1
|
Sevinc Ozdemir N, Belyaev D, Castro MN, Balakin S, Opitz J, Wihadmadyatami H, Anggraeni R, Yucel D, Kenar H, Beshchasna N, Ana ID, Hasirci V. Advances in In Vitro Blood-Air Barrier Models and the Use of Nanoparticles in COVID-19 Research. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:82-96. [PMID: 37597193 DOI: 10.1089/ten.teb.2023.0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2023]
Abstract
Respiratory infections caused by coronaviruses (CoVs) have become a major public health concern in the past two decades as revealed by the emergence of SARS-CoV in 2002, MERS-CoV in 2012, and SARS-CoV-2 in 2019. The most severe clinical phenotypes commonly arise from exacerbation of immune response following the infection of alveolar epithelial cells localized at the pulmonary blood-air barrier. Preclinical rodent models do not adequately represent the essential genetic properties of the barrier, thus necessitating the use of humanized transgenic models. However, existing monolayer cell culture models have so far been unable to mimic the complex lung microenvironment. In this respect, air-liquid interface models, tissue engineered models, and organ-on-a-chip systems, which aim to better imitate the infection site microenvironment and microphysiology, are being developed to replace the commonly used monolayer cell culture models, and their use is becoming more widespread every day. On the contrary, studies on the development of nanoparticles (NPs) that mimic respiratory viruses, and those NPs used in therapy are progressing rapidly. The first part of this review describes in vitro models that mimic the blood-air barrier, the tissue interface that plays a central role in COVID-19 progression. In the second part of the review, NPs mimicking the virus and/or designed to carry therapeutic agents are explained and exemplified.
Collapse
Affiliation(s)
- Neval Sevinc Ozdemir
- Acibadem University (ACU) Biomaterials A&R Center, Atasehir, Istanbul, Turkey
- Department of Medical Biotechnology, ACU Graduate School of Health Sciences, Istanbul, Turkey
- ACU Department of Pharmaceutical Basic Sciences, School of Pharmacy, Istanbul, Turkey
| | - Dmitry Belyaev
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Manuel Nieto Castro
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Sascha Balakin
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Joerg Opitz
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Hevi Wihadmadyatami
- Department of Tissue Engineering and Regenerative Medicine, Research Collaboration Center for Biomedical Scaffolds, National Research and Innovation Agency (BRIN) and Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
| | - Rahmi Anggraeni
- Department of Tissue Engineering and Regenerative Medicine, Research Collaboration Center for Biomedical Scaffolds, National Research and Innovation Agency (BRIN) and Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
| | - Deniz Yucel
- Acibadem University (ACU) Biomaterials A&R Center, Atasehir, Istanbul, Turkey
- ACU Graduate Department of Biomaterials, Istanbul, Turkey
- Department of Histology and Embryology, ACU School of Medicine, Istanbul, Turkey
| | - Halime Kenar
- Acibadem University (ACU) Biomaterials A&R Center, Atasehir, Istanbul, Turkey
- ACU Graduate Department of Biomaterials, Istanbul, Turkey
- ACU Faculty of Engineering Sciences, Department of Biomedical Engineering, Istanbul, Turkey
| | - Natalia Beshchasna
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Ika Dewi Ana
- Department of Tissue Engineering and Regenerative Medicine, Research Collaboration Center for Biomedical Scaffolds, National Research and Innovation Agency (BRIN) and Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
| | - Vasif Hasirci
- Acibadem University (ACU) Biomaterials A&R Center, Atasehir, Istanbul, Turkey
- ACU Graduate Department of Biomaterials, Istanbul, Turkey
- ACU Faculty of Engineering Sciences, Department of Biomedical Engineering, Istanbul, Turkey
- BIOMATEN, METU Ctr. of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey
| |
Collapse
|
2
|
Al-Rekabi Z, Dondi C, Faruqui N, Siddiqui NS, Elowsson L, Rissler J, Kåredal M, Mudway I, Larsson-Callerfelt AK, Shaw M. Uncovering the cytotoxic effects of air pollution with multi-modal imaging of in vitro respiratory models. ROYAL SOCIETY OPEN SCIENCE 2023; 10:221426. [PMID: 37063998 PMCID: PMC10090883 DOI: 10.1098/rsos.221426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/17/2023] [Indexed: 06/19/2023]
Abstract
Annually, an estimated seven million deaths are linked to exposure to airborne pollutants. Despite extensive epidemiological evidence supporting clear associations between poor air quality and a range of short- and long-term health effects, there are considerable gaps in our understanding of the specific mechanisms by which pollutant exposure induces adverse biological responses at the cellular and tissue levels. The development of more complex, predictive, in vitro respiratory models, including two- and three-dimensional cell cultures, spheroids, organoids and tissue cultures, along with more realistic aerosol exposure systems, offers new opportunities to investigate the cytotoxic effects of airborne particulates under controlled laboratory conditions. Parallel advances in high-resolution microscopy have resulted in a range of in vitro imaging tools capable of visualizing and analysing biological systems across unprecedented scales of length, time and complexity. This article considers state-of-the-art in vitro respiratory models and aerosol exposure systems and how they can be interrogated using high-resolution microscopy techniques to investigate cell-pollutant interactions, from the uptake and trafficking of particles to structural and functional modification of subcellular organelles and cells. These data can provide a mechanistic basis from which to advance our understanding of the health effects of airborne particulate pollution and develop improved mitigation measures.
