Next Article in Journal
Cancer Therapy Guided by Mutation Tests: Current Status and Perspectives
Previous Article in Journal
TMEM16A, a Homoharringtonine Receptor, as a Potential Endogenic Target for Lung Cancer Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechanism of Phosgene-Induced Acute Lung Injury and Treatment Strategy

1
Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
2
Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
*
Authors to whom correspondence should be addressed.
Qianying Lu and Siyu Huang contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(20), 10933; https://doi.org/10.3390/ijms222010933
Submission received: 20 August 2021 / Revised: 24 September 2021 / Accepted: 29 September 2021 / Published: 10 October 2021
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Phosgene (COCl2) was once used as a classic suffocation poison and currently plays an essential role in industrial production. Due to its high toxicity, the problem of poisoning caused by leakage during production, storage, and use cannot be ignored. Phosgene mainly acts on the lungs, causing long-lasting respiratory depression, refractory pulmonary edema, and other related lung injuries, which may cause acute respiratory distress syndrome or even death in severe cases. Due to the high mortality, poor prognosis, and frequent sequelae, targeted therapies for phosgene exposure are needed. However, there is currently no specific antidote for phosgene poisoning. This paper reviews the literature on the mechanism and treatment strategies to explore new ideas for the treatment of phosgene poisoning.

1. Introduction

Since the Cornish chemist John Davy first synthesized phosgene (COCl2, CG), it has been present in the world for more than 200 years. It was originally widely known as a “Chemical Warfare Agent” (CWA) during World War I [1]. It is 3.5 times denser than air (ρ = 3.5 g/mL), which makes it easy to deposit in low-lying areas and slow to dissipate. According to Fries’s description, the odor of phosgene is described as “hay or grass”, which is different from the pungent odor of chlorine, and easily leads to identification failure. On 19 December 1915, Germany used phosgene as a CWA for the first time, causing injuries to more than 1000 British soldiers and more than 100 deaths [2]. According to statistics, the use of phosgene by armies around the world caused hundreds of thousands of deaths during World War I [3]. Surprisingly, the production of phosgene only accounts for 25% of all CWA production, but accounts for 85% of CWA deaths [4]. Although the toxicity of phosgene makes people afraid, its value in industrial production has attracted much attention. Currently, phosgene is mostly used to synthesize polyurethane foam monomers such as toluene diisocyanate (TDI) and methylene diphenyl diisocyana1e (MDI) [5]. Additionally, as an important organic intermediate, phosgene is widely used in modern industries such as chemicals, pesticides, and organic synthesis [6]. Although phosgene is no longer used as a CWA, accidents caused by improper operation still result in casualties. For example, in 1984, a phosgene leak at a pesticide factory in India caused the deaths of 2500 people [7]. The threat of phosgene still exists, so there is a need to seriously look for effective treatment strategies.
Inhalation is the main mode of phosgene exposure. After inhaling phosgene, the patient initially shows a mild dry cough, accompanied by skin and mucous membrane irritation. Then, typical symptoms of respiratory tract irritation quickly develop within a few hours, which mostly present with coughing, chest tightness, and wheezing. In severe cases, typical clinical symptoms of phosgene-induced acute lung injury (P-ALI) appears (includes pulmonary edema, difficulty breathing, and hypoxemia), and some may even develop into acute respiratory distress syndrome (ARDS) [8]. Since the molecular mechanisms of P-ALI are not fully elucidated, the clinical treatment of P-ALI is still mainly based on symptomatic measures and supportive treatment (including extracorporeal membrane oxygenation (ECMO), positive end expiratory pressure (PEEP), and delayed low-dose oxygen supplementation) [9,10,11]. However, these treatments can achieve only symptom alleviation, but cannot be completely cured, and there is still no specific antidote for phosgene exposure. For these reasons, the mortality rate of phosgene poisoning remains high. Thus, identifying the mechanisms of P-ALI and searching for effective therapeutics and treatment regimens are crucial. This paper reviews the physiological mechanisms and medical treatment of P-ALI and considers the possible applications to provide a reference for the treatment of P-ALI.

2. Toxicology Studies

After phosgene inhalation exposure, due to the poor water solubility, very little phosgene can be dissolved in the water on the surface of the bronchial wall, most of which migrates through the upper respiratory tract and finally accumulated in the alveolar lumen of the lower respiratory tract. According to Haber’s law “Dose (C) × time (t) = constant”, the toxicity of phosgene exposure is proportional to the dose and exposure duration. Therefore, a chronic exposure to a low concentration of phosgene may be more toxic than an acute exposure to a high concentration [12]. However, according to the Acute Exposure Guideline Levels (AEGL) from the EPA (2002), Haber’s law has a certain range of adaptation for phosgene poisoning. At a high concentration of phosgene (>30 ppm × min for dogs or rats), a short exposure can also result in pulmonary edema and a breakdown in the blood–air barrier. [13] However, due to the pathophysiological differences between humans and animals, these animal models do not fully reflect the real condition in the human body after phosgene poisoning, as data from clinical studies of phosgene poisoning are still lacking. Based on the available accident report, human exposure to a sub-lethal dose of phosgene (30–150 ppm × min) could result in neutrophil infiltration, pulmonary edema, and oxidative stress in the lungs [14,15]. Earlier reports have found that phosgene inhalation in concentrations greater than 1 ppm may produce a transient bioprotective vagus reflex with rapid shallow breathing, and moderate irritation of the eyes and upper respiratory tract may occur at phosgene concentrations greater than 3 ppm. When the inhalation dose is greater than 300 ppm, it may result in the death of some individuals [16]. At the dose of 500 ppm × min, significant pulmonary edema appears within 3 h, with death following within the next 24 h [13], and a higher dose of phosgene (~600 ppm × min) may cause death within minutes [17]. These data are from a limited number of individuals, and more reliable toxicological data rely on more reliable zoological validation.

3. Pathophysiology Mechanisms

Pulmonary edema is an important pathophysiological change of P-ALI, the reasons for which are multifold. On one hand, phosgene interacts with the alveolar surfactant rapidly after inhalation. After the surfactant is exhausted, phosgene reacts with proteins, lipids, and nucleic acids in the alveolar tissue, resulting in plasma membrane impairment, which in turn leads to the destruction of the pulmonary blood–gas barrier, eventually leading to pulmonary edema. Meanwhile, this process is accompanied by the release of a large number of inflammatory mediators and reactive oxygen species, resulting in the in-crease in alveolar capillary permeability, ultimately aggravating the formation of pulmonary edema. The molecular mechanisms of inflammatory mediators and oxygen free radical release are highlighted in later sections. The main mechanisms of P-ALI are shown in Figure 1.
In addition to pulmonary edema caused by impaired air–blood barrier, the neurogenic pulmonary edema is also an important cause of P-ALI. However, the mechanisms of neurogenic pulmonary edema in P-ALI remain speculative. There is a general view that its mechanism may be partly related to the excitation of vagal C-fibers, a vagal afferent nerve that is primarily responsible for innervating the lower respiratory tract [18]. A study has shown that after exposure to high concentrations (~360 ppm × min) of phosgene, the vagal C-fibers in rat continued to excite, resulting in various symptoms accompanying the vagal reflex such as apnea and bradycardia. In addition, this hyperexcitability of vagal C-fibers further results in the abrogation of the pulmonary sympathetic tone and the failure of vasodilatation mechanisms [19]. As previous studies have pointed out, the abrogation of the pulmonary sympathetic tone leads to systemic pulmonary vasoconstriction and lung cavity closure, thus causing a large amount of blood retained in the pulmonary circulation [13,20]. This process causes an increase in pulmonary venous pressure and subsequently develops with the exudation of fluids and proteins, finally forming a pulmonary edema [21].

