1
|
Yost EE, Galizia A, Kapraun DF, Persad AS, Vulimiri SV, Angrish M, Lee JS, Druwe IL. Health Effects of Naphthalene Exposure: A Systematic Evidence Map and Analysis of Potential Considerations for Dose-Response Evaluation. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:76002. [PMID: 34251878 PMCID: PMC8274693 DOI: 10.1289/ehp7381] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 05/19/2023]
Abstract
BACKGROUND Naphthalene is a polycyclic aromatic hydrocarbon that has been associated with health effects, including cancer. As the state of the science on naphthalene toxicity continues to evolve, updated toxicity reference value(s) may be required to support human health risk assessment. OBJECTIVES We present a systematic evidence map of studies that could be used to derive toxicity reference value(s) for naphthalene. METHODS Human and animal health effect studies and physiologically based pharmacokinetic (PBPK) models were identified from a literature search based on populations, exposures, comparators, and outcomes (PECO) criteria. Human and animal studies meeting PECO criteria were refined to a smaller subset considered most informative for deriving chronic reference value(s), which are preferred for assessing risk to the general public. This subset was evaluated for risk of bias and sensitivity, and the suitability of each study for dose-response analysis was qualitatively assessed. Lowest observed adverse effect levels (LOAELs) were extracted and summarized. Other potentially relevant studies (e.g., mechanistic and toxicokinetic studies) were tracked as supplemental information but not evaluated further. Existing reference values for naphthalene are also summarized. RESULTS We identified 26 epidemiology studies and 16 animal studies that were considered most informative for further analysis. Eleven PBPK models were identified. The available epidemiology studies generally had significant risk of bias and/or sensitivity concerns and were mostly found to have low suitability for dose-response analysis due to the nature of the exposure measurements. The animal studies had fewer risk of bias and sensitivity concerns and were mostly found to be suitable for dose-response analysis. CONCLUSION Although both epidemiological and animal studies of naphthalene provide weight of evidence for hazard identification, the available animal studies appear more suitable for reference value derivation. PBPK models and mechanistic and toxicokinetic data can be applied to extrapolate these animal data to humans, considering mode of action and interspecies metabolic differences. https://doi.org/10.1289/EHP7381.
Collapse
Affiliation(s)
- Erin E. Yost
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Audrey Galizia
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Dustin F. Kapraun
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Amanda S. Persad
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Suryanarayana V. Vulimiri
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Michelle Angrish
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Janice S. Lee
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| | - Ingrid L. Druwe
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Washington, District of Columbia, USA
| |
Collapse
|
2
|
Malfatti MA, Buchholz BA, Enright HA, Stewart BJ, Ognibene TJ, McCartt AD, Loots GG, Zimmermann M, Scharadin TM, Cimino GD, Jonas BA, Pan CX, Bench G, Henderson PT, Turteltaub KW. Radiocarbon Tracers in Toxicology and Medicine: Recent Advances in Technology and Science. TOXICS 2019; 7:E27. [PMID: 31075884 PMCID: PMC6631948 DOI: 10.3390/toxics7020027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 01/09/2023]
Abstract
This review summarizes recent developments in radiocarbon tracer technology and applications. Technologies covered include accelerator mass spectrometry (AMS), including conversion of samples to graphite, and rapid combustion to carbon dioxide to enable direct liquid sample analysis, coupling to HPLC for real-time AMS analysis, and combined molecular mass spectrometry and AMS for analyte identification and quantitation. Laser-based alternatives, such as cavity ring down spectrometry, are emerging to enable lower cost, higher throughput measurements of biological samples. Applications covered include radiocarbon dating, use of environmental atomic bomb pulse radiocarbon content for cell and protein age determination and turnover studies, and carbon source identification. Low dose toxicology applications reviewed include studies of naphthalene-DNA adduct formation, benzo[a]pyrene pharmacokinetics in humans, and triclocarban exposure and risk assessment. Cancer-related studies covered include the use of radiocarbon-labeled cells for better defining mechanisms of metastasis and the use of drug-DNA adducts as predictive biomarkers of response to chemotherapy.
Collapse
Affiliation(s)
- Michael A Malfatti
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.
| | - Bruce A Buchholz
- Center for Accelerator Mass Spectrometry, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.
| | - Heather A Enright
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.
| | - Benjamin J Stewart
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.
| | - Ted J Ognibene
- Center for Accelerator Mass Spectrometry, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.
| | - A Daniel McCartt
- Center for Accelerator Mass Spectrometry, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.
| | - Gabriela G Loots
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.
| | - Maike Zimmermann
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis Medical School, Sacramento, CA 95817, USA.
