Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Clin Immunol ; 197: 34-39, 2018 12.
Article in English | MEDLINE | ID: mdl-30170030

ABSTRACT

וֹndoleamine-2,3-dioxygenase (IDO) plays a role in immune regulation. Increased IDO activity was reported in systemic lupus erythematosus (SLE). We investigated the effects of the tolerogenic peptide hCDR1, shown to ameliorate lupus manifestations, on IDO gene expression. mRNA was prepared from splenocytes of hCDR1- treated SLE-afflicted (NZBxNZW)F1 mice, from blood samples of lupus patients, collected before and after their in vivo treatment with hCDR1 and from peripheral blood mononuclear cells (PBMC) of patients incubated with hCDR1. IDO gene expression was determined by real-time RT-PCR. hCDR1 significantly down-regulated IDO expression in SLE-affected mice and in lupus patients (treated in vivo and in vitro). No effects were observed in healthy donors or following treatment with a control peptide. Diminished IDO gene expression was associated with hCDR1 beneficial effects. Our results suggest that the hCDR1-induced FOXP3 expressing regulatory T cells in lupus are not driven by IDO but rather by other hCDR1 regulated pathways.


Subject(s)
Antibodies, Monoclonal/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/drug effects , Lupus Erythematosus, Systemic/genetics , Peptide Fragments/pharmacology , T-Lymphocytes, Regulatory/drug effects , Adult , Aged , Animals , Antibodies, Monoclonal/therapeutic use , Clinical Trials, Phase II as Topic , Down-Regulation , Female , Forkhead Transcription Factors/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/metabolism , Male , Mice , Middle Aged , Peptide Fragments/therapeutic use , Real-Time Polymerase Chain Reaction , Spleen/cytology , T-Lymphocytes, Regulatory/metabolism
2.
Clin Immunol ; 192: 85-91, 2018 07.
Article in English | MEDLINE | ID: mdl-29730433

ABSTRACT

Primary Sjogren's syndrome (pSS) is an autoimmune disease characterized by lymphocytic infiltration of exocrine glands. We investigated whether the tolerogenic peptide, hCDR1, that ameliorates lupus manifestations would have beneficial effects on pSS as well. The in vitro effects of hCDR1 on gene expression of pro-inflammatory cytokines and regulatory molecules were tested in peripheral blood mononuclear cells (PBMC) of 16 pSS patients. hCDR1, but not a control peptide, significantly reduced gene expression of IL-1ß, TNF-α, MX-1 and BlyS and up-regulated immunosuppressive (TGF-ß, FOXP3) molecules in PBMC of pSS patients. hCDR1 did not affect gene expression in patients with rheumatoid arthritis and anti-phospholipid syndrome. Further, hCDR1 up-regulated the expression of Indoleamine 2,3-dioxygenase (IDO) via elevation of TGF-ß. IDO inhibition led to a significant decrease in the expression of FOXP3 which is crucial for the induction of T regulatory cells. Thus, hCDR1 is potential candidate for the specific treatment of pSS patients.


Subject(s)
Cytokines/immunology , Gene Expression Regulation/drug effects , Leukocytes, Mononuclear/drug effects , Peptides/pharmacology , Sjogren's Syndrome/immunology , Adult , Aged , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Cytokines/genetics , Cytokines/metabolism , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/immunology , Humans , Immunologic Factors/pharmacology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptides/genetics , Sjogren's Syndrome/blood , Sjogren's Syndrome/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
4.
J Autoimmun ; 54: 60-71, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24958634

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic multisystem disease in which various cell types and immunological pathways are dysregulated. Current therapies for SLE are based mainly on the use of non-specific immunosuppressive drugs that cause serious side effects. There is, therefore, an unmet need for novel therapeutic means with improved efficacy and lower toxicity. Based on recent better understanding of the pathogenesis of SLE, targeted biological therapies are under different stages of development. The latter include B-cell targeted treatments, agents directed against the B lymphocyte stimulator (BLyS), inhibitors of T cell activation as well as cytokine blocking means. Out of the latter, Belimumab was the first drug approved by the FDA for the treatment of SLE patients. In addition to the non-antigen specific agents that may affect the normal immune system as well, SLE-specific therapeutic means are under development. These are synthetic peptides (e.g. pConsensus, nucleosomal peptides, P140 and hCDR1) that are sequences of conserved regions of molecules involved in the pathogenesis of lupus. The peptides are tolerogenic T-cell epitopes that immunomodulate only cell types and pathways that play a role in the pathogenesis of SLE without interfering with normal immune functions. Two of the peptides (P140 and hCDR1) were tested in clinical trials and were reported to be safe and well tolerated. Thus, synthetic peptides are attractive potential means for the specific treatment of lupus patients. In this review we discuss the various biological treatments that have been developed for lupus with a special focus on the tolerogenic peptides.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Drug Delivery Systems , Epitopes, T-Lymphocyte/therapeutic use , Immunosuppressive Agents/therapeutic use , Lupus Erythematosus, Systemic , Peptides/therapeutic use , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Clinical Trials as Topic , Epitopes, T-Lymphocyte/immunology , Humans , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
5.
BMC Med ; 11: 90, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23556432

