1
|
Hu X, Zhang N, Zhong Y, Liu T, Zhu X. Mechanisms of Apoptosis and Pulmonary Fibrosis Resulting From Sulfur Mustard-Induced Acute Pulmonary Injury in Rats. Int J Toxicol 2025:10915818251315907. [PMID: 39888856 DOI: 10.1177/10915818251315907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
Abstract
Sulfur mustard (SM) is a highly toxic bifunctional alkylating agent that inflicts severe damage on the respiratory tract. Although numerous studies have examined the mechanisms underlying SM-induced pulmonary injury, the exact pathways involved remain unclear. This study aims to investigate an acute pulmonary injury model, with SM administered as a single intraperitoneal injection (8 mg/kg) or single intratracheal instillation (2 mg/kg) at equal toxicity doses (1LD50). The results revealed that epithelial cells in the alveolar septa of the intraperitoneal SM group exhibited a significantly higher expression of apoptotic markers, including pro-apoptotic protein Bax, caspase-3, and caspase-9 proteins, than those in the tracheal SM group. Conversely, the expression of the anti-apoptotic protein Bcl-2 was significantly lower in the intraperitoneal SM group than in the tracheal SM group, as confirmed by TUNEL staining and immunohistochemical staining. The intraperitoneal SM group exhibited markedly higher expression of fibrosis-related proteins, including MMP-2, MMP-9, TIMP-1, TIMP-2, collagen type I, collagen type III, TGF-β1, and Smad7, than the tracheal SM group. These markers, detected through immunohistochemical immunolabeling, indicate a more significant fibrotic response in the intraperitoneal group. In summary, this study demonstrates that intraperitoneal exposure to SM results in increased apoptosis, elevated expression of pro-apoptotic proteins, and fibrosis-related proteins in the alveolar epithelial cells compared with intratracheal exposure, even at equivalent toxicity levels. Our findings highlight the suitability of the intraperitoneal route for further investigation and identify apoptotic and fibrosis-related proteins as potential targets for intervention in SM-induced pulmonary injury.
Collapse
Affiliation(s)
- Xiaoxuan Hu
- Weifang No. 2 People's Hospital, Weifang Respiratory Disease Hospital, Weifang, China
- State Key Laboratory of Antitoxic Drugs and Toxicology, Institute of Toxicology and Pharmacology, Academy of Military Medical Sciences, Beijing, China
| | - Na Zhang
- Department of Respiration, The 80th Group Army Hospital of People's Liberation Army, Weifang, China
| | - Yuxu Zhong
- State Key Laboratory of Antitoxic Drugs and Toxicology, Institute of Toxicology and Pharmacology, Academy of Military Medical Sciences, Beijing, China
| | - Tao Liu
- Department of Respiration, The 80th Group Army Hospital of People's Liberation Army, Weifang, China
| | - Xiaoji Zhu
- Department of Respiration, Jiaozhou Branch of Shanghai East Hospital, Tongji University, Qingdao, China
| |
Collapse
|
2
|
Ye F, Dan G, Zhao Y, Yu W, Cheng J, Chen M, Sai Y, Zou Z. Small-interfering RNA for c-Jun attenuates cell death by preventing JNK-dependent PARP1 cleavage and DNA fragmentation in nitrogen mustard-injured immortalized human bronchial epithelial cells. Toxicol Res (Camb) 2021; 10:1034-1044. [PMID: 34733488 DOI: 10.1093/toxres/tfab081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 06/29/2021] [Accepted: 07/27/2021] [Indexed: 11/14/2022] Open
Abstract
Sulfur mustard (a type of vesicant) can directly damage lung bronchial epithelium via aerosol inhalation, and prevalent cell death is an early event that obstructs the respiratory tract. JNK/c-Jun is a stress response pathway, but its role in cell death of the injured cells is not clear. Here, we report that JNK/c-Jun was activated in immortalized human bronchial epithelial (HBE) cells exposed to a lethal dose (20 μM) of nitrogen mustard (NM, a sulfur mustard analog). c-Jun silencing using small-interfering RNA (siRNA) rendered the cells resistant to NM-mediated cell death by blocking poly(ADP-ribose) polymerase 1 (PARP1) cleavage and DNA fragmentation. In addition, the transduction of upstream extrinsic (Fasl-Fas-caspase-8) and intrinsic (loss of Bcl-2 and mitochondrial membrane potential, ΔΨm) apoptosis pathways, as well as phosphorylated (p)-H2AX (Ser139), an epigenetic marker contributing to DNA fragmentation and PARP1 activity, was partially suppressed. To mimic the detachment of cells by NM, HBE cells were trypsinized and seeded on culture plates that were pre-coated with poly-HEMA to prevent cell adhesion. The JNK/c-Jun pathway was found to be activated in the detached cells. In conclusion, our results indicate that JNK/c-Jun pathway activation is necessary for NM-caused HBE cell death and further suggest that c-Jun silencing may be a potential approach to protect HBE cells from vesicant damage.
Collapse
Affiliation(s)
- Feng Ye
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Guorong Dan
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuanpeng Zhao
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Wenpei Yu
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jin Cheng
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mingliang Chen
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yan Sai
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhongmin Zou
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
3
|
Gilardoni M, Léonço D, Caffin F, Gros-Désormeaux F, Eldin C, Béal D, Ouzia S, Junot C, Fenaille F, Piérard C, Douki T. Evidence for the systemic diffusion of (2-chloroethyl)-ethyl-sulfide, a sulfur mustard analog, and its deleterious effects in brain. Toxicology 2021; 462:152950. [PMID: 34534560 DOI: 10.1016/j.tox.2021.152950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/03/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
Sulfur mustard, a chemical warfare agent known to be a vesicant of skin, readily diffuses in the blood stream and reaches internal organs. In the present study, we used the analog (2-chloroethyl)-ethyl-sulfide (CEES) to provide novel data on the systemic diffusion of vesicants and on their ability to induce brain damage, which result in neurological disorders. SKH-1 hairless mice were topically exposed to CEES and sacrificed at different time until 14 days after exposure. A plasma metabolomics study showed a strong systemic impact following a self-protection mechanism to alleviate the injury of CEES exposure. This result was confirmed by the quantification of specific biomarkers in plasma. Those were the conjugates of CEES with glutathione (GSH-CEES), cysteine (Cys-CEES) and N-acetyl-cysteine (NAC-CEES), as well as the guanine adduct (N7Gua-CEES). In brain, N7Gua-CEES could be detected both in DNA and in organ extracts. Similarly, GSH-CEES, Cys-CEES and NAC-CEES were present in the extracts until day14. Altogether, these results, based on novel exposure markers, confirm the ability of vesicants to induce internal damage following dermal exposure. The observation of alkylation damage to glutathione and DNA in brain provides an additional mechanism to the neurological insult of SM.
