Skip to main content
The Journal of Pharmacology and Experimental Therapeutics logoLink to The Journal of Pharmacology and Experimental Therapeutics
. 2024 Feb;388(2):560–567. doi: 10.1124/jpet.123.001822

Countermeasures against Pulmonary Threat Agents

Jacqui Marzec 1, Srikanth Nadadur 1,
PMCID: PMC10801713  PMID: 37863486

Abstract

Inhaled toxicants are used for diverse purposes, ranging from industrial applications such as agriculture, sanitation, and fumigation to crowd control and chemical warfare, and acute exposure can induce lasting respiratory complications. The intentional release of chemical warfare agents (CWAs) during World War I caused life-long damage for survivors, and CWA use is outlawed by international treaties. However, in the past two decades, chemical warfare use has surged in the Middle East and Eastern Europe, with a shift toward lung toxicants. The potential use of industrial and agricultural chemicals in rogue activities is a major concern as they are often stored and transported near populated areas, where intentional or accidental release can cause severe injuries and fatalities. Despite laws and regulatory agencies that regulate use, storage, transport, emissions, and disposal, inhalational exposures continue to cause lasting lung injury. Industrial irritants (e.g., ammonia) aggravate the upper respiratory tract, causing pneumonitis, bronchoconstriction, and dyspnea. Irritant gases (e.g., acrolein, chloropicrin) affect epithelial barrier integrity and cause tissue damage through reactive intermediates or by direct adduction of cysteine-rich proteins. Symptoms of CWAs (e.g., chlorine gas, phosgene, sulfur mustard) progress from airway obstruction and pulmonary edema to acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), which results in respiratory depression days later. Emergency treatment is limited to supportive care using bronchodilators to control airway constriction and rescue with mechanical ventilation to improve gas exchange. Complications from acute exposure can promote obstructive lung disease and/or pulmonary fibrosis, which require long-term clinical care.

SIGNIFICANCE STATEMENT

Inhaled chemical threats are of growing concern in both civilian and military settings, and there is an increased need to reduce acute lung injury and delayed clinical complications from exposures. This minireview highlights our current understanding of acute toxicity and pathophysiology of a select number of chemicals of concern. It discusses potential early-stage therapeutic development as well as challenges in developing countermeasures applicable for administration in mass casualty situations.

Introduction

Over two hundred chemicals of concern (CoC) have been identified by the US Department of Homeland Security as high-consequence public health threats, and one-fourth of these toxicants affect the lungs. Chemical warfare agents (CWAs) are high-priority health concerns with short exposure windows that cause immediate casualties and lung injury and can induce progressive lung disease. Industrial and agricultural chemicals (irritants) are of significant concern as accidental or intentional exposures affect civilian populations. Lung irritants cause reversible nonimmunologic reactions after direct contact with the nose, throat, and respiratory tract.

Lung tissue in the lower respiratory tract is composed of numerous alveoli that mediate gas exchange and is highly sensitive to inhaled toxicants due its extensive surface area. Toxicants are inhaled through absorption of chemical vapors, particulates, and incomplete combustion byproducts into the lungs. Clinical symptoms and severity depend on dose and proximity, water solubility, and toxicant size. Highly water-soluble (hydrophilic, e.g., ammonia) and larger compounds (>10 μm) mainly affect the upper airways, stimulating the trigeminal nerves of the nasal passages, with rapid symptom onset that presents as burning, irritation, and cough. Those of intermediate size and solubility (5–10 μm, e.g., chlorine gas) cause greater parenchymal injury, and small, poorly water-soluble (hydrophobic) toxicants <5 μm (e.g., phosgene gas) penetrate to the respiratory bronchioles and alveoli of the lower respiratory tract to cause detrimental lung injury (Parkes, 1994; Muskat, 2008; Nelson and Hoffman, 2014). Oxidant gases (e.g., chlorine, phosgene, ammonia) react initially with antioxidants in the epithelial lining fluid, and during acute, high-dose toxicant exposures, this first line of defense can be overwhelmed (Addis et al., 2021). Understudied chemicals and some of the acute toxicities that lead to lung complications are described in Table 1.

TABLE 1.

Acute toxicity of pulmonary chemicals of concern

Chemical Ammonia Acrolein Chloropicrin Chlorine Phosgene
Formula NH3 C3H4O CCl3NO2 Cl2 COCl2
Deposition URT URT URT URT/LRT LRT
Industrial/agricultural use Refrigerant
Fertilizer
Biocide
Livestock feed
Disinfectant
Soil fumigant
Disinfectant
Sanitation
Plastics
Paper
Chemicals
Adhesives
Plastics
Reactive by-products NH4OH C3H4O2, COx COCl2, Cl2, NOx, (intermediates: HOCl, CH3NO2) HCl, HOCl HCl
Pathophysiology Irritant, exothermically reacts with water, damages tissue Irritant, strong electrophile, damages tissue Irritant, induces ER stress, mutagen Reacts with epithelial lining, lyses cells, causes fibrin deposition Hydrolyses with mucus layer, acylates biomolecules, depletes surfactant
Acute toxicity Bronchiectasis, BO, COPD, ILD, respiratory failure Bronchial obstruction, LRT necrosis Bronchitis, pulmonary edema, respiratory failure ALI/ARDS, RADS, pulmonary fibrosis, respiratory failure Pulmonary edema, hypoxemia, ALI/ARDS, respiratory failure

AKG, α-ketoglutarate; C3H4O, acrolein; CCl3NO2, chloropicrin; CH3NO2, nitromethane; Cl2, chlorine gas; COCl2, phosgene; COx, carbon oxide; ER, endoplasmic reticulum; HCl, hydrogen chloride; HOCl, hypochlorous acid; LRT, lower respiratory tract; NH3, anhydrous ammonia; NH4OH, ammonium hydroxide; NOx, nitrogen oxide; URT, upper respiratory tract.

Mechanisms of early lung injury are shared among irritants and CWAs, whereas the development and progression of delayed lung complications are toxicant specific. CWAs cause respiratory distress, rapid airway constriction, and pulmonary edema that can lead to respiratory failure. Similarly, CWAs can cause bronchoconstriction and airway smooth muscle activation that accelerates cardiopulmonary injury. Exposure induces two-phase effects, which initially present as pneumonitis and bronchitis, with latent symptoms of pulmonary edema, bronchiolitis, and acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) days to weeks later (Muskat, 2008). High-dose irritants initially cause persistent rhinosinusitis, airway hyperresponsiveness (AHR), and accelerated lung function decline. Complications from exposure may proceed from airway obstruction to persistent reactive airways disease (RADS) and, at high doses, from pulmonary edema to ALI/ARDS (Leduc et al., 1992; Lu et al., 2017; Pesonen and Vähäkangas, 2020).

