Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 565
Filter
1.
Drug Test Anal ; 14(1): 80-91, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34397154

ABSTRACT

For the verification of exposure to the banned blister agent sulfur mustard (SM) and the better understanding of its pathophysiology, protein adducts formed with endogenous proteins represent an important field of toxicological research. SM and its analogue 2-chloroethyl ethyl sulfide (CEES) are well known to alkylate nucleophilic amino acid side chains, for example, free-thiol groups of cysteine residues. The specific two-dimensional thiol difference gel electrophoresis (2D-thiol-DIGE) technique making use of maleimide dyes allows the staining of free cysteine residues in proteins. As a consequence of alkylation by, for example, SM or CEES, this staining intensity is reduced. 2D-thiol-DIGE analysis of human plasma incubated with CEES and subsequent matrix-assisted laser desorption/ionization time-of-flight (tandem) mass-spectrometry, MALDI-TOF MS(/MS), revealed transthyretin (TTR) as a target of alkylating agents. TTR was extracted from SM-treated plasma by immunomagnetic separation (IMS) and analyzed after tryptic cleavage by microbore liquid chromatography-electrospray ionization high-resolution tandem-mass spectrometry (µLC-ESI MS/HR MS). It was found that the Cys10 -residue of TTR present in the hexapeptide C(-HETE)PLMVK was alkylated by the hydroxyethylthioethyl (HETE)-moiety, which is characteristic for SM exposure. It was shown that alkylated TTR is stable in plasma in vitro at 37°C for at least 14 days. In addition, C(-HETE)PLMVK can be selectively detected, is stable in the autosampler over 24 h, and shows linearity in a broad concentration range from 15.63 µM to 2 mM SM in plasma in vitro. Accordingly, TTR might represent a complementary protein marker molecule for the verification of SM exposure.


Subject(s)
Chemical Warfare Agents/analysis , Mustard Gas/analogs & derivatives , Prealbumin/metabolism , Alkylation , Biomarkers/metabolism , Chemical Warfare Agents/poisoning , Chromatography, Liquid/methods , Electrophoresis/methods , Humans , Mustard Gas/analysis , Mustard Gas/poisoning , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Time Factors
2.
Toxicology ; 463: 152967, 2021 11.
Article in English | MEDLINE | ID: mdl-34619302

ABSTRACT

Blister agents damage the skin, eyes, mucous membranes and subcutaneous tissues. Other toxic effects may occur after absorption. The response of the Scientific Advisory Board (SAB) of the Organisation for the Prohibition of Chemical Weapons (OPCW) to a request from the OPCW Director-General in 2013 on the status of medical countermeasures and treatments to blister agents is updated through the incorporation of the latest information. The physical and toxicological properties of sulfur mustard and clinical effects and treatments are summarised. The information should assist medics and emergency responders who may be unfamiliar with the toxidrome of sulfur mustard and its treatment.


Subject(s)
Chemical Warfare Agents/poisoning , Mustard Gas/poisoning , Animals , Humans , Medical Countermeasures
3.
Anal Bioanal Chem ; 413(28): 6973-6985, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34549323

ABSTRACT

Chemical warfare agents continue to pose a real threat to humanity, despite their prohibition under the Chemical Weapons Convention. Sarin is one of the most toxic and lethal representatives of nerve agents. The methodology for the targeted analysis of known sarin metabolites has reached great heights, but little attention has been paid to the untargeted analysis of biological samples of victims exposed to this deadly poisonous substance. At present, the development of computational and statistical methods of analysis offers great opportunities for finding new metabolites or understanding the mechanisms of action or effect of toxic substances on the organism. This study presents the targeted LC-MS/MS determination of methylphosphonic acid and isopropyl methylphosphonic acid in the urine of rats exposed to a non-lethal dose of sarin, as well as the untarget urine analysis by LC-HRMS. Targeted analysis of polar acidic sarin metabolites was performed on a mixed-mode reversed-phase anion-exchange column, and untargeted analysis on a conventional reversed-phase C18 column. Isopropyl methylphosphonic acid was detected and quantified within 5 days after subcutaneous injection of sarin at a dose of 1/4 LD50. A combination of generalized additive mixed models and dose-response analysis with database searches using accurate mass of precursor ions and corresponding MS/MS spectra enabled us to propose new six potential biomarkers of biological response to exposure. The results confirm the well-known fact that sarin poisoning has a significant impact on the victims' metabolome, with inhibition of acetylcholinesterase being just the first step and trigger of the complex toxicodynamic response.