Collapse
Affiliation(s)
- Zeinab Al-Rekabi
- Department of Chemical and Biological Sciences, National Physical Laboratory, Teddington, UK
| | - Camilla Dondi
- Department of Chemical and Biological Sciences, National Physical Laboratory, Teddington, UK
| | - Nilofar Faruqui
- Department of Chemical and Biological Sciences, National Physical Laboratory, Teddington, UK
| | - Nazia S. Siddiqui
- Faculty of Medical Sciences, University College London, London, UK
- Kingston Hospital NHS Foundation Trust, Kingston upon Thames, UK
| | - Linda Elowsson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jenny Rissler
- Bioeconomy and Health, RISE Research Institutes of Sweden, Lund, Sweden
- Ergonomics and Aerosol Technology, Lund University, Lund, Sweden
| | - Monica Kåredal
- Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Ian Mudway
- MRC Centre for Environment and Health, Imperial College London, London, UK
- National Institute of Health Protection Research Unit in Environmental Exposures and Health, London, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | | | - Michael Shaw
- Department of Chemical and Biological Sciences, National Physical Laboratory, Teddington, UK
- Department of Computer Science, University College London, London, UK
| |
Collapse
|
3
|
Herminghaus A, Kozlov AV, Szabó A, Hantos Z, Gylstorff S, Kuebart A, Aghapour M, Wissuwa B, Walles T, Walles H, Coldewey SM, Relja B. A Barrier to Defend - Models of Pulmonary Barrier to Study Acute Inflammatory Diseases. Front Immunol 2022; 13:895100. [PMID: 35874776 PMCID: PMC9300899 DOI: 10.3389/fimmu.2022.895100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022] Open
Abstract
Pulmonary diseases represent four out of ten most common causes for worldwide mortality. Thus, pulmonary infections with subsequent inflammatory responses represent a major public health concern. The pulmonary barrier is a vulnerable entry site for several stress factors, including pathogens such as viruses, and bacteria, but also environmental factors e.g. toxins, air pollutants, as well as allergens. These pathogens or pathogen-associated molecular pattern and inflammatory agents e.g. damage-associated molecular pattern cause significant disturbances in the pulmonary barrier. The physiological and biological functions, as well as the architecture and homeostatic maintenance of the pulmonary barrier are highly complex. The airway epithelium, denoting the first pulmonary barrier, encompasses cells releasing a plethora of chemokines and cytokines, and is further covered with a mucus layer containing antimicrobial peptides, which are responsible for the pathogen clearance. Submucosal antigen-presenting cells and neutrophilic granulocytes are also involved in the defense mechanisms and counterregulation of pulmonary infections, and thus may directly affect the pulmonary barrier function. The detailed understanding of the pulmonary barrier including its architecture and functions is crucial for the diagnosis, prognosis, and therapeutic treatment strategies of pulmonary diseases. Thus, considering multiple side effects and limited efficacy of current therapeutic treatment strategies in patients with inflammatory diseases make experimental in vitro and in vivo models necessary to improving clinical therapy options. This review describes existing models for studyying the pulmonary barrier function under acute inflammatory conditions, which are meant to improve the translational approaches for outcome predictions, patient monitoring, and treatment decision-making.
