Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Lupus (Los Angel) ; 2(1)2017 Apr.
Article in English | MEDLINE | ID: mdl-28620656

ABSTRACT

OBJECTIVES: Lupus flares are triggered by environmental agents that cause oxidative stress, but the mechanisms involved are unclear. The flares are characterized by oxidative modifications of proteins by 4-hydroxynonenals, malondialdehydes, carbonyls and nitration. These modifications have been proposed to induce and perpetuate lupus flares by "altered self" mechanisms. An epigenetically altered CD4+CD28+ T cell subset, caused at least in part by nitration of T cell signaling molecules, is found in patients with active lupus, and nitrated T cells are sufficient to cause lupus-like autoimmunity in animal models. The relation of protein 4-hydroxynonenals, malondialdehydes, carbonyls and nitration to lupus flares though, is unknown. We tested if the size of the epigenetically altered subset is related to disease activity and one or more of these oxidative modifications in lupus patients. We also tested the relationship between subset size, disease activity and the same oxidative modifications in Sjogren's syndrome, another autoimmune disease also associated with oxidative stress and characterized by anti-nuclear antibodies and the presence of the subset. METHODS: Lupus flare severity was quantitated using the Systemic Lupus Erythematosus Disease Activity Index, and Sjogren's flare severity using the European Sjogren's Syndrome Disease Activity Index. Subset size was determined by flow cytometry. Protein modifications were determined by ELISA. RESULTS: Only protein nitration correlated with the size of the subset in lupus and Sjogren's syndrome. CONCLUSIONS: These results support a role for protein nitration in subset size and lupus flare severity. Protein nitration may also contribute to autoantibody formation in Sjogren's syndrome.

2.
Lupus ; 23(6): 568-76, 2014 May.
Article in English | MEDLINE | ID: mdl-24763540

ABSTRACT

A dose-dependent combination of environmental exposures, estrogenic hormones and genetic predisposition is thought to be required for lupus to develop and flare, but how the environment modifies the immune system in genetically predisposed people is unclear. Current evidence indicates that environmental agents that inhibit DNA methylation can convert normal antigen-specific CD4+ T lymphocytes into autoreactive, cytotoxic, pro-inflammatory cells that are sufficient to cause lupus-like autoimmunity in animal models, and that the same changes in DNA methylation characterize CD4+ T cells from patients with active lupus. Environmental agents implicated in inhibiting T-cell DNA methylation include the lupus-inducing drugs procainamide and hydralazine, as well as diet, and agents causing oxidative stress, such as smoking, UV light exposure, and infections, which have been associated with lupus onset or disease activity. Other studies demonstrate that demethylated T cells cause only anti-DNA antibodies in mice lacking a genetic predisposition to lupus, but are sufficient to cause lupus-like autoimmunity in genetically predisposed mice and likely people, and that estrogens augment the disease. Collectively, these studies suggest that environmental agents that inhibit DNA methylation, together with lupus genes and estrogens or endocrine disruptors, combine in a dose-dependent fashion to cause lupus flares.


Subject(s)
Endocrine Disruptors/adverse effects , Environmental Exposure/adverse effects , Environmental Pollutants/adverse effects , Epigenesis, Genetic/drug effects , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/genetics , Animals , Autoantibodies/blood , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Genetic Predisposition to Disease , Humans , Lupus Erythematosus, Systemic/immunology , Prognosis , Risk Assessment , Risk Factors , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Lupus ; 19(13): 1492-501, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20647250

ABSTRACT

Activated lymphocytes proliferate, secrete cytokines, and can make antibodies. Normally activated B and T cells meet the bioenergetic demand for these processes by up-regulating aerobic glycolysis. In contrast, several lines of evidence suggest that pathogenic lymphocytes in autoimmune diseases like lupus meet ATP demands through oxidative phosphorylation. Using (13)C-glucose as a stable tracer, we found that splenocytes from mice with lupus derive the same fraction of lactate from glucose as control animals, suggesting comparable levels of glycolysis and non-oxidative ATP production. However, lupus splenocytes increase glucose oxidation by 40% over healthy control animals. The ratio between pentose phosphate cycle (PPC) activity and glycolysis is the same for each group, indicating that increased glucose oxidation is due to increased activity of the TCA cycle in lupus splenocytes. Repetitive stimulation of cultured human T cells was used to model chronic lymphocyte activation, a phenotype associated with lupus. Chronically activated T cells rely primarily on oxidative metabolism for ATP synthesis suggesting that chronic antigen stimulation may be the basis for the metabolic findings observed in lupus mice. Identification of disease-related bioenergetic phenotypes should contribute to new diagnostic and therapeutic strategies for immune diseases.