4. Cellular and Molecular Mechanisms

4.1. Plasma Membrane Impairment

Phosgene-induced pulmonary toxicity is associated with membrane rupture and alveolar surfactant damage [22]. The carbonyl group in phosgene has high reactivity. After penetrating the lung tissue, phosgene reacts with –NH2, –SH, and –OH, which are important functional groups in proteins, lipids, and nucleic acids: [COCl2 + 2R-NH2 = CO(NH-R)2 + 2HCl]. These reactions result in the acylation of proteins and phosphatidylcholine, which are the main components of alveolar surfactants and further impact enzymatic systems involved in glycolysis in the lung tissues. Enzymatic system disorders mainly manifest as abnormal cell metabolism and physiological activities. The depletion in the alveolar surfactant leads to pulmonary blood–gas barrier destruction and the in-creased permeability of alveolar capillaries. The acylation reaction of phosgene with certain biological macromolecules and hydrolysis can also generate hydrochloric acid, which further exacerbates damage to the alveolo–capillary barrier. Increased pulmonary capillary permeability caused by these factors destroys the vascular-tissue homeostasis, eventually leading to the formation of noncardiogenic edema. In addition, a high concentration of phosgene can also penetrate the pulmonary surfactant layer of alveoli and deplete glutathione (GSH), leading to the increased production of reactive oxygen species (ROS), which can diffuse to the tissue layer and damage deeper cells.

4.2. Inflammation

ALI is a life-threatening syndrome characterized by inflammation and increased vascular permeability [23]. A feature of phosgene exposure is the explosive cascade of inflammation in the lungs. Pro-inflammatory cytokines such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) are several times higher than those under normal physiological conditions [24,25]. The outbreak of inflammation is related to the activation of some inflammatory signaling pathways. The p38 mitogen-activated protein kinase (p38 MAPK) is one of the most important kinases in inflammatory signals [26]. After inhaling phosgene in rats, the phosphorylation of p38 MAPK increases, and NF-kB, an important nuclear transcription factor in the cell, is activated by regulating the phosphorylation of p65 NF-kB, which leads to the coordinated expression of various inflammatory mediators and continuous inflammatory response [27]. He’s research confirmed that inhibition of MAPK and NF-kB pathways can help reduce the inflammatory response in P-ALI [28]. Nod-like receptor protein 3 (NLRP3) inflammasome is a multi-protein complex that participates in ALI induced by multiple factors such as lipopolysaccharide and bleomycin (BLM) by promoting the maturation and secretion of inflammatory factors. NLRP3 is activated in a classic caspase-1-dependent manner, promoting the release of downstream inflammatory mediators and pyrolysis [29]. After rats inhaled 8.33 g/m3 of phosgene, the NLRP3 inflammasome was activated, triggering a cascade of inflammation, resulting in a significant increase in the levels of IL-1β, interleukin-18 (IL-18), and interleukin-33 (IL-33) [30]. It was further discovered that inhibiting NLRP3 through gene silencing can inhibit the upregulation of inflammatory factors and shift the cytokine balance to anti-inflammatory, which is related to histopathological changes [31]. In summary, anti-inflammatory therapy might be used as a therapeutic modality for the treatment of phosgene-induced ALI.

4.3. Oxidative Stress

ROS are unstable molecules that can readily attack critical cellular biomolecules and finally cause tissue cell damage. Under physiological conditions, the content of ROS in lung tissues is kept at a low level by maintaining the balance between ROS production and elimination [32]. However, after exposure to phosgene, ROS levels in the lung rise dramatically. The reason for this is due to the carbonyl, an active group of phosgene, and its dissociated product carbamoyl chloride reacts with biological molecules such as lipids, sugars, phospholipids, and proteins, thus producing large amounts of ROS [33]. When these excessive ROS are insufficiently scavenged, the pulmonary endothelial cells and alveolar epithelial cells are damaged. At the same time, this is accompanied by the increase in pulmonary capillary permeability and reduction in alveolar surfactant, which resulted in pulmonary edema and atelectasis, and finally the development of ALI.
At the molecular biological level, this process is usually accompanied by the abnormal expression of antioxidative enzymes such as catalase (CAT), glutathione peroxidase (GPx), and superoxide dismutase (SOD) including compensatory increase or decrease [34,35]. NF-E2-related factor 2 (Nrf2) is a ubiquitous transcription factor that is mainly responsible for maintaining the intracellular redox balance [36]. The inhibition of Nrf2 leads to disorders of the Nrf2-mediated antioxidant enzyme system, causing oxidative stress and further exacerbating lung damage [37]. The administration of antioxidants can improve the symptoms of pulmonary edema and the mortality rate, suggesting the critical role of oxidative damage in pulmonary edema. Overall, although there is insufficient evidence on the significant role in P-ALI, it may still be an indispensable link in mediating the early phase of P-ALI. Therefore, antioxidant stress therapy may be a potential therapeutic strategy to prevent or improve P-ALI.

5. Medical Treatments

At present, the clinical treatment of phosgene poisoning mainly focuses on the relief of clinical symptoms in poisoning anaphase including mechanical ventilation, oxygen therapy, extracorporeal membrane oxygenation (ECMO) and fluid resuscitation [38,39]. Although these measures can relieve pulmonary edema and improve lung ventilation, they cannot improve or reverse the pathophysiological changes or prevent the progression of P-ALI. Furthermore, these treatment measures require a lot of medical resources so are not suitable for large-scale phosgene poisoning events [9]. Thus, seeking proactive prevention strategies in the early stages of exposure is the focus of the treatment of phosgene poisoning. The following section reviews the current drug treatment methods available in P-ALI animal models and provides references for follow-up studies.

5.1. Anti-Inflammatory Drugs

5.1.1. Glucocorticoids

Glucocorticoids (GCs) have a rapid, powerful, but nonspecific anti-inflammatory effect [40]. Previous studies have shown that GC is effective in the early treatment of lung inflammation. It is generally believed that GC mainly exerts anti-inflammatory effects through many mechanisms such as inhibiting the release of inflammatory factors, neutrophil activation, reducing capillary permeability, and promoting the differentiation of alveolar macrophages into the M2 phenotype.
In the clinical treatment of phosgene poisoning, the application of adrenal GCs remains controversial. It is generally accepted that early, adequate, and short-term application of GCs can effectively protect pulmonary blood capillary endothelial cells and promote the absorption of pulmonary edema [41]. In the early stage of P-ALI, the application of some GCs also confirmed this finding. Injection of dexamethasone during the early stage of P-ALI significantly reduces the number of neutrophils and protein levels in BALF, inhibits MMP-9 expression, and effectively reduces phosgene inhalation-induced lung injury in rats. [42] Zhou et al. found that low-dose methylprednisolone can also protect against lung injury by inhibiting the activation of NLRP3 inflammasomes in lung tissue cells, which subsequently downregulates the expression of IL-1β and other inflammatory factors [43]. However, a few studies have suggested that GCs may play opposing roles in P-ALI [44]. Liu et al. confirmed that single high-dose dexamethasone treatments may exacerbate the pulmonary toxicity of phosgene [45]. Moreover, some GCs such as mometasone do not significantly improve survival or lung edema in P-ALI [46]. These conflicting results may be due to the application of GC at different time points after phosgene exposure. In clinical cases, early- and short-term use of dexamethasone is the primary treatment in preventing pulmonary edema caused by phosgene. However, the large-scale use of GCs may cause side effects such as femoral head necrosis [47].

5.1.2. Ulinastatin

Ulinastatin (UTI) is a broad-spectrum serine protease inhibitor that has been used clinically to treat acute inflammation in many organs including the lungs [48,49]. In the P-ALI rat model, intraperitoneal injection of UTI can reduce inflammation in lung tissue by downregulating the synthesis and release of proinflammatory factors including interleukin-15 (IL-15), intercellular cell adhesion molecule-1 (ICAM-1), and IL-1β and reducing blood cell infiltration. Furthermore, the regulation of UTI on inflammation is dose-dependent [50,51].