Collapse
Affiliation(s)
- Anna Herminghaus
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Andrey V. Kozlov
- L Boltzmann Institute for Traumatology in Cooperation with AUVA and Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Department of Human Pathology , IM Sechenov Moscow State Medical University, Moscow, Russia
| | - Andrea Szabó
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Zoltán Hantos
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Severin Gylstorff
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Kuebart
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Mahyar Aghapour
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Bianka Wissuwa
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Thorsten Walles
- Department of Thoracic Surgery, Magdeburg University Medicine, Magdeburg, Germany
| | - Heike Walles
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- Core Facility Tissue Engineering, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sina M. Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- *Correspondence: Borna Relja,
| |
Collapse
|
4
|
Albano GD, Montalbano AM, Gagliardo R, Anzalone G, Profita M. Impact of Air Pollution in Airway Diseases: Role of the Epithelial Cells (Cell Models and Biomarkers). Int J Mol Sci 2022; 23:2799. [PMID: 35269941 PMCID: PMC8911203 DOI: 10.3390/ijms23052799] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 02/05/2023] Open
Abstract
Biomedical research is multidisciplinary and often uses integrated approaches performing different experimental models with complementary functions. This approach is important to understand the pathogenetic mechanisms concerning the effects of environmental pollution on human health. The biological activity of the substances is investigated at least to three levels using molecular, cellular, and human tissue models. Each of these is able to give specific answers to experimental problems. A scientific approach, using biological methods (wet lab), cell cultures (cell lines or primary), isolated organs (three-dimensional cell cultures of primary epithelial cells), and animal organisms, including the human body, aimed to understand the effects of air pollution on the onset of diseases of the respiratory system. Biological methods are divided into three complementary models: in vitro, ex vivo, and in vivo. In vitro experiments do not require the use of whole organisms (in vivo study), while ex vivo experiments use isolated organs or parts of organs. The concept of complementarity and the informatic support are useful tools to organize, analyze, and interpret experimental data, with the aim of discussing scientific notions with objectivity and rationality in biology and medicine. In this scenario, the integrated and complementary use of different experimental models is important to obtain useful and global information that allows us to identify the effect of inhaled pollutants on the incidence of respiratory diseases in the exposed population. In this review, we focused our attention on the impact of air pollution in airway diseases with a rapid and descriptive analysis on the role of epithelium and on the experimental cell models useful to study the effect of toxicants on epithelial cells.
Collapse
Affiliation(s)
- Giusy Daniela Albano
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Angela Marina Montalbano
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Rosalia Gagliardo
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Giulia Anzalone
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Mirella Profita
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| |
Collapse
|
5
|
Viola H, Washington K, Selva C, Grunwell J, Tirouvanziam R, Takayama S. A High-Throughput Distal Lung Air-Blood Barrier Model Enabled By Density-Driven Underside Epithelium Seeding. Adv Healthc Mater 2021; 10:e2100879. [PMID: 34174173 DOI: 10.1002/adhm.202100879] [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: 05/04/2021] [Indexed: 12/18/2022]
Abstract
High-throughput tissue barrier models can yield critical insights on how barrier function responds to therapeutics, pathogens, and toxins. However, such models often emphasize multiplexing capability at the expense of physiologic relevance. Particularly, the distal lung's air-blood barrier is typically modeled with epithelial cell monoculture, neglecting the substantial contribution of endothelial cell feedback in the coordination of barrier function. An obstacle to establishing high-throughput coculture models relevant to the epithelium/endothelium interface is the requirement for underside cell seeding, which is difficult to miniaturize and automate. Therefore, this paper describes a scalable, low-cost seeding method that eliminates inversion by optimizing medium density to float cells so they attach under the membrane. This method generates a 96-well model of the distal lung epithelium-endothelium barrier with serum-free, glucocorticoid-free air-liquid differentiation. The polarized epithelial-endothelial coculture exhibits mature barrier function, appropriate intercellular junction staining, and epithelial-to-endothelial transmission of inflammatory stimuli such as polyinosine:polycytidylic acid (poly(I:C)). Further, exposure to influenza A virus PR8 and human beta-coronavirus OC43 initiates a dose-dependent inflammatory response that propagates from the epithelium to endothelium. While this model focuses on the air-blood barrier, the underside seeding method is generalizable to various coculture tissue models for scalable, physiologic screening.