Subject(s)
Adenosine Triphosphate/biosynthesis , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/metabolism , Animals , Antigens/metabolism , Cells, Cultured , Female , Glucose/metabolism , Glycolysis , Humans , Lactic Acid/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NZB , Oxidation-Reduction , Phenotype , Spleen/cytology
4.
Genes Immun ; 10(5): 509-16, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19279650

ABSTRACT

Women develop chronic inflammatory autoimmune diseases more often than men. The mechanisms causing the increased susceptibility are incompletely understood. Chronic immune stimulation characterizes many autoimmune disorders. We hypothesized that repeated stimulation may cause a different T-cell response in women than in men. Microarrays were used to compare gene expression in T cells from healthy men and women with and without repeated stimulation. Four days after a single stimulation, only 25% of differentially expressed, gender-biased genes were expressed at higher levels in women. In contrast, after restimulation, 72% were more highly expressed in women. Immune response genes were significantly over-represented among the genes upregulated in women and among the immune response genes, the inflammatory/cytotoxic effector genes interferon-gamma (IFN-gamma), lymphotoxin beta (LTbeta), granzyme A (GZMA), interleukin-12 receptor beta2 (IL12Rbeta2), and granulysin (GNLY) were among those overexpressed to the highest degree. In contrast, IL17A was the only effector gene more highly expressed in men. Estrogen response elements were identified in the promoters of half the overexpressed immune genes in women, and in <10% of the male-biased genes. The differential expression of inflammatory/cytotoxic effector molecules in restimulated female T cells may contribute to the differences in autoimmune diseases between women and men.


Subject(s)
Gene Expression Profiling , Sex Characteristics , T-Lymphocytes, Cytotoxic/immunology , Adult , Female , Humans , Leukocytes, Mononuclear/immunology , Male , Oligonucleotide Array Sequence Analysis , Response Elements , T-Lymphocytes, Cytotoxic/metabolism
5.
Ageing Res Rev ; 5(4): 449-67, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16965942

ABSTRACT

Heritable changes in gene expression that do not involve coding sequence modifications are referred to as "epigenetic". Epigenetic mechanisms principally include DNA methylation and a variety of histone modifications, of which the best characterized is acetylation. DNA hypermethylation and histone hypoacetylation are hallmarks of gene silencing, while DNA hypomethylation and acetylated histones promote active transcription. Aberrant DNA methylation and histone acetylation have been linked to a number of age related disorders including cancer, autoimmune disorders and others. Since epigenetic alterations are reversible, modifying epigenetic marks contributing to disease development may provide an approach to designing new therapies. Herein we review the role of epigenetic changes in disease development, and recent advances in the therapeutic modification of epigenetic marks.


Subject(s)
Aging/genetics , DNA Methylation , Epigenesis, Genetic , Neoplasms/genetics , Neoplasms/therapy , Humans
6.
Cell Mol Life Sci ; 59(2): 241-57, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11915942

ABSTRACT

The regulation of gene transcription is not simply dependent on the presence or absence of DNA-binding transcription factors that turn genes on or off, but also involves processes determining the ability of transcription factors to gain access to and bind their target DNA. Methylation of DNA cytosine bases leads to the inaccessibility of DNA regulatory elements to their transcription factors by a number of mechanisms. Our understanding of DNA methylation has advanced rapidly in recent years with the identification of an increasingly large number of novel proteins involved in this process. These include methylcytosine-binding proteins as well as additional members of the DNA methyltransferase family. The creation of mice with targeted deletions in a number of genes involved in DNA methylation has further elucidated the functions of many of these proteins. The characterization of complexes that contain proteins known to be involved in DNA methylation has led to the identification of additional proteins, especially those involved in histone deacetylation, indicating that DNA methylation and histone deacetylation very likely act in a synergistic fashion to regulate gene transcription. Finally, the implication of DNA methylation in tumorigenesis and the realization that some congenital diseases are caused by deficiency of proteins involved in DNA methylation has confirmed the importance of this process in regulating gene expression.


