Hostname: page-component-848d4c4894-5nwft Total loading time: 0 Render date: 2024-05-14T16:10:09.752Z Has data issue: false hasContentIssue false

Targeting Tumor Necrosis Factor Alpha to Mitigate Lung Injury Induced by Mustard Vesicants and Radiation

Published online by Cambridge University Press:  18 October 2023

Rama Malaviya
Affiliation:
Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy; Rutgers University, Piscataway, NJ, USA
Jeffrey D. Laskin
Affiliation:
Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, NJ, USA
Rita Businaro
Affiliation:
Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
Debra L. Laskin*
Affiliation:
Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy; Rutgers University, Piscataway, NJ, USA
*
Corresponding author: Debra L. Laskin; Email: laskin@eohsi.rutgers.edu
Rights & Permissions [Opens in a new window]

Abstract

Pulmonary injury induced by mustard vesicants and radiation is characterized by DNA damage, oxidative stress, and inflammation. This is associated with increases in levels of inflammatory mediators, including tumor necrosis factor (TNF)α in the lung and upregulation of its receptor TNFR1. Dysregulated production of TNFα and TNFα signaling has been implicated in lung injury, oxidative and nitrosative stress, apoptosis, and necrosis, which contribute to tissue damage, chronic inflammation, airway hyperresponsiveness, and tissue remodeling. These findings suggest that targeting production of TNFα or TNFα activity may represent an efficacious approach to mitigating lung toxicity induced by both mustards and radiation. This review summarizes current knowledge on the role of TNFα in pathologies associated with exposure to mustard vesicants and radiation, with a focus on the therapeutic potential of TNFα-targeting agents in reducing acute injury and chronic disease pathogenesis.

Type
Systematic Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© Debra Laskin, 2023. Published by Cambridge University Press on behalf of Society for Disaster Medicine and Public Health, Inc

Mustard vesicants and ionizing radiation are cytotoxic to the lung, causing progressive injury at low to moderate doses and lethality at high doses. Reference Graham and Schoneboom1Reference Rajan Radha and Chandrasekharan3 Acute lung injury, pulmonary edema, respiratory epithelial necrosis and sloughing, and pneumonitis are noted within days to weeks of exposure, whereas chronic bronchitis, asthma, bronchiectasis, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, and/or cancer are observed 6–12 months post-radiation or 10–30 years after mustard exposure. Reference Rajan Radha and Chandrasekharan3,Reference Weinberger, Malaviya and Sunil4 A common feature of acute injury and chronic disease induced by mustards and radiation is an accumulation of inflammatory cells within the lung and release of cytokines such as tumor necrosis factor (TNF)α. Reference Malaviya, Heck, Casillas, Lukey, Romano and Salem5Reference Smith, Venosa and Gow7 TNFα is an early response cytokine important in initiating inflammatory responses; it also promotes cellular proliferation and tissue regeneration. Reference Holbrook, Lara-Reyna, Jarosz-Griffiths and McDermott8 Excessive production of TNFα is associated with uncontrolled inflammation and disease pathogenesis. In this context, elevated levels of TNFα have been identified in a number of inflammatory diseases, including COPD, rheumatoid arthritis, psoriasis, and inflammatory bowel disease; moreover, the administration of biologics, which block TNFα, has demonstrated therapeutic efficacy against various pathologies and diseases. Reference Bahia and Silakari9,Reference Jang, Lee and Shin10 In this review, we discuss the role of TNFα in mustard vesicant- and radiation-induced pulmonary disease pathogenesis, with a focus on the therapeutic potential of TNFα-targeting agents in mitigating toxicity.

TNFα Production, Receptors, and Biological Activity

TNFα is primarily produced by macrophages in response to tissue injury or infection. Reference Aggarwal, Gupta and Kim11,Reference Zelová and Hošek12 Synthesized as a transmembrane homotrimer consisting of three 26 KDa subunits (mTNFα), it is cleaved by TNFα-converting enzyme (TACE) to soluble TNFα (sTNFα), a homotrimer consisting of three 17 KDa subunits. Reference Horiuchi, Mitoma and Harashima13 The activity of both mTNFα and sTNFα is mediated by binding to cell surface receptors identified as type 1 (TNFR1) and type 2 (TNFR2) (Figure 1). TNFR1 is expressed on the surface of most cell types, whereas TNFR2 is largely restricted to immune cells and endothelial cells. Reference Zelová and Hošek12 Both forms of TNFα bind to TNFR1 and TNFR2. However, TNFR2 binds to TNFα with lower affinity and may easily dissociate from the ligand. Reference Zelová and Hošek12,Reference Tracey, Klareskog and Sasso14 Thus, it appears that the biological activity of TNFα mainly involves TNFR1 signaling. Reference Jang, Lee and Shin10,Reference Aggarwal15,Reference Grell, Douni and Wajant16 Reports also suggest that signaling pathways activated by these 2 receptors overlap or they transduce signaling cooperatively as genetic deletion of either receptor blocks signaling initiated by TNFα. Reference Aggarwal15,Reference Mukhopadhyay, Suttles, Stout and Aggarwal17 Ligand binding to TNFR1 initiates signaling, resulting in the activation of mitogen activated protein (MAP) kinases and transcription factors, including AP-1 and nuclear factor-kappa B (NF-κB), which regulate genes involved in inflammation, cell proliferation, and differentiation. Reference Jang, Lee and Shin10,Reference Aggarwal15 TNFR1 also recruits TNFR1-associated death domain (TRADD) protein, which promotes cell death. TNFR2 recruits TNFR-associated factor (TRAF)-1 and TRAF-2 proteins, resulting in the activation of MAP kinases, NF-κB, and protein kinase B. Reference Jang, Lee and Shin10,Reference Aggarwal15 Functionally, TNFR1 activation is associated with the induction of cytotoxic and proinflammatory responses of TNFα, whereas TNFR2 mediates homeostatic bioactivities, including tissue regeneration, cell proliferation, and cell survival. Reference Aggarwal15,Reference Probert18

Figure 1. Tumor necrosis factor (TNF)α signaling. Binding of soluble or membrane bound TNFα to TNFR1 and TNFR2 initiates signaling events associated with apoptosis or activation of transcription factors, NFκB and AP-1. TNFα binding to TNFR1 and/or TNFR2 can also activate protein kinase B/Akt, which leads to prolonged NF-κB activation. Together, these responses contribute to inflammation, leukocyte trafficking, cell death, cell proliferation, and tissue remodeling.

AP-1, activator protein-1; IKK, IκB kinase; MAPKs, mitogen-associated protein kinases; mTNF, membrane-bound TNFα; NFκB, nuclear factor kappa B; sTNF, soluble TNFα; TACE, TNFα-converting enzyme; TNFR1, TNFα receptor 1; TNFR2, TNFα receptor 2; TRADD, TNFR associated death domain; TRAF1, TNFR associated factor 1; TRAF2, TNFR associated factor 2.

TNFα is a master regulator of inflammation generated early after injury or infection in response to bacterially derived lipopolysaccharide, as well as interleukin (IL)-1, interferon-γ, granulocyte macrophage colony stimulating factor, platelet derived growth factor, and TNFα itself. Reference Malaviya, Laskin and Laskin19Reference Sabio and Davis21 The biological actions of TNFα are varied and summarized in Table 1. TNFα promotes inflammation by upregulating adhesion molecules important in leukocyte trafficking to inflammatory sites, including intracellular leukocyte adhesion molecule, endothelial leukocyte adhesion molecule-1, and vascular cell adhesion molecule-1, and by stimulating the release of macrophage and neutrophil chemokines, such as CXCL8 (IL-8), CCL2 (MCP)-1, interferon-inducible protein 10 (IP-10) (CXCL10), as well as bioactive lipids (eg, eicosanoids and platelet activating factor), which promote vasodilatation, leukocyte adhesion, and chemotaxis. Reference Zelová and Hošek12,Reference Semenzato22Reference Kelly, Hwang and Kubes25 TNFα also stimulates phagocytic cells to release proinflammatory cytokines (eg, IL-1, IL-6, IL-12, IL-15, IL-23, and TNFα) and reactive oxygen and nitrogen species. In the lung, these cytotoxic/proinflammatory mediators cause alveolar epithelial cell injury, denudation of the basement membrane, hyalin membrane formation, impaired surfactant activity, and altered pulmonary functioning. Reference Malaviya, Laskin and Laskin19,Reference Shimabukuro, Sawa and Gropper26,Reference Laskin, Malaviya and Laskin27

Table 1. Biological activities of tumor necrosis factor (TNF)α in mustard or radiation-induced lung injury

Arg, arginase; CCL, C-C chemokine ligand; CCR, C-C chemokine receptor; COX, cyclooxygenase; CTGF, connective tissue growth factor; CXCL, C-X-C chemokine ligand; CXCR, C-X-C chemokine receptor; Fas L, Fas ligand; Fas R, Fas receptor; FGF, fibroblast growth factor; γH2A.X, histone variant H2A.X; HMGB, high mobility group box; HO, heme oxygenase; H2O2, hydrogen peroxide; IFN, interferon; IL, interleukin; iNOS, inducible nitric oxide synthase; KC/GRO, keratinocyte chemoattractant/human growth-regulated oncogenes; LC3B, light chain 3B; MMP, matrix metalloproteinase; MR, mannose receptor; Gal, galactin; NO, nitric oxide; PARP, poly (ADP-ribose) polymerase; PCNA, proliferating cell nuclear antigen; PDGF, platelet-derived growth factor; PGI2, prostacyclin; SMA, smooth muscle actin; SOD, superoxide dismutase; sRAGE, soluble receptor for advanced glycation end product; TGF, tumor growth factor; TIMP, tissue inhibitors of metalloproteinases; TNF, tumor necrosis factor; TXA, thromboxane A.