Collapse
Affiliation(s)
- Hannah Viola
- School of Chemical and Biomolecular Engineering Georgia Institute of Technology 311 Ferst Dr. NW Atlanta GA 30308 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30332 USA
| | - Kendra Washington
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30308 USA
| | - Cauviya Selva
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30308 USA
| | - Jocelyn Grunwell
- Division of Critical Care Medicine Children's Healthcare of Atlanta at Egleston 1405 Clifton Road NE Atlanta GA 30322 USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics Emory University School of Medicine and Center for CF & Airways Disease Research 2015 Uppergate Dr NE, Rm 344 Atlanta GA 30322 USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30308 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30332 USA
| |
Collapse
|
6
|
Editor's Highlight: Pulmonary Vascular Thrombosis in Rats Exposed to Inhaled Sulfur Mustard. Toxicol Sci 2018; 159:461-469. [PMID: 28962529 DOI: 10.1093/toxsci/kfx151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Sulfur mustard (SM) is a chemical warfare agent. When inhaled, SM causes significant injury to the respiratory tract. Although the mechanism involved in acute airway injury after SM inhalation has been well described previously, the mechanism of SM's contribution to distal lung vascular injury is not well understood. We hypothesized that acute inhalation of vaporized SM causes activated systemic coagulation with subsequent pulmonary vascular thrombi formation after SM inhalation exposure. Sprague Dawley rats inhaled SM ethanolic vapor (3.8 mg/kg). Barium/gelatin CT pulmonary angiograms were performed to assess for pulmonary vascular thrombi burden. Lung immunohistochemistry was performed for common procoagulant markers including fibrin(ogen), von Willebrand factor, and CD42d in control and SM-exposed lungs. Additionally, systemic levels of d-dimer and platelet aggregometry after adenosine diphosphate- and thrombin-stimulation were measured in plasma after SM exposure. In SM-exposed lungs, chest CT angiography demonstrated a significant decrease in the distal pulmonary vessel density assessed at 6 h postexposure. Immunohistochemistry also demonstrated increased intravascular fibrin(ogen), vascular von Willebrand factor, and platelet CD42d in the distal pulmonary vessels (<200 µm diameter). Circulating d-dimer levels were significantly increased (p < .001) at 6, 9, and 12 h after SM inhalation versus controls. Platelet aggregation was also increased in both adenosine diphosphate - (p < .01) and thrombin- (p < .001) stimulated platelet-rich plasma after SM inhalation. Significant pulmonary vascular thrombi formation was evident in distal pulmonary arterioles following SM inhalation in rats assessed by CT angiography and immunohistochemistry. Enhanced systemic platelet aggregation and activated systemic coagulation with subsequent thrombi formation likely contributed to pulmonary vessel occlusion.
Collapse
|
7
|
Faber SC, McCullough SD. Through the Looking Glass: In Vitro Models for Inhalation Toxicology and Interindividual Variability in the Airway. ACTA ACUST UNITED AC 2018; 4:115-128. [PMID: 31380467 DOI: 10.1089/aivt.2018.0002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
With 7 million deaths reported annually from air pollution alone, it is evident that adverse effects of inhaled toxicant exposures remain a major public health concern in the 21st century. Assessment and characterization of the impacts of air pollutants on human health stems from epidemiological and clinical studies, which have linked both outdoor and indoor air contaminant exposure to adverse pulmonary and cardiovascular health outcomes. Studies in animal models support epidemiological findings and have been critical in identifying systemic effects of environmental chemicals on cognitive abilities, liver disease, and metabolic dysfunction following inhalation exposure. Likewise, traditional monoculture systems have aided in identifying biomarkers of susceptibility to inhaled toxicants and served as a screening platform for safety assessment of pulmonary toxicants. Despite their contributions, in vivo and classic in vitro models have not been able to accurately represent the heterogeneity of the human population and account for interindividual variability in response to inhaled toxicants and susceptibility to the adverse health effects. Development of new technologies that can investigate genetic predisposition, are cost and time efficient, and are ethically sound, will enhance elucidation of mechanisms of inhalation toxicity, and aid in the development of novel pharmaceuticals and/or safety evaluation. This review will describe the classic and novel cell-based inhalation toxicity models and how these emerging technologies can be incorporated into regulatory or nonregulatory testing to address interindividual variability and improve overall human health.
