efficacious countermeasure. Here, we provide a proposed mechanism of phosgene-induced lung injury based on the literature and from studies conducted in our lab, as well as provide results from studies designed to evaluate survival efficacy of potential therapies following whole-body phosgene exposure in mice. Several therapies were able to significantly increase 24h survival following an LCt50-70 exposure to phosgene; however, no treatment was able to fully protect against phosgene-induced mortality. These studies provide evidence that mortality following phosgene toxicity can be mitigated by neuro- and calcium-regulators, antioxidants, phosphodiesterase and endothelin receptor antagonists, angiotensin converting enzymes, and transient receptor potential cation channel inhibitors. However, because the mechanism of phosgene toxicity is multifaceted, we conclude that a single therapeutic is unlikely to be sufficient to ameliorate the multitude of direct and secondary toxic effects caused by phosgene inhalation.


Assuntos
Antídotos/uso terapêutico , Substâncias para a Guerra Química , Lesão Pulmonar/tratamento farmacológico , Pulmão/efeitos dos fármacos , Fosgênio , Animais , Modelos Animais de Doenças , Exposição por Inalação , Pulmão/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/diagnóstico , Lesão Pulmonar/metabolismo , Lesão Pulmonar/fisiopatologia , Masculino , Camundongos , Terapia de Alvo Molecular , Transdução de Sinais/efeitos dos fármacos
4.
Arch Toxicol ; 90(2): 375-83, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25537185

RESUMO

Sulfur mustard [SM, bis-(2-chloroethyl) sulfide] is a highly reactive bifunctional alkylating agent that has been used as a vesicating agent in warfare scenarios to induce severe lung, skin, and eye injury. SM cutaneous lesions are characterized by both vesication and severe inflammation, but the molecular mechanisms that lead to these signs and symptoms are not well understood. There is a pressing need for effective therapeutics to treat this injury. An understanding of the molecular mechanisms of injury and identification of potential therapeutic targets is necessary for rational therapeutic development. We have applied a high-throughput small interfering RNA (siRNA) screening approach to the problem of SM cutaneous injury in an effort to meet these needs. Our siRNA screening efforts have initially focused on SM-induced inflammation in cutaneous injury since chronic inflammation after exposure appears to play a role in progressive clinical pathology, and intervention may improve clinical outcome. Also, targets that mitigate cellular injury should reduce the inflammatory response. Historical microarray data on this injury were mined for targets and pathways implicated in inflammation, and a siRNA library of 2,017 targets was assembled for screening. Primary screening and library deconvolution were performed using human HaCaT keratinocytes and focused on cell death and inflammatory markers as end points. Using this in vitro approach, we have identified and validated novel targets for the potential treatment of SM-induced cutaneous injury.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Irritantes/toxicidade , Terapia de Alvo Molecular/métodos , Gás de Mostarda/toxicidade , RNA Interferente Pequeno , Linhagem Celular , Substâncias para a Guerra Química/toxicidade , Dermatite de Contato/etiologia , Humanos , Queratinócitos/efeitos dos fármacos , Pele/efeitos dos fármacos
5.
Brain Res ; 1618: 136-48, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26049129

RESUMO

Exposure to organophosphate (OP) nerve agents, such as sarin, may lead to uncontrolled seizures and irreversible brain injury and neuropathology. In rat studies, a median lethal dose of sarin leads to approximately half of the animals developing seizures. Whereas previous studies analyzed transcriptomic effects associated with seizing sarin-exposed rats, our study focused on the cohort of sarin-exposed rats that did not develop seizures. We analyzed the genomic changes occurring in sarin-exposed, non-seizing rats and compared differentially expressed genes and pathway activation to those of seizing rats. At the earliest time point (0.25 h) and in multiple sarin-sensitive brain regions, defense response genes were commonly expressed in both groups of animals as compared to the control groups. All sarin-exposed animals activated the MAPK signaling pathway, but only the seizing rats activated the apoptotic-associated JNK and p38 MAPK signaling sub-pathway. A unique phenotype of the non-seizing rats was the altered expression levels of genes that generally suppress inflammation or apoptosis. Importantly, the early transcriptional response for inflammation- and apoptosis-related genes in the thalamus showed opposite trends, with significantly down-regulated genes being up-regulated, and vice versa, between the seizing and non-seizing rats. These observations lend support to the hypothesis that regulation of anti-inflammatory genes might be part of an active and sufficient response in the non-seizing group to protect against the onset of seizures. As such, stimulating or activating these responses via pretreatment strategies could boost resilience against nerve agent exposures.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Inibidores da Colinesterase/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Sarina/farmacologia , Transcrição Gênica/efeitos dos fármacos , Animais , Ratos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
6.
Toxicol In Vitro ; 27(7): 2067-75, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23851002

