Skip to main content
Erschienen in: Journal of Occupational Medicine and Toxicology 1/2011

Open Access 01.12.2011 | Research

Immune sensitization to methylene diphenyl diisocyanate (MDI) resulting from skin exposure: albumin as a carrier protein connecting skin exposure to subsequent respiratory responses

verfasst von: Adam V Wisnewski, Lan Xu, Eve Robinson, Jian Liu, Carrie A Redlich, Christina A Herrick

Erschienen in: Journal of Occupational Medicine and Toxicology | Ausgabe 1/2011

Abstract

Background

Methylene diphenyl diisocyanate (MDI), a reactive chemical used for commercial polyurethane production, is a well-recognized cause of occupational asthma. The major focus of disease prevention efforts to date has been respiratory tract exposure; however, skin exposure may also be an important route for inducing immune sensitization, which may promote subsequent airway inflammatory responses. We developed a murine model to investigate pathogenic mechanisms by which MDI skin exposure might promote subsequent immune responses, including respiratory tract inflammation.

Methods

Mice exposed via the skin to varying doses (0.1-10% w/v) of MDI diluted in acetone/olive oil were subsequently evaluated for MDI immune sensitization. Serum levels of MDI-specific IgG and IgE were measured by enzyme-linked immunosorbant assay (ELISA), while respiratory tract inflammation, induced by intranasal delivery of MDI-mouse albumin conjugates, was evaluated based on bronchoalveolar lavage (BAL). Autologous serum IgG from "skin only" exposed mice was used to detect and guide the purification/identification of skin proteins antigenically modified by MDI exposure in vivo.

Results

Skin exposure to MDI resulted in specific antibody production and promoted subsequent respiratory tract inflammation in animals challenged intranasally with MDI-mouse albumin conjugates. The degree of (secondary) respiratory tract inflammation and eosinophilia depended upon the (primary) skin exposure dose, and was maximal in mice exposed to 1% MDI, but paradoxically limited in mice receiving 10-fold higher doses (e.g. 10% MDI). The major antigenically-modified protein at the local MDI skin exposure site was identified as albumin, and demonstrated biophysical changes consistent with MDI conjugation.

Conclusions

MDI skin exposure can induce MDI-specific immune sensitivity and promote subsequent respiratory tract inflammatory responses and thus, may play an important role in MDI asthma pathogenesis. MDI conjugation and antigenic modification of albumin at local (skin/respiratory tract) exposure sites may represent the common antigenic link connecting skin exposure to subsequent respiratory tract inflammation.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1745-6673-6-6) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

AVW drafted the manuscript and supervised the in vitro immunology/biochemistry experiments. LX and ER performed in vivo skin and respiratory tract exposure studies, as well as BAL, and cell counts/differentials. JL performed the in vitro immunology/biochemistry experiments; ELISAs for MDI-specific IgG/IgE and total IgE, SDS-PAGE, Western blot, protein purification, and MDI-mouse albumin conjugate preparation. CAR organized the project and edited the manuscript. CAH conceived the original hypotheses underlying the overall project and supervised all aspects of the in vivo mouse studies. AVW, CAR, and CAH were together responsible for experiment design and data interpretation. All authors reviewed and approved the final manuscript.

Background

Isocyanates, the reactive chemicals used in the production of polyurethane foams, coatings, and adhesives remain a leading cause of occupational asthma world-wide, despite substantial efforts at disease prevention [1]. MDI has become the most commonly used isocyanate for multiple reasons, including its relatively low volatility at room temperature, which has been presumed to make it "safer" than other major isocyanates, e.g. hexamethylene and toluene diisocyanate (HDI and TDI respectively) [2, 3]. However, respirable forms of MDI are inherent to its common applications, which often involve heating and/or spraying the chemical, thus creating vapor and aerosols. The number of people at risk from MDI exposure continues to increase with increasing demand for polyurethane containing products; for example, "environmentally-friendly" or "green" construction using MDI-based spray-foam insulation made with soybean (vs. petroleum)-derived polyols [2, 4, 5]. A better understanding of MDI asthma pathogenesis is central to multiple approaches toward protecting the health of occupationally exposed individuals, including hygiene, engineering controls, personal protective equipment, exposure/disease surveillance and treatment [69].
Despite decades of research, the pathogenesis of MDI, and other isocyanate (TDI, HDI)-induced asthma remains unclear; however, contemporary theories suggest one important step involves the chemical's reactivity with "self" proteins in the respiratory tract, causing antigenic changes in their structure/conformation, which trigger an immune response [10, 11]. The self-proteins crucial to this process remain incompletely defined, however in animal models, the major target for isocyanate in the airways has been identified as albumin, by multiple investigators using several distinct approaches (immunochemical, radiotracing) [1215]. Albumin has also been found conjugated with isocyanate in vivo in occupationally exposed humans, and is the only known "carrier" protein for human antibody recognition and binding (e.g. IgE/IgG from exposed individuals specifically bind to isocyanate conjugates with human albumin, but not other proteins) [16]. Furthermore, in animal models of TDI and HDI asthma, albumin conjugates have been shown to induce asthma-like airway inflammation and/or physiologic responses in previously (isocyanate) sensitized animals [1722]. Thus, while the pathogenesis of MDI (and other isocyanate-induced) asthma remains unclear, previous studies support an important role for chemical conjugation with albumin present in the airways.
Given the airway localization of inflammation in isocyanate asthma patients, inhalation was originally assumed to be the primary exposure route responsible for the immune activation associated with exposure. However, evidence continues to increase in support of an alternative hypothesis; that skin exposure is equally (if not more) effective for isocyanate immune sensitization. Skin exposure to isocyanates is relatively common during polyurethane production (likely more common than airway exposure for "low volatility" isocyanates such as MDI) and thus could play a major role in sensitizing workers, despite appropriate respiratory tract protection, and without "warning" (methods for monitoring skin exposure remain poorly developed, and skin reactions are rare). Once immune sensitization to isocyanate occurs, extremely low airborne levels (below OSHA established permissible exposure levels) can trigger asthmatic reactions [23, 24]. Thus, while research, practice and regulation have focused almost exclusively on understanding and preventing inhalation exposures [6, 2527], skin exposure may be an equally critical, yet, under-recognized target for isocyanate asthma prevention [6, 8, 28, 29].
In this study, we developed a murine model to investigate the capacity of MDI skin exposure to induce systemic immune sensitization, and to identify key "MDI antigens" in this process. The investigation builds upon previous studies in guinea pigs and rats, which pioneered the hypothesis that isocyanate skin exposure might promote airway inflammation/asthma [3033]. The investigation also builds upon more recent mouse models of HDI and TDI asthma, which developed techniques for effectively delivering isocyanates (as mouse albumin conjugates) to the lower airways; thus overcoming technical challenges imposed by species difference between humans and mice ("scrubbing" action of nasal cavities and obligatory nasal breathing of mice), as well as respiratory tract irritation/toxicity by organic solvents (acetone, toluene) typically used for diluting isocyanate [15, 22, 31, 3437]. The findings of the present study are discussed in the context of disease (MDI asthma) pathogenesis and prevention.

Materials and methods

Reagents

Mouse and bovine albumin, triton X-100, sodium chloride, dithiothreitol (DTT), MDI, protease inhibitor cocktail and Tween 20 were from Sigma (St. Louis, MO). Urea and Tris-HCl were from American Bioanalytical (Natick, MA). Nonidet P40 substitute (Igepal CA-360) was from USB Corporation (Cleveland, OH). Acetone was from J.T. Baker (Phillipsburg, NJ). Ethylenediaminetetraacetic acid (EDTA) and phosphate buffered saline (PBS) were from Gibco (Grand Island, NY). Nunc Maxisorp™ microtiter plates were obtained through VWR International (Bridgeport, NJ). SuperSignal West Femto Maximum Sensitivity enhanced chemiluminescence substrate was obtained through Thermo Fisher Scientific (Rochester, NY). Tetramethylbenzidine (TMB) substrate was from BD Bioscience (San Jose, CA). Streptavidin conjugated alkaline phosphatase and p-nitrophenyl phosphate (pNPP) substrate were from Kirkegaard & Perry Laboratories (Gaithersburg, MD). Peroxidase conjugated rat anti-mouse anti-IgG1, and anti-IgG2a were from Pharmingen (San Diego, CA). Protein G Sepharose 4 Fast Flow was from GE Healthcare (Piscataway, NJ). Biotin-labeled rat anti-mouse IgE was from BioSource International, Inc. (Camarillo, CA). Imperial protein stain and rabbit anti-mouse IgG were from Pierce (Rockford, IL). Nitrocellulose and reducing gel electrophoresis buffer were from Bio-Rad (Hurcules, CA). Rabbit anti-tropomyosin, rabbit anti-collagen type 1/α2, and mouse anti-cytokeratin 14 were from Santa Cruz Biotechnology, Inc (Santa Cruz, CA).