Subject(s)
Chemical Warfare Agents/analysis , Chemical Warfare Agents/poisoning , Chromatography, Liquid/methods , Sarin/poisoning , Sarin/urine , Tandem Mass Spectrometry/methods , Animals , Biomarkers/urine , Chemical Warfare Agents/standards , Limit of Detection , Male , Metabolomics/methods , Rats , Reference Standards , Reproducibility of Results , Sarin/standards
4.
Anal Bioanal Chem ; 413(19): 4907-4916, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34215915

ABSTRACT

Sulfur mustard (SM) is a banned chemical warfare agent recently used in the Syrian Arab Republic conflict causing erythema and blisters characterized by complicated and delayed wound healing. For medical and legal reasons, the proof of exposure to SM is of high toxicological and forensic relevance. SM reacts with endogenous human serum albumin (HSA adducts) alkylating the thiol group of the cysteine residue C34, thus causing the addition of the hydroxyethylthioethyl (HETE) moiety. Following proteolysis with pronase, the biomarker dipeptide C(-HETE)P is produced. To expand the possibilities for verification of exposure, we herein introduce a novel biomarker produced from that alkylated dipeptide by derivatization with propionic anhydride inducing the selective propionylation of the N-terminus yielding PA-C(-HETE)P. Quantitative derivatization is carried out at room temperature in aqueous buffer within 10 s. The biomarker was found to be stable in the autosampler at 15 °C for at least 24 h, thus documenting its suitability even for larger sets of samples. Selective and sensitive detection is done by micro liquid chromatography-electrospray ionization tandem-mass spectrometry (µLC-ESI MS/MS) operating in the selected reaction monitoring (SRM) mode detecting product ions of the single protonated PA-C(-HETE)P (m/z 379.1) at m/z 116.1, m/z 137.0, and m/z 105.0. The lower limit of detection corresponds to 32 nM SM in plasma in vitro and the limit of identification to 160 nM. The applicability to real exposure scenarios was proven by analyzing samples from the Middle East confirming poisoning with SM.


Subject(s)
Albumins/chemistry , Anhydrides/chemistry , Chemical Warfare Agents/poisoning , Dipeptides/chemistry , Mustard Gas/poisoning , Propionates/chemistry , Alkylation , Biomarkers , Humans
5.
Drug Test Anal ; 13(9): 1593-1602, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34145783

ABSTRACT

Sulfur mustard (SM, bis[2-chloroethyl]-sulfide) is a banned chemical warfare agent that was frequently used in recent years and led to numerous poisoned victims who developed painful erythema and blisters. Post-exposure analysis of SM incorporation can be performed by the detection of human serum albumin (HSA)-derived peptides. HSA alkylated by SM contains a hydroxyethylthioethyl (HETE)-moiety bound to the cysteine residue C34 yielding the dipeptide biomarker C(-HETE)P after pronase-catalyzed proteolysis. We herein present a novel procedure for the selective precolumn nicotinylation of its N-terminus using 1-nicotinoyloxy-succinimide. The reaction was carried out for 2 h at ambient temperature with a yield of 81%. The derivative NA-C(-HETE)P was analyzed by micro liquid chromatography-electrospray ionization tandem-mass spectrometry working in the selected reaction monitoring mode (µLC-ESI MS/MS SRM). The derivative was shown to be stable in the autosampler at 15°C for at least 24 h. The single protonated precursor ion (m/z 428.1) was subjected to collision-induced dissociation yielding product ions at m/z 116.1, m/z 137.0, and m/z 105.0 used for selective monitoring without any plasma-derived interferences. NA-C(-HETE)P showed a mass spectrometric response superior to the non-derivatized dipeptide thus yielding larger peak areas (factor 1.3 ± 0.2). The lower limit of identification corresponded to 80 nM SM spiked to plasma in vitro. The presented procedure was applied to real case plasma samples from 2015 collected in the Middle East confirming SM poisoning.