RESUMO

Uncontrolled inflammation contributes to cutaneous damage following exposure to the warfare agent bis(2-chloroethyl) sulfide (sulfur mustard, SM). Activation of the p38 mitogen activated protein kinase (MAPK) precedes SM-induced cytokine secretion in normal human epidermal keratinocytes (NHEKs). This study examined the role of p38-regulated MAPK activated kinase 2 (MK2) during this process. Time course analysis studies using NHEK cells exposed to 200µM SM demonstrated rapid MK2 activation via phosphorylation that occurred within 15 min. p38 activation was necessary for MK2 phosphorylation as determined by studies using the p38 inhibitor SB203580. To compare the role of p38 and MK2 during SM-induced cytokine secretion, small interfering RNA (siRNA) targeting these proteins was utilized. TNF-α, IL-1ß, IL-6 and IL-8 secretion was evaluated 24h postexposure, while mRNA changes were quantified after 8h. TNF-α, IL-6 and IL-8 up regulation at the protein and mRNA level was observed following SM exposure. IL-1ß secretion was also elevated despite unchanged mRNA levels. p38 knockdown reduced SM-induced secretion of all the cytokines examined, whereas significant reduction in SM-induced cytokine secretion was only observed with TNF-α and IL-6 following MK2 knockdown. Our observations demonstrate potential activation of other p38 targets in addition to MK2 during SM-induced cytokine secretion.


Assuntos
Substâncias para a Guerra Química/toxicidade , Citocinas/metabolismo , Fármacos Dermatológicos/toxicidade , Peptídeos e Proteínas de Sinalização Intracelular/agonistas , Queratinócitos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Gás de Mostarda/toxicidade , Anti-Inflamatórios não Esteroides/farmacologia , Células Cultivadas , Substâncias para a Guerra Química/química , Citocinas/química , Citocinas/genética , Fármacos Dermatológicos/antagonistas & inibidores , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Irritantes/antagonistas & inibidores , Irritantes/toxicidade , Queratinócitos/citologia , Queratinócitos/imunologia , Queratinócitos/metabolismo , Cinética , Gás de Mostarda/química , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/química , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
J Neuroinflammation ; 8: 83, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21777429

RESUMO

BACKGROUND: Organophosphorus nerve agents irreversibly inhibit acetylcholinesterase, causing a toxic buildup of acetylcholine at muscarinic and nicotinic receptors. Current medical countermeasures to nerve agent intoxication increase survival if administered within a short period of time following exposure but may not fully prevent neurological damage. Therefore, there is a need to discover drug treatments that are effective when administered after the onset of seizures and secondary responses that lead to brain injury. METHODS: To determine potential therapeutic targets for such treatments, we analyzed gene expression changes in the rat piriform cortex following sarin (O-isopropyl methylphosphonofluoridate)-induced seizure. Male Sprague-Dawley rats were challenged with 1 × LD50 sarin and subsequently treated with atropine sulfate, 2-pyridine aldoxime methylchloride (2-PAM), and the anticonvulsant diazepam. Control animals received an equivalent volume of vehicle and drug treatments. The piriform cortex, a brain region particularly sensitive to neural damage from sarin-induced seizures, was extracted at 0.25, 1, 3, 6, and 24 h after seizure onset, and total RNA was processed for microarray analysis. Principal component analysis identified sarin-induced seizure occurrence and time point following seizure onset as major sources of variability within the dataset. Based on these variables, the dataset was filtered and analysis of variance was used to determine genes significantly changed in seizing animals at each time point. The calculated p-value and geometric fold change for each probeset identifier were subsequently used for gene ontology analysis to identify canonical pathways, biological functions, and networks of genes significantly affected by sarin-induced seizure over the 24-h time course. RESULTS: A multitude of biological functions and pathways were identified as being significantly altered following sarin-induced seizure. Inflammatory response and signaling pathways associated with inflammation were among the most significantly altered across the five time points examined. CONCLUSIONS: This analysis of gene expression changes in the rat brain following sarin-induced seizure and the molecular pathways involved in sarin-induced neurodegeneration will facilitate the identification of potential therapeutic targets for the development of effective neuroprotectants to treat nerve agent exposure.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Inibidores da Colinesterase/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Sarina/farmacologia , Convulsões/induzido quimicamente , Transcrição Gênica/efeitos dos fármacos , Animais , Anticonvulsivantes/uso terapêutico , Atropina/uso terapêutico , Encéfalo/anatomia & histologia , Reativadores da Colinesterase/uso terapêutico , Diazepam/uso terapêutico , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Masculino , Análise em Microsséries , Antagonistas Muscarínicos/uso terapêutico , Compostos de Pralidoxima/uso terapêutico , Análise de Componente Principal , Ratos , Ratos Sprague-Dawley , Convulsões/tratamento farmacológico , Transdução de Sinais/fisiologia
8.
J Neuroinflammation ; 8: 84, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21777430