5.1.3. NOS-2 Inhibitors

Excessive production of nitric oxide (NO) is a major factor that contributes to ALI [52]. Nitric oxide synthase (NOS-2), a principal enzyme, can produce high-levels of sustained NO. NOS-2 is upregulated in most ALI models including P-ALI [53,54,55,56]. The inhibition of NOS-2 is considered as a potential treatment for ALI. A previous study confirmed that 1400 W, a NOS-2 inhibitor, could downregulate NO production, further enhancing the expression of surfactant protein-B (SP-B) and zonula occludens protein-1 (ZO-1), thereby decreasing the disruption of the alveolar epithelial barrier [57]. Similarly, aminoguanidine (AG), another NOS-2 inhibitor, exhibited a significant effect on ALI, and its protective effect was more pronounced in low-dose phosgene poisoning than high-dose poisoning [45]. In addition, ethyl pyruvate (EP) could alleviate phosgene-induced ALI by regulating NOS-2 expression and reducing NO production by inhibiting MAPK activation [56]. These studies suggest that the delivery of NOS-2-specific inhibitors offers a novel strategy for treating P-ALI.

5.1.4. Melatonin

Melatonin (MT), a hormone that is secreted by the pineal gland in the brain, is a circadian regulator of different organ systems [58]. Additionally, MT is a well-known anti-inflammatory molecule that has been shown to be effective in ALI induced by many conditions [59,60]. In the P-ALI model, MT can regulate the release of inflammatory factors and have an anti-inflammatory effect by activating the wnt/β-catenin pathway and inhibiting p38 MAPK activation and NOS-2 and NF-κB expression [61,62,63]. These studies show that MT is also a potential anti-inflammatory drug for the treatment of P-ALI.

5.1.5. Angiopoietin-1

Angiopoietin-1 (Ang1) is an oligomeric glycoprotein that plays a key role in the regeneration, maturation, and stabilization of vessels [64]. Ang-1 and its receptor tyrosine kinase receptor 2 (Tie2) constitute the Ang1/Tie2 system, which plays an important role in maintaining endothelial barrier function and vascular integrity [65]. Studies have shown that upregulating Ang1 can effectively improve tissue inflammation and reduce vascular leakage in many animal models of ALI [66,67]. In the P-ALI model, the expression level of Ang was significantly changed [68]. Shen et al. used adenovirus to deliver Ang1 and found that Ang1 significantly reduced the levels of pneumonia factors and vascular permeability in a P-ALI rat model [69]. Further studies have shown that Ang1 reduces P-ALI-associated inflammation by inhibiting the NF-κB and p38 MAPK pathways [28]. This beneficial effect is also mediated in part by inhibiting the activation of NLRP3 inflammasomes [30]. Therefore, Ang1 seems to be a new therapeutic target for phosgene-induced ALI.

5.2. Antioxidant Drugs

5.2.1. N-Acetylcysteine

N-acetylcysteine (NAC), a sulfhydryl compound, has been shown to play antioxidant and ROS-scavenging roles in ALI animal models [70,71]. Lin et al. found that NAC protected against oxidative stress by acting on the Nrf2/GR/GSH pathway, through which NAC elevated the biosynthesis of GSH to repair the defense system that had been destroyed by phosgene [72]. However, Rendell et al. demonstrated that the administration of multiple nebulized doses of NAC was not an effective therapy for P-ALI. This outcome may have been a result of the development of pulmonary edema fluid within the lungs, which prevented the delivery of NAC to the damaged lung tissues [33]. These findings have led to controversy over the clinical effects of NAC.

5.2.2. Caffeic Acid Phenethyl Ester

Caffeic acid phenethyl ester (CAPE) is a natural flavonoid extracted from propolis that has antioxidant, anti-inflammatory, antiviral, and immunomodulatory activities [73]. It has been reported that CAPE affects antioxidant stress in P-ALI, and the effect is mainly achieved by downregulating the levels of MDA and SOD and upregulating the levels of the antioxidant enzyme GSH [34].

5.2.3. Ibuprofen

Ibuprofen (IBU) is one of the most widely used analgesic, antipyretic, and anti-inflammatory drugs today [74,75]. Sciuto et al. first showed that pre- and post-treatment with IBU could significantly reduce lung edema in rats exposed to phosgene [76]. Another study revealed that IBU could alleviate oxidative damage in P-ALI by preventing iron-mediated generation of oxidants or iron-mediated lipid peroxidation after phosgene exposure [77].

5.2.4. Bio300

Bio300 is an isoflavone with antioxidant properties that was originally used as a radio-protectant. Bio300 is able to upregulate antioxidant enzyme expression through the Keap1-Nrf2-ARE pathway, ultimately reducing mortality in P-ALI mice [78].

5.2.5. 5,8,11,14-Eicosatetraynoic acid

5,8,11,14-Eicosatetraynoic acid (ETYA) is an arachidonic acid analog that has antioxidant effects on many types of ALI [79]. In phosgene-exposed guinea pigs, ETYA posttreatment decreased pulmonary edema by increasing the GSH/thiobarbituric acid-reactive substance (TBARS) protection ratio by functioning in an antioxidant-like capacity [80].

5.3. Others

5.3.1. TRP Channel Inhibitors

Transient receptor potential (TRP) channels are a type of nonselective cation channel that are present in the mammalian respiratory tract. TRP channels are specific chemical sensor molecules that control adaptive responses and initiate a cascade of harmful signals [81]. The activation of the TRP subtypes TRPA and TRPV can cause respiratory tract irritation and inflammation [82]. When toxic gases invade (such as phosgene and chlorine), TRP channels are activated, and a large amount of intracellular calcium ([Ca2+]i) is released, causing neuroinflammation, airway vasoconstriction, and vascular membrane barrier permeability, which ultimately leads to pulmonary edema [83]. A study showed that the universal TRP channel inhibitor SKF-96365 [84], TRPA1 inhibitor HC-030031, and pan-TRP inhibitor RR could all inhibit [Ca2+]i efflux in lung epithelial cells and increase the survival rates of P-ALI mice [78]. This finding shows that TRP channel blockers may be potential therapeutic agents for phosgene poisoning, and identifying additional specific TRP channel blockers is the focus of future research.

5.3.2. Mesenchymal Stem Cells

Similar to most stem cells, MSCs have self-renewal and multidirectional differentiation capabilities [85]. In addition, MSCs have strong immunoregulatory abilities and “immune privilege”, which enables these cells to evade host immune system clearance through multiple mechanisms, making the practical application of MSCs possible [86]. In recent years, clinical/preclinical research on MSC therapy has been highly developed [87,88,89]. Clinical and animal experiments have proven that MSCs have therapeutic effects on ALI and other pneumonia-associated diseases [90]. Based on this information, a series of studies on MSC therapy for P-ALI have been conducted.
Chen’s study was the first to confirm the role of MSCs in P-ALI, possibly through participating in pulmonary air–blood barrier repair and regulating inflammatory reactions [91]. Zhang et al. found that wnt3a/β-catenin signaling is inhibited by TGF-β1 and wnt5a in P-ALI and that exogenously administered MSCs home to sites of tissue injury and abrogate the inhibition of wnt3/β-catenin signaling and epithelial permeability in P-ALI [92]. Another way that MSCs work is to directly affect the proliferation and differentiation of various types of lung cells including lung epithelial cells and endogenous lung stem cells [93]. MSCs affect the proliferation of endogenous lung stem cells (club cells) by activating the Notch pathway to promote P-ALI repair [94].
However, due to rejection by the immune system, the homing and survival of MSCs after entering the body has always been a difficult point in treatment [95]. Modifying MSCs to enhance their homing and migration abilities has achieved initial success in a P-ALI animal model. Jin et al. used MSCs overexpressing heat shock protein 70 (Hsp70) to treat P-ALI and surprisingly found that Hsp70 activated the PI3K/AKT pathway to promote MSC survival and migration [96]. In addition, by upregulating the level of the SDF1-specific receptor CXCR7, the ability of MSCs to directionally migrate and differentiate into ATII could be enhanced [97,98].
Although MSCs have good effects on P-ALI, there is a potential risk of iatrogenic tumors [99]. Cell-free replacement therapy using MSC-derived exosomes may be a good way to solve this problem [100]. Exosomes carry proteins, mRNAs, microRNAs, and other factors to target cells and have a similar role as MSCs. Xu’s team found that MSC-derived exosomes can treat P-ALI by inhibiting MMP-9 synthesis and upregulating SP-C [101]. Additionally, miR-28-5p in pulmonary exosomes counteracts the limitations of MSCs through the PI3K/Akt pathway, improving the proliferation, migration, immune regulation, and paracrine effects of MSCs [102]. Although these studies have improved the references for the treatment of P-ALI, the dose, time, and safety issues of MSCs/exosomes have hindered the translation of experimental research into clinical practice. The function of MSCs in P-ALI is shown in Figure 2.

