Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Autoimmun ; 97: 59-69, 2019 02.
Article in English | MEDLINE | ID: mdl-30416032

ABSTRACT

Female sex is a risk factor for lupus. Sex hormones, sex chromosomes and hormone receptors are implicated in the pathogenic pathways in lupus. Estrogen receptor alpha (ERα) knockout (KO) mice are used for defining hormone receptor effects in lupus. Prior studies of ERα KO in lupus have conflicting results, likely due to sex hormone levels, different lupus strains and different ERα KO constructs. Our objective was to compare a complete KO of ERα vs. the original functional KO of ERα (expressing a short ERα) on disease expression and immune phenotype, while controlling sex hormone levels. We studied female lupus prone NZM2410 WT and ERα mutant mice. All mice (n = 44) were ovariectomized (OVX) for hormonal control. Groups of each genotype were estrogen (E2)-repleted after OVX. We found that OVXed NZM mice expressing the truncated ERα (ERα short) had significantly reduced nephritis and prolonged survival compared to both wildtype and the complete ERαKO (ERα null) mice, but surprisingly only if E2-repleted. ERα null mice were not protected regardless of E2 status. We observed significant differences in splenic B cells and dendritic cells and a decrease in cDC2 (CD11b+CD8-) dendritic cells, without a concomitant decrease in cDC1 (CD11b-CD8a+) cells comparing ERα short to ERα null or WT mice. Our data support a protective role for the ERα short protein. ERα short is similar to an endogenously expressed ERα variant (ERα46). Modulating its expression/activity represents a potential approach for treating female-predominant autoimmune diseases.


Subject(s)
Disease Susceptibility , Estrogen Receptor alpha/genetics , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Animals , Autoimmunity/genetics , Biomarkers , Biopsy , Complement C3/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Immunoglobulin G/immunology , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/etiology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Mice , Mice, Knockout , Proteinuria/etiology , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Survival Rate
2.
Clin Immunol ; 183: 132-141, 2017 10.
Article in English | MEDLINE | ID: mdl-28822833

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic and potentially severe autoimmune disease that disproportionately affects women. Despite a known role for hormonal factors impacting autoimmunity and disease pathogenesis, the specific mechanisms of action remain poorly understood. Our laboratory previously backcrossed "estrogen receptor alpha knockout (ERαKO)" mice onto the NZM2410 lupus prone background to generate NZM/ERαKO mice. This original ERαKO mouse, developed in the mid-1990s and utilized in hundreds of published studies, is not in fact ERα null. They express an N-terminally truncated ERα, and are considered a functional KO. They have physiologic deficiencies including infertility due to disruption of a critical activation domain (AF-1) at the N terminus of ERα, required for most classic estrogen (E2) actions. We demonstrated that female NZM/ERαKO mice had significantly less renal disease and significantly prolonged survival compared to WT littermates despite similar serum levels of autoantibodies and glomerular immune complex deposition. Herein, we present results of experiments using a lupus prone true ERα-/- mice (deletional KO mice on the NZM2410 background), surprisingly finding that these animals were not protected if they were ovariectomized, suggesting that another hormonal component confers protection, possibly testosterone, rather than the absence of the full-length ERα.


Subject(s)
Antibodies, Antinuclear/immunology , Estrogen Receptor alpha/genetics , Lupus Erythematosus, Systemic/genetics , Animals , Antibody Formation , Autoantibodies/immunology , DNA/immunology , Estradiol/blood , Estrogen Receptor alpha/immunology , Female , Genetic Predisposition to Disease , Kidney/pathology , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Knockout , Ovariectomy , Testosterone/blood
3.
Clin Immunol ; 175: 109-114, 2017 02.
Article in English | MEDLINE | ID: mdl-28041989