RESUMO

BACKGROUND: Although the acute toxicity of organophosphorus nerve agents is known to result from acetylcholinesterase inhibition, the molecular mechanisms involved in the development of neuropathology following nerve agent-induced seizure are not well understood. To help determine these pathways, we previously used microarray analysis to identify gene expression changes in the rat piriform cortex, a region of the rat brain sensitive to nerve agent exposure, over a 24-h time period following sarin-induced seizure. We found significant differences in gene expression profiles and identified secondary responses that potentially lead to brain injury and cell death. To advance our understanding of the molecular mechanisms involved in sarin-induced toxicity, we analyzed gene expression changes in four other areas of the rat brain known to be affected by nerve agent-induced seizure (amygdala, hippocampus, septum, and thalamus). METHODS: We compared the transcriptional response of these four brain regions to sarin-induced seizure with the response previously characterized in the piriform cortex. In this study, rats were challenged with 1.0 × LD50 sarin and subsequently treated with atropine sulfate, 2-pyridine aldoxime methylchloride, and diazepam. The four brain regions were collected at 0.25, 1, 3, 6, and 24 h after seizure onset, and total RNA was processed for microarray analysis. RESULTS: Principal component analysis identified brain region and time following seizure onset as major sources of variability within the dataset. Analysis of variance identified genes significantly changed following sarin-induced seizure, and gene ontology analysis identified biological pathways, functions, and networks of genes significantly affected by sarin-induced seizure over the 24-h time course. Many of the molecular functions and pathways identified as being most significant across all of the brain regions were indicative of an inflammatory response. There were also a number of molecular responses that were unique for each brain region, with the thalamus having the most distinct response to nerve agent-induced seizure. CONCLUSIONS: Identifying the molecular mechanisms involved in sarin-induced neurotoxicity in these sensitive brain regions will facilitate the development of novel therapeutics that can potentially provide broad-spectrum protection in five areas of the central nervous system known to be damaged by nerve agent-induced seizure.


Assuntos
Tonsila do Cerebelo , Córtex Cerebral , Inibidores da Colinesterase/farmacologia , Hipocampo , Sarina/farmacologia , Septo do Cérebro , Tálamo , Transcrição Gênica/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Animais , Anticonvulsivantes/uso terapêutico , Atropina/uso terapêutico , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Substâncias para a Guerra Química/farmacologia , Diazepam/uso terapêutico , Perfilação da Expressão Gênica , Redes Reguladoras de Genes/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Análise em Microsséries , Antagonistas Muscarínicos/uso terapêutico , Oximas/uso terapêutico , Análise de Componente Principal , Piridinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Convulsões/fisiopatologia , Septo do Cérebro/efeitos dos fármacos , Septo do Cérebro/fisiologia , Tálamo/efeitos dos fármacos , Tálamo/fisiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
9.
Toxicol Lett ; 198(3): 297-303, 2010 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20708669

RESUMO

Medical research on the effects of chemical warfare agents (CWAs) has been ongoing for nearly 100 years, yet these agents continue to pose a serious threat to deployed military forces and civilian populations. CWAs are extremely toxic, relatively inexpensive, and easy to produce, making them a legitimate weapon of choice for terrorist organizations. While the mechanisms of action for many CWAs have been known for years, questions about their molecular effects following acute and chronic exposure remain largely unanswered. Global approaches that can pinpoint which cellular pathways are altered in response to CWAs and characterize long-term toxicity have not been widely used. Fortunately, innovations in genomics and proteomics technologies now allow for thousands of genes and proteins to be identified and subsequently quantified in a single experiment. Advanced bioinformatics software can also help decipher large-scale changes observed, leading to mapping of signaling pathways, functional characterization, and identification of potential therapeutic targets. Here we present an overview of how genomics and proteomics technologies have been applied to CWA research and also provide a series of questions focused on how these techniques could further our understanding of CWA toxicity.