Collapse
Affiliation(s)
- Marie Gilardoni
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, F-38000, Grenoble, France
| | - Daniel Léonço
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif sur Yvette, France
| | - Fanny Caffin
- Institut de Recherche Biomédicale des Armées (IRBA), Place Général Valérie André, BP 73, 91223, Brétigny-sur-Orge Cedex, France
| | - Fanny Gros-Désormeaux
- Institut de Recherche Biomédicale des Armées (IRBA), Place Général Valérie André, BP 73, 91223, Brétigny-sur-Orge Cedex, France
| | - Camille Eldin
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, F-38000, Grenoble, France
| | - David Béal
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, F-38000, Grenoble, France
| | - Sadia Ouzia
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif sur Yvette, France
| | - Christophe Junot
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif sur Yvette, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif sur Yvette, France
| | - Christophe Piérard
- Institut de Recherche Biomédicale des Armées (IRBA), Place Général Valérie André, BP 73, 91223, Brétigny-sur-Orge Cedex, France
| | - Thierry Douki
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, F-38000, Grenoble, France.
| |
Collapse
|
4
|
Emadi SN, Abtahi-Naeini B, Rahmat Pour Rokni G, Emadi SE, Hamzelou S. The mucocutaneous manifestations of sulfur mustard gas on 1024 Iraqi victims: ISIS chemical attack in the South of Kirkuk. TOXIN REV 2021. [DOI: 10.1080/15569543.2021.1942064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Seyed Naser Emadi
- Skin Research Center, Razi and Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Iranian Red Crescent Society, Tehran, Iran
| | - Bahareh Abtahi-Naeini
- Skin Diseases and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ghasem Rahmat Pour Rokni
- Clinical Research Development Unit of Bou-Ali Sina Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Emad Emadi
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahin Hamzelou
- Department of Dermatology, Imam Khomeini and Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Shahriary A, Sabzevari M, Jadidi K, Yazdani F, Aghamollaei H. The Role of Inflammatory Cytokines in Neovascularization of Chemical Ocular Injury. Ocul Immunol Inflamm 2021; 30:1149-1161. [PMID: 33734925 DOI: 10.1080/09273948.2020.1870148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Aim: Chemical injuries can potentially lead to the necrosis anterior segment of the eye, and cornea in particular. Inflammatory cytokines are the first factors produced after chemical ocular injuries. Inflammation via promoting the angiogenesis factor tries to implement the wound healing mechanism in the epithelial and stromal layer of the cornea. Methods: Narrative review.Results: In our review, we described the patterns of chemical injuries in the cornea and their molecular mechanisms associated with the expression of inflammatory cytokines. Moreover, the effects of inflammation signals on angiogenesis factors and CNV were explained. Conclusion: The contribution of inflammation and angiogenesis causes de novo formation of blood vessels that is known as the corneal neovascularization (CNV). The new vascularity interrupts cornea clarity and visual acuity. Inflammation also depleted the Limbal stem cells (LSCs) in the limbus causing the failure of normal corneal epithelial healing and conjunctivalization of the cornea.
Collapse
Affiliation(s)
- Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Milad Sabzevari
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Khosrow Jadidi
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Farshad Yazdani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Sadeghi S, Tapak M, Ghazanfari T, Mosaffa N. A review of Sulfur Mustard-induced pulmonary immunopathology: An Alveolar Macrophage Approach. Toxicol Lett 2020; 333:115-129. [PMID: 32758513 DOI: 10.1016/j.toxlet.2020.07.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/27/2022]
Abstract
Despite many studies investigating the mechanism of Sulfur Mustard (SM) induced lung injury, the underlying mechanism is still unclear. Inflammatory and subsequent fibroproliferative stages of SM-toxicity are based upon several highly-related series of events controlled by the immune system. The inhalation of SM gas variably affects different cell populations within the lungs. Various studies have shown the critical role of macrophages in triggering a pulmonary inflammatory response as well as its maintenance, resolution, and repair. Importantly, macrophages can serve as either pro-inflammatory or anti-inflammatory populations depending on the present conditions at any pathological stage. Different characteristics of macrophages, including their differentiation, phenotypic, and functional properties, as well as interactions with other cell populations determine the outcomes of lung diseases and the extent of long- or short-term pulmonary damage induced by SM. In this paper, we summarize the current state of knowledge regarding the role of alveolar macrophages and their mediators in the pathogenesis of SM in pulmonary injury. Investigating the specific cells and mechanisms involved in SM-lung injury may be useful in finding new target opportunities for treatment of this injury.
Collapse
Affiliation(s)
- Somaye Sadeghi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahtab Tapak
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran; Department of Immunology, Shahed University, Tehran, Iran.
| | - Nariman Mosaffa
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Jost P, Muckova L, Pejchal J. In vitro stress response induced by sulfur mustard in lung fibroblasts NHLF and human pulmonary epithelial cells A-549. Arch Toxicol 2020; 94:3503-3514. [PMID: 32681189 DOI: 10.1007/s00204-020-02845-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/09/2020] [Indexed: 10/23/2022]
Abstract
Sulfur mustard [bis(2-chloroethyl) sulfide; SM] is a highly poisonous chemical warfare agent. The mechanism of its cytotoxicity affects several pathways, which cause cell damage or death. The main organ affected in case of exposure to both aerosol and vapor is lungs. The present study focuses on time- and concentration-dependent changes in human lung fibroblasts NHLF and lung epithelial cell line A-549. The cells were treated with SM at the concentrations of 5, 10 and 100 µM and signs of stress response were evaluated during 1-72 h post-treatment. Parameters for testing included cell viability and morphology, loss of transmembrane mitochondrial potential, apoptosis, oxidative stress, changes in the cell cycle, and ATM kinase activation. The cytotoxic effect of SM resulted in a time-dependent decrease in viability of A-459 associated with apoptosis more markedly than in NHLF. We did not observe any generation of reactive oxygen species by SM. SM at concentrations of 5 and 10 µM induced the S-phase cell cycle arrest at both cell lines. On the other hand, 100 µM caused nonspecific cell cycle arrest. ATM kinase was activated transiently. The results indicate that NHLF cells are less prone to toxic damage by SM in case of cell viability, apoptosis and loss of transmembrane mitochondrial potential. The analysis provides a time-related cytotoxic profile of A-549 and NHLF cells for further investigation into the prevention of SM toxic effects and their potential treatment.
Collapse
Affiliation(s)
- Petr Jost
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic.
| | - Lubica Muckova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| |
Collapse
|
8
|
Hassanpour M, Hajihassani F, Abdollahpourasl M, Cheraghi O, Aghamohamadzade N, Rahbargazi R, Nouri M, Pilehvar-Soltanahmadi Y, Zarghami N, Akbarzadeh A, Panahi Y, Sahebkar A. Pathophysiological Effects of Sulfur Mustard on Skin and its Current Treatments: Possible Application of Phytochemicals. Comb Chem High Throughput Screen 2020; 24:3-19. [PMID: 32679016 DOI: 10.2174/1386207323666200717150414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/21/2020] [Accepted: 06/03/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Sulfur-(SM) and nitrogen (NM)-based mustards are the mutagenic incapacitating compounds which are widely used in vesicating the chemical warfare and cause toxicity in many organs, especially skin. SM, as a potent vesicating agent, contributes to the destruction of skin in dermis and epidermis layers. The progression of the lesion depends on the concentration of SM and the duration of exposure. Body responses start with pruritus, erythema, edema and xerosis, which lead to the accumulation of immune cells in the target sites and recruitment of mast cells and paracrine-mediated activity. Pro-inflammatory effectors are accumulated in the epidermis, hair follicles, and sebaceous glands resulting in the destruction of the basement membrane beneath the epidermis. There is still no satisfactory countermeasure against SM-induced lesions in clinical therapy, and the symptomatic or supportive treatments are routine management approaches. OBJECTIVE The current review highlights the recent progression of herbal medicines application in SM-induced injuries through the illustrative examples and also demonstrates their efficacies, properties and mechanism of actions as therapeutic agents. CONCLUSION Phytochemicals and herbal extracts with anti-bacterial, anti-inflammatory and antioxidant properties have been recently shown to hold therapeutic promise against the SM-induced cutaneous complications. The present review discusses the possible application of herbal medicines in the healing of SM-induced injuries.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fateme Hajihassani
- Department of Health Management, School of Management and Medical informatics, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Omid Cheraghi
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Nasser Aghamohamadzade
- Endocrine and Metabolism Section, Department of Internal Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbargazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Younes Pilehvar-Soltanahmadi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Akbarzadeh
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yunes Panahi
- Pharmacotherapy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
9
|
NAD + in sulfur mustard toxicity. Toxicol Lett 2020; 324:95-103. [PMID: 32017979 DOI: 10.1016/j.toxlet.2020.01.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/08/2020] [Accepted: 01/25/2020] [Indexed: 12/21/2022]
Abstract
Sulfur mustard (SM) is a toxicant and chemical warfare agent with strong vesicant properties. The mechanisms behind SM-induced toxicity are not fully understood and no antidote or effective therapy against SM exists. Both, the risk of SM release in asymmetric conflicts or terrorist attacks and the usage of SM-derived nitrogen mustards as cancer chemotherapeutics, render the mechanisms of mustard-induced toxicity a highly relevant research subject. Herein, we review a central role of the abundant cellular molecule nicotinamide adenine dinucleotide (NAD+) in molecular mechanisms underlying SM toxicity. We also discuss the potential beneficial effects of NAD+ precursors in counteracting SM-induced damage.