ARDS develops as a cascading consequence of ALI and often progresses to respiratory failure. Current research is aimed at studying ALI mechanisms and intervention strategies that can be applied before ARDS develops. Symptoms of ALI and ARDS overlap, with clinical distinction based on the degree of hypoxemia present [PaO2/FiO2 ratio, or oxygenation index, is the ratio of partial pressure of oxygen (PaO2) to fraction of inspired oxygen (FiO2): ALI ≤ 300 mm Hg; ARDS ≤ 200 mm Hg]. The term ALI/ARDS is now widely used (Butt et al., 2016), and diagnosis pairs deteriorating oxygenation levels with evidence of bilateral lung infiltrates on chest X-ray. ALI/ARDS represents a continuum of physiologic changes that disrupt the integrity of the endothelial and epithelial barriers of the lung, causing diffuse alveolar damage and increased lung permeability that impairs gas exchange. It is characterized by three phases: exudative (days 1–6), proliferative (days 7–14), and fibrotic (>14 days), and diverse chemicals included on the CoC list may induce phenotypically diverse effects along the trajectory of lung injury (Radbel et al., 2020).

The exudative phase is indicated by inflammation, surfactant deficiency, and tissue factor dysfunction, which contribute to intravascular and intra-alveolar coagulation and hypoxemia. During this phase, lymphatic drainage from the lung cannot remove fluid that filters from the lung vasculature, and potentially fatal pulmonary edema develops (Lindsay, 2011). The proliferative phase is largely mediated by proresolution macrophages, which recruit specialized proresolving mediators to initiate resolution. In this phase, alveolar type II cells proliferate due to Wnt/β-catenin signaling and differentiate into alveolar type I cells to repair the damaged epithelial barrier (Radbel et al., 2020). Impaired resolution prompts the debilitating fibrotic phase, which is characterized by collagen deposition and persistent thickening of the alveolar interstitium that impedes gas exchange. This promotes obstructive lung diseases [e.g., bronchiolitis obliterans (BO), chronic obstructive pulmonary disease (COPD)] that require long-term clinical management. For comprehensive reviews of ALI/ARDS, see Radbel et al. (2020) and Butt et al. (2016).

In CWA-induced lung injury, the additional extravascular coagulation and fibrin formation in the alveolar compartment may promote the development of pulmonary fibrosis. Early symptoms include unproductive cough, weight loss, and fatigue, largely due to hypoxia (Savin et al., 2022). Diagnosis is based on lung function measures, and blood and imaging tests are used to rule out other lung-related illnesses. Pulmonary fibrosis is a heterogeneous disease with distinct tissue pathology, encompassing multiple chronic lung outcomes, including interstitial lung disease (ILD) and idiopathic pulmonary fibrosis. Left unresolved, pulmonary fibrosis progressively destroys the alveolar structure of the lung and can lead to respiratory failure. For a comprehensive review of fibrosis, see Savin et al. (2022).

The National Institutes of Health Chemical Countermeasures Research Program is focused on toxicant-induced ALI/ARDS and delayed effects of lung injury. Its mission is to understand molecular and cellular perturbations and identify common mechanisms of lung injury to develop safe and effective medical countermeasures that reduce acute mortality and complications of chronic lung disease.

Recent Advances in Inhalational Research

Irritants: Ammonia.

Anhydrous ammonia (NH3) is a compressed liquid used as an agricultural fertilizer and refrigerant and is widely transported through highly populated areas. Upon release, ammonia reacts with water to form ammonium hydroxide, which irritates the mucosa and, at high concentrations, causes severe upper airways irritation and respiratory failure within 2–5 minutes of exposure (Saeed et al., 2018). Tissue damage occurs through exothermic reactions with body tissues, causing alkali skin burns and liquefactive necrosis (Pangeni et al., 2022). In some cases, survivors may develop complications such as bronchiectasis, AHR, BO, COPD, ILD, and end-stage disease that requires lung transplantation. Diagnosis is based on exposure and by physical examination and body-system focused laboratory tests. α-Ketoglutarate (AKG) and fluticasone propionate (Flonase) have been shown to reduce injury in animal and cell culture models (Ali et al., 2012, 2013); however, there are currently no antidotes or tests for the extent of toxicity.

Recent animal studies have established an LD50 value and mechanisms by which ammonia induces ALI/ARDS and systemic changes (Perkins et al., 2017; Elfsmark et al., 2019). Intratracheal ammonia instillation causes severe lung injury and increased vascular resistance in mice, with persistent respiratory acidosis and alveolar damage at day 1 and interstitial hemorrhage and mortality within 7 days of exposure (Elfsmark et al., 2022). Early activation of surfactant protein D (SP-D), which regulates pulmonary innate immunity and defense against xenobiotics (Madsen et al., 2000), and sustained activation of coagulation factors such as plasminogen activator inhibitor 1 (PAI-1) and fibrinogen are observed. Ammonia induces biphasic injury, with an acute inflammatory phase that deteriorates to obstructive lung disease within days. Comparable symptoms of dyspnea, acute neutrophilic airway inflammation, and pulmonary edema are seen in ammonia-induced ALI/ARDS. Case reports show that some patients develop delayed lung injuries without initial upper respiratory tract obstruction (Close et al., 1980) and persistent RADS after a single high-dose exposure (Brooks et al., 1985; Brooks, 2008).

Irritant Gases: Acrolein and Chloropicrin.

Acrolein (C3H4O) and chloropicrin (CCl3NO2) were deployed as CWAs in World War I and are now used for industrial and agricultural applications. Acrolein is a volatile aldehyde formed during incomplete combustion of fossil fuels, and large quantities are synthesized for industrial use as a biocide (Conklin, 2016). It reacts with the respiratory lining fluid and cellular macromolecules, altering gene regulation and mucociliary transport and compromising endothelial barrier integrity. Acrolein induces AHR and mucus hypersecretion, which obstructs the lumen and leads to airspace enlargement with loss of lung elasticity. At high concentrations, it promotes epithelial cell hyperplasia with lower respiratory tract necrosis that induces pulmonary edema and ALI/ARDS if not resolved (Xiong et al., 2018). Acrolein-exposed lungs have histologic features of squamous cell differentiation and dysfunctional tissue remodeling (Beauchamp et al., 1985).

Mechanistic studies show that acrolein affects energy balance, with profound effects on lipid and mitochondrial metabolism (Fabisiak et al., 2011; Agarwal et al., 2013; Snow et al., 2017). RNA-sequencing analyses indicate sex-dependent differences in injury severity and increased neutrophil extracellular trap formation (Bein et al., 2021). In vitro studies using acrolein vapor suggest that interleukin (IL)-1β, IL-22, and IL-17 pathway genes play a central role in toxicity (Johanson et al., 2020). Potential therapies include chemosensory transient receptor potential (TRP)-A1 antagonists (Conklin, 2016), antioxidants (Hochman et al., 2014), and targeting claudin 5 (CLDN5) to preserve endothelial barrier integrity (Jang et al., 2011). Investigators show increased perivascular edema and reduced CLDN5 expression in acrolein-sensitive mice (Jang et al., 2011), and CLDN5 is being studied in other models of chemically induced ALI/ARDS (Geng et al., 2018, 2020).

