1
|
Day BJ. Oxidative Stress: An Intersection Between Radiation and Sulfur Mustard Lung Injury. Disaster Med Public Health Prep 2024; 18:e86. [PMID: 38706344 PMCID: PMC11218645 DOI: 10.1017/dmp.2023.238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Nuclear and chemical weapons of mass destruction share both a tragic and beneficial legacy in mankind's history and health. The horrific health effects of ionizing radiation and mustard gas exposures unleashed during disasters, wars, and conflicts have been harnessed to treat human health maladies. Both agents of destruction have been transformed into therapies to treat a wide range of cancers. The discovery of therapeutic uses of radiation and sulfur mustard was largely due to observations by clinicians treating victims of radiation and sulfur mustard gas exposures. Clinicians identified vulnerability of leukocytes to these agents and repurposed their use in the treatment of leukemias and lymphomas. Given the overlap in therapeutic modalities, it goes to reason that there may be common mechanisms to target as protective strategies against their damaging effects. This commentary will highlight oxidative stress as a common mechanism shared by both radiation and sulfur mustard gas exposures and discuss potential therapies targeting oxidative stress as medical countermeasures against the devastating lung diseases wrought by these agents.
Collapse
Affiliation(s)
- Brian J Day
- Department of Medicine, National Jewish Health, Denver, CO, USA
| |
Collapse
|
2
|
Armoo A, Diemer T, Donkor A, Fedorchik J, Van Slambrouck S, Willand-Charnley R, Logue BA. Methimazole, an Effective Neutralizing Agent of the Sulfur Mustard Derivative 2-Chloroethyl Ethyl Sulfide. ACS BIO & MED CHEM AU 2023; 3:448-460. [PMID: 37876493 PMCID: PMC10591301 DOI: 10.1021/acsbiomedchemau.2c00087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 10/26/2023]
Abstract
Sulfur mustard (SM), designated by the military as HD, is a highly toxic and dangerous vesicant that has been utilized as a chemical warfare agent since World War I. Despite SM's extensive history, an effective antidote does not exist. The effects of SM are predominantly based on its ability to alkylate important biomolecules. Also, with the potential for a fraction of SM to remain unreacted up to days after initial contact, a window of opportunity exists for direct neutralization of unreacted SM over the days following exposure. In this study, we evaluated the structure-activity relationship of multiple nucleophilic molecules to neutralize the toxic effects of 2-chloroethyl ethyl sulfide (CEES), a monofunctional analogue of SM, on human keratinocyte (HaCaT) cells. Cell viability, relative loss of extracellular matrix adhesions, and apoptosis caused by CEES were measured via MTT, cell-matrix adhesion (CMA), and apoptosis protein marker assays, respectively. A set of five two-carbon compounds with various functional groups served as a preliminary group of first-generation neutralizing agents to survey the correlation between mitigation of CEES's toxic effects and functional group nucleophilicity. Apart from thioacids, which produced additive toxicity, we generally observed the trend of increasing protection from cytotoxicity with increasing nucleophilicity. We extended this treatment strategy to second-generation agents which contained advantageous structural features identified from the first-generation molecules. Our results show that methimazole (MIZ), a currently FDA-approved drug used to treat hyperthyroidism, effectively reduced cytotoxicity, increased CMA, and decreased apoptosis resulting from CEES toxicity. MIZ selectively reacts with CEES to produce 2-(2-(ethylthio)ethylthio)-1-methyl-1H-imidazole (EEMI) in media and cell lysate treatments resulting in the reduction of toxicity. Based on these results, future development of MIZ as an SM therapeutic may provide a viable approach to reduce both the immediate and long-term toxicity of SM and may also help mitigate slower developing SM toxicity due to residual intact SM.
Collapse
Affiliation(s)
- Albert Armoo
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Tanner Diemer
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Abigail Donkor
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Jerrod Fedorchik
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, South Dakota 57007, United States
| | | | - Rachel Willand-Charnley
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Brian A Logue
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, South Dakota 57007, United States
| |
Collapse
|
3
|
The Use of Thiocyanate Formulations to Create Manganese Porphyrin Antioxidants That Supplement Innate Immunity. Antioxidants (Basel) 2022; 11:antiox11071252. [PMID: 35883743 PMCID: PMC9311894 DOI: 10.3390/antiox11071252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/02/2022] [Accepted: 06/09/2022] [Indexed: 01/25/2023] Open
Abstract
The innate immune response to infection results in inflammation and oxidative damage, creating a paradox where most anti-inflammatory and antioxidant therapies can further suppress an already inadequate immune response. We have previously reported the beneficial effects of the exogenous supplementation of innate immunity with small pseudohalide thiocyanate (−SCN) in a mouse model of a cystic fibrosis (CF) lung infection and inflammation. The object of this study was to evaluate the use of −SCN as a counter anion for cationic manganese porphyrin (MnP) catalytic antioxidants, which could increase the parent compound’s antioxidant spectrum against hypohalous acids while supplementing innate immunity. The antioxidant activities of the parent compound were examined, as its chloride salt was compared with the −SCN-anion exchanged compound, (MnP(SCN) versus MnP(Cl)). We measured the superoxide dismutase activity spectrophotometrically and performed hydrogen peroxide scavenging using oxygen and hydrogen peroxide electrodes. Peroxidase activity was measured using an amplex red assay. The inhibition of lipid peroxidation was assessed using a thiobarbituric acid reactive species (TBARS) assay. The effects of the MnP compounds on macrophage phagocytosis were assessed by flow cytometry. The abilities of the MnP(Cl) formulations to protect human bronchiolar epithelial cells against hypochlorite (HOCl) and glycine chloramine versus their MnP(SCN) formulations were assessed using a cell viability assay. We found that anions exchanging out the chloride for −SCN improved the cellular bioavailability but did not adversely affect the cell viability or phagocytosis and that they switched hydrogen-peroxide scavenging from a dismutation reaction to a peroxidase reaction. In addition, the −SCN formulations improved the ability of MnPs to protect human bronchiolar epithelial cells against hypochlorous acid (HOCl) and glycine chloramine toxicity. These novel types of antioxidants may be more beneficial in treating lung disease that is associated with chronic infections or acute infectious exacerbations.