Assuntos
Substâncias para a Guerra Química/química , Biologia Computacional/métodos , Genômica/métodos , Proteômica/métodos , Animais , Substâncias para a Guerra Química/intoxicação , Substâncias para a Guerra Química/toxicidade , Humanos
10.
J Biochem Mol Toxicol ; 24(3): 155-64, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20143454

RESUMO

Cutaneous and ocular injuries caused by sulfur mustard (SM; bis-(2-chloroethyl) sulfide) are characterized by severe inflammation and death of exposed cells. Given the known roles of p38MAPK and NF-kappaB in inflammatory cytokine production, and the known roles of NF-kappaB and p53 in cell fate, these pathways are of particular interest in the study of SM injury. In this study, we utilized inhibitory RNA (RNAi) targeted against p38 alpha, the p50 subunit of NF-kappaB, or p53 to characterize their role in SM-induced inflammation and cell death in normal human epidermal keratinocytes (NHEK). Analysis of culture supernatant from 200 microM SM-exposed cells showed that inflammatory cytokine production was inhibited by p38 alpha RNAi but not by NF-kappaB p50 RNAi. These findings further support a critical role for p38 in SM-induced inflammatory cytokine production in NHEK and suggest that NF-kappaB may not play a role in the SM-induced inflammatory response of this cell type. Inhibition of NF-kappaB by p50 RNAi did, however, partially inhibit SM-induced cell death, suggesting a role for NF-kappaB in SM-induced apoptosis or necrosis. Interestingly, inhibition of p53 by RNAi potentiated SM-induced cell death, suggesting that the role of p53 in SM injury, may be complex and not simply prodeath.


Assuntos
Apoptose/efeitos dos fármacos , Citocinas/biossíntese , Gás de Mostarda/toxicidade , NF-kappa B/fisiologia , Interferência de RNA , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Humanos
11.
Chem Res Toxicol ; 23(1): 20-5, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19845377

RESUMO

Sulfur mustard [SM, bis-(2-chloroethyl) sulfide] is a potent alkylating agent and chemical weapon. While there are no effective treatments for SM-induced injury, current research focuses on understanding the molecular changes upon SM exposure. Indeed, efforts that seek a more comprehensive analysis of proteins and post-translational modifications are critical for understanding SM-induced toxicity on a more global scale. Furthermore, these studies can uncover proteins previously uncharacterized in SM-exposed cells, which in turn leads to potential targets for therapeutic intervention. Here, we apply a quantitative proteomic approach termed stable isotope-labeling with amino acids in cell culture combined with immobilized metal affinity chromatography to study the large-scale protein phosphorylation changes resulting from SM exposure in a human keratinocyte cell culture model. This resulted in the characterization of over 2300 nonredundant phosphorylation sites, many of which exhibit altered levels in response to SM. Our results point toward several proteins previously implicated in SM-induced toxicity as well as many additional proteins previously uncharacterized. Further de novo analysis of these phosphoproteins using interaction mapping software revealed both known and novel pathways that may serve as future therapeutic targets of SM exposure.


Assuntos
Substâncias para a Guerra Química/toxicidade , Gás de Mostarda/toxicidade , Proteômica/métodos , Cromatografia Líquida de Alta Pressão , Fosforilação , Espectrometria de Massas em Tandem , Tripsina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Chem Res Toxicol ; 22(4): 633-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19281266