Chloropicrin is a halogenated fumigant and disinfectant that causes respiratory complications when inhaled. Decomposition releases toxic gases such as phosgene, chlorine, and nitrogen oxides, and it can dehalogenate in aqueous systems to form nitromethane. Although initial exposure irritates the lungs, at higher concentrations chloropicrin induces vomiting and breathing difficulties and airway damage that triggers bronchitis and pulmonary edema. This can progress to respiratory failure or, in some instances, life-long complications such as COPD. Histologic lesions including ulceration and necrosis are seen in animal models, and chloropicrin is toxic to other organs (Pesonen et al., 2017).

Mechanisms of chloropicrin-induced lung damage are not well understood, and animal studies have mainly focused on ocular injury. Chloropicrin-exposed primary human bronchial epithelial cells show elevated levels of extracellular signal-regulated kinase 1 and 2 that induce ER stress (Pesonen et al., 2015) and increase cytoplasmic vacuolization (Pesonen et al., 2017). Human bronchial epithelial cells show deformed cytosketal ultrastructure with weakened cell attachments and mitochondrial dysfunction. This suggests that chloropicrin targets β-tubulin and interferes with the tubulin network, inducing apoptosis. Chloropicrin is mutagenetic and modifies sulfhydryl groups on cysteines and enzymes important for energy metabolism (Sparks et al., 2000). There are no specific biomarkers of exposure, and therapy is limited to supportive care and the use of antioxidants. N-acetylcysteine acts as a thiol-reducing agent to scavenge reactive oxygen species/reactive nitrogen species and stimulate glutathione synthesis and prevents chloropicrin-induced cytotoxicity and vacuolization in cell culture models (Pesonen et al., 2014).

Chemical Warfare Agents: Chlorine, Phosgene, and Sulfur Mustard.

Chlorine gas (Cl2) is used industrially for sanitation and water purification and is also used as a CWA. It reacts with the mucosal lining to form hydrochloric and hypochlorous acids and the oxidative byproduct hypochlorite, which mediates cytotoxic effects in the lungs (Achanta and Jordt, 2021). Chlorine initially causes symptoms of bronchospasm and chest pain and, at high concentrations, destroys lung structure, inducing hemorrhage, pulmonary edema, and respiratory collapse within 1 hour of exposure. Survivors of acute exposure may develop symptoms of pulmonary fibrosis and RADS, with cardiovascular complications that last for years (Zaky et al., 2015; Carlisle et al., 2016).

Animal models of chlorine injury show histologic features of epithelial desquamation, leukocyte infiltration, atelectasis, and necrosis (Balakrishna et al., 2014). Biologic changes observed include protease activation, coagulopathies, compromised barrier integrity, and fibrin deposition (Mo et al., 2015; Musah et al., 2017) as well as increased susceptibility to infection (Gessner et al., 2013; Song et al., 2015). Potential therapeutics to counteract chlorine toxicity include antioxidants, corticosteroids, and combined therapies. TRP inhibitors, nitrite administration, heparin, and autophagy activators also show limited benefits (Achanta and Jordt, 2021). The recent identification of chlorinated lipids as biomarkers of exposure provides a potential diagnostic measure of chlorine toxicity (Spickett, 2007; Ford et al., 2016).

Phosgene (COCl2; CG) is a toxic, colorless gas used for chemical synthesis and as a CWA. CG is hydrophobic and slowly reacts with water to form carbon dioxide and hydrochloric acid, which directly damages the respiratory tract (Pauluhn, 2021; Cao et al., 2022). CG bypasses chemosensory perception through limited retention in the upper airways, and reflex bradypnea is largely absent (Pauluhn, 2021). CG collects in the lower respiratory tract, and noncardiogenic pulmonary edema often goes unnoticed, with development of secondary hypoxemia and deterioration of the blood-gas barrier (Pauluhn et al., 2007). Phosgene-induced ALI is preceded by persistent respiratory depression and cardiogenic pulmonary edema within 15–20 hours of exposure (Pauluhn, 2021). This occult or asymptomatic period is not evident on clinical examination. Fluid shifts from the systemic to the pulmonary circulation induce hypovolemia and hypotension that parallel pulmonary edema onset (Pauluhn, 2021). In late-stage phosgene-induced acute lung injury, survivors often develop fibrosis and chronic obstructive lung disease (Cao et al., 2022).

A consistent LC50 in animal models has been difficult to determine (Hobson et al., 2021), and toxic load depends linearly on dose × exposure duration, such that acute high dose is no more toxic than chronic low levels of exposure. CG is electrophilic and acylates nucleophilic biomolecules (Holmes et al., 2016), inducing pulmonary edema as the blood-gas barrier is permeabilized. CG undergoes hemolytic cleavage to form highly reactive carbamoyl chloride derivatives (Arroyo et al., 1993; Holmes et al., 2016) that further alter surfactant levels. Increased nitrosative stress affects neuronal cells that innervate the lungs as well as epithelial, endothelial, and blood cells. Ongoing debate on the role of inflammation in vasopermeability and pulmonary edema (Russell et al., 2006; Chen et al., 2013; Holmes et al., 2016) has impacted agreement on which biomarkers and therapeutics should be explored.

Anti-inflammatories show little benefit for CG-induced injury, and TRP antagonists are limited to reducing neurogenic inflammation (Pauluhn, 2021). Administration of antioxidants such as N-acetylcysteine increase glutathione availability (Ji et al., 2010), and melatonin attenuates CG-induced lung injury through Wnt/β-catenin signaling (Zhang et al., 2017). Strategies using ulinastatin and NOS-2 inhibitors show promise in small animal models (Shen et al., 2014; Filipczak et al., 2015; Zhang et al., 2017), but early treatment during the asymptomatic phase is critical to minimize the risk of cardiogenic edema (Lu et al., 2021). Mesenchymal stem cells that target miRNAs (Xu et al., 2019; Qu et al., 2020; Jiang et al., 2021) or overexpress angiogenic (Shao et al., 2018) or heat shock proteins (Jin et al., 2020) are shown to counteract toxicity in vitro, and administration of cardioprotective Fv-HSP72 may ameliorate injury (Hobson et al., 2021).

Sulfur mustard (C4H8Cl2S; SM) is a toxic vesicant primarily used as a CWA, with short and long-term lung injury effects. It causes blistering and erythema of the respiratory tract (Schmidt et al., 2018) that destroys bronchial tissue and obstructs the airway minutes to hours after exposure. At high doses, this is followed by hemorrhagic pulmonary edema, secondary pneumonia, and respiratory failure 24 hours to 1 week later (Ghanei et al., 2005). Acute exposures can induce pathogenic lesions and fibrosis that promotes BO and/or ILD decades later (Ghanei et al., 2008). In addition, acute SM exposure causes systemic injury and increases cancer risk (Razavi et al., 2016).