ABSTRACT

Plasmacytoid dendritic cells (pDCs) and their production of type I interferons (IFN) are key pathogenic mediators of systemic lupus erythematosus (SLE). Despite the key role of pDCs in SLE, the mechanism by which pDCs promote disease is not well understood. The first objective for this study was to assess the number and maturation state of pDCs in pre-disease NZM2410 lupus prone mice compared to control mice. Second, we sought to identify mechanisms responsible for the alteration in pDCs in NZM mice prior to onset of clinical disease. We compared the number and percent of pDCs in the spleens and bone marrow (BM) of pre-disease NZM24010 (NZM) mice to C57BL/6 (B6) control mice. In the spleens of pre-disease NZM mice, pDC percent and number were increased. This increase occurs in parallel with a decrease in BM pDC number and percent in the NZM mice. The decrease in BM pDC number suggests the increase in spleen pDCs is a result of altered pDC distribution and not increased production of pDCs in the BM. To determine if pDC developmental potential is altered in lupus prone mice, we cultured BM from NZM and B6 mice in vitro. We found a reduced percentage/number of pDCs developing from the BM of NZM mice compared to B6 mice, which further supports that the increase in pDC number is a result of altered pDC distribution rather than increased pDC production. To better characterize the pDC population, we compared the percentage of mature pDCs in the spleens and BM of NZM mice to controls. In the NZM mice, there is a dramatic reduction in the number of mature pDCs in the BM of NZM mice, suggesting that mature pDCs exit the BM at a higher rate/earlier maturation time compared to healthy mice. We conclude that pDCs contribution to disease pathogenesis in NZM mice may include the alteration of pDC distribution to increase the number of pDCs in the spleen prior to disease onset.


Subject(s)
Dendritic Cells/pathology , Lupus Erythematosus, Systemic/pathology , Animals , Dendritic Cells/metabolism , Female , Interferon Type I/metabolism , Lupus Erythematosus, Systemic/metabolism , Mice , Mice, Inbred C57BL , Spleen/metabolism
4.
Front Immunol ; 7: 31, 2016.
Article in English | MEDLINE | ID: mdl-26913030

ABSTRACT

Ninety percent of those diagnosed with systemic lupus erythematosus are female, with peak incidence between the ages of 15 and 45, when women are most hormonally active. Despite significant research effort, the mechanisms underlying this sex bias remain unclear. We previously showed that a functional knockout of estrogen receptor alpha (ERα) resulted in significantly reduced renal disease and increased survival in murine lupus. Dendritic cell (DC) development, which requires both estrogen and ERα, is impacted, as is activation status and cytokine production. Since both estrogen and testosterone levels have immunomodulating effects, we presently studied the phenotype of NZM2410 lupus-prone mice following post- and prepubertal ovariectomy (OVX) ± estradiol (E2) replacement to determine the impact of hormonal status on disease expression and DC development in these mice. We observed a trend toward survival benefit in addition to decreased proteinuria and improved renal histology in the early OVX, but not late OVX- or E2-repleted WT mice. Interestingly, there was also a significant difference in splenic DC subsets by flow cytometry. Spleens from NZM mice OVX'd early had a significant decrease in proinflammatory CD11c+CD11b+ DCs (vs. unmanipulated WTs, late OVX- and E2-repleted mice). These early OVX'd animals also had a significant increase in tolerogenic CD11c+CD8a+ DCs vs. WT. These data join a growing body of evidence that supports a role for hormone modulation of DCs that likely impacts the penetrance and severity of autoimmune diseases, such as lupus.

5.
J Immunol ; 195(12): 5561-71, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26553076

ABSTRACT

Female lupus-prone NZM2410 estrogen receptor α (ERα)-deficient mice are protected from renal disease and have prolonged survival compared with wild-type littermates; however, the mechanism of protection is unknown. Plasmacytoid dendritic cells (pDCs) and type I IFN drive lupus pathogenesis. Estrogen acting via ERα enhances both pDC development and IFN production. The objectives for this study were to determine if ERα modulates pDC function and IFN activity in predisease NZM2410 mice as a possible protective mechanism of ERα deficiency in lupus-prone mice. We measured the effect of ERα deficiency on spleen pDC frequency, number, maturation, and activation state. ERα deficiency reduced type I IFN activity and the frequency of MHC class II(+) pDCs in the spleen without altering overall pDC frequency, number, or maturation state. Additionally, ERα-deficient NZM2410 mice had a significantly decreased frequency of pDCs expressing PDC-TREM, a modulator of TLR-mediated IFN production. After in vitro TLR9 stimulation, ERα deficiency significantly reduced the expression of PDC-TREM on pDCs from both NZM2410 and C57BL/6 mice. Thus, we have identified a significant effect of ERα deficiency on pDCs in predisease NZM2410 mice, which may represent a mechanism by which ERα deficiency protects NZM2410 mice from lupuslike disease.