- Accelerated Medical Diagnostics Incorporated, Berkeley, CA 94708, USA.
| | - Tiffany M Scharadin
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis Medical School, Sacramento, CA 95817, USA.
- Accelerated Medical Diagnostics Incorporated, Berkeley, CA 94708, USA.
| | - George D Cimino
- Accelerated Medical Diagnostics Incorporated, Berkeley, CA 94708, USA.
| | - Brian A Jonas
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis Medical School, Sacramento, CA 95817, USA.
| | - Chong-Xian Pan
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis Medical School, Sacramento, CA 95817, USA.
| | - Graham Bench
- Center for Accelerator Mass Spectrometry, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.
| | - Paul T Henderson
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis Medical School, Sacramento, CA 95817, USA.
- Accelerated Medical Diagnostics Incorporated, Berkeley, CA 94708, USA.
| | - Kenneth W Turteltaub
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.
| |
Collapse
|
3
|
Robert-Hazotte A, Schoumacker R, Semon E, Briand L, Guichard E, Le Quéré JL, Faure P, Heydel JM. Ex vivo real-time monitoring of volatile metabolites resulting from nasal odorant metabolism. Sci Rep 2019; 9:2492. [PMID: 30792537 PMCID: PMC6385289 DOI: 10.1038/s41598-019-39404-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 01/22/2019] [Indexed: 12/02/2022] Open
Abstract
Odorant-metabolizing enzymes are critically involved in the clearance of odorant molecules from the environment of the nasal neuro-olfactory tissue to maintain the sensitivity of olfactory detection. Odorant metabolism may also generate metabolites in situ, the characterization and function of which in olfaction remain largely unknown. Here, we engineered and validated an ex vivo method to measure odorant metabolism in real-time. Glassware containing an explant of rat olfactory mucosa was continuously flushed with an odorant flow and was coupled to a proton transfer reaction-mass spectrometer for volatile compound analysis. Focusing on carboxylic esters and diketone odorants, we recorded the metabolic uptake of odorants by the mucosa, concomitantly with the release of volatile odorant metabolites in the headspace. These results significantly change the picture of real-time in situ odorant metabolism and represent a new step forward in the investigation of the function of odorant metabolites in the peripheral olfactory process. Our method allows the systematic identification of odorant metabolites using a validated animal model and permits the screening of olfactory endogenously produced chemosensory molecules.
Collapse
Affiliation(s)
- Aline Robert-Hazotte
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS/1324 INRA/Université de Bourgogne Franche-Comté, 9 boulevard Jeanne d'Arc, F-21000, Dijon, France
| | - Rachel Schoumacker
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS/1324 INRA/Université de Bourgogne Franche-Comté, 9 boulevard Jeanne d'Arc, F-21000, Dijon, France
| | - Etienne Semon
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS/1324 INRA/Université de Bourgogne Franche-Comté, 9 boulevard Jeanne d'Arc, F-21000, Dijon, France
| | - Loïc Briand
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS/1324 INRA/Université de Bourgogne Franche-Comté, 9 boulevard Jeanne d'Arc, F-21000, Dijon, France
| | - Elisabeth Guichard
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS/1324 INRA/Université de Bourgogne Franche-Comté, 9 boulevard Jeanne d'Arc, F-21000, Dijon, France
| | - Jean-Luc Le Quéré
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS/1324 INRA/Université de Bourgogne Franche-Comté, 9 boulevard Jeanne d'Arc, F-21000, Dijon, France
| | - Philippe Faure
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS/1324 INRA/Université de Bourgogne Franche-Comté, 9 boulevard Jeanne d'Arc, F-21000, Dijon, France
| | - Jean-Marie Heydel
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS/1324 INRA/Université de Bourgogne Franche-Comté, 9 boulevard Jeanne d'Arc, F-21000, Dijon, France.