Collapse
|
4
|
Batinic-Haberle I, Tovmasyan A, Huang Z, Duan W, Du L, Siamakpour-Reihani S, Cao Z, Sheng H, Spasojevic I, Alvarez Secord A. H 2O 2-Driven Anticancer Activity of Mn Porphyrins and the Underlying Molecular Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6653790. [PMID: 33815656 PMCID: PMC7987459 DOI: 10.1155/2021/6653790] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Mn(III) ortho-N-alkyl- and N-alkoxyalkyl porphyrins (MnPs) were initially developed as superoxide dismutase (SOD) mimics. These compounds were later shown to react with numerous reactive species (such as ONOO-, H2O2, H2S, CO3 •-, ascorbate, and GSH). Moreover, the ability of MnPs to oxidatively modify activities of numerous proteins has emerged as their major mechanism of action both in normal and in cancer cells. Among those proteins are transcription factors (NF-κB and Nrf2), mitogen-activated protein kinases, MAPKs, antiapoptotic bcl-2, and endogenous antioxidative defenses. The lead Mn porphyrins, namely, MnTE-2-PyP5+ (BMX-010, AEOL10113), MnTnBuOE-2-PyP5+ (BMX-001), and MnTnHex-2-PyP5+, were tested in numerous injuries of normal tissue and cellular and animal cancer models. The wealth of the data led to the progression of MnTnBuOE-2-PyP5+ into four Phase II clinical trials on glioma, head and neck cancer, anal cancer, and multiple brain metastases, while MnTE-2-PyP5+ is in Phase II clinical trial on atopic dermatitis and itch.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zhiqing Huang
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Weina Duan
- Departments of Anesthesiology, Neurobiology, and Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Li Du
- Departments of Anesthesiology, Neurobiology, and Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Zhipeng Cao
- Departments of Anesthesiology, Neurobiology, and Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Huaxin Sheng
- Departments of Anesthesiology, Neurobiology, and Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Pharmacokinetics/Pharmacodynamics (PK/PD) Core Laboratory, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Angeles Alvarez Secord
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
5
|
Sin KR, Ko SG, Kim CJ, Maeng TW, Choe SJ, Ri KR. Reduction of peroxynitrite by some manganoporphyrins of AEOL series: DFT approach with dispersion correction and NBO analysis. J Inorg Biochem 2020; 214:111299. [PMID: 33152662 DOI: 10.1016/j.jinorgbio.2020.111299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/21/2020] [Accepted: 10/24/2020] [Indexed: 11/18/2022]
Abstract
The AEOL series of manganoporphyrins (MnP; AEOL compounds were named by US Aeolus pharmaceuticals) designed as superoxide dismutase mimic are well-known for their powerful catalytic activity to neutralize reactive oxygen and nitrogen species. Reductive oxygen atom cleavage from peroxynitrite (ONOO-) to form NO2 in aqueous solution by some AEOL compounds (AEOL-10113, AEOL-10150, AEOL-11114 and AEOL-11203) was studied by DFT/M06-2X computations with D3 dispersion correction and gCP (geometrical counterpoise correction) for basis set superposition error. DFT computation showed that AEOL-10150 can form the most stable association complex {MnP…OONO} among four AEOL models. AEOL-10150 complex with ONOO- has the lowest deformation energy. In AEOL compounds and their association complexes with ONOO-, Mn atom prefered the high spin state (S = 2) to the intermediate spin state (S = 1). Natural bond orbital analysis showed that electron transfer from the most negative oxygen atom in ONOO- to Mn atom in MnP has the biggest interaction energy among all kinds of donor-acceptor interactions between ONOO- and MnP.