Subject(s)
Dendritic Cells/immunology , Estrogen Receptor alpha/deficiency , Kidney/physiopathology , Lupus Erythematosus, Systemic/immunology , Receptors, Cell Surface/metabolism , Animals , Cells, Cultured , Dendritic Cells/drug effects , Estrogens/pharmacology , Female , Gene Expression Regulation/drug effects , Immunomodulation , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/genetics , Toll-Like Receptor 9/metabolism
6.
SOJ Immunol ; 2(1)2014 Jan 20.
Article in English | MEDLINE | ID: mdl-25061615

ABSTRACT

We previously found that a maximum innate inflammatory response induced by stimulation of Toll-like receptors (TLRs) 3, 7 and 9 requires ERα, but does not require estrogen in multiple cell types from both control and lupus-prone mice. Given the estrogen-independence, we hypothesized that ERα mediates TLR signaling by tethering to, and enhancing, the activity of downstream transcription factors such as NFκB, rather than acting classically by binding EREs on target genes. To investigate the mechanism of ERα impact on TLR signaling, we utilized mice with a knock-in ERα mutant that is unable to bind ERE. After stimulation with TLR ligands, both ex vivo spleen cells and bone marrow-derived dendritic cells (BM-DCs) isolated from mutant ERα ("KIKO") mice produced significantly less IL-6 compared with cells from wild-type (WT) littermates. These results suggest that ERα modulation of TLR signaling does indeed require ERE binding for its effect on the innate immune response.

7.
Rheumatol Int ; 33(10): 2677-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23011090

ABSTRACT

Sphingolipids are components of the plasma membrane whose metabolic manipulation is of interest as a potential therapeutic approach in a number of diseases. Sphingosine kinase 1 (SphK1), the major kinase that phosphorylates sphingosine to sphingosine-1-phosphate (S1P), was previously shown by our group and others to modulate inflammation in murine models of inflammatory arthritis, inflammatory bowel disease and asthma. Sphingosine kinase 2's (SphK2) impact on inflammation is less well known, as variable results were reported depending on the disease model. A specific SphK2 inhibitor inhibited inflammatory arthritis in one model, while siRNA knockdown of SphK2 worsened arthritis in another. We previously demonstrated that SphK1 deficient mice are protected against development of hTNF-α-induced arthritis. To investigate the role of SphK2 in TNF-α-induced arthritis, we developed SphK2 deficient hTNF-α overexpressing mice and separately treated hTNF-α mice with ABC294640, a SphK2-specific inhibitor. Our data show that genetic inhibition of SphK2 did not significantly impact the severity or progression of inflammatory arthritis, while pharmacologic inhibition of SphK2 led to significantly more severe arthritis. Compared to vehicle-treated mice, ABC294640 treated mice also had less S1P in whole blood and inflamed joint tissue, although the differences were not significant. ABC294640 treatment did not affect SphK1 activity in the inflamed joint while little SphK2 activity was detected in the joint. We conclude that the differences in the inflammatory phenotype in genetic inhibition versus pharmacologic inhibition of SphK2 can be attributed to the amount of ABC294640 used in the experiments versus the impact of acute inhibition of SphK2 with ABC294640 versus genetically induced life-long SphK2 deficiency. Thus, inhibition of SphK2 appears to be proinflammatory in contrast to the clear anti-inflammatory effects of blocking SphK1. Therapies directed at this sphingosine kinase pathways will need to be specific in their targeting of sphingosine kinases.