| |
Collapse
|
4
|
Carratt SA, Hartog M, Buchholz BA, Kuhn EA, Collette NM, Ding X, Van Winkle LS. Naphthalene genotoxicity: DNA adducts in primate and mouse airway explants. Toxicol Lett 2019; 305:103-109. [PMID: 30684585 DOI: 10.1016/j.toxlet.2019.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 01/05/2019] [Accepted: 01/20/2019] [Indexed: 01/14/2023]
Abstract
Naphthalene (NA) is a ubiquitous environmental pollutant and possible human carcinogen that forms tumors in rodents with tissue/regional and species selectivity. This study seeks to determine whether NA is able to directly adduct DNA in an ex vivo culture system. Metabolically active lung tissue was isolated and incubated in explant culture with carbon-14 labeled NA (0, 25, 250 μM) or 1,2-naphthoquinone (NQ), followed by AMS analyses of metabolite binding to DNA. Despite relatively low metabolic bioactivation in the primate airway, dose-dependent NA-DNA adduct formation was detected. More airway adducts were detected in female mice (4.7-fold) and primates (2.1-fold) than in males of the same species. Few adducts were detected in rat airway or nasal epithelium. NQ, which is a metabolic product of NA, proved to be even more potent, with levels of adduct formation 70-80-fold higher than seen when tissues were incubated with the parent compound NA. This is the first study to demonstrate NA-DNA adduct formation at a site of carcinogenesis, the mouse lung. Adducts were also detected in non-human primate lung and with a NQ metabolite of NA. Taken together, this suggests that NA may contribute to in vivo carcinogenesis through a genotoxic mechanism.
Collapse
Affiliation(s)
- Sarah A Carratt
- Center for Health and the Environment, University of California Davis, Davis, CA 95616, USA
| | - Matthew Hartog
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA
| | - Bruce A Buchholz
- Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Edward A Kuhn
- Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | | | - Xinxin Ding
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA.
| | - Laura S Van Winkle
- Center for Health and the Environment, University of California Davis, Davis, CA 95616, USA; Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
5
|
Buchholz BA, Carratt SA, Kuhn EA, Collette NM, Ding X, Van Winkle LS. Naphthalene DNA Adduct Formation and Tolerance in the Lung. NUCLEAR INSTRUMENTS & METHODS IN PHYSICS RESEARCH. SECTION B, BEAM INTERACTIONS WITH MATERIALS AND ATOMS 2019; 438:119-123. [PMID: 30631217 PMCID: PMC6322674 DOI: 10.1016/j.nimb.2018.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Naphthalene (NA) is a respiratory toxicant and possible human carcinogen. NA is a ubiquitous combustion product and significant component of jet fuel. The National Toxicology Program found that NA forms tumors in two species, in rats (nose) and mice (lung). However, it has been argued that NA does not pose a cancer risk to humans because NA is bioactivated by cytochrome P450 monooxygenase enzymes that have very high efficiency in the lung tissue of rodents but low efficiency in the lung tissue of humans. It is thought that NA carcinogenesis in rodents is related to repeated cycles of lung epithelial injury and repair, an indirect mechanism. Repeated in vivo exposure to NA leads to development of tolerance, with the emergence of cells more resistant to NA insult. We tested the hypothesis that tolerance involves reduced susceptibility to the formation of NA-DNA adducts. NA-DNA adduct formation in tolerant mice was examined in individual, metabolically-active mouse airways exposed ex vivo to 250 μΜ 14C-NA. Ex vivo dosing was used since it had been done previously and the act of creating a radioactive aerosol of a potential carcinogen posed too many safety and regulatory obstacles. Following extensive rinsing to remove unbound 14C-NA, DNA was extracted and 14C-NA-DNA adducts were quantified by AMS. The tolerant mice appeared to have slightly lower NA-DNA adduct levels than non-tolerant controls, but intra-group variations were large and the difference was statistically insignificant. It appears the tolerance may be more related to other mechanisms, such as NA-protein interactions in the airway, than DNA-adduct formation.