Collapse
Affiliation(s)
- Samantha C Faber
- Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Shaun D McCullough
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, North Carolina
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Protective effects of the thiol compounds GSH and NAC against sulfur mustard toxicity in a human keratinocyte cell line. Toxicol Lett 2016; 244:35-43. [DOI: 10.1016/j.toxlet.2015.09.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/02/2015] [Accepted: 09/03/2015] [Indexed: 12/29/2022]
|
11
|
Steinritz D, Möhle N, Pohl C, Papritz M, Stenger B, Schmidt A, Kirkpatrick CJ, Thiermann H, Vogel R, Hoffmann S, Aufderheide M. Use of the Cultex® Radial Flow System as an in vitro exposure method to assess acute pulmonary toxicity of fine dusts and nanoparticles with special focus on the intra- and inter-laboratory reproducibility. Chem Biol Interact 2013; 206:479-90. [PMID: 23669118 DOI: 10.1016/j.cbi.2013.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 04/29/2013] [Accepted: 05/02/2013] [Indexed: 01/04/2023]
Abstract
Exposure of the respiratory tract to airborne particles (including metal-dusts and nano-particles) is considered as a serious health hazard. For a wide range of substances basic knowledge about the toxic properties and the underlying pathomechanisms is lacking or even completely missing. Legislation demands the toxicological characterization of all chemicals placed on the market until 2018 (REACH). As toxicological in vivo data are rare with regard to acute lung toxicity or exhibit distinct limitations (e.g. inter-species differences) and legislation claims the reduction of animal experiments in general ("3R" principle), profound in vitro models have to be established and characterized to meet these requirements. In this paper we characterize a recently introduced advanced in vitro exposure system (Cultex® RFS) showing a great similarity to the physiological in vivo exposure situation for the assessment of acute pulmonary toxicity of airborne materials. Using the Cultex® RFS, human lung epithelial cells (A549 cells) were exposed to different concentrations of airborne metal dusts (nano- and microscale particles) at the air-liquid-interface (ALI). Cell viability (WST-1 assay) as a parameter of toxicity was assessed 24h after exposure with special focus on the intra- and inter-laboratory (three independent laboratories) reproducibility. Our results show the general applicability of the Cultex® RFS with regard to the requirements of the ECVAM (European Centre for the Validation of Alternative Methods) principles on test validity underlining its robustness and stability. Intra- and inter-laboratory reproducibility can be considered as sufficient if predefined quality criteria are respected. Special attention must be paid to the pure air controls that turned out to be a critical parameter for a rational interpretation of the results. Our results are encouraging and future work is planned to improve the inter-laboratory reproducibility, to consolidate the results so far and to develop a valid prediction model.
Collapse
Affiliation(s)
- Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 Munich, Germany; Walther Straub Institute of Pharmacology and Toxicology, University of Munich, Goethestraße 33, 80336 Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Ghasemi H, Owlia P, Jalali-Nadoushan MR, Pourfarzam S, Azimi G, Yarmohammadi ME, Shams J, Fallahi F, Moaiedmohseni S, Moin A, Yaraee R, Vaez-Mahdavi MR, Faghihzadeh S, Mohammad Hassan Z, Soroush MR, Naghizadeh MM, Ardestani SK, Ghazanfari T. A clinicopathological approach to sulfur mustard-induced organ complications: a major review. Cutan Ocul Toxicol 2013; 32:304-24. [PMID: 23590683 DOI: 10.3109/15569527.2013.781615] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Sulfur mustard (SM), with an old manufacturing history still remains as potential threat due to easy production and extensive effects. OBJECTIVES Increasing studies on SM indicates the interest of researchers to this subject. Almost all human body organs are at risk for complications of SM. This study offers organ-by-organ information on the effects of SM in animals and humans. METHODS The data sources were literature reviews since 1919 as well as our studies during the Iraq-Iran war. The search items were SM and its all other nomenclatures in relation to, in vivo, in vitro, humans, animals, eye, ocular, ophthalmic, lungs, pulmonary, skin, cutaneous, organs and systemic. Amongst more than 1890 SM-related articles, 257 more relevant clinicopathologic papers were selected for this review. RESULTS SM induces a vast range of damages in nearly all organs. Acute SM intoxication warrants immediate approach. Among chronic lesions, delayed keratitis and blindness, bronchiolitis obliterans and respiratory distress, skin pruritus, dryness and cancers are the most commonly observed clinical sequelae. CONCLUSION Ocular involvements in a number of patients progress toward a severe, rapid onset form of keratitis. Progressive deterioration of respiratory tract leads to "mustard lung". Skin problems continue as chronic frustrating pruritus on old scars with susceptibility to skin cancers. Due to the multiple acute and chronic morbidities created by SM exposure, uses of multiple drugs by several routes of administrations are warranted.