5.3.3. FV-HSP72

Heat shock protein 72 (HSP72) is a pleiotropic drug with antistress and antiapoptotic effects. HSP72 has been used as a cytoprotective agent to treat lung injury [103]. Hobson et al. suggested that HSP72 exhibits great potential in P-ALI as a novel therapeutic agent [104]. In addition to inhibiting the ATP-dependent/independent apoptotic pathway, HSP72 can also directly bind to and prevent NF-κB from entering the nucleus to reduce the expression of NOS-2 and apoptosis induced by oxidative stress [105,106]. However, HSP72 has a short duration of action and is not suitable for the treatment of acute poisoning. Combining HSP72 with the cell-penetrating antibody 3E10 (called Fv-HSP72) improves cell penetration and targeting, resulting in an effective increase in the absorption of HSP72 at the injured site [107]. A recent maximum tolerated dose study of three Fv-HSP72 variants did not find any signs of gross toxicity in either male or female Sprague-Dawley rats. Based on available evidence, it seems that Fv-HSP72 is a potential drug for P-ALI treatment.

6. Conclusions and Perspectives

Since the advent of phosgene, the threat of phosgene in wars, terrorist attacks, and industrial leaks has attracted wide attention. Despite the accumulated data from P-ALI research, the mechanisms of this condition are still unclear, and there is no specific antidote, which makes the treatment of phosgene very difficult. Therefore, it is important to understand the mechanism and develop potential therapeutic drugs of P-ALI.
After phosgene inhalation, it reacts with lung tissue proteins, lipids, and nucleic acids, leading to plasma membrane impairment, which subsequently causes the pulmonary blood–gas barrier destruction, and the increased permeability of alveolar capillaries. Subsequently, a large number of inflammatory mediators and free radicals are released, causing oxidative stress and promoting alveolar and blood vessel damage, which eventually lead to the formation of noncardiogenic edema. Inflammation and oxidative stress are important mechanisms of sublethal phosgene damage, which is consistent with ALI caused by other factors such as bacteria [108], virus [109], smoke inhalation [110], and ex-plosion [111]. Therefore, anti-inflammatory or antioxidant drugs may have a therapeutic effect on ALI including P-ALI. This article reviewed some available anti-inflammatory or antioxidant drug treatments for P-ALI animal models, some of which have proven to be effective in ALI caused by other factors. UTI has been used to treat a broad spectrum of diseases with its potential anti-inflammatory effects. Studies have shown that UTI combined with lung protective mechanical ventilation can effectively improve the clinical effectiveness of patients with ALI. MT, a well-known anti-inflammatory molecule, has been shown to be effective in ALI induced by many conditions [59,60]. It has been indicated that melatonin usage is significantly associated with a 28% reduced likelihood of a positive laboratory test result for SARS-CoV-2 [112], and melatonin is considered as a potential COVID-19 treatment drug. These drugs above-mentioned have been proven to be effective in the clinical treatment of ALI caused by other factors and P-ALI animal models, and may be potential therapeutic drugs for the clinical treatment of P-ALI.
In addition to anti-inflammatory and antioxidant treatments, TRP ion channels antagonists and MSCs are also potential P-ALI treatment strategies. TRP ion channels have been shown to play an important role in medicating pulmonary injury caused by most of the chemical threat agents such as chlorine gas, tear gas agents, acrolein, phosgene, and ammonia. In vitro and in vivo animal models have proven the effects of TRPA1 and TRPV4 antagonists in various chemical injury models. Additionally, selective antagonists of TRP channels have been shown to reduce phosgene-induced neurogenic inflammation in animal models [24]. It is believed that the TRP ion channels are potential treatment strategies for chemical injury, including P-ALI. In recent years, clinical/preclinical research on MSC therapy has been highly developed [87,88,89], and the therapeutic effect of MSCs in ALI has been proven [22]. Taking COVID-19 as an example, a clinical trial showed that MSCs administration could increase the survival rate and improve clinical symptoms [8], which is achieved by modulating the immune system toward an anti-inflammatory state. Several clinical trials of MSCs for the treatment of COVID-19 are currently underway.
In summary, this review summarized the mechanism of P-ALI and treatment strategies in animal models to provide new ideas for clinical research. As inflammation and oxidative stress are important mechanisms of sublethal phosgene damage, anti-inflammatory or antioxidant drugs may have a therapeutic effect on P-ALI.