Collapse
|
10
|
Rafati-Rahimzadeh M, Rafati-Rahimzadeh M, Kazemi S, Moghadamnia AA. Therapeutic options to treat mustard gas poisoning - Review. CASPIAN JOURNAL OF INTERNAL MEDICINE 2019; 10:241-264. [PMID: 31558985 PMCID: PMC6729161 DOI: 10.22088/cjim.10.3.241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/29/2019] [Accepted: 02/03/2019] [Indexed: 11/14/2022]
Abstract
Among the blistering (vesicant) chemical warfare agents (CWA), sulfur mustard is the most important since it is known as the "King of chemical warfare agents". The use of sulfur mustard has caused serious damages in several organs, especially the eyes, skin, respiratory, central and peripheral nervous systems after short and long term exposure, incapacitating and even killing people and troops. In this review, chemical properties, mechanism of actions and their effects on each organ, clinical manifestations, diagnostic evaluation of the actions triage, and treatment of injuries have been described.
Collapse
Affiliation(s)
- Mehrdad Rafati-Rahimzadeh
- Cancer Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ali Akbar Moghadamnia
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
11
|
Menacher G, Balszuweit F, Lang S, Thiermann H, Kehe K, Gudermann T, Schmidt A, Steinritz D, Popp T. Necrosulfonamide - Unexpected effect in the course of a sulfur mustard intoxication. Chem Biol Interact 2018; 298:80-85. [PMID: 30391637 DOI: 10.1016/j.cbi.2018.10.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/23/2018] [Accepted: 10/31/2018] [Indexed: 11/26/2022]
Abstract
Although its first military use in Ypres was 100 years ago, no causal therapy for sulfur mustard (SM) intoxications exists so far. To improve the therapeutic options for the treatment of SM intoxications, we developed a co-culture of keratinocytes (HaCaT cells) and immunocompetent cells (THP-1 cells) to identify potential substances for further research. Here, we report on the influence of necrosulfonamide (NSA) on the course of a SM intoxication in vitro. The cells were challenged with 100, 200 and 300 μM SM and after 1 h treated with NSA (1, 5, 10 μM). NSA was chosen for its known ability to inhibit necroptosis, a specialized pathway of programmed necrosis. However, in our settings NSA showed only mild effects on necrotic cell death after SM intoxication, whereas it had an immense ability to prevent apoptosis. Furthermore, NSA was able to reduce the production of interleukin-6 and interleukin-8 at certain concentrations. Our data highlight NSA as a candidate compound to address cell death and inflammation in SM exposure.
Collapse
Affiliation(s)
- Georg Menacher
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | | | - Simon Lang
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Kai Kehe
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany; Bundeswehr Medical Academy, Dept. Medical CBRN Defense, 80937, Munich, Germany
| | - Thomas Gudermann
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilian-University Munich, Goethestraße 33, 80336, Munich, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany; Bundeswehr University Munich, Faculty of Human Sciences, 85577, Neubiberg, Germany
| | - Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany; Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilian-University Munich, Goethestraße 33, 80336, Munich, Germany
| | - Tanja Popp
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany; Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilian-University Munich, Goethestraße 33, 80336, Munich, Germany.
| |
Collapse
|
12
|
Tumu HCR, Cuffari BJ, Pino MA, Palus J, Piętka-Ottlik M, Billack B. Ebselen oxide attenuates mechlorethamine dermatotoxicity in the mouse ear vesicant model. Drug Chem Toxicol 2018; 43:335-346. [DOI: 10.1080/01480545.2018.1488858] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hemanta C. Rao Tumu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Jamaica, NY, USA
| | - Benedette J. Cuffari
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Jamaica, NY, USA
| | - Maria A. Pino
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Jamaica, NY, USA
- Department of Clinical Specialties, NYIT College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Jerzy Palus
- Department of Organic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wrocław, Poland
| | - Magdalena Piętka-Ottlik
- Department of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wrocław University of Science and Technology, Wrocław, Poland
| | - Blase Billack
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Jamaica, NY, USA
| |
Collapse
|
13
|
Jost P, Fikrova P, Svobodova H, Pejchal J, Stetina R. Protective potential of different compounds and their combinations with MESNA against sulfur mustard-induced cytotoxicity and genotoxicity. Toxicol Lett 2017; 275:92-100. [PMID: 28495614 DOI: 10.1016/j.toxlet.2017.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/11/2017] [Accepted: 05/05/2017] [Indexed: 12/01/2022]
Abstract
The purpose of this study was to evaluate the efficacy of potential candidate molecules or their combinations against strong alkylation agent sulfur mustard (SM) on the human lung alveolar epithelial cell line A-549. Candidate molecules were chosen on the basis of their previously observed protective effects in vitro. The tested compounds, including antioxidants, sulfhydryl or other sulfur-containing molecules, nitrogen-containing molecules, PARP inhibitors and a NO synthase inhibitor, were applicated 30min before SM treatment. The efficiency of candidate molecules to protect cells against DNA damage and cell death induced by SM was determined using single-cell gel electrophoresis (comet assay) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction by viable cells. The damage of DNA was assessed 1 and 24h after dose 50μM SM. Cell survival was assessed 24 and 72h after the exposure. To achieve maximal cytoprotection, combinations of selected compounds with sodium 2-mercaptoethane sulphonate (MESNA) were tested. We found significant protective effects by several drugs used individually and also in combination with MESNA. High protection was achieved by sodium thiosulphate, which was further potentiated when combined with MESNA. Most of the selected compounds or mixture provided only moderate genoptotection without having any effect towards cell viability.