Collapse
Affiliation(s)
- Kye-Ryong Sin
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea.
| | - Sun-Gyong Ko
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Chol-Jin Kim
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Tae-Won Maeng
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Sung-Jub Choe
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Kum-Ryong Ri
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| |
Collapse
|
6
|
Stawicki S, Le N, Garg M, Izurieta R, Garg S, Papadimos T, Arquilla B, Miller A, Khan A, Worlton T, Firstenberg M, Galwankar S, Raina S, Anderson H, Jeanmonod R, Kaufmann K, Jeanmonod D, De Wulf A, McCallister D, Bloem C, Opara I, Martin N, Asensio J. What's new in Academic International Medicine? International health security agenda – Expanded and re-defined. INTERNATIONAL JOURNAL OF ACADEMIC MEDICINE 2020. [DOI: 10.4103/ijam.ijam_113_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
7
|
Jost P, Pejchal J, Kucera T, Muckova L, Stetina R. Screening of the chemoprotective effect of 13 compounds and their mixtures with sodium 2-mercaptoethanesulfonate against 2-chloroethyl ethyl sulfide. J Appl Biomed 2019; 17:136-145. [DOI: 10.32725/jab.2019.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 06/03/2019] [Indexed: 01/25/2023] Open
|
8
|
Pearson-Smith JN, Patel M. Antioxidant drug therapy as a neuroprotective countermeasure of nerve agent toxicity. Neurobiol Dis 2019; 133:104457. [PMID: 31028872 PMCID: PMC7721294 DOI: 10.1016/j.nbd.2019.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
The use of chemical warfare agents is an ongoing, significant threat to both civilians and military personnel worldwide. Nerve agents are by far the most formidable toxicants in terms of their lethality and toxicity. Nerve agents initiate neurotoxicity by the irreversible inhibition of acetylcholinesterase and resultant accumulation of acetylcholine in excitable tissues. The cholinergic toxidrome presents as miosis, lacrimation, diarrhea, fasciculations, seizures, respiratory arrest and coma. Current medical countermeasures can attenuate acute mortality and confer limited protection against secondary neuronal injury when given rapidly after exposure. However, there is an urgent need for the development of novel, add-on neuroprotective therapies to prevent mortality and long-term toxicity of nerve agents. Increasing evidence suggests that pathways other than direct acetylcholinesterase inhibition contribute to neurotoxicity and secondary neuronal injury. Among these, oxidative stress is emerging as a key therapeutic target for nerve agent toxicity. In this review, we discuss the rationale for targeting oxidative stress in nerve agent toxicity and highlight research investigating antioxidant therapy as a neuroprotective medical countermeasure to attenuate oxidative stress, neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Jennifer N Pearson-Smith
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 80045, United States of America
| | - Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 80045, United States of America.
| |
Collapse
|
9
|
Batinic-Haberle I, Tovmasyan A, Spasojevic I. Mn Porphyrin-Based Redox-Active Drugs: Differential Effects as Cancer Therapeutics and Protectors of Normal Tissue Against Oxidative Injury. Antioxid Redox Signal 2018; 29:1691-1724. [PMID: 29926755 PMCID: PMC6207162 DOI: 10.1089/ars.2017.7453] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE After approximatelty three decades of research, two Mn(III) porphyrins (MnPs), MnTE-2-PyP5+ (BMX-010, AEOL10113) and MnTnBuOE-2-PyP5+ (BMX-001), have progressed to five clinical trials. In parallel, another similarly potent metal-based superoxide dismutase (SOD) mimic-Mn(II)pentaaza macrocycle, GC4419-has been tested in clinical trial on application, identical to that of MnTnBuOE-2-PyP5+-radioprotection of normal tissue in head and neck cancer patients. This clearly indicates that Mn complexes that target cellular redox environment have reached sufficient maturity for clinical applications. Recent Advances: While originally developed as SOD mimics, MnPs undergo intricate interactions with numerous redox-sensitive pathways, such as those involving nuclear factor κB (NF-κB) and nuclear factor E2-related factor 2 (Nrf2), thereby impacting cellular transcriptional activity. An increasing amount of data support the notion that MnP/H2O2/glutathione (GSH)-driven catalysis of S-glutathionylation of protein cysteine, associated with modification of protein function, is a major action of MnPs on molecular level. CRITICAL ISSUES Differential effects of MnPs on normal versus tumor cells/tissues, which support their translation into clinic, arise from differences in their accumulation and redox environment of such tissues. This in turn results in different yields of MnP-driven modifications of proteins. Thus far, direct evidence for such modification of NF-κB, mitogen-activated protein kinases (MAPK), phosphatases, Nrf2, and endogenous antioxidative defenses was provided in tumor, while indirect evidence shows the modification of NF-κB and Nrf2 translational activities by MnPs in normal tissue. FUTURE DIRECTIONS Studies that simultaneously explore differential effects in same animal are lacking, while they are essential for understanding of extremely intricate interactions of metal-based drugs with complex cellular networks of normal and cancer cells/tissues.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- 1 Department of Radiation Oncology, Duke University School of Medicine , Durham, North Carolina
| | - Artak Tovmasyan
- 1 Department of Radiation Oncology, Duke University School of Medicine , Durham, North Carolina
| | - Ivan Spasojevic
- 2 Department of Medicine, Duke University School of Medicine , Durham, North Carolina.,3 PK/PD Core Laboratory, Pharmaceutical Research Shared Resource, Duke Cancer Institute , Durham, North Carolina
| |
Collapse
|
10
|
Beigi Harchegani A, Khor A, Tahmasbpour E, Ghatrehsamani M, Bakhtiari Kaboutaraki H, Shahriary A. Role of oxidative stress and antioxidant therapy in acute and chronic phases of sulfur mustard injuries: a review. Cutan Ocul Toxicol 2018; 38:9-17. [DOI: 10.1080/15569527.2018.1495230] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Asghar Beigi Harchegani
- Chemical Injuries Research Center, Systems biology and poisonings institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abolfazl Khor