Author Contributions

Investigation, S.H., J.Z., S.Y., J.L. and M.S.; writing—original draft preparation, S.H.; writing—review and editing, Q.L.; supervision, Y.Z. and H.F.; project administration, X.M.; funding acquisition, H.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Haojun Fan, grant number BLB19J006.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nicholson-Roberts, T.C. Phosgene use in World War 1 and early evaluations of pathophysiology. J. R. Army Med. Corps 2019, 165, 183–187. [Google Scholar] [CrossRef]
  2. Jones, E. Terror weapons: The British experience of gas and its treatment in the First World War. War Hist. 2014, 21, 355–375. [Google Scholar] [CrossRef] [Green Version]
  3. Fitzgerald, G.J. Chemical warfare and medical response during World War I. Am. J. Public Health 2008, 98, 611–625. [Google Scholar] [CrossRef]
  4. Chauhan, S.; Chauhan, S.; D’Cruz, R.; Faruqi, S.; Singh, K.K.; Varma, S.; Singh, M.; Karthik, V. Chemical warfare agents. Environ. Toxicol. Pharmacol. 2008, 26, 113–122. [Google Scholar] [CrossRef] [PubMed]
  5. Fukumura, T.; Mori, W.; Ogawa, M.; Fujinaga, M.; Zhang, M.R. [(11)C]phosgene: Synthesis and application for development of PET radiotracers. Nucl. Med. Biol. 2021, 92, 138–148. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, L.; Wu, D.; Yoon, J. Recent advances in the development of chromophore-based chemosensors for nerve agents and phosgene. ACS Sens. 2018, 3, 27–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Bessac, B.F.; Jordt, S.E. Sensory detection and responses to toxic gases: Mechanisms, health effects, and countermeasures. Proc. Am. Thorac. Soc. 2010, 7, 269–277. [Google Scholar] [CrossRef]
  8. Hardison, L.S., Jr.; Wright, E.; Pizon, A.F. Phosgene exposure: A case of accidental industrial exposure. J. Med. Toxicol. 2014, 10, 51–56. [Google Scholar] [CrossRef] [Green Version]
  9. Li, W.; Rosenbruch, M.; Pauluhn, J. Effect of PEEP on phosgene-induced lung edema: Pilot study on dogs using protective ventilation strategies. Exp. Toxicol. Pathol. 2015, 67, 109–116. [Google Scholar] [CrossRef] [PubMed]
  10. Grainge, C.; Jugg, B.J.; Smith, A.J.; Brown, R.F.; Jenner, J.; Parkhouse, D.A.; Rice, P. Delayed low-dose supplemental oxygen improves survival following phosgene-induced acute lung injury. Inhal. Toxicol. 2010, 22, 552–560. [Google Scholar] [CrossRef]
  11. Graham, S.; Fairhall, S.; Rutter, S.; Auton, P.; Rendell, R.; Smith, A.; Perrott, R.; Roberts, T.N.; Jugg, B. Continuous positive airway pressure: An early intervention to prevent phosgene-induced acute lung injury. Toxicol. Lett. 2018, 293, 120–126. [Google Scholar] [CrossRef] [PubMed]
  12. Pauluhn, J. Concentration x time analyses of sensory irritants revisited: Weight of evidence or the toxic load approach. That is the question. Toxicol. Lett. 2019, 316, 94–108. [Google Scholar] [CrossRef] [PubMed]
  13. Li, W.; Pauluhn, J. Phosgene-induced lung edema: Comparison of clinical criteria for increased extravascular lung water content with postmortem lung gravimetry and lavage-protein in rats and dogs. Toxicol. Lett. 2019, 305, 32–39. [Google Scholar] [CrossRef] [PubMed]
  14. Ghio, A.J.; Kennedy, T.P.; Hatch, G.E.; Tepper, J.S. Reduction of neutrophil influx diminishes lung injury and mortality following phosgene inhalation. J. Appl. Physiol. 1991, 71, 657–665. [Google Scholar] [CrossRef]
  15. Russell, D.; Blain, P.G.; Rice, P. Clinical management of casualties exposed to lung damaging agents: A critical review. Emerg. Med. J. 2006, 23, 421–424. [Google Scholar] [CrossRef]
  16. Diller, W.F. Pathogenesis of phosgene poisoning. Toxicol. Ind. Health 1985, 1, 7–15. [Google Scholar] [CrossRef]
  17. Borak, J.; Diller, W.F. Phosgene exposure: Mechanisms of injury and treatment strategies. J. Occup. Environ. Med. 2001, 43, 110–119. [Google Scholar] [CrossRef]
  18. Lee, L.Y. Respiratory sensations evoked by activation of bronchopulmonary C-fibers. Respir. Physiol. Neurobiol. 2009, 167, 26–35. [Google Scholar] [CrossRef] [Green Version]
  19. Li, W.; Liu, F.; Wang, C.; Truebel, H.; Pauluhn, J. Novel insights into phosgene-induced acute lung injury in rats: Role of dysregulated cardiopulmonary reflexes and nitric oxide in lung edema pathogenesis. Toxicol. Sci. 2013, 131, 612–628. [Google Scholar] [CrossRef] [Green Version]
  20. Ivanhoe, F.; Meyers, F.H. Phosgene poisoning as an example of neuroparalytic acute pulmonary edema: The sympathetic vasomotor reflex involved. Dis. Chest 1964, 46, 211–218. [Google Scholar] [CrossRef] [Green Version]
  21. Pauluhn, J. Phosgene inhalation toxicity: Update on mechanisms and mechanism-based treatment strategies. Toxicology 2021, 450, 152682. [Google Scholar] [CrossRef]
  22. Jugg, B.; Jenner, J.; Rice, P. The effect of perfluoroisobutene and phosgene on rat lavage fluid surfactant phospholipids. Hum. Exp. Toxicol. 1999, 18, 659–668. [Google Scholar] [CrossRef] [PubMed]
  23. Wheeler, A.P.; Bernard, G.R. Acute lung injury and the acute respiratory distress syndrome: A clinical review. Lancet 2007, 369, 1553–1564. [Google Scholar] [CrossRef]
  24. Sciuto, A.M.; Clapp, D.L.; Hess, Z.A.; Moran, T.S. The temporal profile of cytokines in the bronchoalveolar lavage fluid in mice exposed to the industrial gas phosgene. Inhal. Toxicol. 2003, 15, 687–700. [Google Scholar] [CrossRef] [PubMed]
  25. He, D.K.; Shao, Y.R.; Shen, J.; Zhang, L.; Zhang, J.; Zhang, F. Significance of the NLRP3 inflammasome expression in rats with acute lung injury induced by phosgene. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2017, 35, 491–496. [Google Scholar]
  26. Qian, F.; Deng, J.; Wang, G.; Ye, R.D.; Christman, J.W. Pivotal role of mitogen-activated protein kinase-activated protein kinase 2 in inflammatory pulmonary diseases. Curr. Protein Pept. Sci. 2016, 17, 332–342. [Google Scholar] [CrossRef]
  27. Oeckinghaus, A.; Ghosh, S. The NF-κB family of transcription factors and its regulation. Cold Spring Harb. Perspect. Biol. 2009, 1, a000034. [Google Scholar] [CrossRef]
  28. He, D.K.; Shao, Y.R.; Zhang, L.; Shen, J.; Zhong, Z.Y.; Wang, J.; Xu, G. Adenovirus-delivered angiopoietin-1 suppresses NF-kappaB and p38 MAPK and attenuates inflammatory responses in phosgene-induced acute lung injury. Inhal. Toxicol. 2014, 26, 185–192. [Google Scholar] [CrossRef]
  29. Valderrama, J.A.; Riestra, A.M.; Gao, N.J.; LaRock, C.N.; Gupta, N.; Ali, S.R.; Hoffman, H.M.; Ghosh, P.; Nizet, V. Group A streptococcal M protein activates the NLRP3 inflammasome. Nat. Microbiol. 2017, 2, 1425–1434. [Google Scholar] [CrossRef] [Green Version]
  30. He, D.K.; Chen, J.F.; Shao, Y.R.; Zhou, F.Q.; Shen, J. Adenovirus-delivered angiopoietin-1 ameliorates phosgene-induced acute lung injury via inhibition of NLRP3 inflammasome activation. Inhal. Toxicol. 2018, 30, 187–194. [Google Scholar] [CrossRef]
  31. He, D.K.; Xu, N.; Shao, Y.R.; Shen, J. NLRP3 gene silencing ameliorates phosgene-induced acute lung injury in rats by inhibiting NLRP3 inflammasome and proinflammatory factors, but not anti-inflammatory factors. J. Toxicol. Sci. 2020, 45, 625–637. [Google Scholar] [CrossRef]