TNFα is known to cause oxidative and nitrosative stress. Reference Rahman28,Reference Blaser, Dostert, Mak and Brenner29 It also depletes intracellular glutathione, which contributes to its pro-oxidant actions. Reference Rahman28,Reference Obrador, Navarro and Mompo30 Oxidative stress is associated with activation of redox sensitive transcription factors, including NF-κB and AP-1 that upregulate proinflammatory genes, further contributing to inflammation and tissue injury. Reference Aggarwal, Gupta and Kim11 TNFα is also a potent mitogen, stimulating epithelial cell proliferation. Reference Lu, Beuerman and Zhao31 This is thought to be due in part to activation of AP-1 and upregulation of cyclin-D1, a cell cycle regulatory protein. Reference Rahman28,Reference Mukhopadhyay, Mukherjee and Stone32 TNFα-induced proliferation leads to epithelial thickening and pulmonary fibrosis. Reference Allen and Spiteri33,Reference Sasaki, Kashima and Ito34 TNFα also promotes fibrosis by inducing focal accumulation of fibroblasts and collagen deposition and by stimulating the production of matrix metalloproteinases (MMPs) and transforming growth factor (TGF)β. Reference Sasaki, Kashima and Ito34Reference Oikonomou, Harokopos and Zalevsky37 In humans, circulating levels of MMP-9 and TGFβ correlate with the extent of fibrosis. Reference Kim, Min and Cho38 Collectively, these data suggest that blocking TNFα may be efficacious in mitigating mustard or radiation-induced acute lung injury and inflammation, as well as their long-term pulmonary complications.

Role of TNFα in Mustard-Induced Lung Injury

Mustard vesicants, including sulfur mustard (SM) and nitrogen mustard (NM), are cytotoxic alkylating agents that cause incapacitating injury to the respiratory tract. Reference Kristinsson and Johannesson39 Toxicity is largely due to its lipophilic nature that allows it to rapidly penetrate tissues and cells, and alkylate and cross-link cellular macromolecules, including nucleic acids, lipids, and proteins. Reference Malaviya, Laskin and Laskin40 Both conducting and respiratory airways are affected by mustards. Reference Graham and Schoneboom1,Reference Ghabili, Agutter and Ghanei41 Early symptoms include cough, hoarseness, sore throat, mucus discharge, loss of smell and taste, and irritation of the nasal mucosa. Reference Graham and Schoneboom1,Reference Kehe, Thiermann and Balszuweit42Reference Sezigen, Ivelik and Ortatatli44 Pulmonary edema and damage to the pharynx induced by acute mustard inhalation result in an inability to speak, moist rales, tachypnea, and tachycardia. Reference Sohrabpour45,Reference Kehe and Szinicz46 At high doses, necrosis of the respiratory epithelium, epithelial sloughing, pseudo-membrane formation, lung lobe collapse, and death occur. Reference Ghabili, Agutter and Ghanei41,Reference Kehe, Balszuweit and Emmler43,Reference Wang and Xia47,Reference Rancourt, Veress and Ahmad48

Chronic clinical and pathological manifestations of mustard exposure have been observed in survivors of chemical attacks and in manufacturing plant workers. The most common symptoms in long-term survivors of mustard gas exposure are chronic cough, dyspnea, increases in sputum and hemoptysis (airway bleeding), progressive airway deterioration, hyperreactivity, and stenosis of the conducting airways. Reference Graham and Schoneboom1,Reference Malaviya, Laskin and Laskin40,Reference Bijani and Moghadamnia49,Reference Weinberger, Laskin and Sunil50 Common pathologies in victims of the Iran–Iraq war include asthma, bronchitis, bronchiectasis, airway narrowing, COPD, and pulmonary fibrosis. Reference Malaviya, Laskin and Laskin40,Reference Weinberger, Laskin and Sunil50 Emphysema, bronchiectasis, centrilobular nodules, bronchial wall thickening, reticular opacity, ground glass opacity, consolidation, honeycombing, and other respiratory pathologies have similarly been reported in survivors of mustard gas exposure in a manufacturing factory. Reference Malaviya, Laskin and Laskin40,Reference Nishimura, Iwamoto and Ishikawa51Reference Mukaida, Hattori and Iwamoto53

Pulmonary injury from mustard exposure is associated with an accumulation of large numbers of inflammatory cells, including macrophages, neutrophils, and eosinophils at sites of injury in the lung, as well as oxidative stress and production of cytotoxic/proinflammatory cytokines, including TNFα. Reference Weinberger, Malaviya and Sunil4,Reference Malaviya, Laskin and Laskin40,Reference Macit, Yaren and Aydin54,Reference Das, Mukherjee, Smith and Chatterjee55 Levels of TNFR1 are also upregulated, suggesting a role of TNFα signaling through this receptor in the pathogenic response to mustards. Reference Sunil, Patel-Vayas and Shen56 Accumulating evidence described below provides support for this activity.

Rodent Models of Mustard Lung Injury Used to Investigate the Impact of Targeting TNFα

In initial mechanistic studies, 2-chloroethyl ethyl sulfide (CEES), a monofunctional analog of SM and NM, was used as a model for mustard lung toxicity. In rodents, CEES causes injury to the alveolar epithelial barrier as measured by increases in cells, protein, and IgM in bronchoalveolar lavage fluid (BAL); fibrinogen and prothrombin levels also increase, a response associated with impairment of fibrin-degrading activity in the lung. Reference Sunil, Patel-Vayas and Shen56,Reference Rancourt, Ahmad and Veress57 Proliferating cell nuclear antigen (PCNA), a marker of cellular proliferation in response to injury, Reference Arbel, Choudhary, Tfilin and Kupiec58 is upregulated after CEES administration to rodents, along with cyclin D1. Reference Mukhopadhyay, Mukherjee, Smith and Das59 CEES also causes pulmonary oxidative stress, characterized by increases in superoxide dismutase (SOD), Ym-1, and lipid peroxidation end products and decreases in intracellular glutathione levels. Reference Ng, Sim and Loke60Reference Sunil, Shen and Patel-Vayas62 Inflammatory proteins, including inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, TNFα, TNFR1, and CCR2 are also increased in the lung after CEES. Reference Sunil, Patel-Vayas and Shen56,Reference Sawale, Ambhore and Pawar61,Reference Sunil, Shen and Patel-Vayas62 These responses are linked to functional alterations, including decreases in lung compliance and increases in elastance. Reference Sunil, Patel-Vayas and Shen56,Reference Sunil, Shen and Patel-Vayas62 Additionally, methacholine-induced alterations in total lung resistance and central airway resistance are dampened following CEES exposure.