- Chemical Injuries Research Center, Systems biology and poisonings institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Eisa Tahmasbpour
- Laboratory of Regenerative Medicine & Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Ghatrehsamani
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hamid Bakhtiari Kaboutaraki
- Chemical Injuries Research Center, Systems biology and poisonings institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems biology and poisonings institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Zhang XR, Zhou WX, Zhang YX. Improvements in SOD mimic AEOL-10150, a potent broad-spectrum antioxidant. Mil Med Res 2018; 5:30. [PMID: 30185231 PMCID: PMC6125955 DOI: 10.1186/s40779-018-0176-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 08/06/2018] [Indexed: 01/07/2023] Open
Abstract
AEOL-10150 is a broad-spectrum metalloporphyrin superoxidase dismutase (SOD) mimic specifically designed to neutralize reactive oxygen and nitrogen species. Research has shown that AEOL-10150 is a potent medical countermeasure against national security threats including sulfur mustard (SM), nerve agent exposure and radiation pneumonitis following a radiological/nuclear incident sufficient to cause acute radiation syndrome (ARS). AEOL-10150 performed well in animal safety studies, and two completed phase 1 safety studies in patients demonstrated that the drug was safe and well tolerated, indicating that AEOL-10150 has potential as a new catalytic antioxidant drug. In this article, we review improvements in AEOL-10150 in preclinical pharmacodynamic studies, especially regarding anti-SM, chlorine gas and radiation exposure studies.
Collapse
Affiliation(s)
- Xiao-Rui Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Wen-Xia Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Yong-Xiang Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| |
Collapse
|
12
|
Chang YC, Soriano M, Hahn RA, Casillas RP, Gordon MK, Laskin JD, Gerecke DR. Expression of cytokines and chemokines in mouse skin treated with sulfur mustard. Toxicol Appl Pharmacol 2018; 355:52-59. [PMID: 29935281 DOI: 10.1016/j.taap.2018.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 01/11/2023]
Abstract
Sulfur mustard (2,2'-dichlorodiethyl sulfide, SM) is a chemical warfare agent that generates an inflammatory response in the skin and causes severe tissue damage and blistering. In earlier studies, we identified cutaneous damage induced by SM in mouse ear skin including edema, erythema, epidermal hyperplasia and microblistering. The present work was focused on determining if SM-induced injury was associated with alterations in mRNA and protein expression of specific cytokines and chemokines in the ear skin. We found that SM caused an accumulation of macrophages and neutrophils in the tissue within one day which persisted for at least 7 days. This was associated with a 2-15 fold increase in expression of the proinflammatory cytokines interleukin-1β, interleukin-6, and tumor necrosis factor α at time points up to 7 days post-SM exposure. Marked increases (20-1000 fold) in expression of chemokines associated with recruitment and activation of macrophages were also noted in the tissue including growth-regulated oncogene α (GROα/CXCL1), monocyte chemoattractant protein 1 (MCP-1/CCL2), granulocyte-colony stimulating factor (GCSF/CSF3), macrophage inflammatory protein 1α (MIP1α/CCL3), and IFN-γ-inducible protein 10 (IP10/CXCL10). The pattern of cytokines/chemokine expression was coordinate with expression of macrophage elastase/MMP12 and neutrophil collagenase/MMP8 suggesting that macrophages and neutrophils were, at least in part, a source of cytokines and chemokines. These data support the idea that inflammatory cell-derived mediators contribute to the pathogenesis of SM induced skin damage. Modulating the infiltration of inflammatory cells and reducing the expression of inflammatory mediators in the skin may be an important strategy for mitigating SM-induced cutaneous injury.
Collapse
Affiliation(s)
- Yoke-Chen Chang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA.
| | - Melannie Soriano
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Rita A Hahn
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | | | - Marion K Gordon
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health, School of Public Health, Rutgers University, Piscataway, New Jersey, USA
| | - Donald R Gerecke
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
13
|
Affiliation(s)
- Gregory R Ciottone
- From Beth Israel Deaconess Medical Center, Harvard Medical School, Boston
| |
Collapse
|
14
|
Advanced biotherapy for the treatment of sulfur mustard poisoning. Chem Biol Interact 2018; 286:111-118. [DOI: 10.1016/j.cbi.2018.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 02/09/2018] [Accepted: 03/19/2018] [Indexed: 01/09/2023]
|
15
|
Goswami DG, Agarwal R, Tewari-Singh N. Phosgene oxime: Injury and associated mechanisms compared to vesicating agents sulfur mustard and lewisite. Toxicol Lett 2017; 293:112-119. [PMID: 29141200 DOI: 10.1016/j.toxlet.2017.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/08/2017] [Accepted: 11/11/2017] [Indexed: 12/24/2022]
Abstract
Phosgene Oxime (CX, Cl2CNOH), a halogenated oxime, is a potent chemical weapon that causes immediate acute injury and systemic effects. CX, grouped together with vesicating agents, is an urticant or nettle agent with highly volatile, reactive, corrosive, and irritating vapor, and has considerably different chemical properties and toxicity compared to other vesicants. CX is absorbed quickly through clothing with faster cutaneous penetration compared to other vesicating agents causing instantaneous and severe damage. For this reason, it could be produced as a weaponized mixture with other chemical warfare agents to enhance their deleterious effects. The immediate devastating effects of CX and easy synthesis makes it a dangerous chemical with both military and terrorist potentials. Although CX is the most potent vesicating agent, it is one of the least studied chemical warfare agents and the pathophysiology as well as long term effects are largely unknown. CX exposure results in immediate pain and inflammation, and it mainly affects skin, eye and respiratory system. There are no antidotes available against CX-induced injury and the treatment is only supportive. This review summarizes existing knowledge regarding exposure, toxicity and the probable underlying mechanisms of CX compared to other important vesicants' exposure.