Although the lungs are a major target of exposure, SM is highly lipophilic and enters the body through multiple routes. It eliminates chloride ions through intramolecular substitution to form cyclic sulfonium ions. These reactive intermediates alkylate DNA and other nuclear components, inducing genotoxicity and cytotoxic damage that suppresses the immune system and inducible nitric oxide synthase (iNOS) signaling (Tahmasbpour et al., 2019). Inhaled SM activates phagocytic leukocytes and inflammatory mediators that alter serum cytokines, C-reactive protein (CRP), and soluble proapoptotic fasciclin 1 (Fas1) expression. Vascular leakage and endothelial permeability are reported in animal models (Calvet et al., 1994; Rancourt et al., 2013; Malaviya et al., 2020) as well as fibrin cast formation that occludes the airways (White et al., 2016). Increased fibrinogen suppresses surfactant and can prolong macrophage activation and inflammation (Wygrecka et al., 2008).

There are currently no approved therapies for SM inhalation; however, combined administration of vitamin E and L-thiocitrulline (L-TC) helps replenish nitric oxide (NO) and manage respiratory symptoms. Therapies being explored include anti-inflammatories, antioxidants, and protease inhibitors, and some benefits from natural products are shown (Boskabady and Farhadi, 2008; Hossein et al., 2008). Studies show restoration of surfactant may protect against SM-induced lung injury (van Helden et al., 2004) and that SP-D, a nonspecific alveolar injury marker, may serve as an indicator of early respiratory disease (Starosta and Griese, 2006; Sorensen, 2018; Malaviya et al., 2020).

Early Mechanisms of Toxicant-Induced Lung Injury

Although ALI/ARDS progression and delayed respiratory complications are toxicant specific, toxicant-induced inflammation and injury share overlapping protective mechanisms to limit lung damage (Fig. 1). Acute exposure induces nitrosative stress and endothelial damage that occurs concurrently with inflammation (Radbel et al., 2020; Elfsmark et al., 2022). Reactive intermediates (e.g., reactive oxygen species/reactive nitrogen species) induce inflammatory cytokines/chemokines that activate iNOS, which generates excess NO that can magnify oxidative stress (Sun et al., 2010). Peripheral lung inflammation stimulates histamine release and endothelial NO production, which amplifies the inflammatory cascade (Branco et al., 2018; Radbel et al., 2020). In CWA models, the overwhelming magnitude of nitrosative stress depletes NO stores from alveolar macrophages (Malaviya et al., 2023) and aggravates histamine intolerance (Branco et al., 2018). It also reduces glutathione levels and alters phospholipid production, exhausting surfactant reserves in multiple models of toxicity (Tahmasbpour et al., 2019; Pesonen and Vähäkangas, 2020; Elfsmark et al., 2022).

Fig. 1.

Fig. 1.

Mechanisms by which inhaled toxicants affect the bronchioles and alveoli of the lung, leading to nitrosative stress and inflammation. Activation, elevation, and depletion of shared signaling pathways enhance injury. DNA damage, protein adduction, and lipid peroxidation, if left unresolved, can develop into bronchitis, pulmonary edema, and ultimately ALI/ARDS. Current therapies for symptoms include supportive care with bronchodilator use and mechanical ventilation.

Chemosensory TRP channels respond to a variety of inhaled toxicants and induce changes in intracellular calcium that alter alveolar and vascular permeability. Activation of TRP channels in the respiratory epithelium prompts membrane depolarization and inflammatory neuropeptide release (Bessac and Jordt, 2010; Achanta and Jordt, 2020; Pesonen and Vähäkangas, 2020). Irritation from chloropicrin and acrolein is mediated by TRPA1 (Conklin, 2016; Pesonen and Vähäkangas, 2020), stimulating afferent airway fibers that trigger reflex bradypnea and respiratory braking. Chlorine and sulfur mustard directly activate TRP channels (Stenger et al., 2015; Zellner and Eyer, 2020), and this complex interplay of inflammatory mediators and nitrosative stress prompts bronchoconstriction that restricts airflow.

For sulfur mustard and analogs, the heightened nitrosative burden desensitizes β-adrenergic receptors in the distal airways, affecting cAMP levels and impairing endothelial barrier function (Ghanei et al., 2007; Kabir et al., 2009; Rambacher and Moniri, 2020). This allows blood plasma to enter the interstitium and exacerbates pulmonary edema and inflammation (Radbel et al., 2020). Inhaled toxicants can induce lung hemorrhage and activation of the coagulation cascade that mediates fibrinolysis, exacerbating ALI/ARDS. Coagulation is elevated in ammonia and chlorine models (Zarogiannis et al., 2014; Elfsmark et al., 2022), and extravascular coagulation and impaired fibrinolysis is pronounced in SM injury (White et al., 2016). In phosgene models, hemolysis destroys red blood cells, which obstructs the pulmonary capillaries (Aggarwal et al., 2019).

Challenges to Medical Countermeasures Development

The development of intervention strategies to counteract acute toxicant exposures presents innumerable challenges. There are ample opportunities to advance our fundamental mechanistic understanding of toxicant-induced ALI and delayed lung effects such as pulmonary fibrosis. As discussed earlier, the resultant lung injury involves endothelial and epithelial damage, alveolar permeability dysfunction, and dysregulated lung inflammation, which reduce lung compliance and compromise respiration. An immediate challenge in mass casualty scenarios is to develop strategies to protect survivors without prior knowledge of the inhaled toxicant. In addition, animal studies using pregnant dams and neonates have shown that vulnerable subgroups are highly susceptible to pulmonary toxicants and may require population-specific care (Addis et al., 2020, 2021).

Immediate treatment of acute exposures involves personal protective equipment for first responders, removal of victims from the area, and decontamination. As there are few antidotes for inhaled toxicants, therapy largely depends on supportive care, with administration of warm, humidified oxygen, antitussives, and bronchodilators (Walker et al., 2015). In some instances, survivors with compromised airways need endotracheal intubation and low tidal volume mechanical ventilation. Those with severe epithelial injury and sloughing may require bronchoscopic lavage to maintain airflow. Inhaled and systemic corticosteroids can be used to reduce inflammation; however, demonstrations of efficacy are limited to small, uncontrolled studies from other airway diseases. Follow-on clinical therapies are based on long-term outcomes (e.g., ALI/ARDS, BO, RADS, idiopathic pulmonary fibrosis).

To date, several laboratories have explored therapeutic approaches to reduce toxicant-induced ALI using antioxidants, angiotensin-converting enzyme inhibitors, neutrophil elastase inhibitors, anti–tumor necrosis factor α therapy, β-agonists, heparin, and phosphodiesterase inhibitors, and some combinations of these drugs have shown limited success. However, the high attrition rate of ARDS drugs may be due to a missed therapeutic window between ALI development and progression to full-blown ARDS. The translation of findings from other lung disease models (e.g., lipopolysaccharide, bleomycin) to high-dose CWA exposures that involve rapid transition of ALI to ARDS is especially difficult.