Subject(s)
Chemical Warfare Agents/analysis , Chromatography, Liquid/methods , Mustard Gas/analysis , Tandem Mass Spectrometry/methods , Biomarkers/analysis , Chemical Warfare Agents/chemistry , Chemical Warfare Agents/poisoning , Dipeptides/chemistry , Humans , Mustard Gas/chemistry , Mustard Gas/poisoning , Niacin/chemistry , Serum Albumin, Human/chemistry , Spectrometry, Mass, Electrospray Ionization
6.
Ann N Y Acad Sci ; 1480(1): 155-169, 2020 11.
Article in English | MEDLINE | ID: mdl-32885420

ABSTRACT

Lewisite and many other similar arsenicals are warfare vesicants developed and weaponized for use in World Wars I and II. These chemicals, when exposed to the skin and other epithelial tissues, cause rapid severe inflammation and systemic damage. Here, we show that topically applied arsenicals in a murine model produce significant acute kidney injury (AKI), as determined by an increase in the AKI biomarkers NGAL and KIM-1. An increase in reactive oxygen species and ER stress proteins, such as ATF4 and CHOP, correlated with the induction of these AKI biomarkers. Also, TUNEL staining of CHOP-positive renal tubular cells suggests CHOP mediates apoptosis in these cells. A systemic inflammatory response characterized by a significant elevation in inflammatory mediators, such as IL-6, IFN-α, and COX-2, in the kidney could be the underlying cause of AKI. The mechanism of arsenical-mediated inflammation involves activation of AMPK/Nrf2 signaling pathways, which regulate heme oxygenase-1 (HO-1). Indeed, HO-1 induction with cobalt protoporphyrin (CoPP) treatment in arsenical-treated HEK293 cells afforded cytoprotection by attenuating CHOP-associated apoptosis and cytokine mRNA levels. These results demonstrate that topical exposure to arsenicals causes AKI and that HO-1 activation may serve a protective role in this setting.


Subject(s)
Acute Kidney Injury , Apoptosis/drug effects , Arsenicals , Chemical Warfare Agents/poisoning , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Activating Transcription Factor 4/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Biomarkers/metabolism , Cyclooxygenase 2/metabolism , Enzyme Activation/drug effects , HEK293 Cells , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Interleukin-6/metabolism , Mice , Mice, Hairless , NF-E2-Related Factor 2/metabolism , Transcription Factor CHOP/metabolism
7.
Ann N Y Acad Sci ; 1480(1): 73-103, 2020 11.
Article in English | MEDLINE | ID: mdl-32892378

ABSTRACT

The lung is highly sensitive to chemical injuries caused by exposure to threat agents in industrial or transportation accidents, occupational exposures, or deliberate use as weapons of mass destruction (WMD). There are no antidotes for the majority of the chemical threat agents and toxic inhalation hazards despite their use as WMDs for more than a century. Among several putative targets, evidence for transient receptor potential (TRP) ion channels as mediators of injury by various inhalational chemical threat agents is emerging. TRP channels are expressed in the respiratory system and are essential for homeostasis. Among TRP channels, the body of literature supporting essential roles for TRPA1, TRPV1, and TRPV4 in pulmonary chemical injuries is abundant. TRP channels mediate their function through sensory neuronal and nonneuronal pathways. TRP channels play a crucial role in complex pulmonary pathophysiologic events including, but not limited to, increased intracellular calcium levels, signal transduction, recruitment of proinflammatory cells, neurogenic inflammatory pathways, cough reflex, hampered mucus clearance, disruption of the integrity of the epithelia, pulmonary edema, and fibrosis. In this review, we summarize the role of TRP channels in chemical threat agents-induced pulmonary injuries and how these channels may serve as medical countermeasure targets for broader indications.