RESUMO

Soman (O-pinacolyl methylphosphonofluoridate) is a potent neurotoxicant. Acute exposure to soman causes acetylcholinesterase inhibition, resulting in excessive levels of acetylcholine. Excessive acetylcholine levels cause convulsions, seizures, and respiratory distress. The initial cholinergic crisis can be overcome by rapid anticholinergic therapeutic intervention, resulting in increased survival. However, conventional treatments do not protect the brain from seizure-related damage, and thus, neurodegeneration of soman-sensitive brain areas is a potential postexposure outcome. We performed gene expression profiling of the rat hippocampus following soman exposure to gain greater insight into the molecular pathogenesis of soman-induced neurodegeneration. Male Sprague-Dawley rats were pretreated with the oxime HI-6 (l-(((4-aminocarbonyl)pyridinio)methoxyl)methyl)-2-((hydroxyimino)methyl)-pyridinium dichloride; 125 mg/kg, ip) 30 min prior to challenge with soman (180 microg/kg, sc). One minute after soman challenge, animals were treated with atropine methyl nitrate (2.0 mg/kg, im). Hippocampi were harvested 1, 3, 6, 12, 24, 48, 72, 96, and 168 h after soman exposure and RNA extracted to generate microarray probes for gene expression profiling. Principal component analysis of the microarray data revealed a progressive alteration in gene expression profiles beginning 1 h postexposure and continuing through 24 h postexposure. At 48 h to 168 h postexposure, the gene expression profiles clustered nearer to controls but did not completely return to control profiles. On the basis of the principal component analysis, analysis of variance was used to identify the genes most significantly changed as a result of soman at each postexposure time point. To gain insight into the biological relevance of these gene expression changes, genes were rank ordered by p-value and categorized using gene ontology-based algorithms into biological functions, canonical pathways, and gene networks significantly affected by soman. Numerous signaling and inflammatory pathways were identified as perturbed by soman. These data provide important insights into the molecular pathways involved in soman-induced neuropathology and a basis for generating hypotheses about the mechanism of soman-induced neurodegeneration.


Assuntos
Inibidores da Colinesterase/toxicidade , Perfilação da Expressão Gênica , Hipocampo/metabolismo , Soman/toxicidade , Animais , Derivados da Atropina/administração & dosagem , Inibidores da Colinesterase/administração & dosagem , Hipocampo/efeitos dos fármacos , Interleucina-6/metabolismo , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Componente Principal , Ratos , Ratos Sprague-Dawley , Soman/administração & dosagem , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
13.
Chem Res Toxicol ; 22(4): 620-5, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19239275

RESUMO

RDX (hexahydro-1,3,5-trinitro-1,3,5-triazine) is a synthetic, high-impact, relatively stable explosive that has been in use since WWII. Exposure to RDX can occur in occupational settings (e.g., during manufacture) or through the inadvertent ingestion of contaminated environmental media such as groundwater. The toxicology of RDX is dominated by acute clonic-tonic seizures at high doses, which remit when exposure is removed and internal RDX levels decrease. Subchronic studies have revealed few other measurable toxic effects. The objective of this study was to examine the acute effects of RDX on the mammalian brain and liver using global gene expression analysis based on a predetermined maximum internal dose. Male Sprague-Dawley rats were given a single, oral, nonseizure-inducing dose of either 3 or 18 mg/kg RDX in a gel capsule. Effects on gene expression in the cerebral cortex and liver were assessed using Affymetrix Rat Genome 230 2.0 whole genome arrays at 0, 3.5, 24, and 48 h postexposure. RDX blood and brain tissue concentrations rapidly increased between 0 and 3.5 h, followed by decreases at 24 h to below the detection limit at 48 h. Pairwise comparison of high and low doses at each time point showed dramatic differential changes in gene expression at 3.5 h, the time of peak RDX in brain and blood. Using Gene Ontology, biological processes that affected neurotransmission were shown to be primarily down-regulated in the brain, the target organ of toxicity, while those that affected metabolism were up-regulated in the liver, the site of metabolism. Overall, these results demonstrate that a single oral dose of RDX is quickly absorbed and transported into the brain where processes related to neurotransmission are negatively affected, consistent with a potential excitotoxic response, whereas in the liver there was a positive effect on biological processes potentially associated with RDX metabolism.


Assuntos
Encéfalo/metabolismo , Substâncias Explosivas/toxicidade , Expressão Gênica/efeitos dos fármacos , Fígado/metabolismo , Triazinas/toxicidade , Administração Oral , Animais , Encéfalo/efeitos dos fármacos , Substâncias Explosivas/administração & dosagem , Regulação da Expressão Gênica , Fígado/efeitos dos fármacos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Triazinas/administração & dosagem
14.
J Pharmacol Exp Ther ; 321(1): 202-12, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17204746