Animals and skin sensitization

Female BALB/c mice, 9 to 12 weeks, from the National Cancer Institute (Frederick, MD), were used in all experiments. The backs of mice were shaved with electric clippers 1 day before exposure to 50 μl of MDI ranging in dose from 0.1%-10% weight/volume (w/v), delivered in a 4:1 acetone/olive oil "vehicle" (approximate surface area 0.5 - 1 cm2 on right side). Control mice were identically exposed to 50 μL of an acetone/olive oil mixture without MDI. Mice were anesthetized during the skin exposure, and 20 minutes after application, the exposed area was cleansed with 70% ethanol. Mice were re-exposed a second time 7 days later on the opposite (left) side of their back. Serum of exposed mice was obtained on day 21 and analyzed by ELISA for MDI-specific antibodies, and used as a probe to detect MDI (exposure)-induced antigenic-modification of "self" mouse skin proteins. In some studies MDI skin exposed mice were subsequently exposed to MDI-albumin conjugates via the respiratory tract (see below). A time line of skin/airway exposures and sample acquisition is shown in Figure 1.

Measurement of serum antibodies

Mouse sera samples were analyzed for MDI-specific antibodies using an enzyme-linked immunosorbant assay (ELISA), similar to that our laboratory has recently developed for measuring MDI-specific human antibodies [38]. Microtiter plates were coated with 1 μg/well of mouse albumin conjugated with MDI (see below), or control "mock exposed" mouse albumin, by overnight incubation at 4°C, in 0.1 M carbonate buffer (pH 9.5). Plates were "blocked" with 3% (w/v) bovine serum albumin before murine serum samples were titrated in blocking buffer. Sera were incubated for 1 hour at 25°C, followed by a 1:2000 dilution of peroxidase conjugated rat anti-mouse anti-IgG1 or anti-IgG2a. MDI-specific IgE was detected with biotin-labeled secondary rat anti-mouse IgE, followed by streptavidin-conjugated alkaline phosphatase. ELISAs were developed with TMB or p-NPP substrate and optical density (OD) measurements were obtained on a Benchmark microtiter plate reader from Bio-Rad. All samples were tested in triplicate to obtain average values expressed in figures.
MDI-specific IgG data are reported as end-titers; the reciprocal of the highest dilution that yields a positive OD reading, > 3 S.D. units above control serum from unexposed mice. Isocyanate-specific IgE data are represented as a binding ratio, as recommended in previous clinical studies, which is calculated as the (OD of wells coated with MDI-albumin) ÷ (OD of wells coated with control albumin) [39]. Total serum IgE levels were measured as previously described [40].

MDI-albumin

MDI-mouse albumin conjugates used for ELISA and respiratory tract challenge were prepared under the reaction conditions recently defined to yield optimally antigenic MDI-conjugates with human albumin [38]. Mouse albumin in phosphates buffered saline (pH 7.2) at 5 mg/ml was mixed with a freshly prepared solution of 10% (w/v) MDI dissolved in acetone, to achieve a final MDI concentration of 0.1% (w/v). The reaction mixture was rotated end-over-end for 2 hours at room temperature, dialyzed against PBS and (0.2 μM) filtered. "Mock exposed" albumin was identically prepared, using only acetone (1% v/v final concentration) for the 2-hr exposure period. MDI conjugation to mouse albumin was verified based on characteristic shift in electrophoretic mobility, and absorbance at 250 nm, due to MDI's double ring structure [41]. In later experiments, for comparative purposes (with albumin purified from skin exposed to MDI in vivo, see below), we generated MDI-mouse albumin conjugates in vitro with varying levels of MDI/protein molecule, by varying the MDI concentration during conjugation reactions.

Respiratory Tract Challenge with MDI-mouse albumin conjugates

Mice were lightly anesthetized with methoxyflurane and exposed to 50 μL of a 2 mg/ml solution of MDI-albumin or control "mock exposed" albumin in PBS by means of an intranasal droplet on days 14, 15, 18, and 19; and sacrificed by means of CO2 asphyxiation on day 21. Bronchoalveolar lavage (BAL) cell counts and differentials were performed as previously described [40].

Processing of skin proteins

Mice were skin exposed to MDI or vehicle for 20 minutes, as described above; following which, the exposed area was wiped clean with 70% ethanol, surgically excised, and snap frozen in liquid nitrogen. Skin samples were then homogenized in a glass tissue grinder in an isotonic, pH buffered, detergent solution (20 mM Tris-HCl, 0.15 M NaCl, 1 mM EDTA, 1% Triton X-100, 0.5% Nonidet P40 and a cocktail of protease inhibitors). The homogenized samples were then microfuged at 16,000 x g for 5 minutes to obtain a "detergent soluble" fraction (supernatant) of skin proteins. Before Western blot analysis, detergent extracted skin samples were depleted of endogenous murine immunoglobulins by incubation with Protein G-coated sepharose beads, and clearance by centrifugation. The detergent insoluble fraction of skin samples was further homogenized in a strong denaturing buffer containing 9M urea and 50 mM DTT, to obtain a urea soluble fraction of skin proteins.

Detection of antigenically modified skin proteins (MDI antigens)

Skin samples from MDI exposed mice were Western blotted with serum IgG from autologous mice that had been "skin-only" exposed to MDI, to detect "self" proteins antigenically modified by MDI exposure. Specificity controls included parallel blots with sera from mice exposed to vehicle only, and irrelevant (anti-ovalbumin) hyperimmune sera. Electrophoresis and Western blot were performed as previously described using pre-cast sodium dodecyl sulfate (SDS) acrylamide gels (4-15% gradient) from BioRad, and nitrocellulose membrane [42, 43]. Nitrocellulose strips were incubated for 2 hrs with a 1:100 dilution of sera, washed extensively with PBS containing 0.05% Tween 20, incubated with a 1:2000 dilution of peroxidase conjugated anti-mouse IgG, and developed with enhanced chemiluminescence substrate.

Purification of "MDI antigens" from exposed skin

Proteins from MDI exposed mouse skin were purified by a 2-step (isoelectric focusing/electroelution) process, guided by serum IgG from "skin only" exposed autologous mice, to detect antigenic modification. Preparative isoelectric focusing was performed using a Rotofor® system from Bio-Rad, according to the manufacturers recommendations, to initially separate skin proteins into 20 fractions between pH 3 and 10, with subsequent re-focusing between pH 3 to 6, to increase resolution. Rotofor fractions containing proteins antigenically modified by MDI exposure were further fractionated and analyzed by parallel Western blot/SDS-PAGE, from which they were excised using a Bio-Rad Model 422 Electro-Eluter run at constant current (8-10 mA/glass tube) for 3-5 hrs. Purified proteins were aliquoted and further analyzed for protein sequence (see below) and confirmation of MDI-antigenicity via immunoblot with serum IgG from exposed mice.

Protein identification

Liquid chromatography (LC) followed by tandem mass spectrometry (MS/MS) was performed by the Yale Keck Center on a Thermo Scientific LTQ-Orbitrap XL mass spectrometer, as previously described [44]. Briefly, purified proteins were reduced and carboxamidomethylated, trypsin digested and desalted with a C18 zip-tip column before MS/MS analysis. From uninterrupted MS/MS spectra, MASCOT compatible files (http://​www.​matrixscience.​com/​home.​html) were generated, and searched against the NCBI non-redundant database [45, 46]. For true positive protein identification, the 95% confidence level was set as a threshold within the MASCOT search engine (for protein hits based on randomness search). In addition, the following criteria must also have been met (1) two or more MS/MS spectra match the same protein entry in the database searched, (2) matched peptides were derived from trypsin digestion of the protein, (3) the peptides be murine in origin, and (4) the electrophoretic mobility must agree with the molecular weight. The identity of the purified proteins was further confirmed by Western blots with commercially available polyclonal or monoclonal antibodies (type I collagen, keratin-14, and tropomyosin), using hyperimmune anti-ovalbumin rabbit or mouse serum as a (negative) specificity control.

Statistical analyses

Statistical significance was determined using ANOVA with a block design for pooled data from more than one experiment. Antibody data, calculated through 2-fold dilutions, were log(2) transformed for analysis.