Subject(s)
Cytosine/analogs & derivatives , DNA Methylation , Gene Silencing , 5-Methylcytosine , Animals , CpG Islands , Cytosine/metabolism , DNA-Binding Proteins/metabolism , DNA-Cytosine Methylases/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , Immunity/genetics , Mice , Models, Genetic , Transcription, Genetic
7.
Arthritis Rheum ; 44(2): 397-407, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11229472

ABSTRACT

OBJECTIVE: Previous studies have shown that inhibiting T cell DNA methylation causes a lupus-like disease by modifying gene expression. T cells from patients with lupus exhibit diminished levels of DNA methyltransferase (MTase) enzyme activity, hypomethylated DNA, and changes in gene expression similar to those exhibited by T cells treated with methylation inhibitors, suggesting that DNA hypomethylation may contribute to human lupus. Since it is known that DNA MTase levels are regulated by the ras-mitogen-activated protein kinase (MAPK) pathway, this study sought to determine whether decreased ras-MAPK signaling could account for the DNA hypomethylation in lupus T cells. METHODS: DNA MTase messenger RNA (mRNA) from lupus patients and from healthy controls was quantitated by Northern analysis, and ras-MAPK signaling was determined by immunoblotting with antibodies to the activated forms of extracellular receptor-associated kinase (ERK). Results were compared with those in T cells in which ras-MAPK signaling was inhibited with a soluble inhibitor of MAPK ERK I (MEK1). RESULTS: T cells from patients with active lupus had diminished DNA MTase mRNA levels and decreased signaling through the ras-MAPK pathway. Inhibiting signaling through the ras-MAPK pathway with the MEK1 inhibitor decreased DNA MTase mRNA and enzyme activity to the levels seen in lupus T cells, and resulted in DNA hypomethylation resembling that seen in lupus T cells. CONCLUSION: These results suggest that a decrease in signaling through the ras-MAPK pathway may be responsible for the decreased MTase activity and DNA hypomethylation in patients with lupus.


Subject(s)
DNA Methylation , Lupus Vulgaris/genetics , Lupus Vulgaris/pathology , MAP Kinase Signaling System/physiology , T-Lymphocytes/metabolism , Adolescent , Adult , Aged , Female , Genes, ras , Humans , MAP Kinase Signaling System/genetics , Male , Middle Aged
8.
J Immunol ; 164(6): 2897-904, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10706675

ABSTRACT

The human marrow produces approximately 1010 monocytes daily, and this production must be balanced by a similar rate of destruction. Monocytes/macrophages can undergo apoptosis after activating CD4+ T cells, suggesting one mechanism that may contribute to macrophage homeostasis. Previous reports indicate that Fas-Fas ligand interactions are the principle molecules mediating this response. However, D10, an Iak-restricted cloned Th2 line, will similarly induce apoptosis in Ag-presenting macrophages, and D10 cells lack Fas ligand. To confirm that D10 cells kill macrophages through Fas-independent pathways, D10 cells were shown to kill MRL lpr/lpr (Iak) macrophages in an Ag-dependent fashion, indicating additional mechanisms. Recent reports demonstrate that TNF-related apoptosis-inducing ligand (TRAIL), interacting with Apo2, and TNF-like weak inducer of apoptosis (TWEAK), interacting with Apo3, will induce apoptosis in some cells. Using Abs to TRAIL and an Apo3-IgG Fc fusion protein, we demonstrated that D10 cells express both TRAIL and TWEAK. The Apo3 fusion protein, but not human IgG, inhibited D10-induced macrophage apoptosis, as did anti-TRAIL. Further studies demonstrated that AE7, a cloned Th1 line, and splenic T cells express TWEAK, TRAIL, and Fas ligand, and inhibiting these molecules also inhibited macrophage killing. These results indicate that D10 cells induce macrophage apoptosis through TRAIL- and TWEAK-dependent pathways. Because normal T cells also express these molecules, these results support the concept that T cells have multiple pathways by which to induce macrophage apoptosis. These pathways may be important in immune processes such as macrophage homeostasis as well as in down-regulation of immune responses and elimination of macrophages infected with intracellular organisms.