Collapse
Affiliation(s)
- Bruce A Buchholz
- Center for Accelerator Mass Spectrometry, Lawrence Livermore National Laboratory, Livermore, CA USA
| | - Sarah A Carratt
- Center for Health and the Environment, University of California, Davis, CA USA
| | - Edward A Kuhn
- Bioscience and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA USA
| | - Nicole M Collette
- Bioscience and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA USA
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ USA
| | - Laura S Van Winkle
- Center for Health and the Environment, University of California, Davis, CA USA
| |
Collapse
|
6
|
Smith GJ, Thrall RS, Cloutier MM, Manautou JE, Morris JB. Acetaminophen Attenuates House Dust Mite-Induced Allergic Airway Disease in Mice. J Pharmacol Exp Ther 2016; 358:569-79. [PMID: 27402277 DOI: 10.1124/jpet.116.233684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/05/2016] [Indexed: 11/22/2022] Open
Abstract
Epidemiologic evidence suggests that N-acetyl-para-aminophenol (APAP) may play a role in the pathogenesis of asthma, likely through pro-oxidant mechanisms. However, no studies have investigated the direct effects of APAP on the development of allergic inflammation. To determine the likelihood of a causal relationship between APAP and asthma pathogenesis, we explored the effects of APAP on inflammatory responses in a murine house dust mite (HDM) model of allergic airway disease. We hypothesized that APAP would enhance the development of HDM-induced allergic inflammation. The HDM model consisted of once daily intranasal instillations for up to 2 weeks with APAP or vehicle administration 1 hour prior to HDM during either week 1 or 2. Primary assessment of inflammation included bronchoalveolar lavage (BAL), cytokine expression in lung tissue, and histopathology. Contrary to our hypothesis, the effects of HDM treatment were substantially diminished in APAP-treated groups compared with controls. APAP-treated groups had markedly reduced airway inflammation: including decreased inflammatory cells in the BAL fluid, lower cytokine expression in lung tissue, and less perivascular and peribronchiolar immune cell infiltration. The anti-inflammatory effect of APAP was not abrogated by an inhibitor of cytochrome P450 (P450) metabolism, suggesting that the effect was due to the parent compound or a non-P450 generated metabolite. Taken together, our studies do not support the biologic plausibility of the APAP hypothesis that APAP use may contribute to the causation of asthma. Importantly, we suggest the mechanism by which APAP modulates airway inflammation may provide novel therapeutic targets for asthma.
Collapse
Affiliation(s)
- Gregory J Smith
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (G.J.S., J.E.M., J.B.M); Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut (R.S.T.); Connecticut Children's Medical Center, Hartford, Connecticut (M.M.C.)
| | - Roger S Thrall
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (G.J.S., J.E.M., J.B.M); Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut (R.S.T.); Connecticut Children's Medical Center, Hartford, Connecticut (M.M.C.)
| | - Michelle M Cloutier
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (G.J.S., J.E.M., J.B.M); Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut (R.S.T.); Connecticut Children's Medical Center, Hartford, Connecticut (M.M.C.)
| | - Jose E Manautou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (G.J.S., J.E.M., J.B.M); Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut (R.S.T.); Connecticut Children's Medical Center, Hartford, Connecticut (M.M.C.)
| | - John B Morris
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (G.J.S., J.E.M., J.B.M); Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut (R.S.T.); Connecticut Children's Medical Center, Hartford, Connecticut (M.M.C.)
| |
Collapse
|
7
|
Smith GJ, Cichocki JA, Doughty BJ, Manautou JE, Jordt SE, Morris JB. Effects of Acetaminophen on Oxidant and Irritant Respiratory Tract Responses to Environmental Tobacco Smoke in Female Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:642-50. [PMID: 26452297 PMCID: PMC4858387 DOI: 10.1289/ehp.1509851] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 10/05/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Although it is known that acetaminophen causes oxidative injury in the liver, it is not known whether it causes oxidative stress in the respiratory tract. If so, this widely used analgesic may potentiate the adverse effects of oxidant air pollutants. OBJECTIVES The goal of this study was to determine if acetaminophen induces respiratory tract oxidative stress and/or potentiates the oxidative stress and irritant responses to an inhaled oxidant: environmental tobacco smoke (ETS). METHODS Acetaminophen [100 mg/kg intraperitoneal (ip)] and/or sidestream tobacco smoke (as a surrogate for ETS, 5 mg/m3 for 10 min) were administered to female C57Bl/6J mice, and airway oxidative stress was assessed by loss of tissue antioxidants [estimated by nonprotein sulfhydryl (NPSH) levels] and/or induction of oxidant stress response genes. In addition, the effects of acetaminophen on airway irritation reflex responses to ETS were examined by plethysmography. RESULTS Acetaminophen diminished NPSH in nasal, thoracic extrapulmonary, and lung tissues; it also induced the oxidant stress response genes glutamate-cysteine ligase, catalytic subunit, and NAD(P)H dehydrogenase, quinone 1, in these sites. ETS produced a similar response. The response to acetaminophen plus ETS was equal to or greater than the sum of the responses to either agent alone. Although it had no effect by itself, acetaminophen greatly increased the reflex irritant response to ETS. CONCLUSIONS At supratherapeutic levels, acetaminophen induced oxidative stress throughout the respiratory tract and appeared to potentiate some responses to environmentally relevant ETS exposure in female C57Bl/6J mice. These results highlight the potential for this widely used drug to modulate responsiveness to oxidant air pollutants. CITATION Smith GJ, Cichocki JA, Doughty BJ, Manautou JE, Jordt SE, Morris JB. 2016. Effects of acetaminophen on oxidant and irritant respiratory tract responses to environmental tobacco smoke in female mice. Environ Health Perspect 124:642-650; http://dx.doi.org/10.1289/ehp.1509851.