More recently, rodent models of lung injury and chronic disease using NM and SM have been developed as they more closely reflect pulmonary responses in humans. Reference Weinberger, Malaviya and Sunil4,Reference Tang and Loke63 In general, injury, oxidative stress, and inflammatory responses are similar to CEES, appearing early (1–3 days) after exposure; however, they are more severe and prolonged. Additionally, pulmonary fibrosis is observed, typically within 28 days of exposure. The increased pathologic response to SM and NM when compared to CEES is likely due to the fact that they are bifunctional alkylating agents, allowing them to induce DNA intrastrand and interstrand cross-links, as well as DNA-protein cross-links. Reference Jan, Heck, Laskin and Laskin64 These cross-links can alter the structure of DNA and interfere with replication and transcription. In contrast, monofunctional alkylation of DNA and proteins caused by CEES can more readily be repaired. Reference Abbotts and Wilson65

Owing to high reactivity of SM and NM with mucosal surfaces of the upper respiratory track, rodent models of lung injury involve intratracheal exposure to ensure delivery to the lower lungs, where most damage occurs in humans. Reference Malaviya, Sunil and Venosa66Reference Anderson, Byers and Vesely69 A single exposure of rats to SM or NM causes dose and time-related histopathological changes in the lungs, including multifocal lesions comprising perivascular and peribronchial edema, blood vessel hemorrhage, patchy mild thickening of alveolar septa, increased numbers of alveolar macrophages and neutrophils, and luminal accumulation of cellular debris and fibrin. Reference Malaviya, Sunil and Venosa66,Reference Malaviya, Abramova and Rancourt67,Reference Xiaoji, Xiao and Rui70Reference Sunil, Patel and Shen73 Bronchiolization of alveolar walls, indicating type I epithelial cell damage and repair by type II epithelial cells, hyperplasia, and hypertrophy of the bronchial epithelium leading to piling of bronchiolar epithelial cells have also been noted. SM also causes severe ulceration of the proximal bronchioles and deposits of fibrillar membranes in bronchiolar lumen, suggesting apoptosis and necrosis. Reference Malaviya, Abramova and Rancourt67,Reference Anderson, Yourick and Moeller74 Consistent with early SM-induced histopathologic evidence of acute lung injury and bronchiolar epithelial denudation, proteins involved in cell apoptosis and autophagy, including caspase-3, caspase-6, caspase-8, caspase-9, poly (ADP-ribose) polymerase (PARP)-1 and LC3BI and LC3BII, are upregulated in the lung; terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) positive cells are also present. Reference McGraw, Rioux and Garlick75Reference Andres, Keyser and Melber77

The chronic phase of respiratory injury caused by SM and NM in rats (beginning 28 days post-exposure) is characterized by a predominance of fibroblasts, neutrophils, lymphocytes, and enlarged foamy macrophages in the alveoli and/or alveolar septal walls, multifocal fibrotic lesions, and collagen deposition. Reference Malaviya, Laskin and Laskin40,Reference Malaviya, Sunil and Venosa78Reference Solopov, Biancatelli and Marinova80 Fibroplasia, squamous metaplasia of the bronchial wall, honeycombing, and emphysema-like changes in alveolar regions of the lung are also evident. Reference Malaviya, Laskin and Laskin40,Reference Malaviya, Abramova and Rancourt67,Reference McGraw, Dysart and Hendry-Hofer68,Reference Mishra, Rir-Sima-Ah and Grotendorst81 Hyperplasia and squamous metaplasia in ulcerated proximal bronchiolar epithelium are also observed after SM exposure, indicative of aberrant wound repair. Reference Malaviya, Abramova and Rancourt67 These structural changes are correlated with impaired pulmonary functioning. Reference Solopov, Biancatelli and Marinova80,Reference Sunil, Vayas and Abramova82 As observed with CEES, NM- and SM-induced lung injury and apoptosis/necrosis of epithelial cells are associated with increases in BAL cells, protein, IgM, fibrinogen, and total phospholipids. Reference Malaviya, Sunil and Venosa66,Reference Malaviya, Abramova and Rancourt67,Reference Sunil, Vayas and Abramova82 Levels of fibrinogen/fibrin and surfactant protein (SP)-D are also increased in the lung and/or BAL. This is evident early after exposure and remains elevated up to 28 days. Reference Malaviya, Abramova and Rancourt67,Reference Sunil, Vayas and Abramova82,Reference McGraw, Osborne and Mastej83

Lung injury induced by SM and NM is associated with oxidative stress characterized by increases in 8-hydroxy-2-deoxyguanosine (8OHdG), 2-deoxyguanosine, malondialdehyde and 4-hydroxynonenal, and decreases in glutathione levels. Reference Malaviya, Sunil and Venosa66,Reference McElroy, Min and Huang84,Reference Yaren, Mollaoglu and Kurt85 Nitrates and nitrites also increase in BAL and urine. Reference McElroy, Min and Huang84,Reference Yaren, Mollaoglu and Kurt85 Additionally, antioxidants such as heme oxygenase (HO)-1, lipocalin-2, Ym-1, and Mn-SOD are upregulated, a response that persists for at least 28 days post-SM or NM exposure. Reference Malaviya, Sunil and Venosa66,Reference Malaviya, Abramova and Rancourt67,Reference Malaviya, Bellomo and Abramova86 A marker of DNA damage, γH2A.X, and PCNA are also detectable in the lungs. Reference Malaviya, Abramova and Rancourt67,Reference Venosa, Smith and Gow87,Reference Malaviya, Venosa and Hall88 Whereas γH2A.X increases 1–3 days after mustard exposure, PCNA is increased 3–28 days in bronchiolar epithelium, alveolar epithelial cells, interstitial cells, and in focal areas exhibiting honeycombing and/or fibrosis.

Inflammatory genes/proteins, including IL-1, IL-2, IL-6, TNFα, KC/GRO, CCR2, CCR5, CCL2, CCL3, CCL5, CCL11, CX3CR1, CX3CL1, high mobility group box (HMGB)1, and MMP-9, are evident in the lungs and/or BAL fluid from mustard-treated rodents within 1–3 days post-exposure. Reference Malaviya, Abramova and Rancourt67,Reference Venosa, Malaviya and Choi79,Reference Mishra, Rir-Sima-Ah and Grotendorst81,Reference McElroy, Min and Huang84 Proinflammatory macrophages expressing TNFα (Figure 2), iNOS, MMP-9, HMGB1, or COX-2 are also present at this time. Reference Malaviya, Abramova and Rancourt67,Reference Sunil, Vayas and Abramova82,Reference Malaviya, Sunil and Venosa89 Whereas NM-induced increases in expression of inflammatory markers are maximum at 3 days post-exposure, persisting at lower levels up to 28 days, the response to SM is biphasic. Reference Malaviya, Abramova and Rancourt67,Reference Malaviya, Venosa and Hall88 Thus, SM exposure causes an early increase in inflammatory markers at 1–3 days, which is followed by a decrease at 7–16 days, and then a generally more robust increase at 28 days. Antiinflammatory/profibrotic genes (IL-10, pentraxin-2, connective tissue growth factor [CTGF], ApoE) have also been identified in the lungs; however, the timing of their appearance varies with the gene. Reference Venosa, Malaviya and Choi79 Antiinflammatory macrophages characterized by expression of CD206 (mannose receptor), CD68, CD163, galectin-3, and arginase-II are also present in histologic lung sections most prominently at later time points. Reference Malaviya, Abramova and Rancourt67,Reference Malaviya, Sunil and Venosa78,Reference Venosa, Malaviya and Choi79 This is correlated with the upregulation of α-smooth muscle actin, TGFβ, platelet-derived growth factor (PDGF), PDGF receptor-α, and CTGF. Reference McGraw, Dysart and Hendry-Hofer68,Reference Malaviya, Sunil and Venosa78,Reference Mishra, Rir-Sima-Ah and Grotendorst81

Figure 2. Effects of sulfur mustard on TNFα expression in the lung. Rats were exposed by intratracheal inhalation to air (CTL) or sulfur mustard (SM, 0.4 mg/kg) as previously described. Reference Malaviya, Abramova and Rancourt67 Lung sections were prepared 3, 7, 16, and 28 days later and immunostained with anti-TNFα antibody. Binding was visualized using a Vectastain kit. Original magnification, 600X. Representative images from 8–9 rats/group are shown.

Loss of TNFR1 Mitigates Half-Mustard-Induced Lung Injury

As indicated above, TNFR1 is the major receptor mediating the proinflammatory actions of TNFα. In initial studies, to assess the potential role of TNFα in mustard-induced lung injury, mice lacking TNFR1 were used. Reference Sunil, Patel-Vayas and Shen56 In these studies, CEES was used as an experimental model vesicant. TNFR1−/− mice exhibited an attenuated response to CEES-induced lung injury, oxidative stress, and inflammation; thus, expression of oxidative stress markers and inflammatory proteins was reduced or delayed. Loss of TNFR1 also blunted aberrant functional responses to CEES. These findings provided initial evidence for a role of TNFα in mustard lung toxicity. Reference Sunil, Patel-Vayas and Shen56

Pharmacologic Inhibition of TNFα Mitigates Mustard-Induced Lung Injury, Inflammation, and Fibrosis

In further studies, the effects of pharmacologic inhibition of TNFα on mustard toxicity were assessed. Pentoxifylline (PTX) is a nonspecific phosphodiesterase inhibitor with anti-inflammatory activity, due largely to its ability to block TNFα synthesis. Reference Fernandes, de Oliveira and Mamoni90Reference Turhan, Atıcı and Muşlu92 PTX has been reported to blunt TNFα production by alveolar macrophages from patients with sarcoidosis. Reference Marques, Zheng and Poulakis93 PTX is also clinically efficacious in a number of inflammatory pathologies characterized by excessive TNFα production, including alcoholic liver disease and rheumatoid arthritis. Reference Queiroz-Junior, Bessoni and Costa94Reference Zaitone, Hassan and El-Orabi96 Treatment of rats with PTX (46.7 mg/kg, ip) daily for 3 days was found to reduce acute lung injury and inflammation induced by NM. Reference Sunil, Vayas and Cervelli97 Thus, granulocyte infiltration into the lung is blunted, along with edema, fibrin deposition, and fibroplasia; BAL protein and cell numbers are also significantly reduced. PTX also reduces NM-induced oxidative stress and numbers of proinflammatory macrophages in the lung, while increasing numbers of antiinflammatory macrophages. This correlates with persistent upregulation of markers of wound repair, including pro-SP-C and PCNA. These data support the idea that targeting TNFα using more specific inhibitors represents a potentially efficacious approach for treating mustard lung toxicity.