Collapse
Affiliation(s)
- Dinesh Giri Goswami
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
16
|
Scavenging reactive oxygen species inhibits status epilepticus-induced neuroinflammation. Exp Neurol 2017; 298:13-22. [PMID: 28822838 DOI: 10.1016/j.expneurol.2017.08.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/26/2017] [Accepted: 08/15/2017] [Indexed: 02/07/2023]
Abstract
Inflammation has been identified as an important mediator of seizures and epileptogenesis. Understanding the mechanisms underlying seizure-induced neuroinflammation could lead to the development of novel therapies for the epilepsies. Reactive oxygen species (ROS) are recognized as mediators of seizure-induced neuronal damage and are known to increase in models of epilepsies. ROS are also known to contribute to inflammation in several disease states. We hypothesized that ROS are key modulators of neuroinflammation i.e. pro-inflammatory cytokine production and microglial activation in acquired epilepsy. The role of ROS in modulating seizure-induced neuroinflammation was investigated in the pilocarpine model of temporal lobe epilepsy (TLE). Pilocarpine-induced status epilepticus (SE) resulted in a time-dependent increase in pro-inflammatory cytokine production in the hippocampus and piriform cortex. Scavenging ROS with a small-molecule catalytic antioxidant decreased SE-induced pro-inflammatory cytokine production and microglial activation, suggesting that ROS contribute to SE-induced neuroinflammation. Scavenging ROS also attenuated phosphorylation of ribosomal protein S6, the downstream target of the mammalian target of rapamycin (mTOR) pathway indicating that this pathway might provide one mechanistic link between SE-induced ROS production and inflammation. Together, these results demonstrate that ROS contribute to SE-induced cytokine production and antioxidant treatment may offer a novel approach to control neuroinflammation in epilepsy.
Collapse
|
17
|
Srivastava RK, Li C, Weng Z, Agarwal A, Elmets CA, Afaq F, Athar M. Defining cutaneous molecular pathobiology of arsenicals using phenylarsine oxide as a prototype. Sci Rep 2016; 6:34865. [PMID: 27725709 PMCID: PMC5057142 DOI: 10.1038/srep34865] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/16/2016] [Indexed: 11/09/2022] Open
Abstract
Arsenicals are painful, inflammatory and blistering causing agents developed as chemical weapons in World War I/II. However, their large stockpiles still exist posing threat to public health. Phenylarsine oxide (PAO), a strong oxidant and a prototype arsenical is tested for its suitability to defining molecular mechanisms underlying arsenicals-mediated tissue injury. Topically applied PAO induces cutaneous erythema, edema and micro-blisters. These gross inflammatory responses were accompanied by the enhanced production of pro-inflammatory cytokines, ROS and unfolded protein response (UPR) signaling activation. To demonstrate the involvement of UPR in the pathobiology of these lesions, we employed chemical chaperone, 4-phenylbutyric acid (4-PBA) which attenuates UPR. 4-PBA significantly reduced PAO-induced inflammation and blistering. Similar to its effects in murine epidermis, a dose- and time-dependent upregulation of ROS, cytokines, UPR proteins (GRP78, p-PERK, p-eIF2α, ATF4 and CHOP) and apoptosis were observed in PAO-treated human skin keratinocytes NHEK and HaCaT. In addition, 4-PBA significantly restored these molecular alterations in these cells. Employing RNA interference (RNAi)-based approaches, CHOP was found to be a key regulator of these responses. These effects are similar to those manifested by lewisite suggesting that PAO could be used as a prototype of arsenicals to define the molecular pathogenesis of chemical injury.