ABSTRACT

BACKGROUND: The 16/6-idiotype (16/6-Id) of the human anti-DNA antibody was found to induce experimental lupus in naïve mice, manifested by production of autoantibodies, leukopenia and elevated inflammatory markers, as well as kidney and brain involvement. We assessed behavior and brain pathology of naive mice injected intra-cerebra-ventricularly (ICV) with the 16/6-Id antibody. METHODS: C3H female mice were injected ICV to the right hemisphere with the human 16/6-Id antibody or commercial human IgG antibodies (control). The mice were tested for depression by the forced swimming test (FST), locomotor and explorative activity by the staircase test, and cognitive functions were examined by the novel object recognition and Y-maze tests. Brain slices were stained for inflammatory processes. RESULTS: 16/6-Id injected mice were cognitively impaired as shown by significant differences in the preference for a new object in the novel object recognition test compared to controls (P = 0.012). Similarly, the preference for spatial novelty in the Y-maze test was significantly higher in the control group compared to the 16/6-Id-injected mice (42% vs. 9%, respectively, P = 0.065). Depression-like behavior and locomotor activity were not significantly different between the16/6-Id-injected and the control mice. Immunohistochemistry analysis revealed an increase in astrocytes and microglial activation in the hippocampus and amygdala, in the 16/6-Id injected group compared to the control. CONCLUSIONS: Passive transfer of 16/6-Id antibodies directly into mice brain resulted in cognitive impairments and histological evidence for brain inflammation. These findings shed additional light on the diverse mosaic pathophysiology of neuropsychiatric lupus.See related Commentary article: http://www.biomedcentral.com/1741-7015/11/91.


Subject(s)
Antibodies/administration & dosage , Antibodies/toxicity , Cognitive Dysfunction/chemically induced , Encephalitis/chemically induced , Animals , Brain/pathology , Female , Histocytochemistry , Humans , Mice , Mice, Inbred C3H
6.
Immunology ; 132(1): 87-95, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20738420

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease that involves dysregulation of B and T cells. A tolerogenic peptide, designated hCDR1, ameliorates disease manifestations in SLE-afflicted mice. In the present study, the effect of treatment with hCDR1 on the CD74/macrophage migration inhibitory factor (MIF) pathway was studied. We report here that B lymphocytes from SLE-afflicted mice express relatively elevated levels of CD74, compared with B cells from healthy mice. CD74 is a receptor found in complex with CD44, and it binds the pro-inflammatory cytokine MIF. The latter components were also up-regulated in B cells from the diseased mice, and treatment with hCDR1 resulted in their down-regulation and in reduced B-cell survival. Furthermore, up-regulation of CD74 and CD44 expression was detected in brain hippocampi and kidneys, two target organs in SLE. Treatment with hCDR1 diminished the expression of those molecules to the levels determined for young healthy mice. These results suggest that the CD74/MIF pathway plays an important role in lupus pathology.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Autoantigens/immunology , B-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Intramolecular Oxidoreductases/immunology , Lupus Erythematosus, Systemic/immunology , Macrophage Migration-Inhibitory Factors/immunology , Nerve Tissue Proteins/immunology , Peptides/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Antigens, Differentiation, B-Lymphocyte/genetics , Apoptosis/drug effects , Apoptosis/immunology , Autoantigens/chemistry , B-Lymphocytes/drug effects , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/genetics , Humans , Immunomodulation , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/genetics , Lupus Erythematosus, Systemic/pathology , Macrophage Migration-Inhibitory Factors/biosynthesis , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Inbred NZB , Nerve Tissue Proteins/chemistry , Peptides/chemistry , Peptides/pharmacology
7.
J Clin Immunol ; 30(1): 34-44, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19756988