Subject(s)
Chemical Warfare Agents/poisoning , Lung Injury , Lung , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Humans , Lung/metabolism , Lung/pathology , Lung/physiopathology , Lung Injury/chemically induced , Lung Injury/metabolism , Lung Injury/physiopathology , Lung Injury/therapy
8.
Ann N Y Acad Sci ; 1480(1): 146-154, 2020 11.
Article in English | MEDLINE | ID: mdl-32767459

ABSTRACT

Nitrogen mustard (NM) and sulfur mustard are cytotoxic alkylating agents that cause severe and progressive damage to the respiratory tract. Evidence indicates that macrophages play a key role in the acute inflammatory phase and the later resolution/profibrotic phase of the pathogenic response. These diverse roles are mediated by inflammatory macrophages broadly classified as M1 proinflammatory and M2 anti-inflammatory that sequentially accumulate in the lung in response to injury. The goal of the present study was to identify signaling mechanisms contributing to macrophage activation in response to mustards. To accomplish this, we used RNA sequencing to analyze the gene expression profiles of lung macrophages isolated 1 and 28 days after intratracheal exposure of rats to NM (0.125 mg/kg) or phosphate-buffered saline control. We identified 641 and 792 differentially expressed genes 1 and 28 days post-NM exposure, respectively. These genes are primarily involved in processes related to cell movement and are regulated by cytokines, including tumor necrosis factor-α, interferon-γ, and interleukin-1ß. Some of the most significantly enriched canonical pathways included STAT3 and NF-κB signaling. These cytokines and pathways may represent potential targets for therapeutic intervention to mitigate mustard-induced lung toxicity.


Subject(s)
Chemical Warfare Agents/poisoning , Gene Expression Regulation/drug effects , Lung Injury/metabolism , Macrophages, Alveolar/metabolism , Mechlorethamine/poisoning , RNA-Seq , Animals , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lung Injury/chemically induced , Lung Injury/pathology , Macrophages, Alveolar/pathology , Male , Rats , Rats, Wistar
9.
Inorg Chem ; 59(16): 11595-11605, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32799468

ABSTRACT

Due to the unpredictable nature of a battlefield environment, in the simultaneous degradation of sulfur mustard and nerve agents it is preferable to use just one decontaminant. Herein, the new composite HPVMo@MOF-808 (HPVMo = H5PV2Mo10O40) was deliberately synthesized via a simple impregnation method and thoroughly characterized. The results showed that the decontamination rate of the composites (30-40 mg) with optimal HPVMo loadings for HD (4 µL) and GD (4 µL) under ambient conditions was 97.2% (within 120 min) and 90.8% (within 30 min), respectively. Due to the combinational/synergistic effect of MOF-808 and encapsulated homogeneously dispersed HPVMo, the composite can very efficiently oxidize HD to nontoxic products in a single system, while retaining the inherent excellence of MOF-808 in hydrolytically degrading GD. The decontamination process was found to follow first-order reaction kinetics, and the rate constant and half-life of the composite for HD and GD were 0.0231 min-1, 30.13 min and 0.0795 min-1, 8.72 min, respectively. In addition, experimental results in guinea pigs and Kunming mice used as animal models showed that the composite provided effective skin protection against HD and GD, showing great potential for application in skin decontamination and protection.


Subject(s)
Chemical Warfare Agents/chemistry , Metal-Organic Frameworks/chemistry , Mustard Gas/chemistry , Soman/chemistry , Tungsten Compounds/chemistry , Animals , Chemical Warfare Agents/poisoning , Guinea Pigs , Mice , Poisoning/prevention & control
10.
Ann N Y Acad Sci ; 1480(1): 44-53, 2020 11.
Article in English | MEDLINE | ID: mdl-32808309

ABSTRACT

The use of chemical warfare agents (CWAs) in military conflicts and against civilians is a recurrent problem. Despite ongoing CWA research using in vitro or in vivo models, progress to elucidate mechanisms of toxicity and to develop effective therapies, decontamination procedures, and general countermeasures is still limited. Novel scientific approaches to address these questions are needed to expand perspectives on existing knowledge and gain new insights. To achieve this, the use of ex vivo techniques like precision-cut tissue slices (PCTSs) can be a valuable approach. Existing studies employing this economical and relatively easy to implement method show model suitability and comparability with the use of in vitro and in vivo models. In this article, we review research on CWAs in PCTSs to illustrate the advantages of the approach and to promote future applications.