Collapse
Affiliation(s)
- Gregory J. Smith
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Joseph A. Cichocki
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Bennett J. Doughty
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Jose E. Manautou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Sven-Eric Jordt
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - John B. Morris
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
8
|
Bailey LA, Nascarella MA, Kerper LE, Rhomberg LR. Hypothesis-based weight-of-evidence evaluation and risk assessment for naphthalene carcinogenesis. Crit Rev Toxicol 2015; 46:1-42. [PMID: 26202831 PMCID: PMC4732411 DOI: 10.3109/10408444.2015.1061477] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/09/2015] [Indexed: 11/13/2022]
Abstract
Inhalation of naphthalene causes olfactory epithelial nasal tumors in rats (but not in mice) and benign lung adenomas in mice (but not in rats). The limited available human data have not identified an association between naphthalene exposure and increased respiratory cancer risk. Assessing naphthalene's carcinogenicity in humans, therefore, depends entirely on experimental evidence from rodents. We evaluated the respiratory carcinogenicity of naphthalene in rodents, and its potential relevance to humans, using our Hypothesis-Based Weight-of-Evidence (HBWoE) approach. We systematically and comparatively reviewed data relevant to key elements in the hypothesized modes of action (MoA) to determine which is best supported by the available data, allowing all of the data from each realm of investigation to inform interpretation of one another. Our analysis supports a mechanism that involves initial metabolism of naphthalene to the epoxide, followed by GSH depletion, cytotoxicity, chronic inflammation, regenerative hyperplasia, and tumor formation, with possible weak genotoxicity from downstream metabolites occurring only at high cytotoxic doses, strongly supporting a non-mutagenic threshold MoA in the rat nose. We also conducted a dose-response analysis, based on the likely MoA, which suggests that the rat nasal MoA is not relevant in human respiratory tissues at typical environmental exposures. Our analysis illustrates how a thorough WoE evaluation can be used to support a MoA, even when a mechanism of action cannot be fully elucidated. A non-mutagenic threshold MoA for naphthalene-induced rat nasal tumors should be considered as a basis to determine human relevance and to guide regulatory and risk-management decisions.
Collapse
|
9
|
Kültz D, Li J, Sacchi R, Morin D, Buckpitt A, Van Winkle L. Alterations in the proteome of the respiratory tract in response to single and multiple exposures to naphthalene. Proteomics 2015; 15:2655-68. [PMID: 25825134 DOI: 10.1002/pmic.201400445] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 01/16/2015] [Accepted: 03/25/2015] [Indexed: 12/12/2022]
Abstract
Protein adduction is considered to be critical to the loss of cellular homeostasis associated with environmental chemicals undergoing metabolic activation. Despite considerable effort, our understanding of the key proteins mediating the pathologic consequences from protein modification by electrophiles is incomplete. This work focused on naphthalene (NA) induced acute injury of respiratory epithelial cells and tolerance which arises after multiple toxicant doses to define the initial cellular proteomic response and later protective actions related to tolerance. Airways and nasal olfactory epithelium from mice exposed to 15 ppm NA either for 4 h (acute) or for 4 h/day × 7 days (tolerant) were used for label-free protein quantitation by LC/MS/MS. Cytochrome P450 2F2 and secretoglobin 1A1 are decreased dramatically in airways of mice exposed for 4 h, a finding consistent with the fact that CYPs are localized primarily in Clara cells. A number of heat shock proteins and protein disulfide isomerases, which had previously been identified as adduct targets for reactive metabolites from several lung toxicants, were upregulated in airways but not olfactory epithelium of tolerant mice. Protein targets that are upregulated in tolerance may be key players in the pathophysiology associated with reactive metabolite protein adduction. All MS data have been deposited in the ProteomeXchange with identifier PXD000846 (http://proteomecentral.proteomexchange.org/dataset/PXD000846).
Collapse
Affiliation(s)
- Dietmar Kültz
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California, Davis, CA, USA
| | - Johnathon Li
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California, Davis, CA, USA
| | - Romina Sacchi
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California, Davis, CA, USA
| | - Dexter Morin
- Depatment of Molecular Biosciences, University of California, Davis, CA, USA
| | - Alan Buckpitt
- Depatment of Molecular Biosciences, University of California, Davis, CA, USA
| | - Laura Van Winkle
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, USA
| |
Collapse
|