Biologics against TNFα are widely used clinically to treat immunoinflammatory diseases, including rheumatoid arthritis, psoriasis, and Crohn’s disease, with minimal toxicity. Reference Hasegawa, Takasaki, Greene and Murali98,Reference Raychaudhuri and Raychaudhuri99 TNFα blocking agents have also proven beneficial in patients with the lung diseases such as severe asthma, COPD, and sarcoidosis. Reference Malaviya, Laskin and Laskin19,Reference Matera, Calzetta and Cazzola100 Based on these findings, anti-TNF antibody has been evaluated as a countermeasure against mustard lung toxicity. Treatment of rats with anti-TNFα antibody (15 mg/kg, IV, every 8 days) blunts mustard-induced structural alterations in the lung at all post-exposure times (3–28 days) examined. Reference Malaviya, Sunil and Venosa78,Reference Malaviya, Bellomo and Abramova86 Thus, parenchymal lesions are reduced in size and intensity, and deposits of plasma proteins decreased; occlusion of the bronchiolar lumen by fibrillar membrane, ulceration of bronchial epithelium, acute inflammation, edema, bronchoalveolar, and goblet cell hyperplasia and hypertrophy, bronchiectasis, interstitial thickening, macrophage accumulation, squamous cell metaplasia, mesothelial cell proliferation, and emphysema are attenuated. Reference Malaviya, Sunil and Venosa78,Reference Malaviya, Bellomo and Abramova86 Anti-TNFα antibody also reduces NM-induced collagen deposition, peribronchial and parenchymal fibrosis, and numbers of fibrotic lesions in the lung. Reference Malaviya, Sunil and Venosa78

Further studies demonstrated that anti-TNFα reduces mustard-induced alveolar-epithelial barrier dysfunction, oxidative stress, and increases in inflammatory proteins and profibrotic cytokines in the lung, along with the numbers of proinflammatory macrophages, while antiinflammatory macrophages important in wound healing are increased or unaffected. Reference Malaviya, Sunil and Venosa78,Reference Malaviya, Bellomo and Abramova86 Small live animal imaging techniques, including magnetic resonance imaging (MRI) and computed tomography (CT) imaging, confirmed the efficacy of anti-TNFα antibody in blunting NM-induced lung injury and fibrosis. Reference Murray, Gow and Venosa101 Thus, anti-TNFα antibody treatment of rats was found to reduce the percentage of injured lung within 1 day of NM exposure and subsequent development of fibrosis. Together, these data demonstrate that inhibiting TNFα represents an efficacious approach to mitigating acute lung injury, inflammatory macrophage activation, oxidative stress, and lung remodeling induced by mustard vesicants.

Role of TNFα in Radiation-Induced Lung Injury

Radiation exposure causes acute lung injury, which progresses to pneumonitis within weeks to months and fibrosis within months to years. Reference Giuranno, Ient, De Ruysscher and Vooijs102 Early injury is characterized by damage to the alveolar wall, interstitial edema, and an accumulation of proteins and inflammatory cells in the lung lining fluid; this is followed by thickening of alveolar walls, solid lesions with collagen deposits, and bronchiectasis as the pathology develops. Mechanistically, radiation-induced lung injury involves DNA damage and the generation of cytotoxic reactive oxygen and nitrogen species. Reference Azzam, Jay-Gerin and Pain103,Reference Beach, Groves, Williams and Finkelstein104 This is associated with loss of epithelial and endothelial barrier function, an accumulation of inflammatory cells in the lung that produce mediators such as IL-1, IL-6, IL-13, IL-17, TNFα, and TGFβ that can further damage the tissue and/or contribute to tissue remodeling and fibrogenesis. Reference Giuranno, Ient, De Ruysscher and Vooijs102,Reference Darby and Hewitson105Reference Arroyo-Hernández, Maldonado and Lozano-Ruiz110 Radiation also affects pulmonary endothelial cell function as measured by decreases in the activity of angiotensin converting enzyme (ACE) and plasminogen activator (PLA). Reference Ward, Kim and Molteni111 This is accompanied by increases in lung wet weight, protein and hydroxyproline content, and eicosanoids.

Radiation-induced lung injury is characterized by increases in TNFα and TNFR1. Reference Przybyszewska, Miloszewska and Rzonca112Reference Malaviya, Gow and Francis114 This is observed early (1–3 hours) after exposure to a single dose of radiation Reference Rübe, Wilfert and Palm6,Reference Zhang, Qian and Xing113 and aligned with increases in numbers of TUNEL-positive epithelial cells and upregulation of cleaved capase-3, markers of apoptosis. Radiation-induced increases in TNFα persist in the lung up to 24 hours post-exposure; subsequently, TNF levels return to baseline. This is followed by secondary, more exaggerated increases 2–24 weeks post-exposure coinciding with radiation-induced histopathological changes in the lung, including diffuse alveolitis, inflammatory cell accumulation, thickening of alveolar walls, depletion of type II epithelial cells, fibroblast proliferation, and interstitial and alveolar deposition of extracellular matrix. Reference Rübe, Wilfert and Palm6,Reference Roy, Salerno and Citrin106,Reference Johnston, Piedboeuf and Rubin108,Reference Malaviya, Gow and Francis114Reference Osterreicher, Mokry and Navrátil118 Fractionated radiation exposure (single high dose divided into low dose radiation over several days) is also associated with increases in TNF levels at early times but at reduced levels when compared to a single high dose. Cumulative TNFα levels after fractionated radiation, however, are greater and more persistent. Reference Zhang, Qian and Xing113 Of note, early increases in TNFα precede radiation-induced increases in IL-1α and IL-6, suggesting that TNFα plays a role in the initiation of the inflammatory cytokine cascade. Reference Rübe, Wilfert and Palm6

Blocking TNFα Mitigates Radiation-Induced Lung Injury, Pneumonitis, and Fibrosis

Treatment of mice with a TNFR1-specific antisense oligonucleotide (ASO) has been reported to reduce radiation-induced increases in TUNEL-positive cells and expression of cleaved caspase-3. Reference Zhang, Qian and Xing113 This correlates with a reduction in collagen deposition and restoration of pulmonary function 8 weeks post-exposure. Mice lacking TNFR1 are also resistant to radiation-induced alterations in lung function. Reference Zhang, Qian and Xing113 Similarly, gene therapy using a plasmid vector encoding mouse soluble TNFR1 (psTNFR1) reduces radiation-induced lung fibrosis and mortality. Reference Przybyszewska, Miloszewska and Rzonca112 In response to radiation, mice bearing mutations in the TNFα signaling pathway also exhibit an attenuated breathing rate. Reference Hill, Zaidi, Mahmood and Jelveh116

Pharmacologic inhibition of TNFα has also been found to mitigate radiation-induced lung injury and inflammation. For example, Rube et al. (2002) Reference Rübe, Wilfert and Uthe119 reported that PTX downregulates radiation-induced increases in lung TNFα. PTX treatment has also been reported to delay radiation-induced apoptosis in the lung. Reference Osterreicher, Mokry and Navrátil118 Relatively greater numbers of SP-D expressing cells, which are important in suppressing pulmonary inflammatory responses, have been noted 1–5 weeks after radiation exposure in mice returning to control levels after 8–12 weeks. Reference Osterreicher, Mokry and Navrátil118 PTX treatment significantly enhances numbers of SP-D expressing cells at all time points examined after radiation exposure. This is associated with a delayed accumulation of neutrophils in the lung and a reduction in radiation-induced alveolar septal thickness up to 12 weeks after exposure. Reference Osterreicher, Mokry and Navrátil118,Reference Osterreicher, Králik and Navrátil120 PTX treatment also reduces radiation-induced increases in lung wet weight and protein content and improves lung perfusion, which enhances tissue oxygenation and wound healing. Reference Ward, Kim and Molteni111,Reference Koh, Stelzer and Peterson121,Reference Stelzer, Koh, Peterson and Griffin122 Taken together, these findings show that TNF plays a role in radiation-induced lung injury; moreover, inhibition of TNFα or TNFR1 can mitigate the deleterious effects of radiation. Further investigations on the efficacy of biologics against TNFα may lead to new treatments for radiation-induced lung pathology.