Subject(s)
Chemical Warfare Agents/poisoning , Microdissection , Animals , Humans
11.
Ann N Y Acad Sci ; 1480(1): 104-115, 2020 11.
Article in English | MEDLINE | ID: mdl-32645215

ABSTRACT

The threat from deliberate or accidental exposure to halogen gases is increasing, as is their industrial applications and use as chemical warfare agents. Biomarkers that can identify halogen exposure, diagnose victims of exposure or predict injury severity, and enable appropriate treatment are lacking. We conducted these studies to determine and validate biomarkers of bromine (Br2 ) toxicity and correlate the symptoms and the extent of cardiopulmonary injuries. Unanesthetized rats were exposed to Br2 and monitored noninvasively for clinical scores and pulse oximetry. Animals were euthanized and grouped at various time intervals to assess brominated fatty acid (BFA) content in the plasma, lung, and heart using mass spectrometry. Bronchoalveolar lavage fluid (BALF) protein content was used to assess pulmonary injury. Cardiac troponin I (cTnI) was assessed in the plasma to evaluate cardiac injury. The blood, lung, and cardiac tissue BFA content significantly correlated with the clinical scores, tissue oxygenation, heart rate, and cardiopulmonary injury parameters. Total (free + esterified) bromostearic acid levels correlated with lung injury, as indicated by BALF protein content, and free bromostearic acid levels correlated with plasma cTnI levels. Thus, BFAs and cardiac injury biomarkers can identify Br2 exposure and predict the severity of organ damage.


Subject(s)
Bromine/poisoning , Chemical Warfare Agents/poisoning , Fatty Acids/blood , Hydrocarbons, Brominated/blood , Inhalation Exposure/adverse effects , Animals , Biomarkers/blood , Lung/metabolism , Lung/pathology , Male , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Troponin I/blood
12.
Ann N Y Acad Sci ; 1480(1): 5-13, 2020 11.
Article in English | MEDLINE | ID: mdl-32725637

ABSTRACT

Exposure to vesicants, including sulfur mustard and nitrogen mustard, causes damage to the epithelia of the respiratory tract and the lung. With time, this progresses to chronic disease, most notably, pulmonary fibrosis. The pathogenic process involves persistent inflammation and the release of cytotoxic oxidants, cytokines, chemokines, and profibrotic growth factors, which leads to the collapse of lung architecture, with fibrotic involution of the lung parenchyma. At present, there are no effective treatments available to combat this pathological process. Recently, much interest has focused on nutraceuticals, substances derived from plants, herbs, and fruits, that exert pleiotropic effects on inflammatory cells and parenchymal cells that may be useful in reducing fibrogenesis. Some promising results have been obtained with nutraceuticals in experimental animal models of inflammation-driven fibrosis. This review summarizes the current knowledge on the putative preventive/therapeutic efficacy of nutraceuticals in progressive pulmonary fibrosis, with a focus on their activity against inflammatory reactions and profibrotic cell differentiation.


Subject(s)
Chemical Warfare Agents/poisoning , Dietary Supplements , Irritants/poisoning , Mechlorethamine/poisoning , Mustard Gas/poisoning , Pulmonary Fibrosis , Animals , Disease Models, Animal , Humans , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/diet therapy , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology
13.
Ann N Y Acad Sci ; 1480(1): 14-29, 2020 11.
Article in English | MEDLINE | ID: mdl-32726497

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a highly morbid lung pathology induced by exposure to chemical warfare agents, including vesicants, phosgene, chlorine, and ricin. In this review, we describe the pathology associated with the development of ARDS in humans and experimental models of acute lung injury following animal exposure to these high-priority threat agents. Potential future approaches to disease-modifying treatment used in preclinical animal studies, including antioxidants, anti-inflammatories, biologics, and mesenchymal stem cells, are also described. As respiratory pathologies, including ARDS, are the major cause of morbidity and mortality following exposure to chemical threat agents, understanding mechanisms of disease pathogenesis is key to the development of efficacious therapeutics beyond the primary intervention principle, which remains mechanical ventilation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Chemical Warfare Agents/poisoning , Respiration, Artificial , Respiratory Distress Syndrome/therapy , Animals , Humans , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology
14.
Leg Med (Tokyo) ; 47: 101761, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32702607