  32. Wiegman, C.H.; Li, F.; Ryffel, B.; Togbe, D.; Chung, K.F. Oxidative stress in ozone-induced chronic lung inflammation and emphysema: A facet of chronic obstructive pulmonary disease. Front. Immunol. 2020, 11, 1957. [Google Scholar] [CrossRef] [PubMed]
  33. Rendell, R.; Fairhall, S.; Graham, S.; Rutter, S.; Auton, P.; Smith, A.; Perrott, R.; Jugg, B. Assessment of N-acetylcysteine as a therapy for phosgene-induced acute lung injury. Toxicol. Lett. 2018, 290, 145–152. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, P.; Ye, X.L.; Liu, R.; Chen, H.L.; Liang, X.; Li, W.L.; Zhang, X.D.; Qin, X.J.; Bai, H.; Zhang, W.; et al. Mechanism of acute lung injury due to phosgene exposition and its protection by cafeic acid phenethyl ester in the rat. Exp. Toxicol. Pathol. 2013, 65, 311–318. [Google Scholar] [CrossRef] [PubMed]
  35. Sciuto, A.M.; Cascio, M.B.; Moran, T.S.; Forster, J.S. The fate of antioxidant enzymes in bronchoalveolar lavage fluid over 7 days in mice with acute lung injury. Inhal. Toxicol. 2003, 15, 675–685. [Google Scholar] [CrossRef]
  36. Sajadimajd, S.; Khazaei, M. Oxidative stress and cancer: The role of Nrf2. Curr. Cancer Drug Targets 2018, 18, 538–557. [Google Scholar] [CrossRef]
  37. Jaskot, R.H.; Grose, E.C.; Richards, J.H.; Doerfler, D.L. Effects of inhaled phosgene on rat lung antioxidant systems. Fundam. Appl. Toxicol. 1991, 17, 666–674. [Google Scholar] [CrossRef]
  38. He, Z.; Yang, X.; Yang, C. Extracorporeal membrane oxygenation for acute respiratory distress syndrome caused by acute phosgene poisoning: A report of 4 cases. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2019, 31, 232–235. [Google Scholar]
  39. Qiu, S.B.; Hu, Y.T.; He, Q.; Zhu, R.K. Investigation and analysis of 7 cases of acute lung injury caused by a welding operation. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2019, 37, 60–62. [Google Scholar]
  40. Vandewalle, J.; Luypaert, A.; De Bosscher, K.; Libert, C. Therapeutic mechanisms of glucocorticoids. Trends Endocrinol. Metab. 2018, 29, 42–54. [Google Scholar] [CrossRef]
  41. Desmet, S.J.; De Bosscher, K. Glucocorticoid receptors: Finding the middle ground. J. Clin. Investig. 2017, 127, 1136–1145. [Google Scholar] [CrossRef]
  42. He, D.K.; Shen, J.; Zhang, L.; Huang, W.B. Effects of dexamethasone pretreatment on expression of matrix metalloproteinase-9 in rats with acute lung injury induced by phosgene. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2011, 29, 289–293. [Google Scholar]
  43. Zhou, F.Q.; He, D.K.; Shao, Y.R.; Shen, J. The effects of methylprednisolone on NLRP3 inflammasome in rats with acute lung injury Induced by Phosgene. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2018, 36, 573–579. [Google Scholar]
  44. Smith, A.; Brown, R.; Jugg, B.; Platt, J.; Mann, T.; Masey, C.; Jenner, J.; Rice, P. The effect of steroid treatment with inhaled budesonide or intravenous methylprednisolone on phosgene-induced acute lung injury in a porcine model. Mil. Med. 2009, 174, 1287–1294. [Google Scholar] [CrossRef] [Green Version]
  45. Liu, F.; Pauluhn, J.; Trubel, H.; Wang, C. Single high-dose dexamethasone and sodium salicylate failed to attenuate phosgene-induced acute lung injury in rats. Toxicology 2014, 315, 17–23. [Google Scholar] [CrossRef]
  46. Luo, S.; Pauluhn, J.; Trubel, H.; Wang, C. Corticosteroids found ineffective for phosgene-induced acute lung injury in rats. Toxicol. Lett. 2014, 229, 85–92. [Google Scholar] [CrossRef]
  47. Liu, B.Z.; Hai, C.X.; Li, W.L. Clinical diagnosis and treatment of acute phosgene poisoning and its research progress. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2020, 38, 66–70. [Google Scholar] [PubMed]
  48. Li, S.T.; Dai, Q.; Zhang, S.X.; Liu, Y.J.; Yu, Q.Q.; Tan, F.; Lu, S.H.; Wang, Q.; Chen, J.W.; Huang, H.Q.; et al. Ulinastatin attenuates LPS-induced inflammation in mouse macrophage RAW264.7 cells by inhibiting the JNK/NF-kappaB signaling pathway and activating the PI3K/Akt/Nrf2 pathway. Acta Pharmacol. Sin. 2018, 39, 1294–1304. [Google Scholar] [CrossRef] [PubMed]
  49. Wei, F.; Liu, S.; Luo, L.; Gu, N.; Zeng, Y.; Chen, X.; Xu, S.; Zhang, D. Anti-inflammatory mechanism of ulinastatin: Inhibiting the hyperpermeability of vascular endothelial cells induced by TNF-alpha via the RhoA/ROCK signal pathway. Int. Immunopharmacol. 2017, 46, 220–227. [Google Scholar] [CrossRef] [PubMed]
  50. Shen, J.; Gan, Z.; Zhao, J.; Zhang, L.; Xu, G. Ulinastatin reduces pathogenesis of phosgene-induced acute lung injury in rats. Toxicol. Ind. Health 2014, 30, 785–793. [Google Scholar] [CrossRef]
  51. Li, J.; Wang, J.; Zhong, Z.; He, D.; Zhang, J.; Shen, J. Dynamic changes of a group of cytokines in phosgene-induced lung injury and the function of ulinastatin. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2014, 32, 813–818. [Google Scholar] [PubMed]
  52. Liu, F.; Li, W.; Pauluhn, J.; Trubel, H.; Wang, C. Rat models of acute lung injury: Exhaled nitric oxide as a sensitive, noninvasive real-time biomarker of prognosis and efficacy of intervention. Toxicology 2013, 310, 104–114. [Google Scholar] [CrossRef]
  53. Zheng, H.; Liang, W.; He, W.; Huang, C.; Chen, Q.; Yi, H.; Long, L.; Deng, Y.; Zeng, M. Ghrelin attenuates sepsis-induced acute lung injury by inhibiting the NF-kappaB, iNOS, and Akt signaling in alveolar macrophages. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 317, L381–L391. [Google Scholar] [CrossRef]
  54. Ma, H.; Mo, S.; Yi, Q.; Lai, J.; Liu, H.; Shi, Z. Role and mechanism of maresin-1 in acute lung injury induced by trauma-hemorrhagic shock. Med. Sci. Monit. 2020, 26, e923518. [Google Scholar] [CrossRef]
  55. Luo, S.; Trubel, H.; Wang, C.; Pauluhn, J. Phosgene- and chlorine-induced acute lung injury in rats: Comparison of cardiopulmonary function and biomarkers in exhaled breath. Toxicology 2014, 326, 109–118. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, H.L.; Bai, H.; Xi, M.M.; Liu, R.; Qin, X.J.; Liang, X.; Zhang, W.; Zhang, X.D.; Li, W.L.; Hai, C.X. Ethyl pyruvate protects rats from phosgene-induced pulmonary edema by inhibiting cyclooxygenase2 and inducible nitric oxide synthase expression. J. Appl. Toxicol. 2013, 33, 71–77. [Google Scholar] [CrossRef]
  57. Filipczak, P.T.; Senft, A.P.; Seagrave, J.; Weber, W.; Kuehl, P.J.; Fredenburgh, L.E.; McDonald, J.D.; Baron, R.M. NOS-2 inhibition in phosgene-induced acute lung injury. Toxicol. Sci. 2015, 146, 89–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Cipolla-Neto, J.; Amaral, F.G.D. Melatonin as a hormone: New physiological and clinical insights. Endocr. Rev. 2018, 39, 990–1028. [Google Scholar] [CrossRef] [Green Version]
  59. Zhang, Y.; Li, X.; Grailer, J.J.; Wang, N.; Wang, M.; Yao, J.; Zhong, R.; Gao, G.F.; Ward, P.A.; Tan, D.X.; et al. Melatonin alleviates acute lung injury through inhibiting the NLRP3 inflammasome. J. Pineal Res. 2016, 60, 405–414. [Google Scholar] [CrossRef]
  60. Aliasgharzadeh, A.; Farhood, B.; Amini, P.; Saffar, H.; Motevaseli, E.; Rezapoor, S.; Nouruzi, F.; Shabeeb, D.H.; Eleojo Musa, A.; Mohseni, M.; et al. Melatonin attenuates upregulation of Duox1 and Duox2 and protects against lung injury following chest irradiation in rats. Cell J. 2019, 21, 236–242. [Google Scholar]