RESUMO

Bifunctional alkylating agents (BFA) such as mechlorethamine (nitrogen mustard) and bis-(2-chloroethyl) sulfide (sulfur mustard; SM) covalently modify DNA and protein. The roles of nuclear factor kappaB (NF-kappaB) and p53, transcription factors involved in inflammatory and cell death signaling, were examined in normal human epidermal keratinocytes (NHEK) and immortalized HaCaT keratinocytes, a p53-mutated cell line, to delineate molecular mechanisms of action of BFA. NHEK and HaCaT cells exhibited classical NF-kappaB signaling as degradation of inhibitor protein of NF-kappaBalpha (IkappaBalpha) occurred within 5 min after exposure to tumor necrosis factor-alpha. However, exposure to BFA induced nonclassical NF-kappaB signaling as loss of IkappaBalpha was not observed until 2 or 6 h in NHEK or HaCaT cells, respectively. Exposure of an NF-kappaB reporter gene-expressing HaCaT cell line to 12.5, 50, or 100 muM SM activated the reporter gene within 9 h. Pretreatment with caffeic acid phenethyl ester (CAPE), a known inhibitor of NF-kappaB signaling, significantly decreased BFA-induced reporter gene activity. A 1.5-h pretreatment or 30-min postexposure treatment with CAPE prevented BFA-induced loss of membrane integrity by 24 h in HaCaT cells but not in NHEK. CAPE disrupted BFA-induced phosphorylation of p53 and p90 ribosomal S6 kinase (p90RSK) in both cell lines. CAPE also increased nuclear factor E2-related factor 2 and decreased aryl hydrocarbon receptor protein expression, both of which are involved in antioxidant/electrophilic response element (ARE/EpRE) signaling. Thus, disruption of p53/p90RSK-mediated NF-kappaB signaling and activation of ARE/EpRE pathways may be effective strategies to delineate mechanisms of action of BFA-induced inflammation and cell death signaling in immortalized versus normal skin systems.


Assuntos
Alquilantes/farmacologia , Antioxidantes/farmacologia , Ácidos Cafeicos/farmacologia , Genes p53/efeitos dos fármacos , Mecloretamina/farmacologia , Gás de Mostarda/farmacologia , NF-kappa B/efeitos dos fármacos , Álcool Feniletílico/análogos & derivados , Elementos de Resposta/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Western Blotting , Morte Celular/efeitos dos fármacos , Eletroforese em Gel de Poliacrilamida , Genes Reporter/genética , Humanos , Queratinócitos/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Luciferases/genética , Álcool Feniletílico/farmacologia , Receptores de Hidrocarboneto Arílico/biossíntese , Receptores de Hidrocarboneto Arílico/genética , Transfecção
15.
Eplasty ; 8: e2, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-18213398

RESUMO

OBJECTIVE: Sulfur mustard (SM) is a potent alkylating agent that can induce severe cutaneous injury. Though much is known regarding the gross pathology of SM injury, the molecular and cellular basis for this pathology is not well understood. General cellular processes such as inflammation, DNA damage response, and apoptosis have been hypothesized to be involved in SM injury. However, the specific molecules, signaling pathways, and gene products involved in the pathogenesis of SM injury have not been elucidated. This review discusses the molecular mechanisms observed in in vivo and in vitro models of cutaneous SM injury. METHODS: The historical literature on the clinical pathology of SM-induced cutaneous injury is summarized, and recent work elucidating molecular signaling pathways involved in SM toxicity is extensively reviewed. In addition, this review focuses the discussion of SM-induced molecular mechanisms on those that have been experimentally validated in models of SM injury. RESULTS: Recent work has uncovered potential roles for a number of signaling molecules. In particular, molecules in inflammatory signaling, DNA damage response, apoptosis signaling, and calcium signaling have been implicated in SM injury. These include signaling molecules involved in inflammation (e.g. p38 MAP kinase), apoptosis (e.g. p53, NF-kappa B, caspases, Fas), and cell stress responses (e.g. calcium, calmodulin). CONCLUSIONS: Many of the molecules and mechanisms implicated in SM injury are now being experimentally validated. Critical questions are proposed that remain to be answered to increase our understanding of SM toxicity and accelerate the development of vesicant therapeutics.