Lung injury intensity is determined by multiple factors, including proximity to the exposure epicenter, duration, and population demographics (e.g., age, sex, preexisting disease status). The National Institutes of Health Chemical Countermeasures Research Program supports the development of therapeutic agents that can be administered in the field to treat ALI and in trauma/critical care centers to reduce the potential for delayed onset lung complications as well as uncontrolled pulmonary edema from low dose exposure. Current research efforts have focused on understanding the acute pathophysiological consequences of exposure to a limited number of CWAs and industrial toxicants. Given the number of potential CoCs, it is prohibitive to develop a countermeasure specific to each chemical.

Instead, research efforts toward medical countermeasure development should focus on identifying common molecular, cellular, and pathophysiological pathways involved in toxicant-induced ALI using available in vitro, in silico, and organoid systems as well as animal models. Ideally, genomic, epigenomic, and metabolomic approaches could be used to establish kinetic models that demonstrate the evolution and progression of disease and fully characterize specific cell types involved throughout the process. Defining mechanisms of ALI after industrial and agricultural toxicant exposures would be beneficial as we have limited understanding of their molecular pathophysiology. High-throughput screens could accelerate the identification of potential therapeutic targets that counteract acute and delayed effects from diverse toxicant insults.

There is compelling need for better mechanistic understanding of the temporal etiology of lung injury following exposure. Advances in the field are largely driven by findings in animal models and are complicated by variability due to species- and strain-specific differences and duration and routes of exposure. There is a renewed need to develop and refine animal models of exposure and to increase the efficacy and specificity of drug candidates. Significant challenges exist due to ethical constraints on human research and testing. Longitudinal population studies that follow large-scale chemical accidents could delineate long-term effects of toxicant exposures, and appropriate global infrastructure is needed to support these efforts. Ultimately, these comprehensive research efforts will further our understanding of toxicant-induced lung injury progression and support the development of countermeasures against pulmonary threats.

Acknowledgments

The authors would like to thank Thaddeus Schug at the NIEHS for his critical review of the manuscript and Lois Wyrick from NIEHS Arts and Graphics for support in generating the figure.

Abbreviations

AHR

airway hyperresponsiveness

ALI

acute lung injury

ARDS

acute respiratory distress syndrome

BO

bronchiolitis obliterans

CG

phosgene

CLDN5

claudin 5

CoC

chemical of concern

COPD

chronic obstructive pulmonary disease

CWA

chemical warfare agent

IL

interleukin

ILD

interstitial lung disease

NO

nitric oxide

RADS

reactive airways disease

SM

sulfur mustard

TRP

transient receptor potential

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Marzec, Nadadur.

Footnotes

This work was supported by National Institutes of Health National Institute of Environmental Health Sciences Division of Extramural Research and Training program. The contents are solely the responsibility of the authors and do not necessarily represent the official views of the NIH.

The authors declare that there are no conflicts of interest.