Collapse
Affiliation(s)
- Hassan Ghasemi
- Immunoregulation Research Center, Shahed University, Tehran, Iran.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Keyser BM, Andres DK, Nealley E, Holmes WW, Benton B, Paradiso D, Appell A, Carpin C, Anderson DR, Smith WJ, Ray R. Postexposure application of Fas receptor small-interfering RNA to suppress sulfur mustard-induced apoptosis in human airway epithelial cells: implication for a therapeutic approach. J Pharmacol Exp Ther 2012; 344:308-16. [PMID: 23129783 DOI: 10.1124/jpet.112.199935] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sulfur mustard (SM) is a vesicant chemical warfare and terrorism agent. Besides skin and eye injury, respiratory damage has been mainly responsible for morbidity and mortality after SM exposure. Previously, it was shown that suppressing the death receptor (DR) response by the dominant-negative Fas-associated death domain protein prior to SM exposure blocked apoptosis and microvesication in skin. Here, we studied whether antagonizing the Fas receptor (FasR) pathway by small-interfering RNA (siRNA) applied after SM exposure would prevent apoptosis and, thus, airway injury. Normal human bronchial/tracheal epithelial (NHBE) cells were used as an in vitro model with FasR siRNA, FasR agonistic antibody CH11, and FasR antagonistic antibody ZB4 as investigative tools. In NHBE cells, both SM (300 µM) and CH11 (100 ng/ml) induced caspase-3 activation, which was inhibited by FasR siRNA and ZB4, indicating that SM-induced apoptosis was via the Fas response. FasR siRNA inhibited SM-induced caspase-3 activation when added to NHBE cultures up to 8 hours after SM. Results using annexin V/propidium iodide-stained cells showed that both apoptosis and necrosis were involved in cell death due to SM; FasR siRNA decreased both apoptotic and necrotic cell populations. Bronchoalveolar lavage fluid (BALF) of rats exposed to SM (1 mg/kg, 50 minutes) revealed a significant (P < 0.05) increase in soluble Fas ligand and active caspase-3 in BALF cells. These findings suggest an intervention of Fas-mediated apoptosis as a postexposure therapeutic strategy with a therapeutic window for SM inhalation injury and possibly other respiratory diseases involving the Fas response.
Collapse
Affiliation(s)
- Brian M Keyser
- Cellular and Molecular Biology Branch, U.S. Army Medical Research Institute of Chemical Defense, 3100 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010-5400, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Mishra NC, Rir-sima-ah J, Grotendorst GR, Langley RJ, Singh SP, Gundavarapu S, Weber WM, Pena-Philippides JC, Duncan MR, Sopori ML. Inhalation of sulfur mustard causes long-term T cell-dependent inflammation: possible role of Th17 cells in chronic lung pathology. Int Immunopharmacol 2012; 13:101-8. [PMID: 22465472 DOI: 10.1016/j.intimp.2012.03.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 03/12/2012] [Accepted: 03/15/2012] [Indexed: 10/28/2022]
Abstract
Sulfur mustard (SM) is a highly toxic chemical warfare agent that remains a threat to human health. The immediate symptoms of pulmonary distress may develop into chronic lung injury characterized by progressive lung fibrosis, the major cause of morbidity among the surviving SM victims. Although SM has been intensely investigated, little is known about the mechanism(s) by which SM induces chronic lung pathology. Increasing evidence suggests that IL-17(+) cells are critical in fibrosis, including lung fibrotic diseases. In this study we exposed F344 rats and cynomolgus monkeys to SM via inhalation and determined the molecular and cellular milieu in their lungs at various times after SM exposure. In rats, SM induced a burst of pro-inflammatory cytokines/chemokines within 72 h, including IL-1β, TNF-α, IL-2, IL-6, CCL2, CCL3, CCL11, and CXCL1 that was associated with neutrophilic infiltration into the lung. At 2 wks and beyond (chronic phase), lymphocytic infiltration and continued elevated expression of cytokines/chemokines were sustained. TGF-β, which was undetectable in the acute phase, was strongly upregulated in the chronic phase; these conditions persisted until the animals were sacrificed. The chronic phase was also associated with myofibroblast proliferation, collagen deposition, and presence of IL-17(+) cells. At ≥30 days, SM inhalation promoted the accumulation of IL-17(+) cells in the inflamed areas of monkey lungs. Thus, SM inhalation causes acute and chronic inflammatory responses; the latter is characterized by the presence of TGF-β, fibrosis, and IL-17(+) cells in the lung. IL-17(+) cells likely play an important role in the pathogenesis of SM-induced lung injury.