Subject(s)
Arthritis, Experimental/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Mice , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Tumor Necrosis Factor-alpha
8.
Clin Immunol ; 144(1): 1-12, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22659029

ABSTRACT

Systemic lupus erythematosus (SLE) is a disease that disproportionately affects females. Despite significant research effort, the mechanisms underlying the female predominance in this disease are largely unknown. Previously, we showed that estrogen receptor alpha knockout (ERαKO) lupus prone female mice had significantly less pathologic renal disease and proteinuria, and significantly prolonged survival. Since autoantibody levels and number and percentage of B/T cells were not significantly impacted by ERα genotype, we hypothesized that the primary benefit of ERα deficiency in lupus nephritis was via modulation of the innate immune response. Using BMDCs and spleen cells/B cells from female wild-type or ERαKO mice, we found that ERαKO-derived cells have a significantly reduced inflammatory response after stimulation with TLR agonists. Our results indicate that the inflammatory response to TLR ligands is significantly impacted by the presence of ERα despite the absence of estradiol, and may partially explain the protective effect of ERα deficiency in lupus-prone animals.


Subject(s)
Estrogen Receptor alpha/immunology , Lupus Erythematosus, Systemic/immunology , Toll-Like Receptors/immunology , Animals , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , CpG Islands/immunology , Cytokines/immunology , Dendritic Cells/immunology , Disease Models, Animal , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Female , Guanosine/analogs & derivatives , Guanosine/pharmacology , Mice , Mice, Knockout , Spleen/cytology , Toll-Like Receptors/agonists
9.
Results Immunol ; 2: 196-203, 2012.
Article in English | MEDLINE | ID: mdl-24371584

ABSTRACT

TLR2 agonists are well known for inducing NF-kB activation and inflammation, while estrogen receptor-alpha (ER-α) is a regulator of estrogen-mediated anti-inflammatory responses. In the present work, we determined the role of ER-α and phosphorylated ER-α in TLR2 agonist-induced MCP1 production in mesangial cells. We found that TLR2 agonists induced nuclear localization of phospho-ER-α (serine 118), and estrogen and TLR2 agonists both induced phosphorylation of ER-α at the serine 118 and 104/106 positions. Incubation of MRL/lpr mesangial cells with estrogen was found to attenuate TLR2 agonist-mediated MCP1 production. To determine the mode of action of ER-α/pER-α (serine-118), we used the ER-α inhibitor MPP and transfected mesangial cells with ER-α siRNA. ER-α inhibition was found to decrease MCP1 production in mesangial cells. Thus, ER-α/pER-α is an intermediate regulator for both TLR2-mediated MCP1 production during inflammation and estrogen-mediated anti-inflammatory signals in mesangial cells.

10.
Toxicol Sci ; 108(1): 110-23, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19141786

ABSTRACT

N,N-diethyl-meta-toluamide (DEET) is a particularly effective broad-spectrum insect repellent used commonly in recreational, occupational and military environments. Due to its widespread use and suggested link to Gulf War Illness, this study examined the immunotoxicity of DEET. Adult female B6C3F1 mice were injected sc for 14 days with DEET at 0, 7.7, 15.5, 31, or 62 mg/kg/day. Due to differences in the dermal absorption of DEET between mice and humans, this study eliminated this confounding factor by utilizing sc injection and measured circulating blood levels of DEET to assess bioavailability from sc administration. Effects on lymphocyte proliferation, natural killer cell activity, thymus and spleen weight and cellularity, the antibody plaque-forming cell (PFC) response, and thymic and splenic CD4/CD8 lymphocyte subpopulations were assessed 24 h after the last dose. No effect was observed in lymphocyte proliferation, natural killer cell activity, thymic weight, splenic weight, thymic cellularity, or splenic cellularity. Significant decreases were observed in the percentage of splenic CD4-/CD8- and CD4+/CD8- lymphocytes but only at the 62 mg DEET/kg/day treatment level and not in absolute numbers of these cells types. Additionally, significant decreases in the antibody PFC response were observed following treatment with 15.5, 31, or 62 mg DEET/kg/day. Pharmacokinetic (PK) data from the current study indicate 95% bioavailability of the administered dose. Therefore, it is likely that DEET exposure ranges applied in this study are comparable to currently reported occupational usage. Together, the evidence for immunosuppression and available PK data suggest a potential human health risk associated with DEET in the occupational or military environments assuming similar sensitivity between human and rodent responses.