Conclusions

TNFα is a key mediator of local damage and inflammation in the lung. Both mustard vesicant- and radiation-induced lung injury are associated with increases in TNFα. A wide range of responses, including apoptosis, mitosis, chemotaxis, angiogenesis, extracellular matrix production, and release of cytokines and chemokines, are triggered when TNFα binds to its receptor on target cells. Given that tissues and cells are exposed to complex mixtures of inflammatory mediators, it is likely that blocking TNFα immediately after the injury may be beneficial. As increases in TNFα have been linked to both acute and chronic manifestations of toxic injury, multiple sequential doses of biologics against TNFα may be required to keep inflammation in check. Anti-TNFα biological therapies have been used to treat immuno-inflammatory diseases of the skin, joints, and gut. These treatments have also been effective to varying degrees in patients with chronic lung diseases. Reference Malaviya, Laskin and Laskin19,Reference Matera, Calzetta and Cazzola100,Reference Taille, Poulet and Marchand-Adam123,Reference Crommelin, Vorselaars and van Moorsel124 Further research and clinical investigation with anti-TNFα therapy in acute and chronic pulmonary toxicity induced by mustard vesicants and radiation may prove useful for the development of successful treatment strategies using TNF targeting agents.

Author contributions

Rama Malaviya: literature search, original manuscript draft writing, review & editing; Jeffrey D. Laskin: manuscript review & editing; Rita Businaro: manuscript review & editing; Debra L. Laskin: conceptualization, funding acquisition, manuscript review & editing.

Funding statement

This work was supported by National Institutes of Health Grants U54AR055073, R01ES004738, R01ES033698, and P30ES005022.

Competing interests

The authors declare no conflicts of interest.