Subject(s)
Antigen-Presenting Cells/immunology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Carrier Proteins/physiology , Cytotoxicity, Immunologic/immunology , Macrophages, Peritoneal/immunology , Membrane Glycoproteins/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/biosynthesis , Cell Line , Cells, Cultured , Cytokine TWEAK , Fas Ligand Protein , Female , Ligands , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/ultrastructure , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred A , Mice, Inbred AKR , Mice, Inbred C57BL , Mice, Inbred MRL lpr , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factors , fas Receptor/metabolism , fas Receptor/physiology
9.
Immunol Invest ; 29(4): 427-42, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11130784

ABSTRACT

Overexpressing LFA-1 (CD11a/CD18) on antigen specific CD4+ T cells makes the cells proliferate to normally subthreshold stimuli, including self-Ia molecules without specific antigen. The mechanisms by which this occurs are unknown, but potentially include transmission of an increased costimulatory signal, overstabilization of normally low affinity TCR-Ia interactions, or both. A role for increased costimulatory signaling was tested by culturing control and CD18-transfected antigen-specific T cells clones with anti-CD3 and anti-CD11a. Minimal calcium fluxes were detected, but increased protein tyrosine phosphorylation was observed in the transfectants. However, the proliferative response to graded amounts of these antibodies were identical in the transfectants and controls, suggesting that increased signaling alone was insufficient to cause the increased responsiveness. To test for overstabilization, transfected and control clones were cultured with syngeneic Mø with or without antigen. The transfected but not control cells downregulated TCR expression in response to Mø alone, thus demonstrating successful TCR signaling to a low affinity interaction. These results indicate that LFA-1 overexpression permits TCR signal transmission to a normally subthreshold stimulus presented by Mø, consistent with overstabilization. LFA-1 overexpression also causes increased tyrosine phosphorylation, but this alone is not sufficient to cause a proliferative response to low level stimuli.


Subject(s)
CD18 Antigens/biosynthesis , Lymphocyte Function-Associated Antigen-1/biosynthesis , T-Lymphocytes/immunology , Animals , Antigens/immunology , CD18 Antigens/genetics , Calcium Signaling , Cell Division , Conalbumin/immunology , Cross-Linking Reagents , Dose-Response Relationship, Drug , Down-Regulation , Gene Expression , Humans , Lymphocyte Function-Associated Antigen-1/genetics , Mice , Mice, Inbred AKR , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
10.
Immunol Invest ; 29(4): 411-25, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11130783

ABSTRACT

Inducing T cell LFA-1 overexpression by transfection, or by treatment with DNA methylation inhibitors including 5-azacytidine, procainamide and hydralazine, causes MHC-specific T cell autoreactivity in vitro and autoimmunity in vivo. How DNA methylation inhibitors increase LFA-1 expression is unknown. In this report we identify a mechanism by which DNA methylation affects LFA-1 expression. Nuclear run-on assays demonstrated that inhibiting DNA methylation increased transcription of CD11a but not CD18 or beta-actin mRNA. CD11a mRNA stability was not affected. Transfection of hypomethylated cells with reporter constructs containing the CD11a promoter showed no role for overexpression of transcription factors. However, the CD11a promoter demethylated following treatment with DNA methyltransferase inhibitors, and in vitro methylation of the construct suppressed its expression. Together, these results indicate that DNA methylation inhibitors can cause LFA-1 overexpression directly by demethylating the CD11a promoter. This mechanism could contribute to T cell autoreactivity, and potentially to autoimmunity.