Collapse
Affiliation(s)
- Petr Jost
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic.
| | - Petra Fikrova
- Department of Research and Development, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Hana Svobodova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Rudolf Stetina
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic
| |
Collapse
|
14
|
Ronayne CT, Solano LN, Nelson GL, Lueth EA, Hubbard SL, Schumacher TJ, Gardner ZS, Jonnalagadda SK, Gurrapu S, Holy J, Mereddy VR. Synthesis and biological evaluation of 2-alkoxycarbonylallyl esters as potential anticancer agents. Bioorg Med Chem Lett 2017; 27:776-780. [DOI: 10.1016/j.bmcl.2017.01.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 11/30/2022]
|
15
|
McElroy CS, Min E, Huang J, Loader JE, Hendry-Hofer TB, Garlick RB, Rioux JS, Veress LA, Smith R, Osborne C, Anderson DR, Holmes WW, Paradiso DC, White CW, Day BJ. From the Cover: Catalytic Antioxidant Rescue of Inhaled Sulfur Mustard Toxicity. Toxicol Sci 2016; 154:341-353. [PMID: 27605419 DOI: 10.1093/toxsci/kfw170] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sulfur mustard (bis 2-chloroethyl ethyl sulfide, SM) is a powerful bi-functional vesicating chemical warfare agent. SM tissue injury is partially mediated by the overproduction of reactive oxygen species resulting in oxidative stress. We hypothesized that using a catalytic antioxidant (AEOL 10150) to alleviate oxidative stress and secondary inflammation following exposure to SM would attenuate the toxic effects of SM inhalation. Adult male rats were intubated and exposed to SM (1.4 mg/kg), a dose that produces an LD50 at approximately 24 h. Rats were randomized and treated via subcutaneous injection with either sterile PBS or AEOL 10150 (5 mg/kg, sc, every 4 h) beginning 1 h post-SM exposure. Rats were euthanized between 6 and 48 h after exposure to SM and survival and markers of injury were determined. Catalytic antioxidant treatment improved survival after SM inhalation in a dose-dependent manner, up to 52% over SM PBS at 48 h post-exposure. This improvement was sustained for at least 72 h after SM exposure when treatments were stopped after 48 h. Non-invasive monitoring throughout the duration of the studies also revealed blood oxygen saturations were improved by 10% and clinical scores were reduced by 57% after SM exposure in the catalytic antioxidant treatment group. Tissue analysis showed catalytic antioxidant therapy was able to decrease airway cast formation by 69% at 48 h post-exposure. To investigate antioxidant induced changes at the peak of injury, several biomarkers of oxidative stress and inflammation were evaluated at 24 h post-exposure. AEOL 10150 attenuated SM-mediated lung lipid oxidation, nitrosative stress and many proinflammatory cytokines. The findings indicate that catalytic antioxidants may be useful medical countermeasure against inhaled SM exposure.
Collapse
Affiliation(s)
- Cameron S McElroy
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045.,Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Elysia Min
- Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Jie Huang
- Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Joan E Loader
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | | | - Rhonda B Garlick
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Jackie S Rioux
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Livia A Veress
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Russell Smith
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Chris Osborne
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Dana R Anderson
- Analytical Toxicology Division, Proving Grounds United States Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen, Maryland 21010
| | - Wesley W Holmes
- Analytical Toxicology Division, Proving Grounds United States Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen, Maryland 21010
| | - Danielle C Paradiso
- Analytical Toxicology Division, Proving Grounds United States Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen, Maryland 21010
| | - Carl W White
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045.,Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Brian J Day
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045 .,Department of Medicine, National Jewish Health, Denver, Colorado 80206
| |
Collapse
|
16
|
Nourani MR, Mahmoodzadeh Hosseini H, Azimzadeh Jamalkandi S, Imani Fooladi AA. Cellular and molecular mechanisms of acute exposure to sulfur mustard: a systematic review. J Recept Signal Transduct Res 2016; 37:200-216. [DOI: 10.1080/10799893.2016.1212374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Mohammad Reza Nourani
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Liu F, Jiang N, Xiao ZY, Cheng JP, Mei YZ, Zheng P, Wang L, Zhang XR, Zhou XB, Zhou WX, Zhang YX. Effects of poly (ADP-ribose) polymerase-1 (PARP-1) inhibition on sulfur mustard-induced cutaneous injuries in vitro and in vivo. PeerJ 2016; 4:e1890. [PMID: 27077006 PMCID: PMC4830333 DOI: 10.7717/peerj.1890] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/14/2016] [Indexed: 01/13/2023] Open
Abstract
Early studies with first-generation poly (ADP-ribose) polymerase (PARP) inhibitors have already indicated some therapeutic potential for sulfur mustard (SM) injuries. The available novel and more potential PARP inhibitors, which are undergoing clinical trials as drugs for cancer treatment, bring it back to the centre of interest. However, the role of PARP-1 in SM-induced injury is not fully understood. In this study, we selected a high potent specific PARP inhibitor ABT-888 as an example to investigate the effect of PARP inhibitor in SM injury. The results showed that in both the mouse ear vesicant model (MEVM) and HaCaT cell model, PARP inhibitor ABT-888 can reduce cell damage induced by severe SM injury. ABT-888 significantly reduced SM induced edema and epidermal necrosis in MEVM. In the HaCaT cell model, ABT-888 can reduce SM-induced NAD(+)/ATP depletion and apoptosis/necrosis. Then, we studied the mechanism of PARP-1 in SM injury by knockdown of PARP-1 in HaCaT cells. Knockdown of PARP-1 protected cell viability and downregulated the apoptosis checkpoints, including p-JNK, p-p53, Caspase 9, Caspase 8, c-PARP and Caspase 3 following SM-induced injury. Furthermore, the activation of AKT can inhibit autophagy via the regulation of mTOR. Our results showed that SM exposure could significantly inhibit the activation of Akt/mTOR pathway. Knockdown of PARP-1 reversed the SM-induced suppression of the Akt/mTOR pathway. In summary, the results of our study indicated that the protective effects of downregulation of PARP-1 in SM injury may be due to the regulation of apoptosis, necrosis, energy crisis and autophagy. However, it should be noticed that PARP inhibitor ABT-888 further enhanced the phosphorylation of H2AX (S139) after SM exposure, which indicated that we should be very careful in the application of PARP inhibitors in SM injury treatment because of the enhancement of DNA damage.
Collapse
Affiliation(s)
- Feng Liu
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Ning Jiang
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Zhi-Yong Xiao
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Jun-Ping Cheng
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Yi-Zhou Mei
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Pan Zheng
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Li Wang
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Xiao-Rui Zhang
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Xin-Bo Zhou
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Wen-Xia Zhou
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Yong-Xiang Zhang
- Beijing Institute of Pharmacology and Toxicology , Beijing , China
| |
Collapse
|
18
|
Steinritz D, Schmidt A, Balszuweit F, Thiermann H, Simons T, Striepling E, Bölck B, Bloch W. Epigenetic modulations in early endothelial cells and DNA hypermethylation in human skin after sulfur mustard exposure. Toxicol Lett 2016; 244:95-102. [DOI: 10.1016/j.toxlet.2015.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/13/2015] [Accepted: 09/16/2015] [Indexed: 12/11/2022]
|
19
|
Impact of topical application of sulfur mustard on mice skin and distant organs DNA repair enzyme signature. Toxicol Lett 2016; 241:71-81. [DOI: 10.1016/j.toxlet.2015.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/02/2015] [Accepted: 11/02/2015] [Indexed: 11/23/2022]
|
20
|
Khazdair MR, Boskabady MH, Ghorani V. Respiratory effects of sulfur mustard exposure, similarities and differences with asthma and COPD. Inhal Toxicol 2015; 27:731-44. [PMID: 26635274 DOI: 10.3109/08958378.2015.1114056] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Previous research has found relationships between sulfur mustard (SM) toxicity and its adverse effects. OBJECTIVE SM is highly toxic to the respiratory system, leading to hacking cough, rhinorrheachest tightness, acute pharyngitis and laryngitis, chronic bronchitis and lung fibrosis. In this review, based on the scientific literature, we provide an updated summary of information on SM exposures and their differences with asthma and COPD. METHOD Information of this review was obtained by searching Medline/PubMed, ScienceDirect, Scopus, Google Scholar, ISI Web of Knowledge and Chemical Abstracts. RESULTS SM exposure can decrease pulmonary function tests (PFTs) values. In addition, inflammatory cell accumulation in the respiratory tract and increased expression of some pro-inflammatory cytokines including tumor necrosis factor-α (TNFα), IL-1a, IL-1β, and reactive oxygen radicals due to SM exposure have been shown. Matrix metalloproteinase (MMP) which degrade extracellular matrix proteins, contributing to inflammatory cell recruitment, tissue injury and fibrosis are also up-regulated in the lung after SM exposure. In the lung, SM exposure also can cause serious pathological changes including airway inflammation, parenchymal tissue destruction and airway obstruction which can lead to asthma or chronic obstructive pulmonary disease (COPD). Following SM poisoning, DNA damage, apoptosis and autophagy are observed in the lung along with the increased expression of activated caspases and DNA repair enzymes. CONCLUSION In the present article, respiratory symptoms, changes in PFTs, lung pathology and lung inflammation due to SM exposure and the similarities and differences between them and those observed in asthma and COPD were reviewed.