References

Graham, JS, Schoneboom, BA. Historical perspective on effects and treatment of sulfur mustard injuries. Chem Biol Interact. 2013;206(3):512-522.CrossRefGoogle ScholarPubMed
Keyser, BM, Andres, DK, Holmes, WW, et al. Mustard gas inhalation injury: therapeutic strategy. Int J Toxicol. 2014;33(4):271-281.CrossRefGoogle ScholarPubMed
Rajan Radha, R, Chandrasekharan, G. Pulmonary injury associated with radiation therapy—assessment, complications and therapeutic targets. Biomed Pharmacother. 2017;89:1092-1104.CrossRefGoogle ScholarPubMed
Weinberger, B, Malaviya, R, Sunil, VR, et al. Mustard vesicant-induced lung injury: advances in therapy. Toxicol Appl Pharmacol. 2016;305:1-11.CrossRefGoogle ScholarPubMed
Malaviya, R, Heck, D, Casillas, RP, et al. Mustard vesicants. In: Lukey, BJ, Romano, JA Jr, Salem, H, eds. Chemical Warfare Agents: Chemistry, Pharmacology, Toxicology, and Countermeasures. 3rd ed. CRC; 2018:131-143.Google Scholar
Rübe, CE, Wilfert, F, Palm, J, et al. Irradiation induces a biphasic expression of pro-inflammatory cytokines in the lung. Strahlenther Onkol. 2004;180(7):442-448.CrossRefGoogle ScholarPubMed
Smith, LC, Venosa, A, Gow, AJ, et al. Transcriptional profiling of lung macrophages during pulmonary injury induced by nitrogen mustard. Ann N Y Acad Sci. 2020;1480(1):146-154.CrossRefGoogle ScholarPubMed
Holbrook, J, Lara-Reyna, S, Jarosz-Griffiths, H, McDermott, M. Tumour necrosis factor signalling in health and disease. F1000Res. 2019;8. doi:10.12688/f1000research.17023.1CrossRefGoogle Scholar
Bahia, MS, Silakari, O. Tumor necrosis factor alpha converting enzyme: an encouraging target for various inflammatory disorders. Chem Biol Drug Des. 2010;75(5):415-443.CrossRefGoogle ScholarPubMed
Jang, DI, Lee, AH, Shin, HY, et al. The role of tumor necrosis factor alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics. Int J Mol Sci. 2021;22(5). doi: 10.3390/ijms22052719 CrossRefGoogle ScholarPubMed
Aggarwal, BB, Gupta, SC, Kim, JH. Historical perspectives on tumor necrosis factor and its superfamily: 25 years later, a golden journey. Blood. 2012;119(3):651-665.CrossRefGoogle Scholar
Zelová, H, Hošek, J. TNF-α signalling and inflammation: interactions between old acquaintances. Inflamm Res. 2013;62(7):641-651.CrossRefGoogle ScholarPubMed
Horiuchi, T, Mitoma, H, Harashima, S, et al. Transmembrane TNF-α: structure, function and interaction with anti-TNF agents. Rheumatology. 2010;49(7):1215-1228.CrossRefGoogle ScholarPubMed
Tracey, D, Klareskog, L, Sasso, EH, et al. Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacol Ther. 2008;117(2):244-279.CrossRefGoogle ScholarPubMed
Aggarwal, BB. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol. 2003;3(9):745-756.CrossRefGoogle ScholarPubMed
Grell, M, Douni, E, Wajant, H, et al. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell. 1995;83(5):793-802.CrossRefGoogle ScholarPubMed
Mukhopadhyay, A, Suttles, J, Stout, RD, Aggarwal, BB. Genetic deletion of the tumor necrosis factor receptor p60 or p80 abrogates ligand-mediated activation of nuclear factor-κB and of mitogen-activated protein kinases in macrophages. J Biol Chem. 2001;276(34):31906-31912.CrossRefGoogle ScholarPubMed
Probert, L. TNF and its receptors in the CNS: the essential, the desirable and the deleterious effects. Neuroscience. 2015;302:2-22.CrossRefGoogle ScholarPubMed
Malaviya, R, Laskin, JD, Laskin, DL. Anti-TNFα therapy in inflammatory lung diseases. Pharmacol Ther. 2017;180:90-98.CrossRefGoogle ScholarPubMed
Suzuki, J, Hamada, E, Shodai, T, et al. Cytokine secretion from human monocytes potentiated by P-selectin-mediated cell adhesion. Int Arch Allergy Immunol. 2013;160(2):152-160.CrossRefGoogle ScholarPubMed
Sabio, G, Davis, RJ. TNF and MAP kinase signalling pathways. Semin Immunol. 2014;26(3):237-245.CrossRefGoogle ScholarPubMed
Semenzato, G. Tumour necrosis factor: a cytokine with multiple biological activities. Br J Cancer. 1990;61(3):354-361.CrossRefGoogle ScholarPubMed
Camussi, G, Albano, E, Tetta, C, Bussolino, F. The molecular action of tumor necrosis factor-α. Eur J Biochem. 1991;202(1):3-14.CrossRefGoogle ScholarPubMed
Michel, O, Dinh, PH, Doyen, V, Corazza, F. Anti-TNF inhibits the airways neutrophilic inflammation induced by inhaled endotoxin in human. BMC Pharmacol Toxicol. 2014;15:60.CrossRefGoogle ScholarPubMed
Kelly, M, Hwang, JM, Kubes, P. Modulating leukocyte recruitment in inflammation. J Allergy Clin Immunol. 2007;120(1):3-10.CrossRefGoogle ScholarPubMed
Shimabukuro, DW, Sawa, T, Gropper, MA. Injury and repair in lung and airways. Crit Care Med. 2003;31(8 Suppl):S524-S531.CrossRefGoogle ScholarPubMed
Laskin, DL, Malaviya, R, Laskin, JD. Role of macrophages in acute lung injury and chronic fibrosis induced by pulmonary toxicants. Toxicol Sci. 2019;168(2):287-301.CrossRefGoogle ScholarPubMed
Rahman, I. Regulation of nuclear factor-κB, activator protein-1, and glutathione levels by tumor necrosis factor-α and dexamethasone in alveolar epithelial cells. Biochem Pharmacol. 2000;60(8):1041-1049.CrossRefGoogle ScholarPubMed
Blaser, H, Dostert, C, Mak, TW, Brenner, D. TNF and ROS crosstalk in inflammation. Trends Cell Biol. 2016;26:249-261.CrossRefGoogle ScholarPubMed
Obrador, E, Navarro, J, Mompo, J, et al. Regulation of tumour cell sensitivity to TNF-induced oxidative stress and cytotoxicity: role of glutathione. Biofactors. 1998;8(1-2):23-26.CrossRefGoogle ScholarPubMed
Lu, G, Beuerman, RW, Zhao, S, et al. Tumor necrosis factor-alpha and interleukin-1 induce activation of MAP kinase and SAP kinase in human neuroma fibroblasts. Neurochem Int. 1997;30(4-5):401-410.CrossRefGoogle ScholarPubMed
Mukhopadhyay, S, Mukherjee, S, Stone, WL, et al. Role of MAPK/AP-1 signaling pathway in the protection of CEES-induced lung injury by antioxidant liposome. Toxicology. 2009;261(3):143-151.CrossRefGoogle ScholarPubMed
Allen, JT, Spiteri, MA. Growth factors in idiopathic pulmonary fibrosis: relative roles. Respir Res. 2002;3:13.CrossRefGoogle ScholarPubMed
Sasaki, M, Kashima, M, Ito, T, et al. Differential regulation of metalloproteinase production, proliferation and chemotaxis of human lung fibroblasts by PDGF, interleukin-1β and TNF-α. Mediators Inflamm. 2000;9(3-4):155-160.CrossRefGoogle ScholarPubMed
Piguet, PF. Is “tumor necrosis factor” the major effector of pulmonary fibrosis? Eur Cytokine Netw. 1990;1(4):257-258.Google Scholar
Sullivan, DE, Ferris, M, Pociask, D, Brody, AR. Tumor necrosis factor-α induces transforming growth factor-β1 expression in lung fibroblasts through the extracellular signal-regulated kinase pathway. Am J Respir Cell Mol Biol. 2005;32(4):342-349.CrossRefGoogle ScholarPubMed
Oikonomou, N, Harokopos, V, Zalevsky, J, et al. Soluble TNF mediates the transition from pulmonary inflammation to fibrosis. PLoS One. 2006;1:e108.CrossRefGoogle ScholarPubMed
Kim, WU, Min, SY, Cho, ML, et al. Elevated matrix metalloproteinase-9 in patients with systemic sclerosis. Arthritis Res Ther. 2005;7(1):R71-R79.CrossRefGoogle ScholarPubMed
Kristinsson, J, Johannesson, T. [Mustard gas bombs found astray in the Faxafloi bay. Mustard gas: usage and poisonings]. Laeknabladid. 2009;95(5):359-365.Google ScholarPubMed
Malaviya, R, Laskin, JD, Laskin, DL. Long-term respiratory effects of mustard vesicants. Toxicol Lett. 2020;319:168-174.CrossRefGoogle ScholarPubMed
Ghabili, K, Agutter, PS, Ghanei, M, et al. Mustard gas toxicity: the acute and chronic pathological effects. J Appl Toxicol. 2010;30(7):627-643.CrossRefGoogle ScholarPubMed
Kehe, K, Thiermann, H, Balszuweit, F, et al. Acute effects of sulfur mustard injury—Munich experiences. Toxicology. 2009;263(1):3-8.CrossRefGoogle ScholarPubMed
Kehe, K, Balszuweit, F, Emmler, J, et al. Sulfur mustard research—strategies for the development of improved medical therapy. Eplasty. 2008;8:e32.Google ScholarPubMed
Sezigen, S, Ivelik, K, Ortatatli, M, et al. Victims of chemical terrorism, a family of four who were exposed to sulfur mustard. Toxicol Lett. 2019;303:9-15.CrossRefGoogle ScholarPubMed
Sohrabpour, H. Clinical manifestations of chemical agents on Iranian combatants during Iran-Iraq conflict. Arch Belg. 1984;Suppl:291-297.Google Scholar
Kehe, K, Szinicz, L. Medical aspects of sulphur mustard poisoning. Toxicology. 2005;214(3):198-209.CrossRefGoogle ScholarPubMed
Wang, GQ, Xia, ZF. Tissue injury by hot fluid containing nitrogen mustard. Burns. 2007;33(7):923-926.CrossRefGoogle ScholarPubMed
Rancourt, RC, Veress, LA, Ahmad, A, et al. Tissue factor pathway inhibitor prevents airway obstruction, respiratory failure and death due to sulfur mustard analog inhalation. Toxicol Appl Pharmacol. 2013;272(1):86-95.CrossRefGoogle ScholarPubMed
Bijani, K, Moghadamnia, AA. Long-term effects of chemical weapons on respiratory tract in Iraq-Iran war victims living in Babol (North of Iran). Ecotoxicol Environ Saf. 2002;53(3):422-424.CrossRefGoogle ScholarPubMed