  61. Zhang, L.; Zhang, F.; He, D.; Xu, D.; Zhong, Z.; Shen, J. Melatonin attenuates phosgene-induced acute lung injury via the upregulation Wnt/beta-catenin pathway. Int. J. Clin. Exp. Pathol. 2017, 10, 11281–11287. [Google Scholar] [PubMed]
  62. Zhang, L.; He, D.; Shao, Y.; Xu, D.; Shen, J. Effect of melatonin on p38MAPKsignaling pathway in rats with phosgene-induced lung injury. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2014, 32, 648–652. [Google Scholar] [PubMed]
  63. Zhang, L.; Shen, J.; Gan, Z.Y.; He, D.K.; Zhong, Z.Y. Protective effect of melatonin in rats with phosgene-induced lung injury. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2012, 30, 834–838. [Google Scholar]
  64. Gutbier, B.; Neuhauss, A.K.; Reppe, K.; Ehrler, C.; Santel, A.; Kaufmann, J.; Scholz, M.; Weissmann, N.; Morawietz, L.; Mitchell, T.J.; et al. Prognostic and pathogenic role of angiopoietin-1 and -2 in pneumonia. Am. J. Respir. Crit. Care Med. 2018, 198, 220–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liu, P.; Ryczko, M.; Xie, X.; Baardsnes, J.; Lord-Dufour, S.; Duroche, Y.; Hicks, E.A.; Taiyab, A.; Sheardown, H.; Quaggin, S.E.; et al. New soluble angiopoietin analog of Hepta-ANG1 prevents pathological vascular leakage. Biotechnol. Bioeng. 2021, 118, 423–432. [Google Scholar] [CrossRef] [PubMed]
  66. Kim, S.R.; Lee, K.S.; Park, S.J.; Min, K.H.; Lee, K.Y.; Choe, Y.H.; Hong, S.H.; Koh, G.Y.; Lee, Y.C. Angiopoietin-1 variant, COMP-Ang1 attenuates hydrogen peroxide-induced acute lung injury. Exp. Mol. Med. 2008, 40, 320–331. [Google Scholar] [CrossRef]
  67. Mei, S.H.; McCarter, S.D.; Deng, Y.; Parker, C.H.; Liles, W.C.; Stewart, D.J. Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med. 2007, 4, e269. [Google Scholar] [CrossRef]
  68. Yuan, Z.; Zhao, J.; Shen, J. The changes of the ratio of angiopoietin-2 to angiopoietin-1 in the acute lung injury induced by phosgene in rats. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2011, 29, 608–610. [Google Scholar]
  69. Shen, J.; Wang, J.; Shao, Y.R.; He, D.K.; Zhang, L.; Nadeem, L.; Xu, G. Adenovirus-delivered angiopoietin-1 treatment for phosgene-induced acute lung injury. Inhal. Toxicol. 2013, 25, 272–279. [Google Scholar] [CrossRef]
  70. Zhang, R.H.; Li, C.H.; Wang, C.L.; Xu, M.J.; Xu, T.; Wei, D.; Liu, B.J.; Wang, G.H.; Tian, S.F. N-acetyl-l-cystine (NAC) protects against H9N2 swine influenza virus-induced acute lung injury. Int. Immunopharmacol. 2014, 22, 1–8. [Google Scholar] [CrossRef]
  71. Wang, Q.; Zhu, H.; Zhou, W.G.; Guo, X.C.; Wu, M.J.; Xu, Z.Y.; Jiang, J.F.; Shen, C.; Liu, H.Q. N-acetylcysteine-pretreated human embryonic mesenchymal stem cell administration protects against bleomycin-induced lung injury. Am. J. Med. Sci. 2013, 346, 113–122. [Google Scholar] [CrossRef]
  72. Ji, L.; Liu, R.; Zhang, X.D.; Chen, H.L.; Bai, H.; Wang, X.; Zhao, H.L.; Liang, X.; Hai, C.X. N-acetylcysteine attenuates phosgene-induced acute lung injury via up-regulation of Nrf2 expression. Inhal. Toxicol. 2010, 22, 535–542. [Google Scholar] [CrossRef]
  73. Zhang, P.; Tang, Y.; Li, N.G.; Zhu, Y.; Duan, J.A. Bioactivity and chemical synthesis of caffeic acid phenethyl ester and its derivatives. Molecules 2014, 19, 16458–16476. [Google Scholar] [CrossRef] [Green Version]
  74. Rainsford, K.D. Ibuprofen: Pharmacology, efficacy and safety. Inflammopharmacology 2009, 17, 275–342. [Google Scholar] [CrossRef]
  75. Kantor, T.G. Ibuprofen. Ann. Intern. Med. 1979, 91, 877–882. [Google Scholar] [CrossRef]
  76. Sciuto, A.M.; Stotts, R.R.; Hurt, H.H. Efficacy of ibuprofen and pentoxifylline in the treatment of phosgene-induced acute lung injury. J. Appl. Toxicol. 1996, 16, 381–384. [Google Scholar] [CrossRef]
  77. Kennedy, T.P.; Rao, N.V.; Noah, W.; Michael, J.R.; Jafri, M.H., Jr.; Gurtner, G.H.; Hoidal, J.R. Ibuprofen prevents oxidant lung injury and in vitro lipid peroxidation by chelating iron. J. Clin. Investig. 1990, 86, 1565–1573. [Google Scholar] [CrossRef] [Green Version]
  78. Holmes, W.W.; Keyser, B.M.; Paradiso, D.C.; Ray, R.; Andres, D.K.; Benton, B.J.; Rothwell, C.C.; Hoard-Fruchey, H.M.; Dillman, J.F.; Sciuto, A.M.; et al. Conceptual approaches for treatment of phosgene inhalation-induced lung injury. Toxicol. Lett. 2016, 244, 8–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Tobias, L.D.; Hamilton, J.G. The effect of 5,8,11,14-eicosatetraynoic acid on lipid metabolism. Lipids 1979, 14, 181–193. [Google Scholar] [CrossRef] [PubMed]
  80. Sciuto, A.M. Posttreatment with ETYA protects against phosgene-induced lung injury by amplifying the glutathione to lipid peroxidation ratio. Inhal. Toxicol. 2000, 12, 347–356. [Google Scholar] [CrossRef] [PubMed]
  81. Banner, K.H.; Igney, F.; Poll, C. TRP channels: Emerging targets for respiratory disease. Pharmacol. Ther. 2011, 130, 371–384. [Google Scholar] [CrossRef]
  82. Prandini, P.; De Logu, F.; Fusi, C.; Provezza, L.; Nassini, R.; Montagner, G.; Materazzi, S.; Munari, S.; Gilioli, E.; Bezzerri, V.; et al. Transient receptor potential ankyrin 1 channels modulate inflammatory response in respiratory cells from patients with cystic fibrosis. Am. J. Respir. Cell Mol. Biol. 2016, 55, 645–656. [Google Scholar] [CrossRef] [PubMed]
  83. Earley, S.; Brayden, J.E. Transient receptor potential channels in the vasculature. Physiol. Rev. 2015, 95, 645–690. [Google Scholar] [CrossRef] [Green Version]
  84. Andres, D.; Keyser, B.; Benton, B.; Melber, A.; Olivera, D.; Holmes, W.; Paradiso, D.; Anderson, D.; Ray, R. Transient receptor potential (TRP) channels as a therapeutic target for intervention of respiratory effects and lethality from phosgene. Toxicol. Lett. 2016, 244, 21–27. [Google Scholar] [CrossRef] [Green Version]
  85. Ding, D.C.; Shyu, W.C.; Lin, S.Z. Mesenchymal stem cells. Cell Transplant. 2011, 20, 5–14. [Google Scholar] [CrossRef] [Green Version]
  86. Lee, J.W.; Fang, X.; Krasnodembskaya, A.; Howard, J.P.; Matthay, M.A. Concise review: Mesenchymal stem cells for acute lung injury: Role of paracrine soluble factors. Stem Cells 2011, 29, 913–919. [Google Scholar] [CrossRef] [Green Version]
  87. Sengupta, V.; Sengupta, S.; Lazo, A.; Woods, P.; Nolan, A.; Bremer, N. Exosomes derived from bone marrow mesenchymal stem cells as treatment for severe COVID-19. Stem Cells Dev. 2020, 29, 747–754. [Google Scholar] [CrossRef]
  88. Le Blanc, K.; Frassoni, F.; Ball, L.; Locatelli, F.; Roelofs, H.; Lewis, I.; Lanino, E.; Sundberg, B.; Bernardo, M.E.; Remberger, M.; et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: A phase II study. Lancet 2008, 371, 1579–1586. [Google Scholar] [CrossRef]
  89. Shi, L.; Huang, H.; Lu, X.; Yan, X.; Jiang, X.; Xu, R.; Wang, S.; Zhang, C.; Yuan, X.; Xu, Z.; et al. Effect of human umbilical cord-derived mesenchymal stem cells on lung damage in severe COVID-19 patients: A randomized, double-blind, placebo-controlled phase 2 trial. Signal Transduct. Target. Ther. 2021, 6, 58. [Google Scholar] [CrossRef] [PubMed]