Collapse
Affiliation(s)
- Neerad C Mishra
- Lovelace CounterACT Research Center of Excellence, Lovelace Respiratory Research Institute, 2425 Ridgecrest Dr. SE, Albuquerque, NM 87108, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Ghasemi H, Ghazanfari T, Yaraee R, Rafii AB, Pourfarzam S, Soroush MR, Babaei M, Faghihzadeh S, Naghizadeh MM, Hassan ZM. Long-term ocular consequences of sulfur mustard in lung-injured war veterans. Cutan Ocul Toxicol 2011; 31:33-7. [DOI: 10.3109/15569527.2011.595751] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
16
|
Seagrave J, Weber WM, Grotendorst GR. Sulfur mustard vapor effects on differentiated human lung cells. Inhal Toxicol 2011; 22:896-902. [PMID: 20569120 DOI: 10.3109/08958378.2010.493901] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
CONTEXT Sulfur mustard (SM) causes skin blistering and long-term pulmonary dysfunction. Its adverse effects have been studied in battlefield-exposed humans, but lack of knowledge regarding confounding factors makes interpretation challenging. Animal studies are critical to understanding mechanisms, but differences between animals and humans must be addressed. Studies of cultured human cells can bridge animal studies and humans. OBJECTIVE Evaluate effects of SM vapor on airway cells. MATERIALS AND METHODS We examined responses of differentiated human tracheal/bronchial epithelial cells, cultured at an air-liquid interface, to SM vapors. SM effects on metabolic activity (Water Soluble Tetrazolium (WST) assay), cytokine and metalloproteinase secretion, and cellular heme oxygenase 1 (HO-1), an oxidative stress indicator, were measured after 24 h. RESULTS At noncytotoxic levels of exposure, interleukin 8 and matrix metalloproteinase-13 were significantly increased in these cultures, but HO-1 was not significantly affected. DISCUSSION AND CONCLUSION Exposure of differentiated airway epithelial cells to sub-cytotoxic levels of SM vapor induced inflammatory and degradative responses that could contribute to the adverse health effects of inhaled SM.
Collapse
|
17
|
Ghabili K, Agutter PS, Ghanei M, Ansarin K, Panahi Y, Shoja MM. Sulfur mustard toxicity: history, chemistry, pharmacokinetics, and pharmacodynamics. Crit Rev Toxicol 2011; 41:384-403. [PMID: 21329486 DOI: 10.3109/10408444.2010.541224] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sulfur mustard (SM) and similar bifunctional agents have been used as chemical weapons for almost 100 years. Victims of high-dose exposure, both combatants and civilians, may die within hours or weeks, but low-dose exposure causes both acute injury to the eyes, skin, respiratory tract and other parts of the body, and chronic sequelae in these organs are often debilitating and have a serious impact on quality of life. Ever since they were first used in warfare in 1917, SM and other mustard agents have been the subjects of intensive research, and their chemistry, pharmacokinetics and mechanisms of toxic action are now fairly well understood. In the present article we review this knowledge and relate the molecular-biological basis of SM toxicity, as far as it has been elucidated, to the pathological effects on exposure victims.