Collapse
Affiliation(s)
- Ritesh K. Srivastava
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Alabama, USA
| | - Changzhao Li
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Alabama, USA
| | - Zhiping Weng
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Alabama, USA
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Craig A. Elmets
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Alabama, USA
| | - Farrukh Afaq
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Alabama, USA
| | - Mohammad Athar
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
18
|
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: 11] [Impact Index Per Article: 1.2] [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
|
19
|
White CW, Rancourt RC, Veress LA. Sulfur mustard inhalation: mechanisms of injury, alteration of coagulation, and fibrinolytic therapy. Ann N Y Acad Sci 2016; 1378:87-95. [PMID: 27384912 DOI: 10.1111/nyas.13130] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 01/02/2023]
Abstract
Acute lung injury due to sulfur mustard (SM) inhalation causes the formation of airway fibrin casts that obstruct airways at multiple levels, leading to acute respiratory failure and death. These pathophysiological effects are seen in rodent models of acute SM vapor inhalation, as well as in human victims of acute SM inhalation. In rat models, the initial steps in activation of the coagulation system at extravascular sites depend on tissue factor (TF) expression by airway cells, especially in the microparticle fraction, and these effects can be inhibited by TF pathway inhibitor protein. Not only does the procoagulant environment of the acutely injured lung contribute to airway cast formation, but these lesions persist in airways because of the activation of multiple antifibrinolytic pathways, including plasminogen activator inhibitor-1, thrombin-activatable fibrinolysis inhibitor, and α2-antiplasmin. Airway administration of tissue plasminogen activator can overwhelm these effects and save lives by preventing fibrin-dependent airway obstruction, gas-exchange abnormalities, and respiratory failure. In human survivors of SM inhalation, fibrotic processes, including bronchiolitis obliterans and interstitial fibrosis of the lung, are among the most disabling chronic lesions. Antifibrotic therapies may prove useful in preventing either or both of these forms of chronic lung damage.
Collapse
Affiliation(s)
- Carl W White
- Pediatric Airway Research Center, Department of Pediatrics, University of Colorado, Aurora, Colorado.
| | - Raymond C Rancourt
- Pediatric Airway Research Center, Department of Pediatrics, University of Colorado, Aurora, Colorado
| | - Livia A Veress
- Pediatric Airway Research Center, Department of Pediatrics, University of Colorado, Aurora, Colorado
| |
Collapse
|
20
|
Williams JP, Calvi L, Chakkalakal JV, Finkelstein JN, O’Banion MK, Puzas E. Addressing the Symptoms or Fixing the Problem? Developing Countermeasures against Normal Tissue Radiation Injury. Radiat Res 2016; 186:1-16. [PMID: 27332954 PMCID: PMC4991354 DOI: 10.1667/rr14473.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Laura Calvi
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Joe V. Chakkalakal
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Jacob N. Finkelstein
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York
| | - M. Kerry O’Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York
| | - Edward Puzas
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
21
|
Tewari-Singh N, Agarwal R. Mustard vesicating agent-induced toxicity in the skin tissue and silibinin as a potential countermeasure. Ann N Y Acad Sci 2016; 1374:184-92. [PMID: 27326543 DOI: 10.1111/nyas.13099] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Exposure to the vesicating agents sulfur mustard (SM) and nitrogen mustard (NM) causes severe skin injury with delayed blistering. Depending upon the dose and time of their exposure, edema and erythema develop into blisters, ulceration, necrosis, desquamation, and pigmentation changes, which persist weeks and even years after exposure. Research advances have generated data that have started to explain the probable mechanism of action of vesicant-induced skin toxicity; however, despite these advances, effective and targeted therapies are still deficient. This review highlights studies on two SM analogs, 2-chloroethyl ethyl sulfide (CEES) and NM, and CEES- and NM-induced skin injury mouse models that have substantially added to the knowledge on the complex pathways involved in mustard vesicating agent-induced skin injury. Furthermore, employing these mouse models, studies under the National Institutes of Health Countermeasures Against Chemical Threats program have identified the flavanone silibinin as a novel therapeutic intervention with the potential to be developed as an effective countermeasure against skin injury following exposure to mustard vesicating agents.
Collapse
Affiliation(s)
- Neera Tewari-Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
22
|
McElroy CS, Day BJ. Antioxidants as potential medical countermeasures for chemical warfare agents and toxic industrial chemicals. Biochem Pharmacol 2016; 100:1-11. [PMID: 26476351 PMCID: PMC4744107 DOI: 10.1016/j.bcp.2015.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/01/2015] [Indexed: 12/18/2022]
Abstract
The continuing horrors of military conflicts and terrorism often involve the use of chemical warfare agents (CWAs) and toxic industrial chemicals (TICs). Many CWA and TIC exposures are difficult to treat due to the danger they pose to first responders and their rapid onset that can produce death shortly after exposure. While the specific mechanism(s) of toxicity of these agents are diverse, many are associated either directly or indirectly with increased oxidative stress in affected tissues. This has led to the exploration of various antioxidants as potential medical countermeasures for CWA/TIC exposures. Studies have been performed across a wide array of agents, model organisms, exposure systems, and antioxidants, looking at an almost equally diverse set of endpoints. Attempts at treating CWAs/TICs with antioxidants have met with mixed results, ranging from no effect to nearly complete protection. The aim of this commentary is to summarize the literature in each category for evidence of oxidative stress and antioxidant efficacy against CWAs and TICs. While there is great disparity in the data concerning methods, models, and remedies, the outlook on antioxidants as medical countermeasures for CWA/TIC management appears promising.