16.
Toxicol Sci ; 92(2): 560-77, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16601082

RESUMO

Although microarray technology has emerged as a powerful tool to explore expression levels of thousands of genes or even complete genomes after exposure to toxicants, the functional interpretation of microarray data sets still represents a time-consuming and challenging task. Gene ontology (GO) and pathway mapping have both been shown to be powerful approaches to generate a global view of biological processes and cellular components impacted by toxicants. However, current methods only allow for comparisons across two experimental settings at one particular time point. In addition, the resulting annotations are presented in extensive gene lists with minimal or limited quantitative information, data that are crucial in the application of toxicogenomic data for risk assessment. To facilitate quantitative interpretation of dose- or time-dependent genomic data, we propose to use combined average raw gene expression values (e.g., intensity or ratio) of genes associated with specific functional categories derived from the GO database. We developed an extended program (GO-Quant) to extract quantitative gene expression values and to calculate the average intensity or ratio for those significantly altered by functional gene category based on MAPPFinder results. To demonstrate its application, we applied this approach to a previously published dose- and time-dependent toxicogenomic data set (J. F. Dillman et al., 2005, Chem. Res. Toxicol. 18, 28-34). Our results indicate that the above systems approach can describe quantitatively the degree to which functional gene systems change across dose or time. Additionally, this approach provides a robust measurement to illustrate results compared to single-gene assessments and enables the user to calculate the corresponding ED(50) for each specific functional GO term, important for risk assessment.


Assuntos
Gás de Mostarda/toxicidade , Toxicogenética , Animais , Perfilação da Expressão Gênica , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Medição de Risco
17.
J Pharmacol Exp Ther ; 317(1): 76-87, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16377760

RESUMO

Bis-(2-chloroethyl) sulfide (sulfur mustard; SM) is a potent alkylating agent. Three treatment compounds have been shown to limit SM damage in the mouse ear vesicant model: dimercaprol, octyl homovanillamide, and indomethacin. Microarrays were used to determine gene expression profiles of biopsies taken from mouse ears after exposure to SM in the presence or absence of treatment compounds. Mouse ears were topically exposed to SM alone or were pretreated for 15 min with a treatment compound and then exposed to SM. Ear tissue was harvested 24 h after exposure for ear weight determination, the endpoint used to evaluate treatment compound efficacy. RNA extracted from the tissues was used to generate microarray probes for gene expression profiling of therapeutic responses. Principal component analysis of the gene expression data revealed partitioning of the samples based on treatment compound and SM exposure. Patterns of gene responses to the treatment compounds were indicative of exposure condition and were phenotypically anchored to ear weight. Pretreatment with indomethacin, the least effective treatment compound, produced ear weights close to those treated with SM alone. Ear weights from animals pretreated with dimercaprol or octyl homovanillamide were more closely associated with exposure to vehicle alone. Correlation coefficients between gene expression level and ear weight revealed genes involved in mediating responses to both SM exposure and treatment compounds. These data provide a basis for elucidating the mechanisms of response to SM and drug treatment and also provide a basis for developing strategies to accelerate development of effective SM medical countermeasures.


Assuntos
Substâncias para a Guerra Química/toxicidade , Orelha Externa/efeitos dos fármacos , Perfilação da Expressão Gênica , Expressão Gênica/efeitos dos fármacos , Gás de Mostarda/toxicidade , Administração Tópica , Animais , Orelha Externa/metabolismo , Orelha Externa/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Análise de Sequência com Séries de Oligonucleotídeos , Tamanho do Órgão/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Chem Res Toxicol ; 18(11): 1654-60, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16300373

RESUMO

Carbonyl chloride (phosgene) is a toxic industrial compound widely used in industry for the production of synthetic products, such as polyfoam rubber, plastics, and dyes. Exposure to phosgene results in a latent (1-24 h), potentially life-threatening pulmonary edema and irreversible acute lung injury. A genomic approach was utilized to investigate the molecular mechanism of phosgene-induced lung injury. CD-1 male mice were exposed whole body to either air or a concentration x time amount of 32 mg/m3 (8 ppm) phosgene for 20 min (640 mg x min/m3). Lung tissue was collected from air- or phosgene-exposed mice at 0.5, 1, 4, 8, 12, 24, 48, and 72 h postexposure. RNA was extracted from the lung and used as starting material for the probing of oligonucleotide microarrays to determine changes in gene expression following phosgene exposure. The data were analyzed using principal component analysis to determine the greatest sources of data variability. A three-way analysis of variance based on exposure, time, and sample was performed to identify the genes most significantly changed as a result of phosgene exposure. These genes were rank ordered by p values and categorized based on molecular function and biological process. Some of the most significant changes in gene expression reflect changes in glutathione synthesis and redox regulation of the cell, including upregulation of glutathione S-transferase alpha-2, glutathione peroxidase 2, and glutamate-cysteine ligase, catalytic subunit (also known as gamma-glutamyl cysteine synthetase). This is in agreement with previous observations describing changes in redox enzyme activity after phosgene exposure. We are also investigating other pathways that are responsive to phosgene exposure to identify mechanisms of toxicity and potential therapeutic targets.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Fosgênio/toxicidade , Administração por Inalação , Animais , Líquido da Lavagem Broncoalveolar/química , Glutationa/metabolismo , Glutationa Redutase/genética , Glutationa Transferase/genética , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Análise de Sequência com Séries de Oligonucleotídeos , Fosgênio/administração & dosagem , Reação em Cadeia da Polimerase
19.
Toxicol Sci ; 87(1): 306-14, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15976184