Subject(s)
DNA Methylation , Gene Expression Regulation , Lymphocyte Function-Associated Antigen-1/genetics , T-Lymphocytes/immunology , Transcription, Genetic , Azacitidine/pharmacology , Cell Line, Transformed , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , Jurkat Cells , Procainamide/pharmacology , Promoter Regions, Genetic , T-Lymphocytes/drug effects , Transcription, Genetic/drug effects
11.
Biol Chem ; 379(8-9): 1113-20, 1998.
Article in English | MEDLINE | ID: mdl-9792444

ABSTRACT

Our group reported that inhibiting DNA methylation in human T cells increases DNA methyltransferase expression and activity, and suggested that this may represent a response to DNA hypomethylation. The increase correlates with increases in Ha-ras and c-jun, suggesting that increased signaling through the ras-MAPK pathway, due to overexpression of some elements, may be responsible. However, whether human DNA MTase is regulated by the ras-MAPK pathway, and whether overexpression of elements in this pathway will increase DNA MTase, is unknown. We report that treating cells with a DNA methylation inhibitor increases transcription regulated by a putative DNA MTase promoter, and that this increase requires AP-1 sites. Additional studies demonstrate that overexpression of an unmutated Ha-ras causes an increase in DNA MTase, and that human T cell DNA MTase can be decreased by inhibiting signaling through the ras-MAPK pathway. Together, these studies suggest that human T cell DNA MTase is regulated through the ras-MAPK pathway, and that overexpression of Ha-ras is sufficient to increase DNA MTase expression. These results thus provide a mechanism for the increase in DNA MTase observed after inducing DNA hypomethylation, a response which may have relevance to some disease states.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA Methylation , DNA-Cytosine Methylases/metabolism , Oncogene Protein p21(ras)/metabolism , Signal Transduction , Animals , Base Sequence , COS Cells , DNA Primers , DNA-Cytosine Methylases/genetics , Humans , Jurkat Cells , Oncogene Protein p21(ras)/genetics , Promoter Regions, Genetic
12.
J Immunol ; 159(3): 1303-9, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9233626

ABSTRACT

DNA methylation, a mechanism modifying gene expression, is mediated in part by the enzyme DNA methyltransferase. Reduced levels of T cell DNA methyltransferase have been observed in lupus-like diseases, and increased levels have been reported in malignancies. Little is known concerning the regulation of human DNA methyltransferase. In this report we demonstrate that mitogenic T cell stimulation causes an increase in DNA methyltransferase mRNA and enzyme activity. We also show that pharmacologic inhibition of T cell DNA methylation causes an increase in the rate of DNA methyltransferase mRNA transcription and a corresponding increase in mRNA levels and enzyme activity. This suggests that DNA methyltransferase is itself regulated in part by DNA methylation status, possibly representing a feedback mechanism. DNA methylation inhibition also resulted in an increase in Ha-ras and c-jun mRNA levels, overexpression of which increases DNA methyltransferase in murine systems. These results thus identify two mechanisms regulating levels of human T cell DNA methyltransferase and raise the possibility that abnormalities in either could contribute to disorders associated with altered DNA methylation.


Subject(s)
DNA-Cytosine Methylases/biosynthesis , Lymphocyte Activation/drug effects , Phytohemagglutinins/pharmacology , Procainamide/pharmacology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Adult , Azacitidine/pharmacology , DNA Methylation/drug effects , DNA-Cytosine Methylases/antagonists & inhibitors , DNA-Cytosine Methylases/genetics , Enzyme Activation/drug effects , Humans , Jurkat Cells , Male , RNA, Messenger/drug effects , RNA, Messenger/genetics , T-Lymphocytes/drug effects , Transcription, Genetic/drug effects
13.
Immunol Lett ; 58(1): 9-14, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9436462