Collapse
Affiliation(s)
- Mohammad Reza Khazdair
- a Pharmaceutical Research Center and Department of Physiology, School of Medicine .,b Student Research Committee , and
| | - Mohammad Hossein Boskabady
- c Neurogenic Inflammation Research Centre and Department of Physiology, School of Medicine, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Vahideh Ghorani
- a Pharmaceutical Research Center and Department of Physiology, School of Medicine
| |
Collapse
|
21
|
Debiak M, Lex K, Ponath V, Burckhardt-Boer W, Thiermann H, Steinritz D, Schmidt A, Mangerich A, Bürkle A. Immunochemical analysis of poly(ADP-ribosyl)ation in HaCaT keratinocytes induced by the mono-alkylating agent 2-chloroethyl ethyl sulfide (CEES): Impact of experimental conditions. Toxicol Lett 2015; 244:72-80. [PMID: 26383632 DOI: 10.1016/j.toxlet.2015.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
Abstract
Sulfur mustard (SM) is a bifunctional alkylating agent with a long history of use as a chemical weapon. Although its last military use is dated for the eighties of the last century, a potential use in terroristic attacks against civilians remains a significant threat. Thus, improving medical therapy of mustard exposed individuals is still of particular interest. PARP inhibitors were recently brought into the focus as a potential countermeasure for mustard-induced pathologies, supported by the availability of efficient compounds successfully tested in cancer therapy. PARP activation after SM treatment was reported in several cell types and tissues under various conditions; however, a detailed characterization of this phenomenon is still missing. This study provides the basis for such studies by developing and optimizing experimental conditions to investigate poly(ADP-ribosyl)ation (PARylation) in HaCaT keratinocytes upon treatment with the monofunctional alkylating agent 2-chloroethyl ethyl sulfide ("half mustard", CEES). By using an immunofluorescence-based approach, we show that optimization of experimental conditions with regards to the type of solvent, dilution factors and treatment procedure is essential to obtain a homogenous PAR staining in HaCaT cell cultures. Furthermore, we demonstrate that different CEES treatment protocols significantly influence the cytotoxicity profiles of treated cells. Using an optimized treatment protocol, our data reveals that CEES induces a dose- and time-dependent dynamic PARylation response in HaCaT cells that could be completely blocked by treating cells with the clinically relevant pharmacological PARP inhibitor ABT888 (also known as veliparib). Finally, siRNA experiments show that CEES-induced PAR formation is predominantly due to the activation of PARP1. In conclusion, this study provides a detailed analysis of the CEES-induced PARylation response in HaCaT keratinocytes, which forms an experimental basis to study the molecular mechanism of PARP1 activation and its functional consequences after mustard treatment in general. Such a study is presented in an accompanying article (Mangerich et al., 2016).
Collapse
Affiliation(s)
- Malgorzata Debiak
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Kirsten Lex
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Viviane Ponath
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Waltraud Burckhardt-Boer
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Germany
| | - Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Germany; Walther Straub Institute of Pharmacology and Toxicology, 80336 Munich, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Germany
| | - Aswin Mangerich
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Alexander Bürkle
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany.
| |
Collapse
|
22
|
Mangerich A, Debiak M, Birtel M, Ponath V, Balszuweit F, Lex K, Martello R, Burckhardt-Boer W, Strobelt R, Siegert M, Thiermann H, Steinritz D, Schmidt A, Bürkle A. Sulfur and nitrogen mustards induce characteristic poly(ADP-ribosyl)ation responses in HaCaT keratinocytes with distinctive cellular consequences. Toxicol Lett 2015; 244:56-71. [PMID: 26383629 DOI: 10.1016/j.toxlet.2015.09.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 11/18/2022]
Abstract
Mustard agents are potent DNA alkylating agents with mutagenic, cytotoxic and vesicant properties. They include bi-functional agents, such as sulfur mustard (SM) or nitrogen mustard (mustine, HN2), as well as mono-functional agents, such as "half mustard" (CEES). Whereas SM has been used as a chemical warfare agent, several nitrogen mustard derivatives, such as chlorambucil and cyclophosphamide, are being used as established chemotherapeutics. Upon induction of specific forms of genotoxic stimuli, several poly(ADP-ribose) polymerases (PARPs) synthesize the nucleic acid-like biopolymer poly(ADP-ribose) (PAR) by using NAD(+) as a substrate. Previously, it was shown that SM triggers cellular poly(ADP-ribosyl) ation (PARylation), but so far this phenomenon is poorly characterized. In view of the protective effects of PARP inhibitors, the latter have been proposed as a treatment option of SM-exposed victims. In an accompanying article (Debiak et al., 2016), we have provided an optimized protocol for the analysis of the CEES-induced PARylation response in HaCaT keratinocytes, which forms an experimental basis to further analyze mustard-induced PARylation and its functional consequences, in general. Thus, in the present study, we performed a comprehensive characterization of the PARylation response in HaCaT cells after treatment with four different mustard agents, i.e., SM, CEES, HN2, and chlorambucil, on a qualitative, quantitative and functional level. In particular, we recorded substance-specific as well as dose- and time-dependent PARylation responses using independent bioanalytical methods based on single-cell immuno-fluorescence microscopy and quantitative isotope dilution mass spectrometry. Furthermore, we analyzed if and how PARylation contributes to mustard-induced toxicity by treating HaCaT cells with CEES, SM, and HN2 in combination with the clinically relevant PARP inhibitor ABT888. As evaluated by a novel immunofluorescence-based protocol for the detection of N7-ETE-guanine DNA adducts, the excision rate of CEES-induced DNA adducts was not affected by PARP inhibition. Furthermore, while CEES induced moderate changes in cellular NAD(+) levels, annexin V/PI flow cytometry analysis revealed that these changes did not affect CEES-induced short-term cytotoxicity 24h after treatment. In contrast, PARP inhibition impaired cell proliferation and clonogenic survival, and potentiated micronuclei formation of HaCaT cells upon CEES treatment. Similarly, PARP inhibition affected clonogenic survival of cells treated with bi-functional mustards such as SM and HN2. In conclusion, we demonstrate that PARylation plays a functional role in mustard-induced cellular stress response with substance-specific differences. Since PARP inhibitors exhibit therapeutic potential to treat SM-related pathologies and to sensitize cancer cells for mustard-based chemotherapy, potential long-term effects of PARP inhibition on genomic stability and carcinogenesis should be carefully considered when pursuing such a strategy.