Subject(s)
DEET/toxicity , Immune Tolerance/drug effects , Immunosuppressive Agents/toxicity , Insect Repellents/toxicity , Analysis of Variance , Animals , Antibody Formation/drug effects , B-Lymphocytes/immunology , Biological Availability , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , DEET/administration & dosage , DEET/pharmacokinetics , Female , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Inactivation, Metabolic , Injections, Intravenous , Injections, Subcutaneous , Insect Repellents/administration & dosage , Insect Repellents/pharmacokinetics , Killer Cells, Natural/immunology , Male , Mice , Models, Animal , Persian Gulf Syndrome , Spleen/cytology , Thymus Gland/cytology
11.
Article in English | MEDLINE | ID: mdl-18780217

ABSTRACT

A genetically-prone murine lupus model was used to assess the developmental effects of trichloroethylene (TCE) exposure on disease symptom onset (e.g., autoantibody production and proteinuria), lymphocyte proliferation, splenic B-cell populations, and thymic and splenic T-cell populations. MRL +/+ mice were exposed to TCE (0, 1,400 or 14,000 ppb) via drinking water beginning on gestation day (GD) 0 and continuing until 12 months of age. With the exception of splenic CD4-/CD8-cells in female mice only, no alterations were observed in splenic T-cell populations, numbers of splenic B220+ cells, or in lymphocyte proliferation at 12 months of age. Furthermore, populations of all thymic T-cell subpopulations were decreased in male but not female mice following exposure to 14,000 ppb TCE. Autoantibody levels (anti-dsDNA and anti-GA) were assessed periodically from 4 to 12 months of age. Over this period, no increase in autoantibody levels as compared to control was detected, suggesting that TCE did not contribute to or accelerate the development of autoimmune disease markers following lifetime exposure. Not only does this study offer encouraging results, but it is the first study to approach the development of autoimmunity in a novel lifetime exposure paradigm, using an autoimmune prone model, at environmentally relevant exposure levels.


Subject(s)
Autoimmune Diseases/chemically induced , Trichloroethylene/toxicity , Animals , Autoantibodies/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Cell Proliferation/drug effects , Female , Genotype , Immunophenotyping , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred MRL lpr , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
12.
Clin Immunol ; 128(2): 259-68, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18514033

ABSTRACT

Systemic lupus erythematosus (SLE) occurs nine times more often in females than males. The purpose of this study was to determine the impact of estrogen receptor (ER) null genotypes on disease in lupus prone NZM2410 (NZM) and MRL/lpr mice, as a method to define the role of estrogen receptor signaling in lupus. ER alpha deficient NZM females, but not males, had significantly prolonged survival, reduced proteinuria, renal pathology scores and serum urea nitrogen levels compared to wildtype mice, despite higher serum anti-dsDNA levels. ER alpha deficient MRL/lpr female, but not male, mice also had significantly less proteinuria and renal pathology scores with no effect on autoantibody levels. Deficiency of ER beta had no effect on disease in either strain or sex. Taken together, these data demonstrate a key role for ER alpha, but not ER beta, in the development of lupus like disease, but not autoimmunity, in female NZM and MRL/lpr mice.


Subject(s)
Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Lupus Erythematosus, Systemic/physiopathology , Signal Transduction , Animals , Autoantibodies/blood , Blood Urea Nitrogen , Estrogen Receptor alpha/deficiency , Estrogen Receptor beta/deficiency , Female , Kidney/pathology , Lupus Erythematosus, Systemic/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteinuria/pathology
13.
Toxicol Sci ; 104(1): 144-54, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18359764