Collapse
Affiliation(s)
- Cameron S McElroy
- Department of Medicine, National Jewish Health, Denver, CO 80206, United States; Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO 80045, United States
| | - Brian J Day
- Department of Medicine, National Jewish Health, Denver, CO 80206, United States; Department of Medicine, University of Colorado Denver, Aurora, CO 80045, United States; Department of Immunology, University of Colorado Denver, Aurora, CO 80045, United States; Department of Environmental & Occupational Health Sciences, University of Colorado Denver, Aurora, CO 80045, United States; Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO 80045, United States.
| |
Collapse
|
23
|
Jang YJ, Kim K, Tsay OG, Atwood DA, Churchill DG. Update 1 of: Destruction and Detection of Chemical Warfare Agents. Chem Rev 2015; 115:PR1-76. [DOI: 10.1021/acs.chemrev.5b00402] [Citation(s) in RCA: 249] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yoon Jeong Jang
- Molecular Logic Gate Laboratory, Department of Chemistry, KAIST, Daejeon, 305-701, Republic of Korea
| | - Kibong Kim
- Molecular Logic Gate Laboratory, Department of Chemistry, KAIST, Daejeon, 305-701, Republic of Korea
| | - Olga G. Tsay
- Molecular Logic Gate Laboratory, Department of Chemistry, KAIST, Daejeon, 305-701, Republic of Korea
| | - David A. Atwood
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506-0055, United States
| | - David G. Churchill
- Molecular Logic Gate Laboratory, Department of Chemistry, KAIST, Daejeon, 305-701, Republic of Korea
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS), 373-1 Guseong-dong, Yuseong-gu, Daejeon, 305−701, Republic of Korea
| |
Collapse
|
24
|
Tahmasbpour E, Reza Emami S, Ghanei M, Panahi Y. Role of oxidative stress in sulfur mustard-induced pulmonary injury and antioxidant protection. Inhal Toxicol 2015; 27:659-72. [DOI: 10.3109/08958378.2015.1092184] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Pearson JN, Rowley S, Liang LP, White AM, Day BJ, Patel M. Reactive oxygen species mediate cognitive deficits in experimental temporal lobe epilepsy. Neurobiol Dis 2015; 82:289-297. [PMID: 26184893 PMCID: PMC4871280 DOI: 10.1016/j.nbd.2015.07.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/06/2015] [Accepted: 07/09/2015] [Indexed: 11/08/2022] Open
Abstract
Cognitive dysfunction is an important comorbidity of temporal lobe epilepsy (TLE). However, no targeted therapies are available and the mechanisms underlying cognitive impairment, specifically deficits in learning and memory associated with TLE remain unknown. Oxidative stress is known to occur in the pathogenesis of TLE but its functional role remains to be determined. Here, we demonstrate that oxidative stress and resultant processes contribute to cognitive decline associated with epileptogenesis. Using a synthetic catalytic antioxidant, we show that pharmacological removal of reactive oxygen species (ROS) prevents 1) oxidative stress, 2) deficits in mitochondrial oxygen consumption rates, 3) hippocampal neuronal loss and 4) cognitive dysfunction without altering the intensity of the initial status epilepticus (SE) or epilepsy development in a rat model of SE-induced TLE. Moreover, the effects of the catalytic antioxidant on cognition persisted beyond the treatment period suggestive of disease-modification. The data implicate oxidative stress as a novel mechanism by which cognitive dysfunction can arise during epileptogenesis and suggest a potential disease-modifying therapeutic approach to target it.
Collapse
Affiliation(s)
- Jennifer N Pearson
- Neuroscience Program, University of Colorado, Anschutz Medical Campus, 80045, USA
| | - Shane Rowley
- Neuroscience Program, University of Colorado, Anschutz Medical Campus, 80045, USA
| | - Li-Ping Liang
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 80045, USA
| | - Andrew M White
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, 80045, USA
| | - Brian J Day
- National Jewish Health, Denver, CO 80206, USA
| | - Manisha Patel
- Neuroscience Program, University of Colorado, Anschutz Medical Campus, 80045, USA; Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 80045, USA.
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Kumar D, Tewari-Singh N, Agarwal C, Jain AK, Inturi S, Kant R, White CW, Agarwal R. Nitrogen mustard exposure of murine skin induces DNA damage, oxidative stress and activation of MAPK/Akt-AP1 pathway leading to induction of inflammatory and proteolytic mediators. Toxicol Lett 2015; 235:161-71. [PMID: 25891025 DOI: 10.1016/j.toxlet.2015.04.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/11/2015] [Accepted: 04/12/2015] [Indexed: 01/01/2023]
Abstract
Our recent studies in SKH-1 hairless mice have demonstrated that topical exposure to nitrogen mustard (NM), an analog of sulfur mustard (SM), triggers the inflammatory response, microvesication and apoptotic cell death. Here, we sought to identify the mechanism/s involved in these NM-induced injury responses. Results obtained show that NM exposure of SKH-1 hairless mouse skin caused H2A.X and p53 phosphorylation and increased p53 accumulation, indicating DNA damage. In addition, NM also induced the activation of MAPKs/ERK1/2, JNK1/2 and p38 as well as that of Akt together with the activation of transcription factor AP1. Also, NM exposure induced robust expression of pro-inflammatory mediators namely cyclooxygenase 2 and inducible nitric oxide synthase and cytokine tumor necrosis factor alpha, and increased the levels of proteolytic mediator matrix metalloproteinase 9. NM exposure of skin also increased lipid peroxidation, 5,5-dimethyl-2-(8-octanoic acid)-1-pyrroline N-oxide protein adduct formation, protein and DNA oxidation indicating an elevated oxidative stress. We also found NM-induced increase in the homologous recombinant repair pathway, suggesting its involvement in the repair of NM-induced DNA damage. Collectively, these results indicate that NM induces oxidative stress, mainly a bi-phasic response in DNA damage and activation of MAPK and Akt pathways, which activate transcription factor AP1 and induce the expression of inflammatory and proteolytic mediators, contributing to the skin injury response by NM. In conclusion, this study for the first time links NM-induced mechanistic changes with our earlier reported murine skin injury lesions with NM, which could be valuable to identify potential therapeutic targets and rescue agents.