Results

Skin exposure induces an MDI-specific antibody (i.e. systemic) response

The capacity of MDI skin exposure to induce an MDI-specific antibody response was evaluated through ELISA analysis of sera from mice exposed to MDI diluted in acetone, at varying concentrations ranging from 0.1-10% weight/volume (w/v). We found that skin exposure to ≥ 1% MDI resulted in the development of high serum levels of MDI-specific antibodies. As shown in Figure 2, the end titers for MDI-specific antibody reached >1:100,000 and >1:30,000 for IgG1 and IgG2a subclasses respectively. MDI-specific IgE and total IgE serum levels were also elevated, up to 6-fold above control levels. The IgG and IgE induced by MDI skin exposure did not bind to unexposed proteins, or other reactive chemical "haptens" such as DNCB or adipoyl chloride (not shown).

Influence of skin exposure on (secondary) respiratory tract exposure

Mice initially exposed to MDI via the skin, were subsequently exposed via the respiratory, to a water soluble derivative of MDI (mouse albumin conjugates), in an adaptation of our murine HDI asthma model [22]. In the present experiments, mice that received only vehicle (acetone/olive oil) skin exposure, exhibited no change in bronchoalveolar lavage (BAL) cell numbers or differentials, when (airway) challenged with MDI-albumin conjugates. However, mice with previous (≥1%) MDI skin exposure developed significant airway inflammatory responses to respiratory challenge. The observed increase in total cell numbers of BAL samples (obtained 48 hours post exposure) was primarily due to increases in eosinophils and lymphocytes (Figure 3). Thus, respiratory tract exposure, to concentrations of MDI (albumin conjugates) that normally do not evoke cellular inflammation, causes pathologic changes (increased number of airway cells with Th2-profile) in mice previously exposed to MDI via the skin.
The initial MDI (skin) exposure dose was found to have a strong affect on the level of airway inflammation subsequently induced by respiratory tract challenge. The largest degree of airway inflammation was observed in mice initially (skin) exposed to MDI at a 1% (w/v) concentration, with more limited, albeit significant, inflammation in mice that had been skin exposed to 10% (w/v). The reason for the paradoxically limited airway inflammation in mice (skin) exposed to the highest test dose of MDI (10% w/v) remains unclear; however, analogous findings have been reported in HDI exposed mice [22]. A similar (non-linear dose-response) phenomenon is well-described for contact sensitization to many other reactive chemicals, e.g. formaldehyde, picryl chloride, DNCB [47].

Respiratory tract exposure boosts serum levels of MDI-specific antibodies elicited by primary skin exposure

In mice with prior MDI skin exposure, subsequent respiratory tract exposure to MDI-albumin conjugates was found to boost MDI-immune sensitization, based on levels of MDI-specific serum IgG and IgE. As shown in Figure 4, statistically significant increases were detectable among Th2-associated subclasses/isotypes, IgG1 and IgE, but not in the Th1-associated subclass, IgG2a. Thus, in mice previously exposed to MDI via the skin, subsequent respiratory tract exposure to MDI (albumin conjugates) further boosts MDI immune sensitivity.

Identification of MDI antigens in exposed skin

As shown in Figure 5A, detergent extracts from 1% MDI exposed skin contained a single antigenically-modified protein, specifically recognized by antibodies from autologous MDI skin (only) exposed mice, but not control mouse sera. The "MDI antigen" was purified from exposed skin by a 2-step process (Figure 5B, and 6A), and identified as albumin through LC-MS/MS analysis (see Additional file 1). The antigenically modified albumin from exposed skin exhibited biophysical properties consistent with MDI conjugation, when compared with albumin purified from vehicle-only exposed skin, or MDI-mouse albumin conjugates prepared in vitro; specifically, alterations in electrophoretic migration and change in absorbance at 250 nm (Figure 6A&6B).
Additional "MDI antigens", specifically recognized by antibodies from MDI skin (only) exposed autologous mice, but not control mouse sera, were detectable in urea extracts from skin exposed to the highest test dose of MDI (10%), as shown (Figure 7A). Among these antigenically-modified proteins, the most prominent, based on recognition by serum IgG from skin exposed autologous mice, were purified through elecrophoretic fractionation methods, and identified by LC-MS/MS as pro-collagen type 1/α2, keratin 14, and tropomyosin (see Additional file 1). Their (MDI) antigenicity and identity were further confirmed by Western blot with autologous serum IgG from skin exposed mice (Figure 7B) and commercially available protein-specific (collagen, keratin, tropomysosin) antibodies (not shown).

Discussion

In the present study, we utilized a murine MDI exposure model to demonstrate the capacity of skin exposure to induce immune sensitization to MDI, and promote airway inflammation upon subsequent respiratory tract exposure. The degree of secondary (respiratory tract) inflammation was found to depend upon the primary (skin) exposure dose, and exhibited a non-linear relationship that peaked when mice were skin exposed to 1% (w/v) MDI, and was paradoxically limited at 10-fold higher (skin) exposure doses; a phenomenon similar to that reported for HDI. Albumin in exposed skin was found to undergo antigenic as well as structural/conformational changes, consistent with MDI conjugation. Furthermore, MDI-mouse albumin conjugates were specifically recognized by serum IgE and IgG, and triggered heightened respiratory tract responses, in previously skin exposed mice. The data highlight mechanisms by which MDI skin exposure might contribute to the development of systemic immune sensitization and possibly MDI asthma.
The present findings are consistent with limited reports on MDI skin exposure in mice, despite differences in exposure protocols, and methods of assessing immunologic responses [4851]. The findings are also consistent with data on the smaller, more volatile 6-carbon isocyanates, HDI and TDI, including, the non-linear "(skin) dose/(respiratory tract) response" and mixed Th1/Th2-like response to skin exposure [22, 31, 34, 36, 52]. Importantly, in all of these studies, the isocyanate concentrations found to induce immune responses via skin exposure (≤%1 w/v) were within the range commonly used in polyurethane production, and are likely experienced by workers in multiple occupational settings [8, 28, 53].
The presently described mouse model possesses distinct strengths as well as limitations compared with previously published animal studies of MDI and/or other isocyanate-induced asthma. One major strength is the use of skin as the primary exposure route for inducing a state of MDI-specific immune sensitization in which subsequent respiratory tract exposure leads to asthma-like inflammation. In this regard, the present investigation differs from prior studies attempting to model isocyanate-induced airway inflammation through "respiratory tract only" exposure, which have met limited success [15, 31, 49, 5460]. Another strength of the present study is the use of autologous serum IgG from skin exposed mice to identify immunologically-relevant protein targets for MDI conjugation and (antigenic) modification. The major weakness of the study, as viewed a priori, was the use of MDI-albumin conjugates, rather than MDI itself, for respiratory tract exposure (see Introduction for rationale), thus bypassing a major step between inhalation and inflammation. Retrospectively, however, the data suggest that albumin conjugates may be uniquely suited as antigens in modeling isocyanate asthma, especially secondary to initial skin exposure.
The data provide new insight into the reactivity of MDI with proteins present in the skin, which likely contributes to the development of MDI immune sensitization. At the 1% MDI exposure dose (which promoted the strongest secondary respiratory tract responses), only 1 skin protein, albumin, exhibited changes consistent with MDI conjugation (charge/conformation, ultraviolet light absorbance, antigenicity). Albumin is a major protein of the extracellular compartment of the skin, but has not been previously recognized as a target for isocyanate at that anatomical location [61]. However, albumin in airway fluid has been described as a major target for isocyanate conjugation in vivo following respiratory tract exposure [1214, 16, 43, 62]. Furthermore, albumin is the only known human protein whose conjugation with isocyanate confers specific recognition by human antibodies from exposed individuals [43, 63]. Thus, the present data suggest that MDI conjugation to albumin in exposed skin creates an antigenic trigger that promotes subsequent airway inflammatory responses to respiratory tract exposure [22, 35].
While albumin was the only MDI antigen detectable in skin exposed to 1% MDI, additional proteins were found to be antigenically-modified in skin samples exposed to the highest test dose (10%) of MDI. The significance of these proteins in response to MDI skin exposure will require further investigation. However, it is interesting to speculate the possibility that reactivity with MDI may alter their normal conformation in a manner that breaks "immune tolerance" given the reported association of anti-keratin antibodies with isocyanate asthma, and the pan-allergenicity of non-mammalian tropomyosin [6466].
If the present data translate across species, they will provide important insight into pathogenic mechanisms of MDI asthma as well as practical guidance for disease prevention, among occupationally exposed individuals. The murine model will facilitate investigation of the role of specific genes, through transgenic technology, and provide a system for evaluating the effectiveness of different exposure interventions. The ELISA assay for MDI-specific IgG, described herein, may be helpful in assessing workplace skin exposure, which currently goes largely undetected, due to the lack of practical methodology for measuring. Furthermore, recognition of the ability to generate systemic immune sensitization to MDI via skin exposure, may promote increased awareness and use of personal (skin) protection, including gloves, overalls and head coverings.