ABSTRACT

Adult male and female B6C3F1 mice were exposed to perfluorooctane sulfonate (PFOS) daily via gavage for 28 days (0, 0.005, 0.05, 0.1, 0.5, 1, or 5 mg/kg total administered dose [TAD]). Following exposure, various immune parameters were assessed and serum PFOS concentrations were determined. Lymphocyte proliferation was not altered in either gender. Natural killer cell activity was increased compared with control at 0.5, 1, and 5 mg/kg TAD in male mice but was not altered in female mice. At these treatment levels, splenic T-cell immunophenotypes were minimally altered in females, but all T-cell subpopulations were significantly modulated in males beginning at 0.1 mg/kg TAD. The sheep red blood cell (SRBC) plaque-forming cell (PFC) response was suppressed in male mice beginning at 0.05 mg/kg TAD and in females at 0.5 mg/kg TAD. Serum trinitrophenyl (TNP)-specific IgM titers were also decreased by PFOS after TNP-LPS (TNP conjugated to lipopolysacharide) challenge suggesting that the humoral immune effects may be attributed to the B-cell rather than T-cell because both T-dependent (SRBC) and T-independent (TI) (TNP-LPS) antigens result in suppressed IgM production. Based on the PFC response, the low observed effect level (LOEL) for males was 0.05 mg/kg TAD (ED(50) = 0.021 mg/kg TAD) and for females was 0.5 mg/kg TAD (ED(50) = 0.59 mg/kg TAD). Measured PFOS serum concentrations at these dose levels were 91.5 +/- 22.2 ng/g and 666 +/- 108 ng/g (mean +/- SD), respectively. The male LOEL serum level was approximately 14-fold lower than reported mean blood levels from occupationally exposed humans and fell in the upper range of concentrations reported for the general population. Overall, this study provides a profile of PFOS immunotoxicity showing effects at levels reported in humans and identifies the B-cells as a potential target.


Subject(s)
Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Lymphocytes/drug effects , Alkanesulfonic Acids/blood , Alkanesulfonic Acids/pharmacokinetics , Animals , Antibody Formation/drug effects , Cell Proliferation/drug effects , Erythrocytes/immunology , Female , Fluorocarbons/blood , Fluorocarbons/pharmacokinetics , Hemolytic Plaque Technique , Immunoglobulin M/blood , Immunoglobulin M/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Male , Mice , Mice, Inbred Strains , Muramidase/blood , No-Observed-Adverse-Effect Level , Spleen/drug effects , Spleen/growth & development , Spleen/immunology , Trinitrobenzenes/immunology
14.
J Toxicol Environ Health A ; 70(13): 1130-41, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17558808

ABSTRACT

Perfluorinated hydrocarbons have been manufactured for over 40 yr and have numerous applications in industry. This group of compounds has recently generated much interest, as some of these compounds such as perfluorooctane sulfonate (PFOS) and perfluoroctanic acid (PFOA) are persistent in the environment and detectable in blood samples of both wildlife and humans. Studies show that these perfluorinated compounds induce peroxisomal proliferation, induce hepatomegaly, alter steroidogenesis, and decrease body weight, accompanied by a wasting syndrome; however, effects on immune function have not been addressed at length. This study examined sulfluramid, a perfluorinated pesticide that is currently available in the marketplace and is a representative member of this class of chemicals. Adult female B6C3F1 mice were exposed via gavage to either an oil carrier control or sulfluramid for 14 d (1, 3, 10, or 30 mg/kg/d) or 28 d (0.3, 1, 3, or 10 mg/kg/d). Although responses were normal in natural killer cell activity and lymphocyte proliferation, dose-responsive suppression was noted in the plaque forming cell (PFC) response at exposure levels as low as 3 mg/kg/d in the 14-d exposure and 0.3 mg/kg/d for 28 d. Dose-responsive increases in liver mass were observed following treatment with 1, 3, 10, or 30 mg/kg/d for 14 d and 0.3, 1, 3, or 10 mg/kg/d for 28 d. A significant reduction in body weight was observed at the highest dose level in each study. Novel findings in this study indicate that sulfluramid suppresses immunoglobulin (Ig) M production. Additional immunotoxicity studies are required to understand potential mechanisms of suppression and determine potential health risks associated with exposure to perfluorinated hydrocarbons.