ABSTRACT

The CD6 glycoprotein is expressed by T lymphocytes and is hypothesized to interact with one or more ligands expressed on antigen presenting cells (APCs). We show that CD6 mediates binding of the transformed CD4+ T cell line Hut 78 to gamma-interferon activated keratinocytes (KCs). A recombinant CD6-Ig fusion protein has been reported to bind to a CD6 ligand ALCAM, but this is the first demonstration that cell-cell adhesion of human T lymphocytes can be CD6 dependent. The known CD6 ligand ALCAM (CD166) is expressed on cultured KCs but does not appear to mediate KC-Hut 78 binding, suggesting the existence of additional CD6 ligands expressed on KCs. In functional studies using autologous KCs as APCs for tetanus toxoid specific T cell clones, KCs +/- gamma-interferon are unable to stimulate autologous T cells with recall antigen. Therefore interaction of T cell CD6 with CD6 ligands on KCs does not provide sufficient co-stimulation of primed T cells to support responses to nominal antigen.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Glycoproteins/immunology , Interferon-gamma/pharmacology , Keratinocytes/immunology , T-Lymphocytes/immunology , Activated-Leukocyte Cell Adhesion Molecule , Cell Adhesion , Glycoproteins/analysis , Humans , Keratinocytes/drug effects , Lymphocyte Activation , Thymus Gland/immunology
14.
J Cogn Neurosci ; 9(2): 277-93, 1997 Mar.
Article in English | MEDLINE | ID: mdl-23962017

ABSTRACT

Many neuropsychological investigations of human memory have focused on the amnesic deficits of alcoholic Korsakoff's syndrome. Structural neuroimaging suggests that the syndrome results from midline diencephalic damage, but functional neuroimaging has the potential to reveal additional neuropathology that may be responsible for cognitive dysfunction. Accordingly, high-resolution positron emission tomography (PET) was used to measure regional cerebral metabolic rates for glucose utilization in five alcoholic Korsakoff patients and nine alcoholic control subjects. Results from a continuous recognition test administered during the radiotracer uptake period indicated that all subjects performed normally with respect to immediate memory, whereas Korsakoff patients demonstrated a marked memory impairment in delayed recognition. PET results from the Korsakoff group showed a widespread decline in glucose metabolism in frontal, parietal, and cingulate regions, suggesting that these functional abnormalities in the cerebral cortex contribute to the memory impairment. Hippocampal glucose metabolism did not differ between the groups. Thus, the evidence did not support the hypothesis that parallel brain dysfunctions are responsible for the similar amnesic symptomatology after hippocampal and diencephalic damage. We hypothesize that the amnesic dysfunction of Korsakoff's syndrome depends on a disruption of thalamocortical interactions that mediate a function critical for normal memory storage.

15.
Immunology ; 88(4): 537-43, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8881754

ABSTRACT

CD6 is a 130 000 MW T-cell surface glycoprotein that can deliver coactivating signals to mature T lymphocytes. Studies using monoclonal antibodies (mAb) have defined at least four epitopes on CD6, and distinct functional responses are elicited by mAb to the different epitopes. The function of CD6 is unknown. Multiple CD6 ligands are predicted, based on data that a soluble CD6 fusion protein precipitates at least three peptides. A cDNA clone for one of these ligands, termed activated leucocyte-cell adhesion molecule (ALCAM) has recently been isolated. In order to further characterize the role of CD6 in cell-cell interactions, we have examined the role of CD6 in a variety of responses by tetanus toxoid (TT) specific human T-cell clones. Anti-CD6 mAb UMCD6 (epitope 3) inhibits antigen-specific responses of such clones to TT, but not to the superantigen SEA. Responses of clones to nominal antigen are CD6-dependent using either peripheral blood mononuclear cells (PBMC) or macrophage-depleted E rosette negative cells as the antigen-presenting cell (APC) population. Furthermore, these clones made autoreactive with DNA methyltransferase inhibitors express increased CD6, and autoreactivity is inhibited by UMCD6. Taken together, the data suggests the existence of a functional CD6 ligand in peripheral blood which is expressed by APC, including cells other than macrophages. Interactions between CD6 and CD6 ligands may regulate both antigen specific and autoreactive responses of human T lymphocytes.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Autoimmunity , Epitopes/immunology , T-Lymphocytes/immunology , Antigen-Presenting Cells/immunology , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , B-Lymphocytes/immunology , Cell Division/immunology , Cell Line , Clone Cells/immunology , Enterotoxins/immunology , Humans , RNA, Messenger/metabolism , Superantigens/immunology , Up-Regulation/immunology
17.
J Clin Invest ; 97(7): 1748-60, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8601641