Weinberger, B, Laskin, JD, Sunil, VR, et al. Sulfur mustard-induced pulmonary injury: therapeutic approaches to mitigating toxicity. Pulm Pharmacol Ther. 2011;24(1):92-99.CrossRefGoogle ScholarPubMed
Nishimura, Y, Iwamoto, H, Ishikawa, N, et al. Long-term pulmonary complications of chemical weapons exposure in former poison gas factory workers. Inhal Toxicol. 2016;28(8):343-348.CrossRefGoogle ScholarPubMed
Doi, M, Hattori, N, Yokoyama, A, et al. Effect of mustard gas exposure on incidence of lung cancer: a longitudinal study. Am J Epidemiol. 2011;173(6):659-666.CrossRefGoogle ScholarPubMed
Mukaida, K, Hattori, N, Iwamoto, H, et al. Mustard gas exposure and mortality among retired workers at a poisonous gas factory in Japan: a 57-year follow-up cohort study. Occup Environ Med. 2017;74(5):321-327.CrossRefGoogle Scholar
Macit, E, Yaren, H, Aydin, I, et al. The protective effect of melatonin and S-methylisothiourea treatments in nitrogen mustard induced lung toxicity in rats. Environ Toxicol Pharmacol. 2013;36(3):1283-1290.CrossRefGoogle ScholarPubMed
Das, SK, Mukherjee, S, Smith, MG, Chatterjee, D. Prophylactic protection by N-acetylcysteine against the pulmonary injury induced by 2-chloroethyl ethyl sulfide, a mustard analogue. J Biochem Mol Toxicol. 2003;17(3):177-184.CrossRefGoogle Scholar
Sunil, VR, Patel-Vayas, K, Shen, J, et al. Role of TNFR1 in lung injury and altered lung function induced by the model sulfur mustard vesicant, 2-chloroethyl ethyl sulfide. Toxicol Appl Pharmacol. 2011;250(3):245-255.CrossRefGoogle Scholar
Rancourt, RC, Ahmad, A, Veress, LA, et al. Antifibrinolytic mechanisms in acute airway injury after sulfur mustard analog inhalation. Am J Respir Cell Mol Biol. 2014;51(4):559-567.CrossRefGoogle ScholarPubMed
Arbel, M, Choudhary, K, Tfilin, O, Kupiec, M. PCNA loaders and unloaders—one ring that rules them all. Genes (Basel). 2021;12(11). doi: 10.3390/genes12111812 CrossRefGoogle Scholar
Mukhopadhyay, S, Mukherjee, S, Smith, M, Das, SK. Activation of MAPK/AP-1 signaling pathway in lung injury induced by 2-chloroethyl ethyl sulfide, a mustard gas analog. Toxicol Lett. 2008;181(2):112-117.CrossRefGoogle ScholarPubMed
Ng, ET, Sim, MK, Loke, WK. Protective actions of des-aspartate-angiotensin I in mice model of CEES-induced lung intoxication. J Appl Toxicol. 2011;31(6):568-578.CrossRefGoogle ScholarPubMed
Sawale, SD, Ambhore, PD, Pawar, PP, et al. Ameliorating effect of S-2(ω-aminoalkylamino) alkylaryl sulfide (DRDE-07) on sulfur mustard analogue, 2-chloroethyl ethyl sulfide-induced oxidative stress and inflammation. Toxicol Mech Methods. 2013;23(9):702-710.CrossRefGoogle Scholar
Sunil, VR, Shen, J, Patel-Vayas, K, et al. Role of reactive nitrogen species generated via inducible nitric oxide synthase in vesicant-induced lung injury, inflammation and altered lung functioning. Toxicol Appl Pharmacol. 2012;261:22-30.CrossRefGoogle ScholarPubMed
Tang, FR, Loke, WK. Sulfur mustard and respiratory diseases. Crit Rev Toxicol. 2012;42(8):688-702.CrossRefGoogle ScholarPubMed
Jan, YH, Heck, DE, Laskin, DL, Laskin, JD. DNA damage signaling in the cellular responses to mustard vesicants. Toxicol Lett. 2020;326:78-82.CrossRefGoogle ScholarPubMed
Abbotts, R, Wilson, DM 3rd. Coordination of DNA single strand break repair. Free Radic Biol Med. 2017;107:228-244.CrossRefGoogle ScholarPubMed
Malaviya, R, Sunil, VR, Venosa, A, et al. Inflammatory mechanisms of pulmonary injury induced by mustards. Toxicol Lett. 2016;244:2-7.CrossRefGoogle ScholarPubMed
Malaviya, R, Abramova, EV, Rancourt, RC, et al. Progressive lung injury, inflammation, and fibrosis in rats following inhalation of sulfur mustard. Toxicol Sci. 2020;178(2):358-374.CrossRefGoogle ScholarPubMed
McGraw, MD, Dysart, MM, Hendry-Hofer, TB, et al. Bronchiolitis obliterans and pulmonary fibrosis after sulfur mustard inhalation in rats. Am J Respir Cell Mol Biol. 2018;58(6):696-705.CrossRefGoogle ScholarPubMed
Anderson, DR, Byers, SL, Vesely, KR. Treatment of sulfur mustard (HD)-induced lung injury. J Appl Toxicol. 2000;20 Suppl 1:S129-S132.Google Scholar
Xiaoji, Z, Xiao, M, Rui, X, et al. Mechanism underlying acute lung injury due to sulfur mustard exposure in rats. Toxicol Ind Health. 2016;32(8):1345-1357.CrossRefGoogle ScholarPubMed
Perry, MR, Neal, M, Hawks, R, et al. A novel sulfur mustard (HD) vapor inhalation exposure model of pulmonary toxicity for the efficacy evaluation of candidate medical countermeasures. Inhal Toxicol. 2021;33(6-8):221-233.CrossRefGoogle ScholarPubMed
Gao, X, Anderson, DR, Brown, AW, et al. Pathological studies on the protective effect of a macrolide antibiotic, roxithromycin, against sulfur mustard inhalation toxicity in a rat model. Toxicol Pathol. 2011;39(7):1056-1064.CrossRefGoogle ScholarPubMed
Sunil, VR, Patel, KJ, Shen, J, et al. Functional and inflammatory alterations in the lung following exposure of rats to nitrogen mustard. Toxicol Appl Pharmacol. 2011;250(1):10-18.CrossRefGoogle ScholarPubMed
Anderson, DR, Yourick, JJ, Moeller, RB, et al. Pathologic changes in rat lungs following acute sulfur mustard inhalation. Inhal Toxicol. 1996;8(3):285-297.CrossRefGoogle Scholar
McGraw, MD, Rioux, JS, Garlick, RB, et al. From the cover: Impaired proliferation and differentiation of the conducting airway epithelium associated with bronchiolitis obliterans after sulfur mustard inhalation injury in rats. Toxicol Sci. 2017;157(2):399-409.CrossRefGoogle ScholarPubMed
Malaviya, R, Sunil, VR, Cervelli, J, et al. Inflammatory effects of inhaled sulfur mustard in rat lung. Toxicol Appl Pharmacol. 2010;248(2):89-99.CrossRefGoogle ScholarPubMed
Andres, DK, Keyser, BM, Melber, AA, et al. Apoptotic cell death in rat lung following mustard gas inhalation. Am J Physiol Lung Cell Mol Physiol. 2017;312(6):L959-L968.CrossRefGoogle ScholarPubMed
Malaviya, R, Sunil, VR, Venosa, A, et al. Attenuation of nitrogen mustard-induced pulmonary injury and fibrosis by anti-tumor necrosis factor-α antibody. Toxicol Sci. 2015;148(1):71-88.CrossRefGoogle ScholarPubMed
Venosa, A, Malaviya, R, Choi, H, et al. Characterization of distinct macrophage subpopulations during nitrogen mustard-induced lung injury and fibrosis. Am J Respir Cell Mol Biol. 2016;54(3):436-446.CrossRefGoogle ScholarPubMed
Solopov, P, Biancatelli, R, Marinova, M, et al. The HSP90 inhibitor, AUY-922, ameliorates the development of nitrogen mustard-induced pulmonary fibrosis and lung dysfunction in mice. Int J Mol Sci. 2020;21(13). doi: 10.3390/ijms21134740 CrossRefGoogle ScholarPubMed
Mishra, NC, Rir-Sima-Ah, J, Grotendorst, GR, et al. Inhalation of sulfur mustard causes long-term T cell-dependent inflammation: possible role of Th17 cells in chronic lung pathology. Int Immunopharmacol. 2012;13(1):101-108.CrossRefGoogle ScholarPubMed
Sunil, VR, Vayas, KN, Abramova, EV, et al. Lung injury, oxidative stress and fibrosis in mice following exposure to nitrogen mustard. Toxicol Appl Pharmacol. 2020;387:114798.CrossRefGoogle ScholarPubMed
McGraw, MD, Osborne, CM, Mastej, EJ, et al. Editor’s highlight: Pulmonary vascular thrombosis in rats exposed to inhaled sulfur mustard. Toxicol Sci. 2017;159(2):461-469.CrossRefGoogle ScholarPubMed
McElroy, CS, Min, E, Huang, J, et al. From the cover: Catalytic antioxidant rescue of inhaled sulfur mustard toxicity. Toxicol Sci. 2016;154(2):341-353.CrossRefGoogle ScholarPubMed
Yaren, H, Mollaoglu, H, Kurt, B, et al. Lung toxicity of nitrogen mustard may be mediated by nitric oxide and peroxynitrite in rats. Res Vet Sci. 2007;83(1):116-122.CrossRefGoogle ScholarPubMed
Malaviya, R, Bellomo, A, Abramova, E, et al. Pulmonary injury and oxidative stress in rats induced by inhaled sulfur mustard is ameliorated by anti-tumor necrosis factor-α antibody. Toxicol Appl Pharmacol. 2021;428:115677.CrossRefGoogle ScholarPubMed
Venosa, A, Smith, LC, Gow, AJ, et al. Macrophage activation in the lung during the progression of nitrogen mustard induced injury is associated with histone modifications and altered miRNA expression. Toxicol Appl Pharmacol. 2021;423:115569.CrossRefGoogle ScholarPubMed
Malaviya, R, Venosa, A, Hall, L, et al. Attenuation of acute nitrogen mustard-induced lung injury, inflammation and fibrogenesis by a nitric oxide synthase inhibitor. Toxicol Appl Pharmacol. 2012;265(3):279-291.CrossRefGoogle ScholarPubMed
Malaviya, R, Sunil, VR, Venosa, A, et al. Macrophages and inflammatory mediators in pulmonary injury induced by mustard vesicants. Ann N Y Acad Sci. 2016;1374(1):168-175.CrossRefGoogle ScholarPubMed
Fernandes, JL, de Oliveira, RT, Mamoni, RL, et al. Pentoxifylline reduces pro-inflammatory and increases anti-inflammatory activity in patients with coronary artery disease—a randomized placebo-controlled study. Atherosclerosis. 2008;196(1):434-442.CrossRefGoogle ScholarPubMed
