Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Vis Exp ; (159)2020 05 29.
Article in English | MEDLINE | ID: mdl-32538903

ABSTRACT

Drug-induced autoimmune hepatitis (DIH) is the most common hepatic drug-induced hypersensitization process observed in approximately 9 to 12% of patients with autoimmune hepatitis. The overwhelming majority of patients with DIH are women. The underlying mechanisms of these sex differences in prevalence are unclear because of the paucity of animal models that mimic human disease. Even so, underlying mechanisms are widely believed to be associated with human leukocyte antigen haplotypes and sex hormones. In contrast, using a DIH mouse model, we have uncovered that IL-4 initiated CD4+ T cells directed against an epitope of cytochrome P450 2E1 induces influx of neutrophils, macrophages and mast cells into the livers of female BALB/c mice. Using this model, we have also shown that IL-33-induced FoxP3+regulatory T cells confer protection against DIH in female and male mice. This DIH model is induced by immunizing mice with an epitope of CYP2E1 that has been covalently altered with a drug metabolite that has been associated with DIH. This epitope is recognized by patients with DIH. Our method induces robust and reproducible hepatitis and autoantibodies that can be utilized to study the pathogenesis of DIH. While in vivo studies can cause undue pain and distress in mice when done improperly, the advantage of an in vivo model is the ability to evaluate the pathogenesis of disease in a large number of mice. Additionally, biological effects of the altered liver proteins can be studied using invasive procedures. The addition of in vitro studies to the experimental design allows rapid repetition and mechanistic analysis at a cellular level. Thus, we will demonstrate our model protocol and how it can be utilized to study in vivo and in vitro mechanisms of DIH.


Subject(s)
Chemical and Drug Induced Liver Injury/diagnosis , Hepatitis, Autoimmune/etiology , Animals , Chemical and Drug Induced Liver Injury/pathology , Hepatitis, Autoimmune/pathology , Mice , Mice, Inbred BALB C
2.
Cell Mol Immunol ; 16(8): 706-717, 2019 08.
Article in English | MEDLINE | ID: mdl-30030493

ABSTRACT

Immune-mediated, drug-induced hepatitis is a rare complication of halogenated volatile anesthetic administration. IL-4-regulated Th2-polarized reactions initiate this type and other types of hepatitis, while the mechanisms that regulate the severity remain elusive. IL-33 is an innate, IL-4-inducing, Th2-polarizing cytokine that has been detected in patients with liver failure and has been associated with upregulated ST2+Foxp3+CD4+CD25+ T cells; however, roles for IL-33 in drug-induced hepatitis are unclear. We investigated IL-33 in an anesthetic, immune-mediated hepatitis modeled in BALB/c, IL-33-/- and ST2-/- mice, as well as in patients with anesthetic hepatitis. The hepatic IL-33 and ST2 levels were elevated in BALB/c mice (p < 0.05) with hepatitis, and anti-IL-33 diminished hepatitis (p < 0.05) without reducing IL-33 levels. The complete absence of IL-33 reduced IL-10 (p < 0.05) and ST2+Foxp3+CD4+CD25+ T cells (p < 0.05), as well as reduced the overall survival (p < 0.05), suggesting suppressive roles for IL-33 in anesthetic, immune-mediated hepatitis. All of the mice demonstrated similar levels of CD4+ T-cell proliferation following direct T-cell receptor stimulation, but we detected splenic IL-33 and ST2-negative Foxp3+CD4+CD25+ T cells in ST2-/- mice that developed less hepatitis than BALB/c mice (p < 0.05), suggesting that ST2-negative Foxp3+CD4+CD25+ T cells reduced hepatitis. In patients, serum IL-33 and IPEX levels were correlated in controls (r2 = 0.5, p < 0.05), similar to the levels in mice, but not in anesthetic hepatitis patients (r2 = 0.01), who had elevated IL-33 (p < 0.001) and decreased IPEX (p < 0.01). Our results suggest that, in anesthetic, immune-mediated hepatitis, IL-33 does not regulate the CD4+ T-cell proliferation that initiates hepatitis, but IL-33, likely independent of ST2, reduces hepatitis via upregulation of Foxp3+CD4+CD25+ T cells. Further studies are needed to translate the role of IL-33 to human liver disease.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/immunology , Forkhead Transcription Factors/blood , Forkhead Transcription Factors/metabolism , Interleukin-33/blood , Interleukin-33/metabolism , Animals , Cell Proliferation/genetics , Chemical and Drug Induced Liver Injury/mortality , Cytochrome P-450 CYP2E1/immunology , Disease Models, Animal , Epitopes/chemistry , Epitopes/pharmacology , Female , Fluoroacetates/chemistry , Fluoroacetates/pharmacology , Humans , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-33/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout
3.
J Immunol ; 196(8): 3470-8, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26936880

ABSTRACT

Tumor-induced myeloid-derived suppressor cells (MDSC) contribute to immune suppression in tumor-bearing individuals and are a major obstacle to effective immunotherapy. Reactive oxygen species (ROS) are one of the mechanisms used by MDSC to suppress T cell activation. Although ROS are toxic to most cells, MDSC survive despite their elevated content and release of ROS. NF erythroid 2-related factor 2 (Nrf2) is a transcription factor that regulates a battery of genes that attenuate oxidative stress. Therefore, we hypothesized that MDSC resistance to ROS may be regulated by Nrf2. To test this hypothesis, we used Nrf2(+/+)and Nrf2(-/-)BALB/c and C57BL/6 mice bearing 4T1 mammary carcinoma and MC38 colon carcinoma, respectively. Nrf2 enhanced MDSC suppressive activity by increasing MDSC production of H2O2, and it increased the quantity of tumor-infiltrating MDSC by reducing their oxidative stress and rate of apoptosis. Nrf2 did not affect circulating levels of MDSC in tumor-bearing mice because the decreased apoptotic rate of tumor-infiltrating MDSC was balanced by a decreased rate of differentiation from bone marrow progenitor cells. These results demonstrate that Nrf2 regulates the generation, survival, and suppressive potency of MDSC, and that a feedback homeostatic mechanism maintains a steady-state level of circulating MDSC in tumor-bearing individuals.