Conclusions

In summary, we developed a murine model to investigate the potential consequences of MDI skin exposure, which is relatively common in the numerous industries that utilize MDI to make polyurethane products. The present data demonstrate that MDI skin exposure can induce systemic immune sensitization and asthmatic-like inflammatory responses to subsequent respiratory tract exposure. Albumin was found to be a major target for MDI conjugation in exposed skin, and MDI-albumin conjugates were also shown to trigger heightened respiratory tract inflammation in previously skin exposed mice (vs. unexposed controls). The data may help explain the development of new MDI asthma cases despite extremely low workplace airborne MDI levels and provide practical guidance for exposure and disease prevention.

Acknowledgements

The authors would like to Acknowledge Dr. Kathy Stone and Tom Abbot for their expert help with the LC/MS-MS studies. Funding was provided by grant support from the National Institutes of Health (NIH), the National Institute of Environmental Health Safety (NIEHS), and the National Institute for Occupational Safety and Health (NIOSH).
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

AVW drafted the manuscript and supervised the in vitro immunology/biochemistry experiments. LX and ER performed in vivo skin and respiratory tract exposure studies, as well as BAL, and cell counts/differentials. JL performed the in vitro immunology/biochemistry experiments; ELISAs for MDI-specific IgG/IgE and total IgE, SDS-PAGE, Western blot, protein purification, and MDI-mouse albumin conjugate preparation. CAR organized the project and edited the manuscript. CAH conceived the original hypotheses underlying the overall project and supervised all aspects of the in vivo mouse studies. AVW, CAR, and CAH were together responsible for experiment design and data interpretation. All authors reviewed and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Dykewicz MS: Occupational asthma: current concepts in pathogenesis, diagnosis, and management. J Allergy Clin Immunol 2009, 123: 519–28. quiz 529–30 10.1016/j.jaci.2009.01.061CrossRefPubMed Dykewicz MS: Occupational asthma: current concepts in pathogenesis, diagnosis, and management. J Allergy Clin Immunol 2009, 123: 519–28. quiz 529–30 10.1016/j.jaci.2009.01.061CrossRefPubMed
2.
Zurück zum Zitat Redlich CA, Wisnewski AV, Bello D: Environmental and Occupational Medicine. Edited by: Rom, W. N. Lippincott, Williams and Wilkins, Philadelphia, PA; 2007. Redlich CA, Wisnewski AV, Bello D: Environmental and Occupational Medicine. Edited by: Rom, W. N. Lippincott, Williams and Wilkins, Philadelphia, PA; 2007.
3.
Zurück zum Zitat Allport DC, Gilbert DS, Outterside SM, (Eds): MDI and TDI: Safety, Health and the Environment: A Source Book and Practical Guide. Wiley, Wiley, Chichester Wiley; 2003.CrossRef Allport DC, Gilbert DS, Outterside SM, (Eds): MDI and TDI: Safety, Health and the Environment: A Source Book and Practical Guide. Wiley, Wiley, Chichester Wiley; 2003.CrossRef
4.
Zurück zum Zitat ACC Center for the Polyurethane Industry: End Use Market Survey on the Polyurethanes Industry. 2008. ACC Center for the Polyurethane Industry: End Use Market Survey on the Polyurethanes Industry. 2008.
6.
Zurück zum Zitat Petsonk EL, Wang ML, Lewis DM, Siegel PD, Husberg BJ: Asthma-like symptoms in wood product plant workers exposed to methylene diphenyl diisocyanate. Chest 2000, 118: 1183–93. 10.1378/chest.118.4.1183CrossRefPubMed Petsonk EL, Wang ML, Lewis DM, Siegel PD, Husberg BJ: Asthma-like symptoms in wood product plant workers exposed to methylene diphenyl diisocyanate. Chest 2000, 118: 1183–93. 10.1378/chest.118.4.1183CrossRefPubMed
7.
Zurück zum Zitat Sabbioni G, Wesp H, Lewalter J, Rumler R: Determination of isocyanate biomarkers in construction site workers. Biomarkers 2007, 12: 468–83. 10.1080/13547500701395636CrossRefPubMed Sabbioni G, Wesp H, Lewalter J, Rumler R: Determination of isocyanate biomarkers in construction site workers. Biomarkers 2007, 12: 468–83. 10.1080/13547500701395636CrossRefPubMed
8.
Zurück zum Zitat Liljelind I, Norberg C, Egelrud L, Westberg H, Eriksson K, Nylander-French LA: Dermal and inhalation exposure to methylene bisphenyl isocyanate (MDI) in iron foundry workers. Ann Occup Hyg 2010, 54: 31–40. 10.1093/annhyg/mep067CrossRefPubMed Liljelind I, Norberg C, Egelrud L, Westberg H, Eriksson K, Nylander-French LA: Dermal and inhalation exposure to methylene bisphenyl isocyanate (MDI) in iron foundry workers. Ann Occup Hyg 2010, 54: 31–40. 10.1093/annhyg/mep067CrossRefPubMed
9.
Zurück zum Zitat Chester DA, Hanna EA, Pickelman BG, Rosenman KD: Asthma death after spraying polyurethane truck bedliner. Am J Ind Med 2005, 48: 78–84. 10.1002/ajim.20183CrossRefPubMed Chester DA, Hanna EA, Pickelman BG, Rosenman KD: Asthma death after spraying polyurethane truck bedliner. Am J Ind Med 2005, 48: 78–84. 10.1002/ajim.20183CrossRefPubMed
10.
Zurück zum Zitat Bernstein JA: Overview of diisocyanate occupational asthma. Toxicology 1996, 111: 181–9. 10.1016/0300-483X(96)03375-6CrossRefPubMed Bernstein JA: Overview of diisocyanate occupational asthma. Toxicology 1996, 111: 181–9. 10.1016/0300-483X(96)03375-6CrossRefPubMed
11.
Zurück zum Zitat Chen SE, Bernstein IL: The guinea pig model of diisocyanate sensitization. I. Immunologic studies. J Allergy Clin Immunol 1982, 70: 383–92. 10.1016/0091-6749(82)90029-XCrossRefPubMed Chen SE, Bernstein IL: The guinea pig model of diisocyanate sensitization. I. Immunologic studies. J Allergy Clin Immunol 1982, 70: 383–92. 10.1016/0091-6749(82)90029-XCrossRefPubMed
12.
Zurück zum Zitat Kennedy AL, Stock MF, Alarie Y, Brown WE: Uptake and distribution of 14C during and following inhalation exposure to radioactive toluene diisocyanate. Toxicol Appl Pharmacol 1989, 100: 280–92. 10.1016/0041-008X(89)90314-1CrossRefPubMed Kennedy AL, Stock MF, Alarie Y, Brown WE: Uptake and distribution of 14C during and following inhalation exposure to radioactive toluene diisocyanate. Toxicol Appl Pharmacol 1989, 100: 280–92. 10.1016/0041-008X(89)90314-1CrossRefPubMed
13.
Zurück zum Zitat Jin R, Day BW, Karol MH: Toluene diisocyanate protein adducts in the bronchoalveolar lavage of guinea pigs exposed to vapors of the chemical. Chem Res Toxicol 1993, 6: 906–12. 10.1021/tx00036a023CrossRefPubMed Jin R, Day BW, Karol MH: Toluene diisocyanate protein adducts in the bronchoalveolar lavage of guinea pigs exposed to vapors of the chemical. Chem Res Toxicol 1993, 6: 906–12. 10.1021/tx00036a023CrossRefPubMed
14.
Zurück zum Zitat Kennedy AL, Wilson TR, Stock MF, Alarie Y, Brown WE: Distribution and reactivity of inhaled 14C-labeled toluene diisocyanate (TDI) in rats. Arch Toxicol 1994, 68: 434–43. 10.1007/s002040050094CrossRefPubMed Kennedy AL, Wilson TR, Stock MF, Alarie Y, Brown WE: Distribution and reactivity of inhaled 14C-labeled toluene diisocyanate (TDI) in rats. Arch Toxicol 1994, 68: 434–43. 10.1007/s002040050094CrossRefPubMed
15.
Zurück zum Zitat Kennedy AL, Singh G, Alarie Y, Brown WE: Autoradiographic analyses of guinea pig airway tissues following inhalation exposure to 14C-labeled methyl isocyanate. Fundam Appl Toxicol 1993, 20: 57–67. 10.1006/faat.1993.1007CrossRefPubMed Kennedy AL, Singh G, Alarie Y, Brown WE: Autoradiographic analyses of guinea pig airway tissues following inhalation exposure to 14C-labeled methyl isocyanate. Fundam Appl Toxicol 1993, 20: 57–67. 10.1006/faat.1993.1007CrossRefPubMed
16.
Zurück zum Zitat Liu Q, Wisnewski AV: Recent developments in diisocyanate asthma. Ann Allergy Asthma Immunol 2003, 90: 35–41. 10.1016/S1081-1206(10)61647-XCrossRefPubMed Liu Q, Wisnewski AV: Recent developments in diisocyanate asthma. Ann Allergy Asthma Immunol 2003, 90: 35–41. 10.1016/S1081-1206(10)61647-XCrossRefPubMed
17.
Zurück zum Zitat Bernstein IL, Splansky GL, Chen SE, Vinegar A: The guinea pig model of diisocyanate sensitization. II. Physiologic studies. J Allergy Clin Immunol 1982, 70: 393–8. 10.1016/0091-6749(82)90030-6CrossRefPubMed Bernstein IL, Splansky GL, Chen SE, Vinegar A: The guinea pig model of diisocyanate sensitization. II. Physiologic studies. J Allergy Clin Immunol 1982, 70: 393–8. 10.1016/0091-6749(82)90030-6CrossRefPubMed