ABSTRACT

On the battlefields of Syria, many innocent civilians have been killed or injured by sarin poisoning. In Malaysia in February 2017, a North Korean man was assassinated with VX at Kuala Lumpur International Airport. In the face of such threats, a more effective antidote against organophosphonate acetylcholinesterase (AChE) inhibitors is needed, one that can freely penetrate into the central nervous system (CNS) through the blood-brain barrier (BBB). In the 1995 Tokyo subway sarin attack, which produced more than 6,000 victims, 2-pyridinealdoxime methiodide was the most commonly used antidote in hospitals, but it was unable to prevent CNS damage and no other oximes have been approved for use in Japan. Ultimately, 12 people died, and many victims had severe neurological injuries or sequelae. Although more than 25 years have passed since the incident, progress has been slow in the development of a new antidote that can penetrate the BBB, restore AChE activity in the CNS, and definitely prevent brain injury. From the perspectives of countering terrorism and protecting innocent people from nerve agent attacks, the search for nerve agent antidotes should be accelerated with the goals of improving both survival and quality of life. This review gives an overview of a series of our studies on the development of a new antidote since the Tokyo subway sarin attack and emphasizes that there is unfortunately still no promising antidote for saving the CNS in Japan.


Subject(s)
Antidotes , Chemical Terrorism , Chemical Warfare Agents/poisoning , Cholinesterase Inhibitors/poisoning , Drug Development , Railroads , Sarin/poisoning , Blood-Brain Barrier/metabolism , Chemical Terrorism/prevention & control , Chemical Warfare Agents/metabolism , Cholinesterase Inhibitors/metabolism , Drug Development/trends , Humans , Pralidoxime Compounds , Sarin/metabolism , Time Factors , Tokyo
15.
PLoS One ; 15(6): e0234967, 2020.
Article in English | MEDLINE | ID: mdl-32574198

ABSTRACT

OBJECTIVES: The Tokyo subway sarin attack in 1995 was an unprecedented act of terrorism that killed 13 people and sickened more than 6,000. The long-term somatic and psychological effects on its victims remain unknown. METHODS: We conducted analyses on the self-rating questionnaire collected annually by the Recovery Support Center (RSC) during the period from 2000 to 2009. The RSC is the only organization that has large-scale follow-up data about sarin attack victims. The prevalence of self-reported symptoms was calculated over 10 years. We also evaluated the prevalence of posttraumatic stress response (PTSR), defined as a score ≥ 25 on the Japanese-language version of the Impact of Event Scale-Revised. The multivariate Poisson regression model was applied to estimate the risk ratios of age, gender, and year factor on the prevalence of PTSR. RESULTS: Subjects were 747 survivors (12% of the total) who responded to the annual questionnaire once or more during the study period. The prevalence of somatic symptoms, especially eye symptoms, was 60-80% and has not decreased. PTSR prevalence was 35.1%, and again there was no change with time. The multivariate Poisson regression model results revealed "old age" and "female" as independent risk factors, but the passage of time did not decrease the risk of PTSR. CONCLUSIONS: Although symptoms in most victims of the Tokyo subway sarin were transient, this large-scale follow-up data analysis revealed that survivors have been suffering from somatic and psychological long-term effects.


Subject(s)
Chemical Terrorism , Chemical Warfare Agents/poisoning , Miosis/epidemiology , Sarin/poisoning , Stress Disorders, Post-Traumatic/epidemiology , Survivors/statistics & numerical data , Adolescent , Adult , Aged , Female , Follow-Up Studies , Humans , Male , Middle Aged , Miosis/chemically induced , Prevalence , Railroads , Risk Factors , Self Report/statistics & numerical data , Stress Disorders, Post-Traumatic/psychology , Survivors/psychology , Tokyo/epidemiology , Young Adult
16.
Arch Iran Med ; 23(4Suppl1): S33-S37, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32349506