RESUMO

Gene expression profiling is an important tool in the development of medical countermeasures against chemical warfare agents (CWAs). Non-human primates (NHPs), specifically the rhesus macaque (Macaca mulatta), the cynomologus macaque (Macaca fascicularis), and the African green monkey (Chlorocebus aethiops), are vital models in the development of CWA prophylactics, therapeutics, and diagnostics. However, gene expression profiling of these NHPs is complicated by the fact their genomes are not completely sequenced, and that no commercially available oligonucleotide microarrays (genechips) exist. We, therefore, sought to determine whether gene expression profiling of NHPs could be performed using human genechips. Whole blood RNA was isolated from each species and used to generate genechip probes. Hybridization of the NHP samples to human genechips (Affymetrix Human U133 Plus 2.0) resulted in comparable numbers of transcripts detected compared with human samples. Statistical analysis revealed intraspecies reproducibility of genechip quality control metrics; interspecies comparison between NHPs and humans showed little significant difference in the quality and reproducibility of data generated using human genechips. Expression profiles of each species were compared using principal component analysis (PCA) and hierarchical clustering to determine the similarity of the expression profiles within and across the species. The cynomologus group showed the least intraspecies variability, and the human group showed the greatest intraspecies variability. Intraspecies comparison of the expression profiles identified probe sets that were reproducibly detected within each species. Each NHP species was found to be dissimilar to humans; the cynomologus group was the most dissimilar. Interspecies comparison of the expression profiles revealed probe sets that were reproducibly detected in all species examined. These results show that human genechips can be used for expression profiling of NHP samples and provide a foundation for the development of tools for comparing human and NHP gene expression profiles.


Assuntos
Perfilação da Expressão Gênica , RNA Mensageiro/sangue , Animais , Chlorocebus aethiops , Feminino , Humanos , Macaca fascicularis , Macaca mulatta , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Sondas RNA , Especificidade da Espécie
20.
Chem Res Toxicol ; 18(1): 28-34, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15651846

RESUMO

Bis-(2-chloroethyl) sulfide (sulfur mustard, SM) is a carcinogenic alkylating agent that has been utilized as a chemical warfare agent. To understand the mechanism of SM-induced lung injury, we analyzed global changes in gene expression in a rat lung SM exposure model. Rats were injected in the femoral vein with liquid SM, which circulates directly to the pulmonary vein and then to the lung. Rats were exposed to 1, 3, or 6 mg/kg of SM, and lungs were harvested at 0.5, 1, 3, 6, and 24 h postinjection. Three biological replicates were used for each time point and dose tested. RNA was extracted from the lungs and used as the starting material for the probing of replicate oligonucleotide microarrays. The gene expression data were analyzed using principal component analysis and two-way analysis of variance to identify the genes most significantly changed across time and dose. These genes were ranked by p value and categorized based on molecular function and biological process. Computer-based data mining algorithms revealed several biological processes affected by SM exposure, including protein catabolism, apoptosis, and glycolysis. Several genes that are significantly upregulated in a dose-dependent fashion have been reported as p53 responsive genes, suggesting that cell cycle regulation and p53 activation are involved in the response to SM exposure in the lung. Thus, SM exposure induces transcriptional changes that reveal the cellular response to this potent alkylating agent.


Assuntos
Substâncias para a Guerra Química/toxicidade , Perfilação da Expressão Gênica , Genômica , Pulmão/efeitos dos fármacos , Gás de Mostarda/análogos & derivados , Gás de Mostarda/toxicidade , Animais , Relação Dose-Resposta a Droga , Genes cdc , Genes p53 , Injeções Intravenosas , Pulmão/metabolismo , Masculino , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...