ABSTRACT

Although anti-DNA autoantibodies are an important hallmark of lupus, the relationships among anti-DNA structure, reactivity, and pathogenicity have not been fully elucidated. To further investigate these relationships, we compare the variable genes and primary structure of eight anti-DNA mAbs previously obtained from an MRL/MpJ-lpr/lpr mouse along with the ability of three representative mAbs to induce nephritis in nonautoimmune mice using established adoptive transfer protocols. One monospecific anti-single-stranded (ss) DNA (11F8) induces severe diffuse proliferative glomerulonephritis in nonautoimmune mice whereas another anti-ssDNA with apparently similar in vitro binding properties (9F11) and an anti-double-stranded DNA (4B2) are essentially benign. These results establish a murine model of anti-DNA-induced glomerular injury resembling the severe nephritis seen in lupus patients and provide direct evidence that anti-ssDNA can be more pathogenic than anti-double-stranded DNA. In vitro binding experiments using both protein-DNA complexes and naive kidney tissue indicate that glomerular localization of 11F8 may occur by recognition of a planted antigen in vivo. Binding to this antigen is DNase sensitive which suggests that DNA or a DNA-containing molecule is being recognized.


Subject(s)
Antibodies, Antinuclear/genetics , Antibodies, Antinuclear/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , DNA/immunology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA/genetics , DNA Primers/genetics , Genes, Immunoglobulin , Hybridomas/immunology , Immunoglobulin Variable Region/genetics , Kidney/immunology , Kidney/pathology , Ligands , Lupus Nephritis/etiology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Mice , Mice, Mutant Strains , Microscopy, Electron , Molecular Sequence Data , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
18.
Lab Invest ; 73(6): 746-59, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8558836

ABSTRACT

Although the importance of DNA methylation in normal cellular development and hereditary disease states has been appreciated for some time, the role of environmental agents in causing DNA methylation abnormalities and the effects of DNA hypomethylation on T cells have only recently been examined. This review summarizes current knowledge about the role of DNA methylation in regulating T function and gene expression and highlights a novel mechanism causing autoimmunity, in which epigenetic modification of T cell DNA by environmental agents plays an important role in triggering lupus-like diseases. The observations that DNA methylation inhibitors modify gene expression and induce autoreactivity in cloned, Ag-specific CD4+ cells in vitro, that the modified cells cause autoimmunity in vivo, and that similar changes are found in patients with active lupus provide a new approach to understanding how some forms of autoimmunity develop and may lead to new and more effective treatments.


Subject(s)
Lupus Vulgaris/chemically induced , Acetylation , Animals , Cell Communication , DNA/metabolism , Gene Expression , Humans , Macrophages/physiology , Methylation , T-Lymphocytes/physiology
19.
J Rheumatol ; 22(11): 2126-31, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8596155

ABSTRACT

OBJECTIVE: Having previously found that treating small areas of synovitis within the knees of patients with rheumatoid arthritis (RA) with 8-methoxypsoralen (8-MOP) and laser-derived ultraviolet A (PUVA) resulted in decreases in adhesion molecule expression, we sought to determine the effect of PUVA on expression of vascular cellular adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and E-selection by human umbilical vein endothelial cells (HUVEC). METHODS: Expression of VCAM-1, ICAM-1, and E-selectin on the surface of HUVEC was measured using specific antibodies and flow cytometry or a fluorescence plate reader, following treatment of cells with 8-MOP and UVA, before and after tumor necrosis factor (TNF) stimulation. RESULTS: PUVA led to significant dose dependent decreases in the expression of VCAM-1 and E-selectin that had been induced with TNF before PUVA treatment. Pretreatment with PUVA was also able to prevent subsequent TNF induction of VCAM-1 expression. TNF-induced ICAM-1 expression was not decreased by PUVA, however, and pretreatment only partially decreased ICAM-1 expression. CONCLUSION: The in vivo effects of PUVA may be explained, in part, by down regulation of adhesion molecule expression. The relative resistance of ICAM-1 to PUVA suggests some specificity to the effect on adhesion molecule expression.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/radiation effects , Intercellular Adhesion Molecule-1/metabolism , Methoxsalen/pharmacology , Photosensitizing Agents/pharmacology , Ultraviolet Rays , Vascular Cell Adhesion Molecule-1/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...