References

  1. Achanta S, Jordt SE (2020) Transient receptor potential channels in pulmonary chemical injuries and as countermeasure targets. Ann N Y Acad Sci 1480:73–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Achanta S, Jordt SE (2021) Toxic effects of chlorine gas and potential treatments: a literature review. Toxicol Mech Methods 31:244–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Addis DR, Aggarwal S, Lazrak A, Jilling T, Matalon S (2021) Halogen-Induced Chemical Injury to the Mammalian Cardiopulmonary Systems. Physiology (Bethesda) 36:272–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Addis DR, Molyvdas A, Ambalavanan N, Matalon S, Jilling T (2020) Halogen exposure injury in the developing lung. Ann N Y Acad Sci 1480:30–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Agarwal AR, Yin F, Cadenas E (2013) Metabolic shift in lung alveolar cell mitochondria following acrolein exposure. Am J Physiol Lung Cell Mol Physiol 305:L764–L773. [DOI] [PubMed] [Google Scholar]
  6. Aggarwal SJilling TDoran SAhmad IEagen JEGu SGillespie MAlbert CJFord DOh JY, et al. (2019) Phosgene inhalation causes hemolysis and acute lung injury. Toxicol Lett 312:204–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Ali R, Mittal G, Ali R, Kumar M, Kishan Khar R, Ahmad FJ, Bhatnagar A (2013) Development, characterisation and pharmacoscintigraphic evaluation of nano-fluticasone propionate dry powder inhalation as potential antidote against inhaled toxic gases. J Microencapsul 30:546–558. [DOI] [PubMed] [Google Scholar]
  8. Ali R, Mittal G, Sultana S, Bhatnagar A (2012) Ameliorative potential of alpha-ketoglutaric acid (AKG) on acute lung injuries induced by ammonia inhalation in rats. Exp Lung Res 38:435–444. [DOI] [PubMed] [Google Scholar]
  9. Arroyo CM, Feliciano F, Kolb DL, Keeler JR, Millette SR, Stotts RR (1993) Autoionization reaction of phosgene (OCCl2) studied by electron paramagnetic resonance/spin trapping techniques. J Biochem Toxicol 8:107–110. [DOI] [PubMed] [Google Scholar]
  10. Balakrishna SSong WAchanta SDoran SFLiu BKaelberer MMYu ZSui ACheung MLeishman E, et al. (2014) TRPV4 inhibition counteracts edema and inflammation and improves pulmonary function and oxygen saturation in chemically induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 307:L158–L172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Beauchamp RO Jr, Andjelkovich DA, Kligerman AD, Morgan KT, Heck HD (1985) A critical review of the literature on acrolein toxicity. Crit Rev Toxicol 14:309–380. [DOI] [PubMed] [Google Scholar]
  12. Bein K, Birru RL, Wells H, Larkin TP, Ge T, Leikauf GD (2021) Sex-dependent acrolein sensitivity in mice is associated with differential lung cell, protein, and transcript changes. Physiol Rep 9:e14997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bessac BF, Jordt SE (2010) Sensory detection and responses to toxic gases: mechanisms, health effects, and countermeasures. Proc Am Thorac Soc 7:269–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Boskabady MH, Farhadi J (2008) The possible prophylactic effect of Nigella sativa seed aqueous extract on respiratory symptoms and pulmonary function tests on chemical war victims: a randomized, double-blind, placebo-controlled trial. J Altern Complement Med 14:1137–1144. [DOI] [PubMed] [Google Scholar]
  15. Branco ACCC, Yoshikawa FSY, Pietrobon AJ, Sato MN (2018) Role of Histamine in Modulating the Immune Response and Inflammation. Mediators Inflamm 2018:9524075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Brooks SM (2008) Irritant-induced chronic cough: irritant-induced TRPpathy. Lung 186 (Suppl 1):S88–S93. [DOI] [PubMed] [Google Scholar]
  17. Brooks SM, Weiss MA, Bernstein IL (1985) Reactive airways dysfunction syndrome (RADS). Persistent asthma syndrome after high level irritant exposures. Chest 88:376–384. [DOI] [PubMed] [Google Scholar]
  18. Butt Y, Kurdowska A, Allen TC (2016) Acute Lung Injury: A Clinical and Molecular Review. Arch Pathol Lab Med 140:345–350. [DOI] [PubMed] [Google Scholar]
  19. Calvet JH, Jarreau PH, Levame M, D’Ortho MP, Lorino H, Harf A, Macquin-Mavier I (1994). Acute and chronic respiratory effects of sulfur mustard intoxication in guinea pig. J Appl Physiol (1985) 76:681–688. [DOI] [PubMed] [Google Scholar]
  20. Cao C, Zhang L, Shen J (2022) Phosgene-Induced acute lung injury: Approaches for mechanism-based treatment strategies. Front Immunol 13:917395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Carlisle M, Lam A, Svendsen ER, Aggarwal S, Matalon S (2016) Chlorine-induced cardiopulmonary injury. Ann N Y Acad Sci 1374:159–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chen HL, Bai H, Xi MM, Liu R, Qin XJ, Liang X, Zhang W, Zhang XD, Li WL, Hai CX (2013) Ethyl pyruvate protects rats from phosgene-induced pulmonary edema by inhibiting cyclooxygenase2 and inducible nitric oxide synthase expression. J Appl Toxicol 33:71–77. [DOI] [PubMed] [Google Scholar]
  23. Close LG, Catlin FI, Cohn AM (1980) Acute and chronic effects of ammonia burns on the respiratory tract. Arch Otolaryngol 106:151–158. [DOI] [PubMed] [Google Scholar]
  24. Conklin DJ (2016) Acute cardiopulmonary toxicity of inhaled aldehydes: role of TRPA1. Ann N Y Acad Sci 1374:59–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Elfsmark L, Ågren L, Akfur C, Jonasson S (2022) Ammonia exposure by intratracheal instillation causes severe and deteriorating lung injury and vascular effects in mice. Inhal Toxicol 34:145–158. [DOI] [PubMed] [Google Scholar]
  26. Elfsmark L, Ågren L, Akfur C, Wigenstam E, Bergström U, Jonasson S (2019) Comparisons of acute inflammatory responses of nose-only inhalation and intratracheal instillation of ammonia in rats. Inhal Toxicol 31:107–118. [DOI] [PubMed] [Google Scholar]
  27. Fabisiak JPMedvedovic MAlexander DCMcDunn JEConcel VJBein KJang ASBerndt AVuga LJBrant KA, et al. (2011) Integrative metabolome and transcriptome profiling reveals discordant energetic stress between mouse strains with differential sensitivity to acrolein-induced acute lung injury. Mol Nutr Food Res 55:1423–1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Filipczak PT, Senft AP, Seagrave J, Weber W, Kuehl PJ, Fredenburgh LE, McDonald JD, Baron RM (2015) NOS-2 Inhibition in Phosgene-Induced Acute Lung Injury. Toxicol Sci 146:89–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Ford DA, Honavar J, Albert CJ, Duerr MA, Oh JY, Doran S, Matalon S, Patel RP (2016) Formation of chlorinated lipids post-chlorine gas exposure. J Lipid Res 57:1529–1540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Geng P, Ma T, Xing J, Jiang L, Sun H, Zhu B, Zhang H, Xiao H, Wang J, Zhang J (2018) Dexamethasone ameliorates H2S-induced acute lung injury by increasing claudin-5 expression via the PI3K pathway. Hum Exp Toxicol 37:626–635. [DOI] [PubMed] [Google Scholar]
  31. Geng P, Yu F, Tan D, Xu J, Yang Y, Xu M, Wang H, Ling B (2020) Involvement of claudin-5 in H2S-induced acute lung injury. J Toxicol Sci 45:293–304. [DOI] [PubMed] [Google Scholar]
  32. Gessner MA, Doran SF, Yu Z, Dunaway CW, Matalon S, Steele C (2013) Chlorine gas exposure increases susceptibility to invasive lung fungal infection. Am J Physiol Lung Cell Mol Physiol 304:L765–L773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Ghanei M, Khalili AR, Arab MJ, Mojtahedzadeh M, Aslani J, Lessan-Pezeshki M, Panahi Y, Alaeddini F (2005) Diagnostic and therapeutic value of short-term corticosteroid therapy in exacerbation of mustard gas-induced chronic bronchitis. Basic Clin Pharmacol Toxicol 97:302–305. [DOI] [PubMed] [Google Scholar]