González-Espinoza, L, Rojas-Campos, E, Medina-Pérez, M, et al. Pentoxifylline decreases serum levels of tumor necrosis factor alpha, interleukin 6 and C-reactive protein in hemodialysis patients: results of a randomized double-blind, controlled clinical trial. Nephrol Dial Transplant. 2012;27(5):2023-2028.CrossRefGoogle ScholarPubMed
Turhan, AH, Atıcı, A, Muşlu, N, et al. The effects of pentoxifylline on lung inflammation in a rat model of meconium aspiration syndrome. Exp Lung Res. 2012;38(5):250-255.CrossRefGoogle Scholar
Marques, LJ, Zheng, L, Poulakis, N, et al. Pentoxifylline inhibits TNF-α production from human alveolar macrophages. Am J Respir Crit Care Med. 1999;159(2):508-511.CrossRefGoogle ScholarPubMed
Queiroz-Junior, CM, Bessoni, RL, Costa, VV, et al. Preventive and therapeutic anti-TNF-α therapy with pentoxifylline decreases arthritis and the associated periodontal co-morbidity in mice. Life Sci. 2013;93(9-11):423-428.CrossRefGoogle ScholarPubMed
Sidhu, SS, Goyal, O, Singla, M, et al. Pentoxifylline in severe alcoholic hepatitis: a prospective, randomised trial. J Assoc Physicians India. 2012;60:20-22.Google ScholarPubMed
Zaitone, S, Hassan, N, El-Orabi, N, El-Awady el S. Pentoxifylline and melatonin in combination with pioglitazone ameliorate experimental non-alcoholic fatty liver disease. Eur J Pharmacol. 2011;662(1-3):70-77.CrossRefGoogle Scholar
Sunil, VR, Vayas, KN, Cervelli, JA, et al. Pentoxifylline attenuates nitrogen mustard-induced acute lung injury, oxidative stress and inflammation. Exp Mol Pathol. 2014;97(1):89-98.CrossRefGoogle ScholarPubMed
Hasegawa, A, Takasaki, W, Greene, MI, Murali, R. Modifying TNFα for therapeutic use: a perspective on the TNF receptor system. Mini Rev Med Chem. 2001;1(1):5-16.CrossRefGoogle ScholarPubMed
Raychaudhuri, SP, Raychaudhuri, SK. Biologics: target-specific treatment of systemic and cutaneous autoimmune diseases. Indian J Dermatol. 2009;54(2):100-109.CrossRefGoogle ScholarPubMed
Matera, MG, Calzetta, L, Cazzola, M. TNF-α inhibitors in asthma and COPD: we must not throw the baby out with the bath water. Pulm Pharmacol Ther. 2010;23(2):121-128.CrossRefGoogle Scholar
Murray, A, Gow, AJ, Venosa, A, et al. Assessment of mustard vesicant lung injury and anti-TNF-α efficacy in rodents using live-animal imaging. Ann N Y Acad Sci. 2020;1480(1):246-256.CrossRefGoogle ScholarPubMed
Giuranno, L, Ient, J, De Ruysscher, D, Vooijs, MA. Radiation-induced lung injury (RILI). Front Oncol. 2019;9:877.CrossRefGoogle ScholarPubMed
Azzam, EI, Jay-Gerin, JP, Pain, D. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 2012;327(1-2):48-60.CrossRefGoogle ScholarPubMed
Beach, TA, Groves, AM, Williams, JP, Finkelstein, JN. Modeling radiation-induced lung injury: lessons learned from whole thorax irradiation. Int J Radiat Biol. 2020;96(1):129-144.CrossRefGoogle ScholarPubMed
Darby, IA, Hewitson, TD. Fibroblast differentiation in wound healing and fibrosis. Int Rev Cytol. 2007;257:143-179.CrossRefGoogle ScholarPubMed
Roy, S, Salerno, KE, Citrin, DE. Biology of radiation-induced lung injury. Semin Radiat Oncol. 2021;31(2):155-161.CrossRefGoogle ScholarPubMed
Trott, KR, Herrmann, T, Kasper, M. Target cells in radiation pneumopathy. Int J Radiat Oncol Biol Phys. 2004;58(2):463-469.CrossRefGoogle ScholarPubMed
Johnston, CJ, Piedboeuf, B, Rubin, P, et al. Early and persistent alterations in the expression of interleukin-1α, interleukin-1β and tumor necrosis factor α mRNA levels in fibrosis-resistant and sensitive mice after thoracic irradiation. Radiat Res. 1996;145(6):762-767.CrossRefGoogle Scholar
Lierova, A, Jelicova, M, Nemcova, M, et al. Cytokines and radiation-induced pulmonary injuries. J Radiat Res. 2018;59(6):709-753.Google ScholarPubMed
Arroyo-Hernández, M, Maldonado, F, Lozano-Ruiz, F, et al. Radiation-induced lung injury: current evidence. BMC Pulm Med. 2021;21(1):9. doi: 10.1186/s12890-020-01376-4 CrossRefGoogle ScholarPubMed
Ward, WF, Kim, YT, Molteni, A, et al. Pentoxifylline does not spare acute radiation reactions in rat lung and skin. Radiat Res. 1992;129(1):107-111.CrossRefGoogle Scholar
Przybyszewska, M, Miloszewska, J, Rzonca, S, et al. Soluble TNF-α receptor I encoded on plasmid vector and its application in experimental gene therapy of radiation-induced lung fibrosis. Arch Immunol Ther Exp (Warsz). 2011;59(4):315-326.CrossRefGoogle ScholarPubMed
Zhang, M, Qian, J, Xing, X, et al. Inhibition of the tumor necrosis factor-α pathway is radioprotective for the lung. Clin Cancer Res. 2008;14(6):1868-1876.CrossRefGoogle ScholarPubMed
Malaviya, R, Gow, AJ, Francis, M, et al. Radiation-induced lung injury and inflammation in mice: role of inducible nitric oxide synthase and surfactant protein D. Toxicol Sci. 2015;144(1):27-38.CrossRefGoogle ScholarPubMed
Hong, JH, Chiang, CS, Tsao, CY, et al. Rapid induction of cytokine gene expression in the lung after single and fractionated doses of radiation. Int J Radiat Biol. 1999;75(11):1421-1427.Google ScholarPubMed
Hill, RP, Zaidi, A, Mahmood, J, Jelveh, S. Investigations into the role of inflammation in normal tissue response to irradiation. Radiother Oncol. 2011;101(1):73-79.CrossRefGoogle Scholar
Niu, S, Zhang, Y, Cong, C, et al. Comparative study of radiation-induced lung injury model in two strains of mice. Health Phys. 2022;122(5):579-585.CrossRefGoogle ScholarPubMed
Osterreicher, J, Mokry, J, Navrátil, L, et al. The alveolar septal thickness and type II pneumocytes number in irradiated lungs, time expression and the effect of pentoxifylline. Acta Medica (Hradec Kralove). 2001;44(1):15-19.CrossRefGoogle ScholarPubMed
Rübe, CE, Wilfert, F, Uthe, D, et al. Modulation of radiation-induced tumour necrosis factor α (TNF-α) expression in the lung tissue by pentoxifylline. Radiother Oncol. 2002;64(2):177-187.CrossRefGoogle ScholarPubMed
Osterreicher, J, Králik, M, Navrátil, L, et al. Apoptosis and bcl-2 expression in irradiated lungs and the effect of pentoxifylline. Acta Medica (Hradec Kralove). 2001;44(4):125-130.CrossRefGoogle ScholarPubMed
Koh, WJ, Stelzer, KJ, Peterson, LM, et al. Effect of pentoxifylline on radiation-induced lung and skin toxicity in rats. Int J Radiat Oncol Biol Phys. 1995;31(1):71-77.CrossRefGoogle ScholarPubMed
Stelzer, KJ, Koh, WJ, Peterson, LM, Griffin, TW. Effect of high-dose pentoxifylline on acute radiation-induced lung toxicity in a rat lung perfusion model. Int J Radiat Oncol Biol Phys. 1996;34(1):111-115.CrossRefGoogle Scholar
Taille, C, Poulet, C, Marchand-Adam, S, et al. Monoclonal anti-TNF-α antibodies for severe steroid-dependent asthma: a case series. Open Respir Med J. 2013;7:21-25.CrossRefGoogle ScholarPubMed
Crommelin, HA, Vorselaars, AD, van Moorsel, CH, et al. Anti-TNF therapeutics for the treatment of sarcoidosis. Immunotherapy. 2014;6(10):1127-1143.CrossRefGoogle ScholarPubMed
Sunil, VR, Vayas, KN, Cervelli, JA, et al. Protective role of surfactant protein-D against lung injury and oxidative stress induced by nitrogen mustard. Toxicol Sci. 2018;166(1):108-122.Google ScholarPubMed
Elzayat, MA, Bayoumi, AMA, Abdel-Bakky, MS, et al. Ameliorative effect of 2-methoxyestradiol on radiation-induced lung injury. Life Sci. 2020;255:117743.CrossRefGoogle ScholarPubMed
Flockerzi, E, Schanz, S, Rübe, CE. Even low doses of radiation lead to DNA damage accumulation in lung tissue according to the genetically-defined DNA repair capacity. Radiother Oncol. 2014;111(2):212-218.CrossRefGoogle Scholar
Rajan, V, Pandey, BN. Cytoproliferative effect of low dose alpha radiation in human lung cancer cells is associated with connexin 43, caveolin-1, and survivin pathway. Int J Radiat Biol. 2021;97(3):356-366.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Tumor necrosis factor (TNF)α signaling. Binding of soluble or membrane bound TNFα to TNFR1 and TNFR2 initiates signaling events associated with apoptosis or activation of transcription factors, NFκB and AP-1. TNFα binding to TNFR1 and/or TNFR2 can also activate protein kinase B/Akt, which leads to prolonged NF-κB activation. Together, these responses contribute to inflammation, leukocyte trafficking, cell death, cell proliferation, and tissue remodeling.AP-1, activator protein-1; IKK, IκB kinase; MAPKs, mitogen-associated protein kinases; mTNF, membrane-bound TNFα; NFκB, nuclear factor kappa B; sTNF, soluble TNFα; TACE, TNFα-converting enzyme; TNFR1, TNFα receptor 1; TNFR2, TNFα receptor 2; TRADD, TNFR associated death domain; TRAF1, TNFR associated factor 1; TRAF2, TNFR associated factor 2.

Figure 1

Table 1. Biological activities of tumor necrosis factor (TNF)α in mustard or radiation-induced lung injury

Figure 2

Figure 2. Effects of sulfur mustard on TNFα expression in the lung. Rats were exposed by intratracheal inhalation to air (CTL) or sulfur mustard (SM, 0.4 mg/kg) as previously described.67 Lung sections were prepared 3, 7, 16, and 28 days later and immunostained with anti-TNFα antibody. Binding was visualized using a Vectastain kit. Original magnification, 600X. Representative images from 8–9 rats/group are shown.