18.
Zurück zum Zitat Patterson R, Zeiss CR, Harris KE: Immunologic and respiratory responses to airway challenges of dogs with toluene diisocyanate. J Allergy Clin Immunol 1983, 71: 604–11. 10.1016/0091-6749(83)90443-8CrossRefPubMed Patterson R, Zeiss CR, Harris KE: Immunologic and respiratory responses to airway challenges of dogs with toluene diisocyanate. J Allergy Clin Immunol 1983, 71: 604–11. 10.1016/0091-6749(83)90443-8CrossRefPubMed
19.
Zurück zum Zitat Pauluhn J: Assessment of respiratory hypersensitivity in guinea pigs sensitized to toluene diisocyanate: improvements on analysis of respiratory response. Fundam Appl Toxicol 1997, 40: 211–9. 10.1006/faat.1997.2393CrossRefPubMed Pauluhn J: Assessment of respiratory hypersensitivity in guinea pigs sensitized to toluene diisocyanate: improvements on analysis of respiratory response. Fundam Appl Toxicol 1997, 40: 211–9. 10.1006/faat.1997.2393CrossRefPubMed
20.
Zurück zum Zitat Sugawara Y, Okamoto Y, Sawahata T, Tanaka K: An asthma model developed in the guinea pig by intranasal application of 2,4-toluene diisocyanate. Int Arch Allergy Immunol 1993, 101: 95–101. 10.1159/000236504CrossRefPubMed Sugawara Y, Okamoto Y, Sawahata T, Tanaka K: An asthma model developed in the guinea pig by intranasal application of 2,4-toluene diisocyanate. Int Arch Allergy Immunol 1993, 101: 95–101. 10.1159/000236504CrossRefPubMed
21.
Zurück zum Zitat Huang J, Millecchia LL, Frazer DG, Fedan JS: Airway hyperreactivity elicited by toluene diisocyanate (TDI)-albumin conjugate is not accompanied by airway eosinophilic infiltration in guinea pigs. Arch Toxicol 1998, 72: 141–6. 10.1007/s002040050480CrossRefPubMed Huang J, Millecchia LL, Frazer DG, Fedan JS: Airway hyperreactivity elicited by toluene diisocyanate (TDI)-albumin conjugate is not accompanied by airway eosinophilic infiltration in guinea pigs. Arch Toxicol 1998, 72: 141–6. 10.1007/s002040050480CrossRefPubMed
22.
Zurück zum Zitat Herrick CA, Xu L, Wisnewski AV, Das J, Redlich CA, Bottomly K: A novel mouse model of diisocyanate-induced asthma showing allergic-type inflammation in the lung after inhaled antigen challenge. J Allergy Clin Immunol 2002, 109: 873–8. 10.1067/mai.2002.123533CrossRefPubMed Herrick CA, Xu L, Wisnewski AV, Das J, Redlich CA, Bottomly K: A novel mouse model of diisocyanate-induced asthma showing allergic-type inflammation in the lung after inhaled antigen challenge. J Allergy Clin Immunol 2002, 109: 873–8. 10.1067/mai.2002.123533CrossRefPubMed
23.
Zurück zum Zitat Bello D, Herrick CA, Smith TJ, Woskie SR, Streicher RP, Cullen MR, Liu Y, Redlich CA: Skin exposure to isocyanates: reasons for concern. Environ Health Perspect 2007, 115: 328–35. 10.1289/ehp.9557PubMedCentralCrossRefPubMed Bello D, Herrick CA, Smith TJ, Woskie SR, Streicher RP, Cullen MR, Liu Y, Redlich CA: Skin exposure to isocyanates: reasons for concern. Environ Health Perspect 2007, 115: 328–35. 10.1289/ehp.9557PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Redlich CA, Herrick CA: Lung/skin connections in occupational lung disease. Curr Opin Allergy Clin Immunol 2008, 8: 115–9. 10.1097/ACI.0b013e3282f85a31CrossRefPubMed Redlich CA, Herrick CA: Lung/skin connections in occupational lung disease. Curr Opin Allergy Clin Immunol 2008, 8: 115–9. 10.1097/ACI.0b013e3282f85a31CrossRefPubMed
25.
Zurück zum Zitat Tinnerberg H, Mattsson C: Usage of air monitoring and biomarkers of isocyanate exposure to assess the effect of a control intervention. Ann Occup Hyg 2008, 52: 187–94. 10.1093/annhyg/men006CrossRefPubMed Tinnerberg H, Mattsson C: Usage of air monitoring and biomarkers of isocyanate exposure to assess the effect of a control intervention. Ann Occup Hyg 2008, 52: 187–94. 10.1093/annhyg/men006CrossRefPubMed
26.
Zurück zum Zitat Liu Y, Stowe MH, Bello D, Woskie SR, Sparer J, Gore R, Youngs F, Cullen MR, Redlich CA: Respiratory protection from isocyanate exposure in the autobody repair and refinishing industry. J Occup Environ Hyg 2006, 3: 234–49. 10.1080/15459620600628704CrossRefPubMed Liu Y, Stowe MH, Bello D, Woskie SR, Sparer J, Gore R, Youngs F, Cullen MR, Redlich CA: Respiratory protection from isocyanate exposure in the autobody repair and refinishing industry. J Occup Environ Hyg 2006, 3: 234–49. 10.1080/15459620600628704CrossRefPubMed
27.
Zurück zum Zitat Pauluhn J, Woolhiser MR, Bloemen L: Repeated inhalation challenge with diphenylmethane-4,4'-diisocyanate in brown Norway rats leads to a time-related increase of neutrophils in bronchoalveolar lavage after topical induction. Inhal Toxicol 2005, 17: 67–78. 10.1080/08958370590898434CrossRefPubMed Pauluhn J, Woolhiser MR, Bloemen L: Repeated inhalation challenge with diphenylmethane-4,4'-diisocyanate in brown Norway rats leads to a time-related increase of neutrophils in bronchoalveolar lavage after topical induction. Inhal Toxicol 2005, 17: 67–78. 10.1080/08958370590898434CrossRefPubMed
28.
Zurück zum Zitat Liu Y, Stowe MH, Bello D, Sparer J, Gore RJ, Cullen MR, Redlich CA, Woskie SR: Skin exposure to aliphatic polyisocyanates in the auto body repair and refinishing industry: III. A personal exposure algorithm. Ann Occup Hyg 2009, 53: 33–40. 10.1093/annhyg/men070PubMedCentralCrossRefPubMed Liu Y, Stowe MH, Bello D, Sparer J, Gore RJ, Cullen MR, Redlich CA, Woskie SR: Skin exposure to aliphatic polyisocyanates in the auto body repair and refinishing industry: III. A personal exposure algorithm. Ann Occup Hyg 2009, 53: 33–40. 10.1093/annhyg/men070PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat Bello D, Redlich CA, Stowe MH, Sparer J, Woskie SR, Streicher RP, Hosgood HD, Liu Y: Skin exposure to aliphatic polyisocyanates in the auto body repair and refinishing industry: II. A quantitative assessment. Ann Occup Hyg 2008, 52: 117–24. 10.1093/annhyg/mem066CrossRefPubMed Bello D, Redlich CA, Stowe MH, Sparer J, Woskie SR, Streicher RP, Hosgood HD, Liu Y: Skin exposure to aliphatic polyisocyanates in the auto body repair and refinishing industry: II. A quantitative assessment. Ann Occup Hyg 2008, 52: 117–24. 10.1093/annhyg/mem066CrossRefPubMed
30.
Zurück zum Zitat Karol MH, Hauth BA, Riley EJ, Magreni CM: Dermal contact with toluene diisocyanate (TDI) produces respiratory tract hypersensitivity in guinea pigs. Toxicol Appl Pharmacol 1981, 58: 221–30. 10.1016/0041-008X(81)90426-9CrossRefPubMed Karol MH, Hauth BA, Riley EJ, Magreni CM: Dermal contact with toluene diisocyanate (TDI) produces respiratory tract hypersensitivity in guinea pigs. Toxicol Appl Pharmacol 1981, 58: 221–30. 10.1016/0041-008X(81)90426-9CrossRefPubMed
31.
Zurück zum Zitat Ban M, Morel G, Langonne I, Huguet N, Pepin E, Binet S: TDI can induce respiratory allergy with Th2-dominated response in mice. Toxicology 2006, 218: 39–47. 10.1016/j.tox.2005.09.013CrossRefPubMed Ban M, Morel G, Langonne I, Huguet N, Pepin E, Binet S: TDI can induce respiratory allergy with Th2-dominated response in mice. Toxicology 2006, 218: 39–47. 10.1016/j.tox.2005.09.013CrossRefPubMed
32.
Zurück zum Zitat Pauluhn J: Brown Norway rat asthma model of diphenylmethane-4,4'-diisocyanate (MDI): impact of vehicle for topical induction. Regul Toxicol Pharmacol 2008, 50: 144–54. 10.1016/j.yrtph.2007.09.003CrossRefPubMed Pauluhn J: Brown Norway rat asthma model of diphenylmethane-4,4'-diisocyanate (MDI): impact of vehicle for topical induction. Regul Toxicol Pharmacol 2008, 50: 144–54. 10.1016/j.yrtph.2007.09.003CrossRefPubMed
33.
Zurück zum Zitat Pauluhn J: Brown Norway rat asthma model of diphenylmethane-4,4'-diisocyanate (MDI): analysis of the elicitation dose-response relationship. Toxicol Sci 2008, 104: 320–31. 10.1093/toxsci/kfn098CrossRefPubMed Pauluhn J: Brown Norway rat asthma model of diphenylmethane-4,4'-diisocyanate (MDI): analysis of the elicitation dose-response relationship. Toxicol Sci 2008, 104: 320–31. 10.1093/toxsci/kfn098CrossRefPubMed
34.