Collapse
Affiliation(s)
- Kamyar Ghabili
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | | | |
Collapse
|
18
|
Pohl C, Hofmann H, Moisch M, Papritz M, Iris Hermanns M, Dei-Anang J, Mayer E, Kehe K, Kirkpatrick CJ. Acute cytotoxicity and apoptotic effects after l-Pam exposure in different cocultures of the proximal and distal respiratory system. J Biotechnol 2010; 148:31-7. [DOI: 10.1016/j.jbiotec.2009.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 11/20/2009] [Accepted: 12/01/2009] [Indexed: 11/17/2022]
|
19
|
Chowdhury F, Howat WJ, Phillips GJ, Lackie PM. Interactions between endothelial cells and epithelial cells in a combined cell model of airway mucosa: effects on tight junction permeability. Exp Lung Res 2010; 36:1-11. [DOI: 10.3109/01902140903026582] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
20
|
Pohl C, Papritz M, Moisch M, Wübbeke C, Hermanns MI, Uboldi C, Dei-Anang J, Mayer E, Kirkpatrick CJ, Kehe K. Acute Morphological and Toxicological Effects in a Human Bronchial Coculture Model after Sulfur Mustard Exposure. Toxicol Sci 2009; 112:482-9. [DOI: 10.1093/toxsci/kfp211] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
21
|
Kovacic P, Somanathan R. Pulmonary toxicity and environmental contamination: radicals, electron transfer, and protection by antioxidants. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2009; 201:41-69. [PMID: 19484588 DOI: 10.1007/978-1-4419-0032-6_2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The atmosphere is replete with a mixture of toxic substances, both natural and man-made. Inhalation of toxic substances produces a variety of insults to the pulmonary system. Lung poisons include industrial materials, particulates from mining and combustion, agricultural chemicals, cigarette smoke, ozone, and nitrogen oxides, among a large number of other chemicals and environmental contaminants. Many proposals have been advanced to explain the mode of action of pulmonary toxicants. In this review we focus on mechanisms of pulmonary toxicity that involve ET, ROS, and OS. The vast majority of toxicants or their metabolites possess chemical ET functionalities that can undergo redox cycling. Such recycling may generate ROS that can injure various cellular constituents in the lung and in other tissues. ET agents include quinones, metal complexes, aromatic nitro compounds, and conjugated iminium ions. Often, these agents are formed metabolically from parent toxicants. Such metabolic reactions are often catalytic and require only small amounts of the offending material. Oxidative attack is commonly associated with lipid peroxidation and oxidation of DNA, and it may result in strand cleavage and 8-OH-DG production. Toxicity is often accompanied by depletion of natural AOs, which further exacerbates the toxic effect. It is not surprising that the use of AOs, both natural in fruits and vegetables, as well as synthetic, may provide protection from the adverse effects of toxicant exposure. The mechanistic framework described earlier is also applicable to some of the more prominent pulmonary illnesses, such as asthma, COPD, and cancer.
Collapse
Affiliation(s)
- Peter Kovacic
- Department of Chemistry, San Diego State University, San Diego, CA 92182-1030, USA.
| | | |
Collapse
|
22
|
Karacsonyi C, Shanmugam N, Kagan E. A clinically relevant in vitro model for evaluating the effects of aerosolized vesicants. Toxicol Lett 2008; 185:38-44. [PMID: 19110046 DOI: 10.1016/j.toxlet.2008.11.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Revised: 11/20/2008] [Accepted: 11/21/2008] [Indexed: 01/18/2023]
Abstract
The chemical warfare vesicant sulfur mustard (HD) is a known toxic agent to the human respiratory tract and the major airways are considered to be a primary target of HD-induced injury. However, there is no consensus regarding which model systems are most appropriate for studying the effects of aerosolized vesicants on human airway epithelium. In this study, we evaluated the consequences of exposure of differentiated human respiratory epithelial cells in air-liquid interface to mechlorethamine (HN2), an HD functional analog. HN2 challenge was administered via the apical (air) interface over a wide dose range (20-400 microM) to differentiated HBE1 cells. Cultures were observed over 1-48 h for evidence of HN2-induced morphologic abnormalities as well as for possible cellular cytotoxicity, apoptotic changes, and induction of cytokine secretion. HN2 at concentrations of > or =200 microM caused disruption and denudation of the airway epithelial architecture within 24h of exposure. Moreover, HN2-induced cytotoxic and apoptotic changes in HBE1 cells in a dose- and time-dependent fashion. HN2 challenge also induced secretion of chemokines and proinflammatory cytokines including TNF-alpha, IL-1 alpha, IL-1 beta, IL-6, IL-8, RANTES, MCP-1, IP-10, G-CSF, GM-CSF and IL-15. These observations parallel those described in the lungs of HD-exposed victims and underscore the utility and potential applicability of this model to future mechanistic studies of vesicant-induced pulmonary injury.
Collapse
Affiliation(s)
- Claudia Karacsonyi
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4799, United States
| | | | | |
Collapse
|