Collapse
Affiliation(s)
- Aswin Mangerich
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Malgorzata Debiak
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Matthias Birtel
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Viviane Ponath
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Frank Balszuweit
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Germany
| | - Kirsten Lex
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Rita Martello
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Waltraud Burckhardt-Boer
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany
| | - Romano Strobelt
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Germany
| | - Markus Siegert
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Germany
| | - Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Germany; Walther-Straub-Institute of Pharmacology and Toxicology, 80336 Munich, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Germany
| | - Alexander Bürkle
- University of Konstanz, Molecular Toxicology Group, Department of Biology, 78457 Konstanz, Germany.
| |
Collapse
|
23
|
Keyser BM, Andres DK, Holmes WW, Paradiso D, Appell A, Letukas VA, Benton B, Clark OE, Gao X, Ray P, Anderson DR, Ray R. Mustard Gas Inhalation Injury. Int J Toxicol 2014; 33:271-281. [DOI: 10.1177/1091581814532959] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mustard gas (sulfur mustard [SM], bis-[2-chloroethyl] sulfide) is a vesicating chemical warfare agent and a potential chemical terrorism agent. Exposure of SM causes debilitating skin blisters (vesication) and injury to the eyes and the respiratory tract; of these, the respiratory injury, if severe, may even be fatal. Therefore, developing an effective therapeutic strategy to protect against SM-induced respiratory injury is an urgent priority of not only the US military but also the civilian antiterrorism agencies, for example, the Homeland Security. Toward developing a respiratory medical countermeasure for SM, four different classes of therapeutic compounds have been evaluated in the past: anti-inflammatory compounds, antioxidants, protease inhibitors and antiapoptotic compounds. This review examines all of these different options; however, it suggests that preventing cell death by inhibiting apoptosis seems to be a compelling strategy but possibly dependent on adjunct therapies using the other drugs, that is, anti-inflammatory, antioxidant, and protease inhibitor compounds.
Collapse
Affiliation(s)
- Brian M. Keyser
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Devon K. Andres
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Wesley W. Holmes
- Analytical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Danielle Paradiso
- Analytical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Ashley Appell
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Valerie A. Letukas
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Betty Benton
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Offie E. Clark
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Xiugong Gao
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Prabhati Ray
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Dana R. Anderson
- Analytical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Radharaman Ray
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| |
Collapse
|
24
|
Batal M, Boudry I, Mouret S, Cléry-Barraud C, Wartelle J, Bérard I, Douki T. DNA damage in internal organs after cutaneous exposure to sulphur mustard. Toxicol Appl Pharmacol 2014; 278:39-44. [PMID: 24732442 DOI: 10.1016/j.taap.2014.04.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 03/27/2014] [Accepted: 04/03/2014] [Indexed: 11/30/2022]
Abstract
Sulphur mustard (SM) is a chemical warfare agent that attacks mainly skin, eye and lungs. Due to its lipophilic properties, SM is also able to diffuse through the skin and reach internal organs. DNA represents one of the most critical molecular targets of this powerful alkylating agent which modifies DNA structure by forming monoadducts and biadducts. These DNA lesions are involved in the acute toxicity of SM as well as its long-term carcinogenicity. In the present work we studied the formation and persistence of guanine and adenine monoadducts and guanine biadducts in the DNA of brain, lungs, kidneys, spleen, and liver of SKH-1 mice cutaneously exposed to 2, 6 and 60mg/kg of SM. SM-DNA adducts were detected in all studied organs, except in liver at the two lowest doses. Brain and lungs were the organs with the highest level of SM-DNA adducts, followed by kidney, spleen and liver. Monitoring the level of adducts for three weeks after cutaneous exposure showed that the lifetime of adducts were not the same in all organs, lungs being the organ with the longest persistence. Diffusion from skin to internal organs was much more efficient at the highest compared to the lowest dose investigated as the result of the loss of the skin barrier function. These data provide novel information on the distribution of SM in tissues following cutaneous exposures and indicate that brain is an important target.
Collapse
Affiliation(s)
- Mohamed Batal
- Laboratoire « Lésions des Acides Nucléiques », Université Joseph Fourier - Grenoble 1/CEA/Institut Nanoscience et Cryogénie/SCIB, UMR-E3, Grenoble, France; Département de Toxicologie et Risques Chimiques; Unité de Brûlure Chimique, Institut de Recherche Biomédicale des Armées, Antenne de La Tronche, BP87, F-38702 La Tronche Cedex, France
| | - Isabelle Boudry
- Département de Toxicologie et Risques Chimiques; Unité de Brûlure Chimique, Institut de Recherche Biomédicale des Armées, Antenne de La Tronche, BP87, F-38702 La Tronche Cedex, France
| | - Stéphane Mouret
- Département de Toxicologie et Risques Chimiques; Unité de Brûlure Chimique, Institut de Recherche Biomédicale des Armées, Antenne de La Tronche, BP87, F-38702 La Tronche Cedex, France
| | - Cécile Cléry-Barraud
- Département de Toxicologie et Risques Chimiques; Unité de Brûlure Chimique, Institut de Recherche Biomédicale des Armées, Antenne de La Tronche, BP87, F-38702 La Tronche Cedex, France
| | - Julien Wartelle
- Département de Toxicologie et Risques Chimiques; Unité de Brûlure Chimique, Institut de Recherche Biomédicale des Armées, Antenne de La Tronche, BP87, F-38702 La Tronche Cedex, France
| | - Izabel Bérard
- Laboratoire « Lésions des Acides Nucléiques », Université Joseph Fourier - Grenoble 1/CEA/Institut Nanoscience et Cryogénie/SCIB, UMR-E3, Grenoble, France
| | - Thierry Douki
- Laboratoire « Lésions des Acides Nucléiques », Université Joseph Fourier - Grenoble 1/CEA/Institut Nanoscience et Cryogénie/SCIB, UMR-E3, Grenoble, France.
| |
Collapse
|
25
|
Lomash V, Jadhav SE, Vijayaraghavan R, Pant SC. Time course pathogenesis of sulphur mustard-induced skin lesions in mouse model. Int Wound J 2012; 10:441-54. [PMID: 22672652 DOI: 10.1111/j.1742-481x.2012.01003.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Sulphur mustard (SM) is a bifunctional alkylating agent that causes cutaneous blistering in humans and animals. In this study, we have presented closer views on pathogenesis of SM-induced skin injury in a mouse model. SM diluted in acetone was applied once dermally at a dose of 5 or 10 mg/kg to Swiss albino mice. Skin was dissected out at 0, 1, 3, 6, 12, 24, 48, 72 and 168 hours, post-SM exposure for studying histopathological changes and immunohistochemistry of inflammatory-reparative biomarkers, namely, transforming growth factor alpha (TGF-α), fibroblast growth factor (FGF), endothelial nitric oxide synthase (eNOS) and interlukin 6 (IL-6). Histopathological changes were similar to other mammalian species and basal cell damage resembled the histopathological signs observed with vesication in human skin. Inflammatory cell recruitment at the site of injury was supported by differential expressions of IL-6 at various stages. Time-dependent expressions of eNOS played pivotal roles in all the events of wound healing of SM-induced skin lesions. TGF-α and FGF were strongly associated with keratinocyte migration, re-epithelialisation, angiogenesis, fibroblast proliferation and cell differentiation. Furthermore, quantification of the tissue leukocytosis and DNA damage along with semiquantitative estimation of re-epithelialisation, fibroplasia and neovascularisation on histomorphologic scale could be efficiently used for screening the efficacy of orphan drugs against SM-induced skin injury.