Zurück zum Zitat Tarkowski M, Vanoirbeek JA, Vanhooren HM, De Vooght V, Mercier CM, Ceuppens J, Nemery B, Hoet PH: Immunological determinants of ventilatory changes induced in mice by dermal sensitization and respiratory challenge with toluene diisocyanate. Am J Physiol Lung Cell Mol Physiol 2007, 292: L207–14. 10.1152/ajplung.00157.2005CrossRefPubMed Tarkowski M, Vanoirbeek JA, Vanhooren HM, De Vooght V, Mercier CM, Ceuppens J, Nemery B, Hoet PH: Immunological determinants of ventilatory changes induced in mice by dermal sensitization and respiratory challenge with toluene diisocyanate. Am J Physiol Lung Cell Mol Physiol 2007, 292: L207–14. 10.1152/ajplung.00157.2005CrossRefPubMed
35.
Zurück zum Zitat Herrick CA, Das J, Xu L, Wisnewski AV, Redlich CA, Bottomly K: Differential roles for CD4 and CD8 T cells after diisocyanate sensitization: genetic control of TH2-induced lung inflammation. J Allergy Clin Immunol 2003, 111: 1087–94. 10.1067/mai.2003.1413CrossRefPubMed Herrick CA, Das J, Xu L, Wisnewski AV, Redlich CA, Bottomly K: Differential roles for CD4 and CD8 T cells after diisocyanate sensitization: genetic control of TH2-induced lung inflammation. J Allergy Clin Immunol 2003, 111: 1087–94. 10.1067/mai.2003.1413CrossRefPubMed
36.
Zurück zum Zitat Vanoirbeek JA, Tarkowski M, Ceuppens JL, Verbeken EK, Nemery B, Hoet PH: Respiratory response to toluene diisocyanate depends on prior frequency and concentration of dermal sensitization in mice. Toxicol Sci 2004, 80: 310–21. 10.1093/toxsci/kfh155CrossRefPubMed Vanoirbeek JA, Tarkowski M, Ceuppens JL, Verbeken EK, Nemery B, Hoet PH: Respiratory response to toluene diisocyanate depends on prior frequency and concentration of dermal sensitization in mice. Toxicol Sci 2004, 80: 310–21. 10.1093/toxsci/kfh155CrossRefPubMed
37.
Zurück zum Zitat Dearman RJ, Moussavi A, Kemeny DM, Kimber I: Contribution of CD4+ and CD8+ T lymphocyte subsets to the cytokine secretion patterns induced in mice during sensitization to contact and respiratory chemical allergens. Immunology 1996, 89: 502–10. 10.1046/j.1365-2567.1996.d01-778.xPubMedCentralCrossRefPubMed Dearman RJ, Moussavi A, Kemeny DM, Kimber I: Contribution of CD4+ and CD8+ T lymphocyte subsets to the cytokine secretion patterns induced in mice during sensitization to contact and respiratory chemical allergens. Immunology 1996, 89: 502–10. 10.1046/j.1365-2567.1996.d01-778.xPubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Wisnewski AV, Liu J, Redlich CA: Antigenic changes in human albumin caused by reactivity with the occupational allergen diphenylmethane diisocyanate. Anal Biochem 2010, 400: 251–8. 10.1016/j.ab.2010.01.037PubMedCentralCrossRefPubMed Wisnewski AV, Liu J, Redlich CA: Antigenic changes in human albumin caused by reactivity with the occupational allergen diphenylmethane diisocyanate. Anal Biochem 2010, 400: 251–8. 10.1016/j.ab.2010.01.037PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Karol MH, Kramarik JA, Ferguson J: Methods to assess RAST results in patients exposed to chemical allergens. Allergy 1995, 50: 48–54. 10.1111/j.1398-9995.1995.tb02482.xCrossRefPubMed Karol MH, Kramarik JA, Ferguson J: Methods to assess RAST results in patients exposed to chemical allergens. Allergy 1995, 50: 48–54. 10.1111/j.1398-9995.1995.tb02482.xCrossRefPubMed
40.
Zurück zum Zitat Herrick CA, MacLeod H, Glusac E, Tigelaar RE, Bottomly K: Th2 responses induced by epicutaneous or inhalational protein exposure are differentially dependent on IL-4. J Clin Invest 2000, 105: 765–75. 10.1172/JCI8624PubMedCentralCrossRefPubMed Herrick CA, MacLeod H, Glusac E, Tigelaar RE, Bottomly K: Th2 responses induced by epicutaneous or inhalational protein exposure are differentially dependent on IL-4. J Clin Invest 2000, 105: 765–75. 10.1172/JCI8624PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Jin RZ, Karol MH: Intra- and intermolecular reactions of 4,4'-diisocyanatodiphenylmethane with human serum albumin. Chem Res Toxicol 1988, 1: 281–7. 10.1021/tx00005a005CrossRefPubMed Jin RZ, Karol MH: Intra- and intermolecular reactions of 4,4'-diisocyanatodiphenylmethane with human serum albumin. Chem Res Toxicol 1988, 1: 281–7. 10.1021/tx00005a005CrossRefPubMed
42.
Zurück zum Zitat Coligan J, Kruisbeck A, Marguiles D, Sevacch E, Stober W, editors: Current Protocols in Immunology. Wiley and Sons Inc, West Sussex; 1998. Coligan J, Kruisbeck A, Marguiles D, Sevacch E, Stober W, editors: Current Protocols in Immunology. Wiley and Sons Inc, West Sussex; 1998.
43.
Zurück zum Zitat Wisnewski AV, Srivastava R, Herick C, Xu L, Lemus R, Cain H, Magoski NM, Karol MH, Bottomly K, Redlich CA: Identification of human lung and skin proteins conjugated with hexamethylene diisocyanate in vitro and in vivo. Am J Respir Crit Care Med 2000, 162: 2330–6.CrossRefPubMed Wisnewski AV, Srivastava R, Herick C, Xu L, Lemus R, Cain H, Magoski NM, Karol MH, Bottomly K, Redlich CA: Identification of human lung and skin proteins conjugated with hexamethylene diisocyanate in vitro and in vivo. Am J Respir Crit Care Med 2000, 162: 2330–6.CrossRefPubMed
44.
Zurück zum Zitat Stone KL, DeAngelis R, LoPresti M, Jones J, Papov VV, Williams KR: Use of liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) for routine identification of enzymatically digested proteins separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Electrophoresis 1998, 19: 1046–52. 10.1002/elps.1150190620CrossRefPubMed Stone KL, DeAngelis R, LoPresti M, Jones J, Papov VV, Williams KR: Use of liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) for routine identification of enzymatically digested proteins separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Electrophoresis 1998, 19: 1046–52. 10.1002/elps.1150190620CrossRefPubMed
45.
Zurück zum Zitat Perkins DN, Pappin DJ, Creasy DM, Cottrell JS: Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 1999, 20: 3551–67. 10.1002/(SICI)1522-2683(19991201)20:18<3551::AID-ELPS3551>3.0.CO;2-2CrossRefPubMed Perkins DN, Pappin DJ, Creasy DM, Cottrell JS: Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 1999, 20: 3551–67. 10.1002/(SICI)1522-2683(19991201)20:18<3551::AID-ELPS3551>3.0.CO;2-2CrossRefPubMed
46.
Zurück zum Zitat Hirosawa M, Hoshida M, Ishikawa M, Toya T: MASCOT: multiple alignment system for protein sequences based on three-way dynamic programming. Comput Appl Biosci 1993, 9: 161–7.PubMed Hirosawa M, Hoshida M, Ishikawa M, Toya T: MASCOT: multiple alignment system for protein sequences based on three-way dynamic programming. Comput Appl Biosci 1993, 9: 161–7.PubMed
47.
Zurück zum Zitat Andersen KE: Testing for contact allergy in experimental animals. Pharmacol Toxicol 1987, 61: 1–8. 10.1111/j.1600-0773.1987.tb01762.xCrossRefPubMed Andersen KE: Testing for contact allergy in experimental animals. Pharmacol Toxicol 1987, 61: 1–8. 10.1111/j.1600-0773.1987.tb01762.xCrossRefPubMed
48.
Zurück zum Zitat Dearman RJ, Basketter DA, Kimber I: Characterization of chemical allergens as a function of divergent cytokine secretion profiles induced in mice. Toxicol Appl Pharmacol 1996, 138: 308–16. 10.1006/taap.1996.0129CrossRefPubMed Dearman RJ, Basketter DA, Kimber I: Characterization of chemical allergens as a function of divergent cytokine secretion profiles induced in mice. Toxicol Appl Pharmacol 1996, 138: 308–16. 10.1006/taap.1996.0129CrossRefPubMed
49.
Zurück zum Zitat Farraj AK, Boykin E, Haykal-Coates N, Gavett SH, Doerfler D, Selgrade M: Th2 Cytokines in Skin Draining Lymph Nodes and Serum IgE Do Not Predict Airway Hypersensitivity to Intranasal Isocyanate Exposure in Mice. Toxicol Sci 2007, 100: 99–108. 10.1093/toxsci/kfm194CrossRefPubMed Farraj AK, Boykin E, Haykal-Coates N, Gavett SH, Doerfler D, Selgrade M: Th2 Cytokines in Skin Draining Lymph Nodes and Serum IgE Do Not Predict Airway Hypersensitivity to Intranasal Isocyanate Exposure in Mice. Toxicol Sci 2007, 100: 99–108. 10.1093/toxsci/kfm194CrossRefPubMed
50.
Zurück zum Zitat Selgrade M, Boykin EH, Haykal-Coates N, Woolhiser MR, Wiescinski C, Andrews DL, Farraj AK, Doerfler DL, Gavett SH: Inconsistencies between cytokine profiles, antibody responses, and respiratory hyperresponsiveness following dermal exposure to isocyanates. Toxicol Sci 2006, 94: 108–17. 10.1093/toxsci/kfl094CrossRefPubMed Selgrade M, Boykin EH, Haykal-Coates N, Woolhiser MR, Wiescinski C, Andrews DL, Farraj AK, Doerfler DL, Gavett SH: Inconsistencies between cytokine profiles, antibody responses, and respiratory hyperresponsiveness following dermal exposure to isocyanates. Toxicol Sci 2006, 94: 108–17. 10.1093/toxsci/kfl094CrossRefPubMed
51.
Zurück zum Zitat Potter DW, Wederbrand KS: Total IgE antibody production in BALB/c mice after dermal exposure to chemicals. Fundam Appl Toxicol 1995, 26: 127–35. 10.1006/faat.1995.1082CrossRefPubMed Potter DW, Wederbrand KS: Total IgE antibody production in BALB/c mice after dermal exposure to chemicals. Fundam Appl Toxicol 1995, 26: 127–35. 10.1006/faat.1995.1082CrossRefPubMed
52.
Zurück zum Zitat Vanoirbeek JA, De Vooght V, Vanhooren HM, Nawrot TS, Nemery B, Hoet PH: How long do the systemic and ventilatory responses to toluene diisocyanate persist in dermally sensitized mice? J Allergy Clin Immunol 2008, 121: 456–463e5. 10.1016/j.jaci.2007.09.006CrossRefPubMed Vanoirbeek JA, De Vooght V, Vanhooren HM, Nawrot TS, Nemery B, Hoet PH: How long do the systemic and ventilatory responses to toluene diisocyanate persist in dermally sensitized mice? J Allergy Clin Immunol 2008, 121: 456–463e5. 10.1016/j.jaci.2007.09.006CrossRefPubMed
53.
Zurück zum Zitat Lesage J, Stanley J, Karoly WJ, Lichtenberg FW: Airborne methylene diphenyl diisocyanate (MDI) concentrations associated with the application of polyurethane spray foam in residential construction. J Occup Environ Hyg 2007, 4: 145–55. 10.1080/15459620601133779CrossRefPubMed Lesage J, Stanley J, Karoly WJ, Lichtenberg FW: Airborne methylene diphenyl diisocyanate (MDI) concentrations associated with the application of polyurethane spray foam in residential construction. J Occup Environ Hyg 2007, 4: 145–55. 10.1080/15459620601133779CrossRefPubMed
54.
Zurück zum Zitat Vanoirbeek JA, De Vooght V, Nemery B, Hoet PH: Multiple challenges in a mouse model of chemical-induced asthma lead to tolerance: ventilatory and inflammatory responses are blunted, immunologic humoral responses are not. Toxicology 2009, 257: 144–52. 10.1016/j.tox.2008.12.020CrossRefPubMed Vanoirbeek JA, De Vooght V, Nemery B, Hoet PH: Multiple challenges in a mouse model of chemical-induced asthma lead to tolerance: ventilatory and inflammatory responses are blunted, immunologic humoral responses are not. Toxicology 2009, 257: 144–52. 10.1016/j.tox.2008.12.020CrossRefPubMed
55.
Zurück zum Zitat Satoh T, Kramarik JA, Tollerud DJ, Karol MH: A murine model for assessing the respiratory hypersensitivity potential of chemical allergens. Toxicol Lett 1995, 78: 57–66. 10.1016/0378-4274(94)03234-XCrossRefPubMed Satoh T, Kramarik JA, Tollerud DJ, Karol MH: A murine model for assessing the respiratory hypersensitivity potential of chemical allergens. Toxicol Lett 1995, 78: 57–66. 10.1016/0378-4274(94)03234-XCrossRefPubMed
56.
Zurück zum Zitat Pauluhn J, Dearman R, Doe J, Hext P, Landry TD: Respiratory hypersensitivity to diphenylmethane-4,4'-diisocyanate in guinea pigs: comparison with trimellitic anhydride. Inhal Toxicol 1999, 11: 187–214. 10.1080/089583799197140CrossRefPubMed Pauluhn J, Dearman R, Doe J, Hext P, Landry TD: Respiratory hypersensitivity to diphenylmethane-4,4'-diisocyanate in guinea pigs: comparison with trimellitic anhydride. Inhal Toxicol 1999, 11: 187–214. 10.1080/089583799197140CrossRefPubMed
57.
Zurück zum Zitat Nabe T, Yamauchi K, Shinjo Y, Niwa T, Imoto K, Koda A, Kohno S: Delayed-type asthmatic response induced by repeated intratracheal exposure to toluene-2,4-diisocyanate in guinea pigs. Int Arch Allergy Immunol 2005, 137: 115–24. 10.1159/000085466CrossRefPubMed Nabe T, Yamauchi K, Shinjo Y, Niwa T, Imoto K, Koda A, Kohno S: Delayed-type asthmatic response induced by repeated intratracheal exposure to toluene-2,4-diisocyanate in guinea pigs. Int Arch Allergy Immunol 2005, 137: 115–24. 10.1159/000085466CrossRefPubMed
58.
Zurück zum Zitat Karol MH: Concentration-dependent immunologic response to toluene diisocyanate (TDI) following inhalation exposure. Toxicol Appl Pharmacol 1983, 68: 229–41. 10.1016/0041-008X(83)90007-8CrossRefPubMed Karol MH: Concentration-dependent immunologic response to toluene diisocyanate (TDI) following inhalation exposure. Toxicol Appl Pharmacol 1983, 68: 229–41. 10.1016/0041-008X(83)90007-8CrossRefPubMed
59.
Zurück zum Zitat Johnson VJ, Yucesoy B, Reynolds JS, Fluharty K, Wang W, Richardson D, Luster MI: Inhalation of toluene diisocyanate vapor induces allergic rhinitis in mice. J Immunol 2007, 179: 1864–71.CrossRefPubMed Johnson VJ, Yucesoy B, Reynolds JS, Fluharty K, Wang W, Richardson D, Luster MI: Inhalation of toluene diisocyanate vapor induces allergic rhinitis in mice. J Immunol 2007, 179: 1864–71.CrossRefPubMed
60.
Zurück zum Zitat Blaikie L, Morrow T, Wilson AP, Hext P, Hartop PJ, Rattray NJ, Woodcock D, Botham PA: A two-centre study for the evaluation and validation of an animal model for the assessment of the potential of small molecular weight chemicals to cause respiratory allergy. Toxicology 1995, 96: 37–50. 10.1016/0300-483X(94)03007-OCrossRefPubMed Blaikie L, Morrow T, Wilson AP, Hext P, Hartop PJ, Rattray NJ, Woodcock D, Botham PA: A two-centre study for the evaluation and validation of an animal model for the assessment of the potential of small molecular weight chemicals to cause respiratory allergy. Toxicology 1995, 96: 37–50. 10.1016/0300-483X(94)03007-OCrossRefPubMed
61.
Zurück zum Zitat Quinlan GJ, Martin GS, Evans TW: Albumin: biochemical properties and therapeutic potential. Hepatology 2005, 41: 1211–9. 10.1002/hep.20720CrossRefPubMed Quinlan GJ, Martin GS, Evans TW: Albumin: biochemical properties and therapeutic potential. Hepatology 2005, 41: 1211–9. 10.1002/hep.20720CrossRefPubMed
62.
Zurück zum Zitat Wass U, Belin L: Immunologic specificity of isocyanate-induced IgE antibodies in serum from 10 sensitized workers. J Allergy Clin Immunol 1989, 83: 126–35. 10.1016/0091-6749(89)90487-9CrossRefPubMed Wass U, Belin L: Immunologic specificity of isocyanate-induced IgE antibodies in serum from 10 sensitized workers. J Allergy Clin Immunol 1989, 83: 126–35. 10.1016/0091-6749(89)90487-9CrossRefPubMed
63.
Zurück zum Zitat Wisnewski AV, Stowe MH, Cartier A, Liu Q, Liu J, Chen L, Redlich CA: Isocyanate vapor-induced antigenicity of human albumin. J Allergy Clin Immunol 2004, 113: 1178–84. 10.1016/j.jaci.2004.03.009CrossRefPubMed Wisnewski AV, Stowe MH, Cartier A, Liu Q, Liu J, Chen L, Redlich CA: Isocyanate vapor-induced antigenicity of human albumin. J Allergy Clin Immunol 2004, 113: 1178–84. 10.1016/j.jaci.2004.03.009CrossRefPubMed
64.
Zurück zum Zitat Ye YM, Nahm DH, Kim CW, Kim HR, Hong CS, Park CS, Suh CH, Park HS: Cytokeratin autoantibodies: useful serologic markers for toluene diisocyanate-induced asthma. Yonsei Med J 2006, 47: 773–81. 10.3349/ymj.2006.47.6.773PubMedCentralCrossRefPubMed Ye YM, Nahm DH, Kim CW, Kim HR, Hong CS, Park CS, Suh CH, Park HS: Cytokeratin autoantibodies: useful serologic markers for toluene diisocyanate-induced asthma. Yonsei Med J 2006, 47: 773–81. 10.3349/ymj.2006.47.6.773PubMedCentralCrossRefPubMed
65.
Zurück zum Zitat Arlian LG, Morgan MS, Vyszenski-Moher DL, Sharra D: Cross-reactivity between storage and dust mites and between mites and shrimp. Exp Appl Acarol 2009, 47: 159–72. 10.1007/s10493-008-9199-xCrossRefPubMed Arlian LG, Morgan MS, Vyszenski-Moher DL, Sharra D: Cross-reactivity between storage and dust mites and between mites and shrimp. Exp Appl Acarol 2009, 47: 159–72. 10.1007/s10493-008-9199-xCrossRefPubMed
66.
Zurück zum Zitat Reese G, Ayuso R, Lehrer SB: Tropomyosin: an invertebrate pan-allergen. Int Arch Allergy Immunol 1999, 119: 247–58. 10.1159/000024201CrossRefPubMed Reese G, Ayuso R, Lehrer SB: Tropomyosin: an invertebrate pan-allergen. Int Arch Allergy Immunol 1999, 119: 247–58. 10.1159/000024201CrossRefPubMed
Metadaten
Titel
Immune sensitization to methylene diphenyl diisocyanate (MDI) resulting from skin exposure: albumin as a carrier protein connecting skin exposure to subsequent respiratory responses
verfasst von
Adam V Wisnewski
Lan Xu
Eve Robinson
Jian Liu
Carrie A Redlich
Christina A Herrick
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
Journal of Occupational Medicine and Toxicology / Ausgabe 1/2011
Elektronische ISSN: 1745-6673
DOI
https://doi.org/10.1186/1745-6673-6-6

Weitere Artikel der Ausgabe 1/2011

Journal of Occupational Medicine and Toxicology 1/2011 Zur Ausgabe

Neu im Fachgebiet Arbeitsmedizin

Das Geschlechterparadoxon in der gesundheitlich beeinträchtigten Lebenszeit – Ende eines Mythos?

Beginnend mit den 1920er-Jahren hat sich eine Vorstellung über die Geschlechterdifferenzen in Gesundheit und Mortalität etabliert, die von Lorber und Moore in dem einprägsamen Satz: „Women get sicker, but men die quicker“, zusammengefasst wurde [ 1 …, S. 13]. Tatsächlich erscheinen vor dem Hintergrund der höheren Lebenserwartung der Frauen die Studienergebnisse zu den Geschlechterdifferenzen in der Morbidität überraschend, wonach Frauen im Durchschnitt einen schlechteren Gesundheitszustand aufweisen als Männer [

Gesunde Lebenserwartung: Ein kritischer Blick auf Nutzen und Potenziale des demographischen Gesundheitsindikators

Open Access Leitthema

Die demographische Alterung hat vielfältige gesellschaftliche Konsequenzen, deren Ausmaß wesentlich vom Gesundheitszustand der Bevölkerung abhängt. Um diesen analysieren und bewerten zu können, wurden spezielle Kennziffern entwickelt, die in …

Wie hat sich die Lebenserwartung ohne funktionelle Einschränkungen in Deutschland entwickelt? Eine Analyse mit Daten des Deutschen Alterssurveys (DEAS)

Deutschland erfährt, wie andere Hocheinkommensstaaten, aufgrund kontinuierlich rückläufiger Mortalität und niedriger Geburtenraten tiefgreifende demografische Veränderungen. Der demografische Wandel führt in Deutschland zu einem zunehmend höheren …

Hitzeschutz im Fokus der hessischen Betreuungs- und Pflegeaufsicht

Open Access Klimawandel Übersichtsartikel

Im Sommer 2023 kündigte das Bundesministerium für Gesundheit (BMG) einen nationalen Hitzeschutzplan an und forderte die Länder auf, zu prüfen, „ob die Warnstufen des [Deutschen Wetterdienstes] DWD mit der Durchführung von Akutmaßnahmen …