ABSTRACT

BACKGROUND: Limited studies have reported epidemiologic data on the impact of Iran-Iraq war. This study examines the war casualties for both combatants and civilians on Iranians at national level. METHODS: Databases of Veterans and Martyrs Affair Foundation (VMAF), Janbazan Medical and Engineering Research Center (JMERC) and Ministry of Health were used to collect the data. The prevalence of injuries for both civilians and combatants was presented. Casualties were studied based on conventional and unconventional weapons attacks (1980-2018), separately. RESULTS: The Iran-Iraq war led to 183623 lost lives, 554990 injured and 40240 captured. The mean length of captivity was 45.7 months (1 month-19 years) and 2.7% (n = 575) died in captivity. There were 1439180 war related injuries recorded in databanks, mostly affecting men (98.4%). About 1439180 injuries were recorded, most of them related to conventional weapons (938928 [65.24%]). Remaining artillery and mortar fragmentation in the body (39.5%, n = 371236), psychological disorders (15.9%, n = 228944), and exposure to chemical weapons (11%, n = 158817) were the most prevalent war-related injuries. CONCLUSION: Human casualties of the Iran-Iraq war on the Iranian side and the health care system are huge even after more than three decades.


Subject(s)
Chemical Warfare Agents/poisoning , Mental Disorders/epidemiology , War-Related Injuries/epidemiology , Warfare , Humans , Iran/epidemiology , Surveys and Questionnaires , Time Factors , War-Related Injuries/mortality , Weapons of Mass Destruction
17.
Ann N Y Acad Sci ; 1479(1): 210-222, 2020 11.
Article in English | MEDLINE | ID: mdl-32329907

ABSTRACT

Lewisite is a strong vesicating and chemical warfare agent. Because of the rapid transdermal absorption, cutaneous exposure to lewisite can also elicit severe systemic injury. Lewisite (2.5, 5.0, and 7.5 mg/kg) was applied to the skin of Ptch1+/- /SKH-1 mice and acute lung injury (ALI) was assessed after 24 hours. Arterial blood gas measurements showed hypercapnia and hypoxemia in the lewisite-exposed group. Histological evaluation of lung tissue revealed increased levels of proinflammatory neutrophils and a dose-dependent increase in structural changes indicative of injury. Increased inflammation was also confirmed by altered expression of cytokines, including increased IL-33, and a dose-dependent elevation of CXCL1, CXCL5, and GCSF was observed in the lung tissue. In the bronchoalveolar lavage fluid of lewisite-exposed animals, there was a significant increase in HMGB1, a damage-associated molecular pattern molecule, as well as elevated CXCL1 and CXCL5, which coincided with an influx of neutrophils to the lungs. Complete blood cell analysis revealed eosinophilia and altered neutrophil-lymphocyte ratios as a consequence of lewisite exposure. Mean platelet volume and RBC distribution width, which are predictors of lung injury, were also increased in the lewisite group. These data demonstrate that cutaneous lewisite exposure causes ALI and may contribute to mortality in exposed populations.


Subject(s)
Acute Lung Injury , Arsenicals , Chemical Warfare Agents/poisoning , Cytokines/metabolism , Lung , Neutrophil Infiltration/drug effects , Neutrophils , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage , Female , Leukocyte Count , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Hairless , Neutrophils/metabolism , Neutrophils/pathology , Platelet Count , Skin/metabolism , Skin/pathology
18.
Toxicol Appl Pharmacol ; 396: 114994, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32251685

ABSTRACT

Anticholinergic treatment is key for effective medical treatment of nerve agent exposure. Atropine is included at a 2 mg intramuscular dose in so-called autoinjectors designed for self- and buddy-aid. As patient cohorts are not available, predicting and evaluating the efficacy of medical countermeasures relies on animal models. The use of atropine as a muscarinic antagonist is based on efficacy achieved in studies in a variety of species. The dose of atropine administered varies considerably across these studies. This is a complicating factor in the prediction of efficacy in the human situation, largely because atropine dosing also influences therapeutic efficacy of oximes and anticonvulsants generally part of the treatment administered. To improve translation of efficacy of dosing regimens, including pharmacokinetics and physiology provide a promising approach. In the current study, pharmacokinetics and physiological parameters obtained using EEG and ECG were assessed in naïve rats and in sarin-exposed rats for two anticholinergic drugs, atropine and scopolamine. The aim was to find a predictive parameter for therapeutic efficacy. Scopolamine and atropine showed a similar bioavailability, but brain levels reached were much higher for scopolamine. Scopolamine exhibited a dose-dependent loss of beta power in naïve animals, whereas atropine did not show any such central effect. This effect was correlated with an enhanced anticonvulsant effect of scopolamine compared to atropine. These findings show that an approach including pharmacokinetics and physiology could contribute to improved dose scaling across species and assessing the therapeutic potential of similar anticholinergic and anticonvulsant drugs against nerve agent poisoning.