Subject(s)
Antibody Formation/drug effects , Fluorocarbons/toxicity , Insecticides/toxicity , Sulfonamides/toxicity , Animals , Dose-Response Relationship, Drug , Endocrine System/drug effects , Female , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Liver/drug effects , Liver/pathology , Lymphocytes/drug effects , Lymphocytes/immunology , Mice
15.
Article in English | MEDLINE | ID: mdl-16484062

ABSTRACT

This study assessed the developmental immunotoxicity of trichloroethylene (TCE) in B6C3F1 mice exposed via drinking water (0, 1,400, 14,000 ppb) from gestation day 0 (GD0) to either 3 or 8 weeks of age. Lymphocyte proliferation, NK cell activity, SRBC-specific IgM production (PFC response), splenic B220+ cells, and thymic and splenic T-cell immunophenotypes were assessed at 3 and 8 weeks of age. Delayed type hypersensitivity (DTH) and autoantibodies to ds-DNA were assessed in 8-week old animals only. Proliferation and NK cell activity were not affected at either age. Decreased PFC responses were noted in male offspring at both ages and both TCE treatment levels. PFC responses in female offspring were suppressed by treatment with 14,000 ppb TCE at both ages assessed and at 1,400 ppb TCE at 8 weeks of age. Splenic numbers of B220 cells were only decreased in 3-week old pups exposed to 14,000 ppb TCE. The most pronounced alteration in T-cell subpopulations were increases in all thymic (CD4+, CD8+, CD4+/CD8+, and CD4-/CD8-) T-cell types in 8-week old animals. DTH was increased in females at both TCE levels and in males at the high dose only. These results indicate that TCE may be an effective developmental immunotoxicant and suggests that additional studies are required to determine the health risks associated with developmental exposure to TCE.


Subject(s)
Cell Proliferation/drug effects , Killer Cells, Natural/drug effects , Spleen/drug effects , T-Lymphocytes/drug effects , Thymus Gland/drug effects , Trichloroethylene/toxicity , Animals , Dose-Response Relationship, Drug , Immune System/drug effects , Immunophenotyping , Mice , Risk Assessment , Spleen/immunology , Thymus Gland/immunology , Time Factors
16.
Toxicol Sci ; 76(2): 347-56, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14514957

ABSTRACT

Immunological parameters, host resistance, and thyroid hormones were evaluated in F1 mice exposed in utero to jet propulsion fuel-8 (JP-8). C57BL/6 pregnant dams (mated with C3H/HeJ males) were gavaged daily on gestation days 6-15 with JP-8 in a vehicle of olive oil at 0, 1000, or 2000 mg/kg. At weaning (3 weeks of age), no significant differences were observed in body, liver, spleen, or thymus weight, splenic and thymic cellularity, splenic CD4/CD8 lymphocyte subpopulations, or T-cell proliferation. Yet, lymphocytic proliferative responses to B-cell mitogens were suppressed in the 2000 mg/kg treatment group. In addition, thymic CD4-/CD8+ cells were significantly increased. By adulthood (8 weeks of age), lymphocyte proliferative responses and the alteration in thymic CD4-/CD8+ cells had returned to normal. However, splenic weight and thymic cellularity were altered, and the IgM plaque forming cell response was suppressed by 46% and 81% in the 1000 and 2000 mg/kg treatment groups, respectively. Furthermore, a 38% decrease was detected in the total T4 serum hormone level at 2000 mg/kg. In F1 adults, no significant alterations were observed in natural killer cell activity, T-cell lymphocyte proliferation, bone marrow cellularity and proliferative responses, complete blood counts, peritoneal and splenic cellularity, liver, kidney, or thymus weight, macrophage phagocytosis or nitric oxide production, splenic CD4/CD8 lymphocyte subpopulations, or total T3 serum hormone levels. Host resistance models in treated F1 adults demonstrated that immunological responses were normal after challenge with Listeria monocytogenes, but heightened susceptibility to B16F10 tumor challenge was seen at both treatment levels. This study demonstrates that prenatal exposure to JP-8 can target the developing murine fetus and result in impaired immune function and altered T4 levels in adulthood.


Subject(s)
Hydrocarbons/toxicity , Immunity, Maternally-Acquired/drug effects , Petroleum/toxicity , Prenatal Exposure Delayed Effects , Administration, Oral , Animals , Disease Susceptibility/chemically induced , Disease Susceptibility/immunology , Dose-Response Relationship, Drug , Female , Hydrocarbons/administration & dosage , Listeria monocytogenes/immunology , Lymphocyte Activation/drug effects , Male , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Pregnancy , Thyroid Hormones/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...