  90. Wilson, J.G.; Liu, K.D.; Zhuo, H.; Caballero, L.; McMillan, M.; Fang, X.; Cosgrove, K.; Vojnik, R.; Calfee, C.S.; Lee, J.W.; et al. Mesenchymal stem (stromal) cells for treatment of ARDS: A phase 1 clinical trial. Lancet Respir. Med. 2015, 3, 24–32. [Google Scholar] [CrossRef] [Green Version]
  91. Chen, J.; Shao, Y.; Xu, G.; Lim, C.; Li, J.; Xu, D.; Shen, J. Bone marrow-derived mesenchymal stem cells attenuate phosgene-induced acute lung injury in rats. Inhal. Toxicol. 2015, 27, 254–261. [Google Scholar] [CrossRef]
  92. Zhang, J.; Shao, Y.; He, D.; Zhang, L.; Xu, G.; Shen, J. Evidence that bone marrow-derived mesenchymal stem cells reduce epithelial permeability following phosgene-induced acute lung injury via activation of wnt3a protein-induced canonical wnt/beta-catenin signaling. Inhal. Toxicol. 2016, 28, 572–579. [Google Scholar] [CrossRef]
  93. Zepp, J.A.; Zacharias, W.J.; Frank, D.B.; Cavanaugh, C.A.; Zhou, S.; Morley, M.P.; Morrisey, E.E. Distinct mesenchymal lineages and niches promote epithelial self-renewal and myofibrogenesis in the lung. Cell 2017, 170, 1134–1148.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ye, K.; He, D.; Shao, Y.; Xu, N.; Jin, C.; Zhang, L.; Shen, J. Exogenous mesenchymal stem cells affect the function of endogenous lung stem cells (club cells) in phosgene-induced lung injury. Biochem. Biophys. Res. Commun. 2019, 514, 586–592. [Google Scholar] [CrossRef] [PubMed]
  95. Szydlak, R. Mesenchymal stem cells’ homing and cardiac tissue repair. Acta Biochim. Pol. 2019, 66, 483–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Jin, C.; Zhou, F.; Zhang, L.; Shen, J. Overexpression of heat shock protein 70 enhanced mesenchymal stem cell treatment efficacy in phosgene-induced acute lung injury. J. Biochem. Mol. Toxicol. 2020, 34, e22515. [Google Scholar] [CrossRef]
  97. Naumann, U.; Cameroni, E.; Pruenster, M.; Mahabaleshwar, H.; Raz, E.; Zerwes, H.G.; Rot, A.; Thelen, M. CXCR7 functions as a scavenger for CXCL12 and CXCL11. PLoS ONE 2010, 5, e9175. [Google Scholar] [CrossRef] [Green Version]
  98. Shao, Y.; Zhou, F.; He, D.; Zhang, L.; Shen, J. Overexpression of CXCR7 promotes mesenchymal stem cells to repair phosgene-induced acute lung injury in rats. Biomed. Pharmacother. 2019, 109, 1233–1239. [Google Scholar] [CrossRef]
  99. Lou, G.; Chen, Z.; Zheng, M.; Liu, Y. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp. Mol. Med. 2017, 49, e346. [Google Scholar] [CrossRef]
  100. Lee, J.H.; Park, J.; Lee, J.W. Therapeutic use of mesenchymal stem cell-derived extracellular vesicles in acute lung injury. Transfusion 2019, 59, 876–883. [Google Scholar] [CrossRef] [Green Version]
  101. Xu, N.; Shao, Y.; Ye, K.; Qu, Y.; Memet, O.; He, D.; Shen, J. Mesenchymal stem cell-derived exosomes attenuate phosgene-induced acute lung injury in rats. Inhal. Toxicol. 2019, 31, 52–60. [Google Scholar] [CrossRef] [PubMed]
  102. Xu, N.; He, D.; Shao, Y.; Qu, Y.; Ye, K.; Memet, O.; Zhang, L.; Shen, J. Lung-derived exosomes in phosgene-induced acute lung injury regulate the functions of mesenchymal stem cells partially via miR-28–5p. Biomed. Pharmacother. 2020, 121, 109603. [Google Scholar] [CrossRef] [PubMed]
  103. Parseghian, M.H.; Hobson, S.T.; Richieri, R.A. Targeted heat shock protein 72 for pulmonary cytoprotection. Ann. N. Y. Acad. Sci. 2016, 1374, 78–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Hobson, S.T.; Richieri, R.A.; Parseghian, M.H. Phosgene: Toxicology, animal models, and medical countermeasures. Toxicol. Mech. Methods 2021, 31, 293–307. [Google Scholar] [CrossRef] [PubMed]
  105. Beere, H.M.; Wolf, B.B.; Cain, K.; Mosser, D.D.; Mahboubi, A.; Kuwana, T.; Tailor, P.; Morimoto, R.I.; Cohen, G.M.; Green, D.R. Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome. Nat. Cell Biol. 2000, 2, 469–475. [Google Scholar] [CrossRef]
  106. Zheng, Z.; Kim, J.Y.; Ma, H.; Lee, J.E.; Yenari, M.A. Anti-inflammatory effects of the 70 kDa heat shock protein in experimental stroke. J. Cereb. Blood Flow Metab. 2008, 28, 53–63. [Google Scholar] [CrossRef] [Green Version]
  107. Tanimoto, T.; Parseghian, M.H.; Nakahara, T.; Kawai, H.; Narula, N.; Kim, D.; Nishimura, R.; Weisbart, R.H.; Chan, G.; Richieri, R.A.; et al. Cardioprotective Effects of HSP72 administration on ischemia-reperfusion Injury. J. Am. Coll. Cardiol. 2017, 70, 1479–1492. [Google Scholar] [CrossRef]
  108. Lei, J.; Wei, Y.; Song, P.; Li, Y.; Zhang, T.; Feng, Q.; Xu, G. Cordycepin inhibits LPS-induced acute lung injury by inhibiting inflammation and oxidative stress. Eur. J. Pharmacol. 2018, 818, 110–114. [Google Scholar] [CrossRef]
  109. Beltran-Garcia, J.; Osca-Verdegal, R.; Pallardo, F.V.; Ferreres, J.; Rodriguez, M.; Mulet, S.; Sanchis-Gomar, F.; Carbonell, N.; Garcia-Gimenez, J.L. Oxidative stress and inflammation in COVID-19-associated sepsis: The potential role of anti-oxidant therapy in avoiding disease progression. Antioxidants 2020, 9, 936. [Google Scholar] [CrossRef]
  110. De Carvalho, F.O.; Felipe, F.A.; de Melo Costa, A.C.; Teixeira, L.G.; Silva, E.R.; Nunes, P.S.; Shanmugam, S.; de Lucca Junior, W.; Quintans, J.S.; de Souza Araujo, A.A. Inflammatory mediators and oxidative stress in animals subjected to smoke inhalation: A systematic review. Lung 2016, 194, 487–499. [Google Scholar] [CrossRef]
  111. Wang, H.; Zhang, W.; Liu, J.; Gao, J.; Fang, L.E.; Liu, Z.; Xia, B.; Fan, X.; Li, C.; Lu, Q.; et al. NF-kappaB and FosB mediate inflammation and oxidative stress in the blast lung injury of rats exposed to shock waves. Acta Biochim. Biophys. Sin. 2021, 53, 283–293. [Google Scholar] [CrossRef] [PubMed]
  112. Zhou, Y.; Hou, Y.; Shen, J.; Mehra, R.; Kallianpur, A.; Culver, D.A.; Gack, M.U.; Farha, S.; Zein, J.; Comhair, S.; et al. A network medicine approach to investigation and population-based validation of disease manifestations and drug repurposing for COVID-19. PLoS Biol. 2020, 18, e3000970. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The mechanisms of phosgene-induced acute lung injury.
Figure 1. The mechanisms of phosgene-induced acute lung injury.
Ijms 22 10933 g001
Figure 2. The function of MSCs in P-ALI.
Figure 2. The function of MSCs in P-ALI.
Ijms 22 10933 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lu, Q.; Huang, S.; Meng, X.; Zhang, J.; Yu, S.; Li, J.; Shi, M.; Fan, H.; Zhao, Y. Mechanism of Phosgene-Induced Acute Lung Injury and Treatment Strategy. Int. J. Mol. Sci. 2021, 22, 10933. https://doi.org/10.3390/ijms222010933

AMA Style

Lu Q, Huang S, Meng X, Zhang J, Yu S, Li J, Shi M, Fan H, Zhao Y. Mechanism of Phosgene-Induced Acute Lung Injury and Treatment Strategy. International Journal of Molecular Sciences. 2021; 22(20):10933. https://doi.org/10.3390/ijms222010933

Chicago/Turabian Style

Lu, Qianying, Siyu Huang, Xiangyan Meng, Jianfeng Zhang, Sifan Yu, Junfeng Li, Mingyu Shi, Haojun Fan, and Yanmei Zhao. 2021. "Mechanism of Phosgene-Induced Acute Lung Injury and Treatment Strategy" International Journal of Molecular Sciences 22, no. 20: 10933. https://doi.org/10.3390/ijms222010933

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop