Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol Appl Pharmacol ; 288(3): 439-52, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26341290

ABSTRACT

Subchronic and chronic studies in rats of the gasoline oxygenates ethyl tert-butyl ether (ETBE) and tert-butanol (TBA) report similar noncancer kidney and liver effects but differing results with respect to kidney and liver tumors. Because TBA is a major metabolite of ETBE, it is possible that TBA is the active toxic moiety in all these studies, with reported differences due simply to differences in the internal dose. To test this hypothesis, a physiologically-based pharmacokinetic (PBPK) model was developed for ETBE and TBA to calculate internal dosimetrics of TBA following either TBA or ETBE exposure. This model, based on earlier PBPK models of methyl tert-butyl ether (MTBE), was used to evaluate whether kidney and liver effects are consistent across routes of exposure, as well as between ETBE and TBA studies, on the basis of estimated internal dose. The results demonstrate that noncancer kidney effects, including kidney weight changes, urothelial hyperplasia, and chronic progressive nephropathy (CPN), yielded consistent dose-response relationships across routes of exposure and across ETBE and TBA studies using TBA blood concentration as the dose metric. Relative liver weights were also consistent across studies on the basis of TBA metabolism, which is proportional to TBA liver concentrations. However, kidney and liver tumors were not consistent using any dose metric. These results support the hypothesis that TBA mediates the noncancer kidney and liver effects following ETBE administration; however, additional factors besides internal dose are necessary to explain the induction of liver and kidney tumors.


Subject(s)
Ethyl Ethers/toxicity , Kidney/drug effects , Liver/drug effects , tert-Butyl Alcohol/toxicity , Animals , Dose-Response Relationship, Drug , Female , Gasoline/toxicity , Hyperplasia/etiology , Hyperplasia/pathology , Inhalation Exposure , Male , Models, Biological , Organ Size/drug effects , Rats , Urothelium/drug effects , Urothelium/pathology , tert-Butyl Alcohol/blood
2.
J Immunotoxicol ; 10(4): 329-33, 2013.
Article in English | MEDLINE | ID: mdl-23256773

ABSTRACT

The prevalence of anti-nuclear antibodies (ANA) and self-reported systemic autoimmune diseases were increased in residents of Libby, MT, as was the incidence of ANA in Lewis rats exposed to Libby amphibole (LA) asbestos. However, rats induced to develop rheumatoid arthritis (RA) did not develop autoantibodies associated with RA, nor was RA exacerbated by LA exposure, suggesting that increased ANA expression might be related to some other autoimmune process. Libby residents self-reported increased numbers of physician-diagnosed cases of systemic lupus erythematosus (SLE). Thus, the goal of this study was to determine if the increased incidence of ANA in Lewis rats exposed to LA is related to the development of SLE-like disease. Female Lewis rats were intratracheally instilled bi-weekly for 13 weeks with total doses of 0.15, 0.5, 1.5, or 5.0 mg of LA or 0.5 or 1.5 mg of a positive control fiber, amosite. ANA incidence was significantly increased in all asbestos dose groups, although no dose response was observed. The occurrence of proteinuria was increased in LA 0.5, LA 5.0, and amosite 0.5 dose groups; however, the microscopic appearance of the kidneys was normal, no binding of autoimmune complexes to glomerular surfaces was observed, and antibodies to double-stranded DNA were not elevated. Therefore, an increased prevalence of ANA in rats exposed to asbestos does not appear to correlate with disease markers typically observed in SLE. Analysis of ANA specificity for extractable nuclear antigens (ENA) determined that 98% of ENA(+) samples were specific for the Jo-1 antigen. Autoantibodies to Jo-1 have been reported in patients with interstitial lung disease, suggesting that autoantibodies to Jo-1 may be a biomarker for asbestos-related pulmonary disease.


Subject(s)
Asbestos, Amosite/toxicity , Asbestos, Amphibole/toxicity , Kidney/pathology , Lupus Erythematosus, Systemic/epidemiology , Proteinuria/epidemiology , Animals , Antibodies, Antinuclear/blood , Biomarkers/blood , Environmental Exposure/adverse effects , Female , Histidine-tRNA Ligase/immunology , Humans , Incidence , Intubation, Intratracheal , Lupus Erythematosus, Systemic/immunology , Montana , Prevalence , Proteinuria/immunology , Rats , Rats, Inbred Lew
3.
J Toxicol Environ Health A ; 75(6): 351-65, 2012.
Article in English | MEDLINE | ID: mdl-22480172

ABSTRACT

Epidemiological data suggest that occupational exposure to the amphibole-containing vermiculite in Libby, MT, was associated with increased risk for developing autoimmune diseases and had an odds ratio of 3.23 for developing rheumatoid arthritis (RA). The collagen-induced arthritis (CIA) and the peptidoglycan-polysaccharide (PG-PS) models of RA were employed to determine whether exposure to Libby amphibole (LA) induced a more rapid onset, increased expression, or prolonged course of RA. Female Lewis rats were intratracheally instilled with total doses of 0.15, 0.5, 1.5, or 5 mg LA or 0.5 or 1.5 mg amosite asbestos, and arthritis was induced with either the PG-PS or CIA model. Neither LA nor amosite exposure affected the disease course in the CIA model, or the production of rheumatoid factor (RF) or anti-cyclic citrullinated peptide (CCP) antibodies. LA exposure reduced swelling in the PG-PS model and decreased anti-PG-PS and total immunoglobulin M (IgM) antibody titers. Both amosite and LA exposure increased the number of rats with circulating anti-nuclear antibodies (ANA), the majority of which presented a speckled staining pattern. However, this ANA enhancement was not dose responsive. These results failed to show a positive correlation between LA exposure and RA disease in two animal models, although upregulated ANA suggest an altered immunological profile consistent with other systemic autoimmune diseases.


Subject(s)
Arthritis, Experimental/chemically induced , Arthritis, Rheumatoid/chemically induced , Asbestos, Amphibole/toxicity , Aluminum Silicates/chemistry , Animals , Antibodies/blood , Asbestos, Amphibole/administration & dosage , Biomarkers , Dose-Response Relationship, Drug , Female , Immunoglobulin G/blood , Injections, Spinal , Joints/pathology , Lung Diseases/chemically induced , Lung Diseases/pathology , Rats , Rats, Inbred Lew , Time Factors
4.
Proc Am Thorac Soc ; 7(2): 130-3, 2010 May.
Article in English | MEDLINE | ID: mdl-20427585

ABSTRACT

Mesenchymal stem cells (MSCs) have been shown to differentiate into a variety of mesenchymal cell types, including fibroblasts, myofibroblasts, osteoblasts, chondroblasts, adipocytes, and myoblasts, as well as epithelial cells. It has been shown that these cells can be recovered from bone marrow as well as umbilical cord blood, and they can be propagated, stored, and administered to animals and patients in clinical trials. It is clear that the cells engraft in the lung, and several laboratories have demonstrated an ameliorating effect in models of acute injury caused by LPS and in chronic lung injury induced by bleomycin and asbestos. However, it is not at all clear under what conditions these cells must be applied to provide an advantage and when using these cells might cause exacerbation of the lung injury. This brief review focuses on the biology of MSCs in vitro, how the cells have been used in some animal models, and the potential for their use in therapeutic strategies for diseases as diverse as lung cancer and interstitial fibrosis.


Subject(s)
Lung Injury/etiology , Lung Injury/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Animals , Cell Differentiation , Disease Models, Animal , Humans , Lung Injury/pathology , Mesenchymal Stem Cells/cytology
5.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L1002-11, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19734317

ABSTRACT

Studies have been carried out previously to determine whether mesenchymal stem cells (MSC) influence the progression of pulmonary fibrosis. Here, we asked whether MSC (derived from mouse bone marrow and human umbilical cord blood) produce factors that mediate lung fibroblast (LF) growth and matrix production. MSC-conditioned media (CM) were found by ELISA to contain significant amounts of PDGF-AA and transforming growth factor-beta1 (TGF-beta1). Proliferation was increased in a concentration-dependent manner in LF cell lines and primary cells cultured in MSC-CM, but neither anti-PDGF antibodies nor PDGF receptor-specific antibodies affected proliferation, nor did a number of other antibodies to well-known mitogenic factors. However, proliferation was significantly inhibited by the Wnt signaling antagonist, secreted frizzled related protein-1 (sFRP-1). In addition, anti-Wnt1 and anti-Wnt2 antibodies attenuated MSC-CM-induced proliferation, and increased expression of Wnt7b was identified. As would be expected in cells activated by Wnt, nuclear beta-catenin was increased. The amount of TGF-beta1 in MSC-CM and its biological activity were revealed by activation at acidic pH. The stem cells synthesized and released TGF-beta1 that increased alpha1-procollagen gene expression by LF target cells. Addition of anti-TGF-beta to the MSC-CM blocked upregulation of collagen gene expression. These data demonstrate that MSC from mice and humans produce Wnt proteins and TGF-beta1 that respectively stimulate LF proliferation and matrix production, two hallmarks of fibroproliferative lung disease. It will be essential to determine whether these factors can play a role in attempts to use MSC for therapeutic approaches.


Subject(s)
Collagen Type I/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Lung/cytology , Mesenchymal Stem Cells/metabolism , Transforming Growth Factor beta1/metabolism , Wnt Proteins/metabolism , Animals , Antibodies/pharmacology , Bone Marrow Cells/cytology , Cell Proliferation/drug effects , Coculture Techniques , Collagen Type I/genetics , Culture Media, Conditioned/pharmacology , Fetal Blood/cytology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Platelet-Derived Growth Factor/metabolism , Polymerase Chain Reaction , Protein Isoforms/metabolism , Proteins/pharmacology , Solubility/drug effects , Transforming Growth Factor beta1/antagonists & inhibitors , beta Catenin/metabolism
6.
J Toxicol Environ Health B Crit Rev ; 11(8): 630-45, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18821422

ABSTRACT

The pesticide 3,4-dichloropropionanilide (propanil or, alternatively, DCPA) is a member of the acetanilide chemical family and is predominantly used for the control of weeds on commercial rice crops worldwide. This article was written to provide a brief review of the general toxicity of propanil followed by a detailed summary of the immunotoxicity studies that were performed to date in mammalian in vivo and in vitro models. Propanil affects the immune system at organ, cellular, and molecular levels. Studies demonstrated that it produces thymic atrophy and splenomegaly and decreases developing T- and B-cell populations in the thymus and bone marrow. Natural killer (NK) cells and macrophages are critical components of the innate immune system. NK cell cytotoxicity and the ability of macrophages to phagocytose, kill pathogenic bacteria, and produce inflammatory cytokines are suppressed by propanil. Propanil also affects the respiratory burst of macrophages, inhibiting reactive oxygen and nitrogen species production. Molecular mechanisms responsible for propanil's effects have begun to be elucidated and include alterations in nuclear factor (NF)-kappaB transcription factor activity and intracellular Ca(2+) signaling. Propanil exposure alters a number of functions of mature T lymphocytes and B lymphocytes that impacts the adaptive immune response. T-cell cytotoxic activity and cytokine production are major T-cell functions inhibited by propanil. The humoral antibody response to model antigens and intact bacteria is differentially affected after propanil exposure. How these changes in innate and adaptive immune responses impact the host response to bacterial challenge or vaccination has begun to be examined.


Subject(s)
Anilides/toxicity , Herbicides/toxicity , Immunity/drug effects , Agriculture , Anilides/immunology , Animals , Environmental Exposure , Humans , Molecular Structure , Occupational Exposure
7.
Toxicol Sci ; 93(1): 62-74, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16788000

ABSTRACT

Steroid hormones are known to affect the humoral immune response to a variety of antigens. However, the mechanisms regulating these effects are poorly understood. The immunotoxic chemical propanil and estrogen have similar effects on the immune system including augmentation of humoral immune responses. Propanil enhances the number of phosphorylcholine (PC)-specific IgG2b, IgG3, and IgM antibody-secreting cells (ASCs) in the spleen four- to sixfold 7 days after vaccination of female C57BL/6 mice with heat-killed Streptococcus pneumoniae. Several experiments were performed to test the hypothesis that propanil increases the response via an estrogenic pathway. Ovariectomy abrogated the effect of propanil on the PC-specific ASC response. Both in vitro and in vivo assays indicate that propanil does not bind either estrogen receptor (ER) alpha or beta. Exogenous estradiol administration in ovariectomized mice failed to restore the effect of propanil on the PC response. Treatment of female mice with a pure ER antagonist, ICI 182,780, or the progesterone antagonist RU486 did not inhibit the increase in ASCs. These data suggest that estrogen and progesterone do not regulate the effect of propanil. However, complete inhibition of steroid synthesis with the gonadotropin-releasing hormone (GnRH) antagonist antide abrogated the increased response in propanil-treated mice, indicating a necessary role for steroid synthesis. Experiments in male mice demonstrated that propanil increased the number of ASCs comparable to female mice. However, orchiectomy did not inhibit this effect, suggesting that androgens do not regulate the amplification of the humoral response. These data suggest a novel role for the ovarian hormones in the regulation of the PC-specific antibody response.


Subject(s)
Antibody Formation/drug effects , Endocrine Disruptors/toxicity , Ovary/drug effects , Pesticides/toxicity , Propanil/toxicity , Steroids/biosynthesis , Animals , Estradiol/blood , Female , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Ovary/physiology , Receptors, Estrogen/metabolism
8.
Toxicol Sci ; 87(1): 123-33, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15976183

ABSTRACT

Propanil (3,4-dichloropropionanilide) and 2,4-D (2,4-dichlorophenoxyacetic acid) are commonly used herbicides that have toxic effects on the immune system. The present study determined the effect of exposure to these chemicals on the immune response to a bacterial vaccine. The antibody responses to the T-independent type 2 antigen, phosphorylcholine (PC) and the T-dependent antigen, pneumococcal surface protein A (PspA) were characterized in C57BL/6 mice after heat-killed Streptococcus pneumoniae (HKSP) immunization and single or mixture herbicide exposure. Propanil exposure significantly increased the number of PC-specific IgM, IgG2b, and IgG3 antibody-secreting B cells (ASC) in the spleen 4-6-fold over control animals in a dose-dependent manner. However, the number of ASC in the bone marrow and serum titers were comparable in control and propanil-treated mice. In contrast, 2,4-D exposure decreased the number of PC-specific IgM and IgG bone marrow ASC 2-3-fold from control animals. The decrease in bone marrow ASC in 2,4-D-treated mice corresponded to a 3-4-fold decrease in PC-specific IgM, IgG2b, and IgG3 serum titers compared to control mice. The number of ASC in the spleens of 2,4-D-treated mice was, however, comparable to control mice. The antibody response to PspA was not affected by any of the treatments. There were no mixture interactions between the two herbicides in any of the responses measured. These results characterize the primary PC-specific antibody response in the bone marrow, spleen, and serum after HKSP vaccination and herbicide exposure. The differential effects of propanil and 2,4-D on the antibody response to a bacterial vaccine demonstrate the potential of chemical exposure to augment or suppress immune responses to vaccines and infectious diseases.


Subject(s)
2,4-Dichlorophenoxyacetic Acid/toxicity , Antibodies, Bacterial/blood , Herbicides/toxicity , Pneumococcal Vaccines/immunology , Propanil/toxicity , Streptococcus pneumoniae/immunology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bacterial Proteins/immunology , Female , Mice , Mice, Inbred C57BL , Phosphorylcholine/immunology , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...