  34. Ghanei M, Shohrati M, Harandi AA, Eshraghi M, Aslani J, Alaeddini F, Manzoori H (2007) Inhaled corticosteroids and long-acting beta 2-agonists in treatment of patients with chronic bronchiolitis following exposure to sulfur mustard. Inhal Toxicol 19:889–894. [DOI] [PubMed] [Google Scholar]
  35. Ghanei M, Tazelaar HD, Chilosi M, Harandi AA, Peyman M, Akbari HM, Shamsaei H, Bahadori M, Aslani J, Mohammadi A (2008) An international collaborative pathologic study of surgical lung biopsies from mustard gas-exposed patients. Respir Med 102:825–830. [DOI] [PubMed] [Google Scholar]
  36. Hobson ST, Richieri RA, Parseghian MH (2021) Phosgene: toxicology, animal models, and medical countermeasures. Toxicol Mech Methods 31:293–307. [DOI] [PubMed] [Google Scholar]
  37. Hochman DJ, Collaco CR, Brooks EG (2014) Acrolein induction of oxidative stress and degranulation in mast cells. Environ Toxicol 29:908–915. [DOI] [PubMed] [Google Scholar]
  38. Holmes WWKeyser BMParadiso DCRay RAndres DKBenton BJRothwell CCHoard-Fruchey HMDillman JFSciuto AM, et al. (2016) Conceptual approaches for treatment of phosgene inhalation-induced lung injury. Toxicol Lett 244:8–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Hossein BM, Nasim V, Sediqa A (2008) The protective effect of Nigella sativa on lung injury of sulfur mustard-exposed Guinea pigs. Exp Lung Res 34:183–194. [DOI] [PubMed] [Google Scholar]
  40. Jang ASConcel VJBein KBrant KALiu SPope-Varsalona HDopico RA JrDi YPKnoell DLBarchowsky A, et al. (2011) Endothelial dysfunction and claudin 5 regulation during acrolein-induced lung injury. Am J Respir Cell Mol Biol 44:483–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Ji L, Liu R, Zhang XD, Chen HL, Bai H, Wang X, Zhao HL, Liang X, Hai CX (2010) N-acetylcysteine attenuates phosgene-induced acute lung injury via up-regulation of Nrf2 expression. Inhal Toxicol 22:535–542. [DOI] [PubMed] [Google Scholar]
  42. Jiang ZF, Shao Y, Zhang L, Shen J (2021) Lung-derived exosomes regulate the function of mesenchymal stem cells and alleviate phosgene-induced lung injury via miR-34c-3p. J Biochem Mol Toxicol 35:e22851. [DOI] [PubMed] [Google Scholar]
  43. Jin C, Zhou F, Zhang L, Shen J (2020) Overexpression of heat shock protein 70 enhanced mesenchymal stem cell treatment efficacy in phosgene-induced acute lung injury. J Biochem Mol Toxicol 34:e22515. [DOI] [PubMed] [Google Scholar]
  44. Johanson G, Dwivedi AM, Ernstgård L, Palmberg L, Ganguly K, Chen LC, Galdanes K, Gordon T, Upadhyay S (2020) Analysis of Acrolein Exposure Induced Pulmonary Response in Seven Inbred Mouse Strains and Human Primary Bronchial Epithelial Cells Cultured at Air-Liquid Interface. BioMed Res Int 2020:3259723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Kabir SM, Mukherjee S, Rajaratnam V, Smith MG, Das SK (2009) Desensitization of beta-adrenergic receptors in lung injury induced by 2-chloroethyl ethyl sulfide, a mustard analog. J Biochem Mol Toxicol 23:59–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Leduc D, Gris P, Lheureux P, Gevenois PA, De Vuyst P, Yernault JC (1992) Acute and long term respiratory damage following inhalation of ammonia. Thorax 47:755–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Lindsay CD (2011) Novel therapeutic strategies for acute lung injury induced by lung damaging agents: the potential role of growth factors as treatment options. Hum Exp Toxicol 30:701–724. [DOI] [PubMed] [Google Scholar]
  48. Lu Q, Huang S, Meng X, Zhang J, Yu S, Li J, Shi M, Fan H, Zhao Y (2021) Mechanism of Phosgene-Induced Acute Lung Injury and Treatment Strategy. Int J Mol Sci 22:10933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Lu QMundy MChambers ELange TNewton JBorgas DYao HChoudhary GBasak ROldham M, et al. (2017) Alda-1 Protects Against Acrolein-Induced Acute Lung Injury and Endothelial Barrier Dysfunction. Am J Respir Cell Mol Biol 57:662–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Madsen J, Kliem A, Tornoe I, Skjodt K, Koch C, Holmskov U (2000) Localization of lung surfactant protein D on mucosal surfaces in human tissues. J Immunol 164:5866–5870. [DOI] [PubMed] [Google Scholar]
  51. Malaviya RAbramova EVRancourt RCSunil VRNapierala MWeinstock DCroutch CRRoseman JTuttle RPeters E, et al. (2020) Progressive Lung Injury, Inflammation, and Fibrosis in Rats Following Inhalation of Sulfur Mustard. Toxicol Sci 178:358–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Malaviya R, Gardner CR, Rancourt RC, Smith LC, Abramova EV, Vayas KN, Gow AJ, Laskin JD, Laskin DL (2023) Lung injury and oxidative stress induced by inhaled chlorine in mice is associated with proinflammatory activation of macrophages and altered bioenergetics. Toxicol Appl Pharmacol 461:116388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Mo Y, Chen J, Humphrey DM Jr, Fodah RA, Warawa JM, Hoyle GW (2015) Abnormal epithelial structure and chronic lung inflammation after repair of chlorine-induced airway injury. Am J Physiol Lung Cell Mol Physiol 308:L168–L178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Musah S, Schlueter CF, Humphrey DM Jr, Powell KS, Roberts AM, Hoyle GW (2017) Acute lung injury and persistent small airway disease in a rabbit model of chlorine inhalation. Toxicol Appl Pharmacol 315:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Muskat PC (2008) Mass casualty chemical exposure and implications for respiratory failure. Respir Care 53:58–63, discussion 63–66. [PubMed] [Google Scholar]
  56. Nelson LS, Hoffman RS (2014) Inhaled toxins, in Rosen’s Emergency Medicine: Concepts and Clinical Practice (Marx JA, Hockberger RS, and Walls RM eds) pp 2036–2043, Saunders, Elsevier. [Google Scholar]
  57. Pangeni RP, Timilsina B, Oli PR, Khadka S, Regmi PR (2022) A multidisciplinary approach to accidental inhalational ammonia injury: A case report. Ann Med Surg (Lond) 82:104741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Parkes WR, editor (1994) Aerosols: their deposition and clearance. Occupational lung disorders, p 35, Butterworth-Heinemann, Oxford. [Google Scholar]
  59. Pauluhn J (2021) Phosgene inhalation toxicity: Update on mechanisms and mechanism-based treatment strategies. Toxicology 450:152682. [DOI] [PubMed] [Google Scholar]
  60. Pauluhn J, Carson A, Costa DL, Gordon T, Kodavanti U, Last JA, Matthay MA, Pinkerton KE, Sciuto AM (2007) Workshop summary: phosgene-induced pulmonary toxicity revisited: appraisal of early and late markers of pulmonary injury from animal models with emphasis on human significance. Inhal Toxicol 19:789–810. [DOI] [PubMed] [Google Scholar]
  61. Perkins MW, Wong B, Tressler J, Rodriguez A, Sherman K, Andres J, Devorak J, L Wilkins W, Sciuto AM (2017) Adverse respiratory effects in rats following inhalation exposure to ammonia: respiratory dynamics and histopathology. Inhal Toxicol 29:32–41. [DOI] [PubMed] [Google Scholar]
  62. Pesonen M, Häkkinen M, Rilla K, Juvonen R, Kuitunen T, Pasanen M, Vähäkangas K (2014) Chloropicrin-induced toxic responses in human lung epithelial cells. Toxicol Lett 226:236–244. [DOI] [PubMed] [Google Scholar]
  63. Pesonen M, Rysä J, Storvik M, Rilla K, Pasanen M, Vähäkangas K (2017) Molecular targets of chloropicrin in human airway epithelial cells. Toxicol In Vitro 42:247–254. [DOI] [PubMed] [Google Scholar]