Collapse
Affiliation(s)
- Vinay Lomash
- Department of Pharmacology and Toxicology, Defence Research and Development Establishment, Gwalior, Madhya Pradesh, India
| | | | | | | |
Collapse
|
26
|
Jain AK, Tewari-Singh N, Gu M, Inturi S, White CW, Agarwal R. Sulfur mustard analog, 2-chloroethyl ethyl sulfide-induced skin injury involves DNA damage and induction of inflammatory mediators, in part via oxidative stress, in SKH-1 hairless mouse skin. Toxicol Lett 2011; 205:293-301. [PMID: 21722719 DOI: 10.1016/j.toxlet.2011.06.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 06/13/2011] [Accepted: 06/14/2011] [Indexed: 12/20/2022]
Abstract
Bifunctional alkyalating agent, sulfur mustard (SM)-induced cutaneous injury is characterized by inflammation and delayed blistering. Our recent studies demonstrated that 2-chloroethyl ethyl sulfide (CEES), a monofunctional analog of SM that can be used in laboratory settings, induces oxidative stress. This could be the major cause of the activation of Akt/MAP kinase and AP1/NF-κB pathways that are linked to the inflammation and microvesication, and histopathological alterations in SKH-1 hairless mouse skin. To further establish a link between CEES-induced DNA damage and signaling pathways and inflammatory responses, skin samples from mice exposed to 2 mg or 4 mg CEES for 9-48 h were subjected to molecular analysis. Our results show a strong CEES-induced phosphorylation of H2A.X and an increase in cyclooxygenase-2 (COX-2), inducible NOS (iNOS), and matrix metalloproteinase-9 (MMP-9) levels, indicating the involvement of DNA damage and inflammation in CEES-induced skin injury in male and female mice. Since, our recent studies showed reduction in CEES-induced inflammatory responses by glutathione (GSH), we further assessed the role of oxidative stress in CEES-related DNA damage and the induction of inflammatory molecules. Oral GSH (300 mg/kg) administration 1h before CEES exposure attenuated the increase in both CEES-induced H2A.X phosphorylation (59%) as well as expression of COX-2 (68%), iNOS (53%) and MMP-9 (54%). Collectively, our results indicate that CEES-induced skin injury involves DNA damage and an induction of inflammatory mediators, at least in part via oxidative stress. This study could help in identifying countermeasures that alone or in combination, can target the unveiled pathways for reducing skin injury in humans by SM.
Collapse
Affiliation(s)
- Anil K Jain
- Department of Pharmaceutical Sciences, University of Colorado Denver School of Pharmacy, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Paromov V, Brannon M, Kumari S, Samala M, Qui M, Smith M, Stone WL. Sodium Pyruvate Modulates Cell Death Pathways in HaCaT Keratinocytes Exposed to Half-Mustard Gas. Int J Toxicol 2011; 30:197-206. [DOI: 10.1177/1091581810390824] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
2-Chloroethyl ethyl sulfide (CEES) or half-mustard gas, a sulfur mustard (HD) analog, is a genotoxic agent that causes oxidative stress and induces both apoptotic and necrotic cell death. Sodium pyruvate induced a necrosis-to-apoptosis shift in HaCaT cells exposed to CEES levels ≤ 1.5 mmol/L and lowered markers of DNA damage, oxidative stress, and inflammation. This study provides a rationale for the future development of multicomponent therapies for HD toxicity in the skin. We hypothesize that a combination of pyruvates with scavengers/antioxidants encapsulated in liposomes for optimal local delivery should be therapeutically beneficial against HD-induced skin injury. However, the latter suggestion should be verified in animal models exposed to HD.
Collapse
Affiliation(s)
- Victor Paromov
- Department of Pharmacology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Marianne Brannon
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Sudha Kumari
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Mallikarjun Samala
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Min Qui
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - William L. Stone
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| |
Collapse
|
28
|
Sunil VR, Patel-Vayas K, Shen J, Gow AJ, Laskin JD, Laskin DL. 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; 250:245-55. [PMID: 21070800 PMCID: PMC3520488 DOI: 10.1016/j.taap.2010.10.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 10/15/2010] [Accepted: 10/27/2010] [Indexed: 01/08/2023]
Abstract
Lung toxicity induced by sulfur mustard is associated with inflammation and oxidative stress. To elucidate mechanisms mediating pulmonary damage, we used 2-chloroethyl ethyl sulfide (CEES), a model sulfur mustard vesicant. Male mice (B6129) were treated intratracheally with CEES (3 or 6 mg/kg) or control. Animals were sacrificed 3, 7 or 14 days later and bronchoalveolar lavage (BAL) fluid and lung tissue collected. Treatment of mice with CEES resulted in an increase in BAL protein, an indication of alveolar epithelial damage, within 3 days. Expression of Ym1, an oxidative stress marker also increased in the lung, along with inducible nitric oxide synthase, and at 14 days, cyclooxygenase-2 and monocyte chemotactic protein-1, inflammatory proteins implicated in tissue injury. These responses were attenuated in mice lacking the p55 receptor for TNFα (TNFR1-/-), demonstrating that signaling via TNFR1 is key to CEES-induced injury, oxidative stress, and inflammation. CEES-induced upregulation of CuZn-superoxide dismutase (SOD) and MnSOD was delayed or absent in TNFR1-/- mice, relative to WT mice, suggesting that TNFα mediates early antioxidant responses to lung toxicants. Treatment of WT mice with CEES also resulted in functional alterations in the lung including decreases in compliance and increases in elastance. Additionally, methacholine-induced alterations in total lung resistance and central airway resistance were dampened by CEES. Loss of TNFR1 resulted in blunted functional responses to CEES. These effects were most notable in the airways. These data suggest that targeting TNFα signaling may be useful in mitigating lung injury, inflammation and functional alterations induced by vesicants.
Collapse
Affiliation(s)
- Vasanthi R. Sunil
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | - Kinal Patel-Vayas
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | - Jianliang Shen
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | - Andrew J. Gow
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | - Jeffrey D. Laskin
- Department of Environmental and Occupational Medicine, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Debra L. Laskin
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
29
|
Tewari-Singh N, Gu M, Agarwal C, White CW, Agarwal R. Biological and molecular mechanisms of sulfur mustard analogue-induced toxicity in JB6 and HaCaT cells: possible role of ataxia telangiectasia-mutated/ataxia telangiectasia-Rad3-related cell cycle checkpoint pathway. Chem Res Toxicol 2010; 23:1034-44. [PMID: 20469912 DOI: 10.1021/tx100038b] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Effective medical treatment and preventive measures for chemical warfare agent sulfur mustard (HD)-caused incapacitating skin toxicity are lacking, because of limited knowledge of its mechanism of action. The proliferating basal epidermal cells are primary major sites of attack during HD-caused skin injury. Therefore, employing mouse JB6 and human HaCaT epidermal cells, here, we investigated the molecular mechanism of HD analogue 2-chloroethyl ethyl sulfide (CEES)-induced skin cytotoxicity. As compared to the control, up to 1 mM CEES treatment of these cells for 2, 4, and 24 h caused dose-dependent decreases in cell viability and proliferation as measured by DNA synthesis, together with S and G2-M phase arrest in cell cycle progression. Mechanistic studies showed phosphorylation of DNA damage sensors and checkpoint kinases, ataxia telangiectasia-mutated (ATM) at ser1981 and ataxia telangiectasia-Rad3-related (ATR) at ser428 within 30 min of CEES exposure, and modulation of S and G2-M phase-associated cell cycle regulatory proteins, which are downstream targets of ATM and ATR kinases. Hoechst-propidium iodide staining demonstrated that CEES-induced cell death was both necrotic and apoptotic in nature, and the latter was induced at 4 and 24 h of CEES treatment in HaCaT and JB6 cells, respectively. An increase in caspase-3 activity and both caspase-3 and poly(ADP-ribose)polymerase (PARP) cleavage coinciding with CEES-caused apoptosis in both cell lines suggested the involvement of the caspase pathway. Together, our findings suggest a DNA-damaging effect of CEES that activates ATM/ATR cell cycle checkpoint signaling as well as caspase-PARP pathways, leading to cell cycle arrest and apoptosis/necrosis in both JB6 and HaCaT cells. The identified molecular targets, quantitative biomarkers, and epidermal cell models in this study have the potential and usefulness in rapid development of effective prophylactic and therapeutic interventions against HD-induced skin toxicity.