Collapse
Affiliation(s)
- Dileep Kumar
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA
| | - Anil K Jain
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA
| | - Swetha Inturi
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA
| | - Rama Kant
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA
| | - Carl W White
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA.
| |
Collapse
|
29
|
Flavanone silibinin treatment attenuates nitrogen mustard-induced toxic effects in mouse skin. Toxicol Appl Pharmacol 2015; 285:71-8. [PMID: 25791923 DOI: 10.1016/j.taap.2015.03.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 03/06/2015] [Accepted: 03/07/2015] [Indexed: 01/12/2023]
Abstract
Currently, there is no effective antidote to prevent skin injuries by sulfur mustard (SM) and nitrogen mustard (NM), which are vesicating agents with potential relevance to chemical warfare, terrorist attacks, or industrial/laboratory accidents. Our earlier report has demonstrated the therapeutic efficacy of silibinin, a natural flavanone, in reversing monofunctional alkylating SM analog 2-chloroethyl ethyl sulfide-induced toxic effects in mouse skin. To translate this effect to a bifunctional alkylating vesicant, herein, efficacy studies were carried out with NM. Topical application of silibinin (1 or 2mg) 30 min after NM exposure on the dorsal skin of male SKH-1 hairless mice significantly decreased NM-induced toxic lesions at 24, 72 or 120 h post-exposure. Specifically, silibinin treatment resulted in dose-dependent reduction of NM-induced increase in epidermal thickness, dead and denuded epidermis, parakeratosis and microvesication. Higher silibinin dose also caused a 79% and 51%reversal in NM-induced increases in myeloperoxidase activity and COX-2 levels, respectively. Furthermore, silibinin completely prevented NM-induced H2A.X phosphorylation, indicating reversal of DNA damage which could be an oxidative DNA damage as evidenced by high levels of 8-oxodG in NM-exposed mouse skin that was significantly reversed by silibinin. Together, these findings suggest that attenuation of NM-induced skin injury by silibinin is due to its effects on the pathways associated with DNA damage, inflammation, vesication and oxidative stress. In conclusion, results presented here support the optimization of silibinin as an effective treatment of skin injury by vesicants.
Collapse
|
30
|
Goswami DG, Kumar D, Tewari-Singh N, Orlicky DJ, Jain AK, Kant R, Rancourt RC, Dhar D, Inturi S, Agarwal C, White CW, Agarwal R. Topical nitrogen mustard exposure causes systemic toxic effects in mice. ACTA ACUST UNITED AC 2014; 67:161-70. [PMID: 25481215 DOI: 10.1016/j.etp.2014.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/12/2014] [Accepted: 11/17/2014] [Indexed: 10/24/2022]
Abstract
Vesicating agents sulfur mustard (SM) and nitrogen mustard (NM) are reported to be easily absorbed by skin upon exposure causing severe cutaneous injury and blistering. Our studies show that topical exposure of NM (3.2mg) onto SKH-1 hairless mouse skin, not only caused skin injury, but also led to significant body weight loss and 40-80% mortality (120 h post-exposure), suggesting its systemic effects. Accordingly, further studies herein show that NM exposure initiated an increase in circulating white blood cells by 24h (neutrophils, eosinophils and basophils) and thereafter a decrease (neutrophils, lymphocytes and monocytes). NM exposure also reduced both white and red pulp areas of the spleen. In the small intestine, NM exposure caused loss of membrane integrity of the surface epithelium, abnormal structure of glands and degeneration of villi. NM exposure also resulted in the dilation of glomerular capillaries of kidneys, and an increase in blood urea nitrogen/creatinine ratio. Our results here with NM are consistent with earlier reports that exposure to higher SM levels can cause damage to the hematopoietic system, and kidney, spleen and gastrointestinal tract toxicity. These outcomes will add to our understanding of the toxic effects of topical vesicant exposure, which might be helpful towards developing effective countermeasures against injuries from acute topical exposures.
Collapse
Affiliation(s)
- Dinesh G Goswami
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dileep Kumar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David J Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anil K Jain
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rama Kant
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Raymond C Rancourt
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Deepanshi Dhar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Swetha Inturi
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carl W White
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
31
|
Stenger B, Zehfuß F, Mückter H, Schmidt A, Balszuweit F, Schäfer E, Büch T, Gudermann T, Thiermann H, Steinritz D. Activation of the chemosensing transient receptor potential channel A1 (TRPA1) by alkylating agents. Arch Toxicol 2014; 89:1631-43. [DOI: 10.1007/s00204-014-1414-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 11/06/2014] [Indexed: 12/21/2022]
|