  64. Pesonen M, Storvik M, Kokkola T, Rysä J, Vähäkangas K, Pasanen M (2015) Transcriptomic Analysis of Human Primary Bronchial Epithelial Cells after Chloropicrin Treatment. Chem Res Toxicol 28:1926–1935. [DOI] [PubMed] [Google Scholar]
  65. Pesonen M, Vähäkangas K (2020) Chloropicrin-induced toxicity in the respiratory system. Toxicol Lett 323:10–18. [DOI] [PubMed] [Google Scholar]
  66. Qu Y, Zhang L, He D, Xu N, Tang Y, Shao Y, Shen J (2020) Protective role of mesenchymal stem cells transfected with miRNA-378a-5p in phosgene inhalation lung injury. Biochem Biophys Res Commun 530:189–195. [DOI] [PubMed] [Google Scholar]
  67. Radbel J, Laskin DL, Laskin JD, Kipen HM (2020) Disease-modifying treatment of chemical threat agent-induced acute lung injury. Ann N Y Acad Sci 1480:14–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Rambacher KM, Moniri NH (2020) The β2-adrenergic receptor-ROS signaling axis: An overlooked component of β2AR function? Biochem Pharmacol 171:113690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Rancourt RC, Veress LA, Ahmad A, Hendry-Hofer TB, Rioux JS, Garlick RB, White CW (2013) Tissue factor pathway inhibitor prevents airway obstruction, respiratory failure and death due to sulfur mustard analog inhalation. Toxicol Appl Pharmacol 272:86–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Razavi SM, Abdollahi M, Salamati P (2016) Cancer Events After Acute or Chronic Exposure to Sulfur Mustard: A Review of the Literature. Int J Prev Med 7:76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Russell D, Blain PG, Rice P (2006) Clinical management of casualties exposed to lung damaging agents: a critical review. Emerg Med J 23:421–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Saeed O, Boyer NL, Pamplin JC, Driscoll IR, DellaVolpe J, Cannon J, Cancio LC (2018) Inhalation Injury and Toxic Industrial Chemical Exposure. Mil Med 183 (suppl_2):130–132. [DOI] [PubMed] [Google Scholar]
  73. Savin IA, Zenkova MA, Sen’kova AV (2022) Pulmonary Fibrosis as a Result of Acute Lung Inflammation: Molecular Mechanisms, Relevant In Vivo Models, Prognostic and Therapeutic Approaches. Int J Mol Sci 23:14959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Schmidt A, Steinritz D, Rudolf KD, Thiermann H, Striepling E (2018) Accidental sulfur mustard exposure: A case report. Toxicol Lett 293:62–66. [DOI] [PubMed] [Google Scholar]
  75. Shao Y, Shen J, Zhou F, He D (2018) Mesenchymal stem cells overexpressing Ang1 attenuates phosgene-induced acute lung injury in rats. Inhal Toxicol 30:313–320. [DOI] [PubMed] [Google Scholar]
  76. Shen J, Gan Z, Zhao J, Zhang L, Xu G (2014) Ulinastatin reduces pathogenesis of phosgene-induced acute lung injury in rats. Toxicol Ind Health 30:785–793. [DOI] [PubMed] [Google Scholar]
  77. Snow SJ, McGee MA, Henriquez A, Richards JE, Schladweiler MC, Ledbetter AD, Kodavanti UP (2017) Respiratory Effects and Systemic Stress Response Following Acute Acrolein Inhalation in Rats. Toxicol Sci 158:454–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Song W, Yu Z, Doran SF, Ambalavanan N, Steele C, Garantziotis S, Matalon S (2015) Respiratory syncytial virus infection increases chlorine-induced airway hyperresponsiveness. Am J Physiol Lung Cell Mol Physiol 309:L205–L210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Sorensen GL (2018) Surfactant Protein D in Respiratory and Non-Respiratory Diseases. Front Med (Lausanne) 5:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Sparks SE, Quistad GB, Li W, Casida JE (2000) Chloropicrin dechlorination in relation to toxic action. J Biochem Mol Toxicol 14:26–32. [DOI] [PubMed] [Google Scholar]
  81. Spickett CM (2007) Chlorinated lipids and fatty acids: an emerging role in pathology. Pharmacol Ther 115:400–409. [DOI] [PubMed] [Google Scholar]
  82. Starosta V, Griese M (2006) Oxidative damage to surfactant protein D in pulmonary diseases [published correction appears in Free Radic Res (2006) 40:665]. Free Radic Res 40:419–425. [DOI] [PubMed] [Google Scholar]
  83. Stenger B, Zehfuss F, Mückter H, Schmidt A, Balszuweit F, Schäfer E, Büch T, Gudermann T, Thiermann H, Steinritz D (2015) Activation of the chemosensing transient receptor potential channel A1 (TRPA1) by alkylating agents. Arch Toxicol 89:1631–1643. [DOI] [PubMed] [Google Scholar]
  84. Sun J, Druhan LJ, Zweier JL (2010) Reactive oxygen and nitrogen species regulate inducible nitric oxide synthase function shifting the balance of nitric oxide and superoxide production. Arch Biochem Biophys 494:130–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Tahmasbpour E, Ghanei M, Khor A, Panahi Y (2019) Altered expression of cyclooxygenase-2, 12-lipoxygenase, inducible nitric oxide synthase-2 and surfactant protein D in lungs of patients with pulmonary injury caused by sulfur mustard. Drug Chem Toxicol 42:257–263. [DOI] [PubMed] [Google Scholar]
  86. van Helden HP, Kuijpers WC, Diemel RV (2004) Asthmalike symptoms following intratracheal exposure of Guinea pigs to sulfur mustard aerosol: therapeutic efficacy of exogenous lung surfactant curosurf and salbutamol. Inhal Toxicol 16:537–548. [DOI] [PubMed] [Google Scholar]
  87. Walker PF, Buehner MF, Wood LA, Boyer NL, Driscoll IR, Lundy JB, Cancio LC, Chung KK (2015) Diagnosis and management of inhalation injury: an updated review. Crit Care 19:351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. White CW, Rancourt RC, Veress LA (2016) Sulfur mustard inhalation: mechanisms of injury, alteration of coagulation, and fibrinolytic therapy. Ann N Y Acad Sci 1378:87–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Wygrecka M, Jablonska E, Guenther A, Preissner KT, Markart P (2008) Current view on alveolar coagulation and fibrinolysis in acute inflammatory and chronic interstitial lung diseases. Thromb Haemost 99:494–501. [DOI] [PubMed] [Google Scholar]
  90. Xiong R, Wu Q, Muskhelishvili L, Davis K, Shemansky JM, Bryant M, Rosenfeldt H, Healy SM, Cao X (2018) Evaluating Mode of Action of Acrolein Toxicity in an In Vitro Human Airway Tissue Model. Toxicol Sci 166:451–464. [DOI] [PubMed] [Google Scholar]
  91. Xu N, Shao Y, Ye K, Qu Y, Memet O, He D, Shen J (2019) Mesenchymal stem cell-derived exosomes attenuate phosgene-induced acute lung injury in rats. Inhal Toxicol 31:52–60. [DOI] [PubMed] [Google Scholar]
  92. Zaky A, Bradley WE, Lazrak A, Zafar I, Doran S, Ahmad A, White CW, Dell’Italia LJ, Matalon S, Ahmad S (2015) Chlorine inhalation-induced myocardial depression and failure. Physiol Rep 3:e12439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Zarogiannis SG, Wagener BM, Basappa S, Doran S, Rodriguez CA, Jurkuvenaite A, Pittet JF, Matalon S (2014) Postexposure aerosolized heparin reduces lung injury in chlorine-exposed mice. Am J Physiol Lung Cell Mol Physiol 307:L347–L354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Zellner T, Eyer F (2020) Choking agents and chlorine gas - History, pathophysiology, clinical effects and treatment. Toxicol Lett 320:73–79. [DOI] [PubMed] [Google Scholar]
  95. Zhang L, Zhang F, He D, Xu D, Zhong Z, Shen J (2017) Melatonin attenuates phosgene-induced acute lung injury via the upregulation Wnt/β-catenin pathway. Int J Clin Exp Pathol 10:11281–11287. [PMC free article] [PubMed] [Google Scholar]

Articles from The Journal of Pharmacology and Experimental Therapeutics are provided here courtesy of American Society for Pharmacology and Experimental Therapeutics

RESOURCES