Subject(s)
Apoptosis/immunology , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Myeloid Cells/cytology , NF-E2-Related Factor 2/immunology , Tumor Escape/immunology , Animals , Bone Marrow Cells/cytology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Survival/immunology , Colonic Neoplasms/pathology , Female , Hydrogen Peroxide/metabolism , Immune Tolerance/immunology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Stem Cells/cytology
4.
Am J Physiol Endocrinol Metab ; 310(5): E332-45, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26670485

ABSTRACT

C1q/TNF-related protein 3 (CTRP3) is a secreted metabolic regulator whose circulating levels are reduced in human and rodent models of obesity and diabetes. Previously, we showed that CTRP3 infusion lowers blood glucose by suppressing gluconeogenesis and that transgenic overexpression of CTRP3 protects mice against diet-induced hepatic steatosis. Here, we used a genetic loss-of-function mouse model to further address whether CTRP3 is indeed required for metabolic homeostasis under normal and obese states. Both male and female mice lacking CTRP3 had similar weight gain when fed a control low-fat (LFD) or high-fat diet (HFD). Regardless of diet, no differences were observed in adiposity, food intake, metabolic rate, energy expenditure, or physical activity levels between wild-type (WT) and Ctrp3-knockout (KO) animals of either sex. Contrary to expectations, loss of CTRP3 in LFD- or HFD-fed male and female mice also had minimal or no impact on whole body glucose metabolism, insulin sensitivity, and fasting-induced hepatic gluconeogenesis. Unexpectedly, the liver sizes of HFD-fed Ctrp3-KO male mice were markedly reduced despite a modest increase in triglyceride content. Furthermore, liver expression of fat oxidation genes was upregulated in the Ctrp3-KO mice. Whereas the liver and adipose expression of profibrotic TGFß1, as well as its serum levels, was suppressed in HFD-fed KO mice, circulating proinflammatory IL-6 levels were markedly increased; these changes, however, were insufficient to affect systemic metabolic outcome. We conclude that, although it is dispensable for physiological control of energy balance, CTRP3 plays a previously unsuspected role in modulating liver size and circulating cytokine levels in response to obesity.


Subject(s)
Adipokines/genetics , Interleukin-6/metabolism , Liver/pathology , Obesity/genetics , Transforming Growth Factor beta1/metabolism , Adipose Tissue , Animals , Calorimetry, Indirect , Chemokine CCL2/genetics , Diacylglycerol O-Acyltransferase/genetics , Diet, Fat-Restricted , Diet, High-Fat , Female , Gluconeogenesis/genetics , Glucose/metabolism , Glucose Tolerance Test , Glycerol-3-Phosphate O-Acyltransferase/genetics , Inflammation , Insulin Resistance/genetics , Lipid Metabolism/genetics , Liver/metabolism , Male , Matrix Metalloproteinase 12/genetics , Mice , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Organ Size , Oxidation-Reduction , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Tumor Necrosis Factor-alpha/genetics , Weight Gain/genetics
5.
J Leukoc Biol ; 96(6): 1109-18, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25170116

ABSTRACT

MDSC and macrophages are present in most solid tumors and are important drivers of immune suppression and inflammation. It is established that cross-talk between MDSC and macrophages impacts anti-tumor immunity; however, interactions between tumor cells and MDSC or macrophages are less well studied. To examine potential interactions between these cells, we studied the impact of MDSC, macrophages, and four murine tumor cell lines on each other, both in vitro and in vivo. We focused on IL-6, IL-10, IL-12, TNF-α, and NO, as these molecules are produced by macrophages, MDSC, and many tumor cells; are present in most solid tumors; and regulate inflammation. In vitro studies demonstrated that MDSC-produced IL-10 decreased macrophage IL-6 and TNF-α and increased NO. IL-6 indirectly regulated MDSC IL-10. Tumor cells increased MDSC IL-6 and vice versa. Tumor cells also increased macrophage IL-6 and NO and decreased macrophage TNF-α. Tumor cell-driven macrophage IL-6 was reduced by MDSC, and tumor cells and MDSC enhanced macrophage NO. In vivo analysis of solid tumors identified IL-6 and IL-10 as the dominant cytokines and demonstrated that these molecules were produced predominantly by stromal cells. These results suggest that inflammation within solid tumors is regulated by the ratio of tumor cells to MDSC and macrophages and that interactions of these cells have the potential to alter significantly the inflammatory milieu within the tumor microenvironment.


Subject(s)
Cell Communication/physiology , Gene Expression Regulation, Neoplastic/physiology , Macrophages/physiology , Myeloid Cells/physiology , Neoplasms, Experimental/pathology , Tumor Microenvironment/physiology , Animals , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , Inflammation , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-12/biosynthesis , Interleukin-12/genetics , Interleukin-12/metabolism , Interleukin-6/biosynthesis , Interleukin-6/genetics , Interleukin-6/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Stromal Cells/metabolism , Stromal Cells/pathology , Transplantation, Isogeneic , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...