Collapse
Affiliation(s)
- Neera Tewari-Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, 12700 East 19th Avenue, Box C238 P-15, Research 2, Aurora, Colorado 80045, USA
| | | | | | | | | |
Collapse
|
30
|
Sulfur mustard-induced pulmonary injury: therapeutic approaches to mitigating toxicity. Pulm Pharmacol Ther 2010; 24:92-9. [PMID: 20851203 DOI: 10.1016/j.pupt.2010.09.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 09/03/2010] [Accepted: 09/09/2010] [Indexed: 11/23/2022]
Abstract
Sulfur mustard (SM) is highly toxic to the lung inducing both acute and chronic effects including upper and lower obstructive disease, airway inflammation, and acute respiratory distress syndrome, and with time, tracheobronchial stenosis, bronchitis, and bronchiolitis obliterans. Thus it is essential to identify effective strategies to mitigate the toxicity of SM and related vesicants. Studies in animals and in cell culture models have identified key mechanistic pathways mediating their toxicity, which may be relevant targets for the development of countermeasures. For example, following SM poisoning, DNA damage, apoptosis, and autophagy are observed in the lung, along with increased expression of activated caspases and DNA repair enzymes, biochemical markers of these activities. This is associated with inflammatory cell accumulation in the respiratory tract and increased expression of tumor necrosis factor-α and other proinflammatory cytokines, as well as reactive oxygen and nitrogen species. Matrix metalloproteinases are also upregulated in the lung after SM exposure, which are thought to contribute to the detachment of epithelial cells from basement membranes and disruption of the pulmonary epithelial barrier. Findings that production of inflammatory mediators correlates directly with altered lung function suggests that they play a key role in toxicity. In this regard, specific therapeutic interventions currently under investigation include anti-inflammatory agents (e.g., steroids), antioxidants (e.g., tocopherols, melatonin, N-acetylcysteine, nitric oxide synthase inhibitors), protease inhibitors (e.g., doxycycline, aprotinin, ilomastat), surfactant replacement, and bronchodilators. Effective treatments may depend on the extent of lung injury and require a multi-faceted pharmacological approach.
Collapse
|
31
|
Black AT, Hayden PJ, Casillas RP, Heck DE, Gerecke DR, Sinko PJ, Laskin DL, Laskin JD. Expression of proliferative and inflammatory markers in a full-thickness human skin equivalent following exposure to the model sulfur mustard vesicant, 2-chloroethyl ethyl sulfide. Toxicol Appl Pharmacol 2010; 249:178-87. [PMID: 20840853 DOI: 10.1016/j.taap.2010.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 08/31/2010] [Accepted: 09/03/2010] [Indexed: 12/20/2022]
Abstract
Sulfur mustard is a potent vesicant that induces inflammation, edema and blistering following dermal exposure. To assess molecular mechanisms mediating these responses, we analyzed the effects of the model sulfur mustard vesicant, 2-chloroethyl ethyl sulfide, on EpiDerm-FT™, a commercially available full-thickness human skin equivalent. CEES (100-1000 μM) caused a concentration-dependent increase in pyknotic nuclei and vacuolization in basal keratinocytes; at high concentrations (300-1000 μM), CEES also disrupted keratin filament architecture in the stratum corneum. This was associated with time-dependent increases in expression of proliferating cell nuclear antigen, a marker of cell proliferation, and poly(ADP-ribose) polymerase (PARP) and phosphorylated histone H2AX, markers of DNA damage. Concentration- and time-dependent increases in mRNA and protein expression of eicosanoid biosynthetic enzymes including COX-2, 5-lipoxygenase, microsomal PGE₂ synthases, leukotriene (LT) A₄ hydrolase and LTC₄ synthase were observed in CEES-treated skin equivalents, as well as in antioxidant enzymes, glutathione S-transferases A1-2 (GSTA1-2), GSTA3 and GSTA4. These data demonstrate that CEES induces rapid cellular damage, cytotoxicity and inflammation in full-thickness skin equivalents. These effects are similar to human responses to vesicants in vivo and suggest that the full thickness skin equivalent is a useful in vitro model to characterize the biological effects of mustards and to develop potential therapeutics.
Collapse
Affiliation(s)
- Adrienne T Black
- Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Malaviya R, Sunil VR, Cervelli J, Anderson DR, Holmes WW, Conti ML, Gordon RE, Laskin JD, Laskin DL. Inflammatory effects of inhaled sulfur mustard in rat lung. Toxicol Appl Pharmacol 2010; 248:89-99. [PMID: 20659490 DOI: 10.1016/j.taap.2010.07.018] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 07/16/2010] [Accepted: 07/19/2010] [Indexed: 12/19/2022]
Abstract
Inhalation of sulfur mustard (SM), a bifunctional alkylating agent that causes severe lung damage, is a significant threat to both military and civilian populations. The mechanisms mediating its cytotoxic effects are unknown and were investigated in the present studies. Male rats Crl:CD(SD) were anesthetized, and then intratracheally intubated and exposed to 0.7-1.4mg/kg SM by vapor inhalation. Animals were euthanized 6, 24, 48h or 7days post-exposure and bronchoalveolar lavage fluid (BAL) and lung tissue collected. Exposure of rats to SM resulted in rapid pulmonary toxicity, including focal ulceration and detachment of the trachea and bronchial epithelia from underlying mucosa, thickening of alveolar septal walls and increased numbers of inflammatory cells in the tissue. There was also evidence of autophagy and apoptosis in the tissue. This was correlated with increased BAL protein content, a marker of injury to the alveolar epithelial lining. SM exposure also resulted in increased expression of markers of inflammation including cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNFα), inducible nitric oxide synthase (iNOS), and matrix metalloproteinase-9 (MMP-9), each of which has been implicated in pulmonary toxicity. Whereas COX-2, TNFα and iNOS were mainly localized in alveolar regions, MMP-9 was prominent in bronchial epithelium. In contrast, expression of the anti-oxidant hemeoxygenase, and the anti-inflammatory collectin, surfactant protein-D, decreased in the lung after SM exposure. These data demonstrate that SM-induced oxidative stress and injury are associated with the generation of cytotoxic inflammatory proteins which may contribute to the pathogenic response to this vesicant.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kehe K, Balszuweit F, Steinritz D, Thiermann H. Molecular toxicology of sulfur mustard-induced cutaneous inflammation and blistering. Toxicology 2009; 263:12-9. [DOI: 10.1016/j.tox.2009.01.019] [Citation(s) in RCA: 197] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 01/07/2009] [Accepted: 01/08/2009] [Indexed: 02/07/2023]
|