Subject(s)
Atropine/therapeutic use , Chemical Warfare Agents/poisoning , Sarin/poisoning , Scopolamine/therapeutic use , Animals , Atropine/blood , Atropine/pharmacokinetics , Atropine/pharmacology , Brain Chemistry/drug effects , Cholinergic Antagonists , Electrocardiography/drug effects , Electroencephalography/drug effects , Male , Mice , Rats, Wistar , Sarin/antagonists & inhibitors , Scopolamine/blood , Scopolamine/pharmacokinetics , Scopolamine/pharmacology , Telemetry/methods
19.
BMJ Mil Health ; 166(2): 99-102, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32086265

ABSTRACT

Recent uses of nerve agents underline the need of early diagnosis as trigger to react (initiating medical countermeasures, avoiding cross-contamination). As organophosphorus (OP) pesticide poisoning exerts the same pathomechanism, that is, inhibition of the pivotal enzyme acetylcholinesterase (AChE), a portable cholinesterase (ChE) test kit was applied in an emergency room for rapid diagnosis of OP poisoning. OP nerve agents or pesticides result in the inhibition of AChE. As AChE is also expressed on erythrocytes, patient samples are easily available. However, in most clinics only determination of plasma butyrylcholinesterase (BChE) is established which lacks a pathophysiological correlate, shows higher variability in the population and behaves different regarding inhibition by OP and reactivation by oximes. The ChE test kit helped to diagnose atypical cases of OP poisoning, for example, missing of typical muscarinic symptoms, and resulted in administration of pralidoxime, the oxime used in Serbia. The ChE test kit also allows an initial assessment whether an oxime therapy is successful. In one case report, AChE activity increased after oxime administration indicating therapeutic success whereas BChE activity did not. With only BChE at hand, this therapeutic effect would have been missed. As inhibition of AChE or BChE activity is determined, the CE-certified device is a global diagnostic tool for all ChE inhibitors including carbamates which might also be misused as chemical weapon. The ChE test kit is a helpful point-of-care device for the diagnosis of ChE inhibitor poisoning. Its small size and easy menu-driven use advocate procurement where nerve agent and OP pesticide exposure are possible.


Subject(s)
Chemical Warfare Agents/poisoning , Cholinesterase Inhibitors/poisoning , Medical Countermeasures , Nerve Agents/poisoning , Point-of-Care Testing , Early Diagnosis , Humans
20.
Arh Hig Rada Toksikol ; 71(4): 266-284, 2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33410774

ABSTRACT

Phosphylation of the pivotal enzyme acetylcholinesterase (AChE) by nerve agents (NAs) leads to irreversible inhibition of the enzyme and accumulation of neurotransmitter acetylcholine, which induces cholinergic crisis, that is, overstimulation of muscarinic and nicotinic membrane receptors in the central and peripheral nervous system. In severe cases, subsequent desensitisation of the receptors results in hypoxia, vasodepression, and respiratory arrest, followed by death. Prompt action is therefore critical to improve the chances of victim's survival and recovery. Standard therapy of NA poisoning generally involves administration of anticholinergic atropine and an oxime reactivator of phosphylated AChE. Anticholinesterase compounds or NA bioscavengers can also be applied to preserve native AChE from inhibition. With this review of 70 years of research we aim to present current and potential approaches to counteracting NA poisoning.


Subject(s)
Chemical Warfare Agents , Chemical Warfare , Cholinesterase Reactivators , Nerve Agents , Organophosphate Poisoning , Acetylcholinesterase , Chemical Warfare Agents/poisoning , Cholinesterase Inhibitors , Humans , Organophosphate Poisoning/drug therapy , Oximes
SELECTION OF CITATIONS
SEARCH DETAIL
...