ABSTRACT

INTRODUCTION: Systemic lupus erythematosus (SLE) is characterized by a variety of autoantibodies and systemic clinical manifestations. A tolerogenic peptide, hCDR1, ameliorated lupus manifestations in mice models. The objectives of this study were to induce experimental SLE in pigs and to determine the ability of hCDR1 to immunomodulate the disease manifestations. RESULTS AND DISCUSSION: We report here the successful induction, by a monoclonal anti-DNA antibody, of an SLE-like disease in pigs, manifested by autoantibody production and glomerular immune complex deposits. Treatment of pigs with hCDR1 ameliorated the lupus-related manifestations. Furthermore, the treatment downregulated the gene expression of the pathogenic cytokines, interleukin (IL)-1beta, tumor necrosis factor alpha, interferon gamma, and IL-10, and upregulated the expression of the immunosuppressive cytokine transforming growth factor beta, the antiapoptotic molecule Bcl-xL, and the suppressive master gene, Foxp3, hence restoring the expression of the latter to normal levels. Thus, hCDR1 is capable of ameliorating lupus in large animals and is a potential candidate for the treatment of SLE patients.


Subject(s)
Antibodies, Antinuclear/administration & dosage , Complementarity Determining Regions/administration & dosage , Kidney/metabolism , Lupus Erythematosus, Systemic/immunology , Peptide Fragments/administration & dosage , Animals , Antibodies, Antinuclear/chemistry , Antibodies, Monoclonal/immunology , Cells, Cultured , Complementarity Determining Regions/chemistry , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Immune Tolerance , Immunomodulation , Kidney/immunology , Lupus Erythematosus, Systemic/physiopathology , Lupus Erythematosus, Systemic/therapy , Mice , Peptide Fragments/chemistry , Swine/immunology , bcl-X Protein/biosynthesis , bcl-X Protein/genetics
8.
J Autoimmun ; 34(2): 87-95, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19596183

ABSTRACT

Dysregulated expression of Bcl-xL and Bcl-2 may initiate the development of autoimmune diseases including systemic lupus erythematosus (SLE). A tolerogenic peptide designated hCDR1 was shown to ameliorate manifestations of spontaneous and induced murine SLE. Recently, we demonstrated that Bcl-xL plays a critical role in the modulating effects of hCDR1, as manifested by reducing the state of activation of lymphocytes and by down-regulating the secretion of the pathogenic cytokines, IFN-gamma and IL-10. Here we studied the role of Bcl-xL in the development and function of CD4 regulatory T-cells (Treg) from hCDR1-treated, SLE-afflicted (New-Zealand-Black x New-Zealand-White) F1 mice. We report that Bcl-xL was up-regulated in CD4 Treg of tolerized mice, where it played a role in inducing the regulatory/inhibitory molecules Foxp3, CTLA-4, and TGF-beta and in repressing PD-1. Further, Bcl-xL mediated the induction of CTLA-4 and TGF-beta in effector T cells (Teff) by CD4 Treg of the tolerized mice. The induction of Bcl-xL in Teff by Treg was TGF-beta dependent and CTLA-4 independent, leading to inhibition of proliferation and to a decrease in activated Teff. We conclude that Bcl-xL is required for the development and function of CD4 Treg, which ameliorate lupus following treatment with a tolerogenic peptide.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Lupus Erythematosus, Systemic/immunology , Peptide Fragments/administration & dosage , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , bcl-X Protein/metabolism , Animals , Antigens, CD/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Female , Forkhead Transcription Factors/biosynthesis , Humans , Immune Tolerance , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/therapy , Lymphocyte Activation/drug effects , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/metabolism , Up-Regulation , bcl-X Protein/genetics
9.
J Immunol ; 181(5): 3243-51, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18713995

ABSTRACT

A peptide, designated human CDR1 (hCDR1), that is based on the CDR1 of an anti-DNA Ab ameliorates systemic lupus erythematosus (SLE) in murine models via the induction of CD4(+)CD25(+) regulatory T cells (Tregs). In the present study, the involvement of CD8 Tregs in the mode of action of hCDR1 was investigated in SLE-afflicted (NZB x NZW)F1 mice and in SJL mice following immunization with the lupus-inducing anti-DNA mAb that bears a common Id, 16/6Id. Treatment with hCDR1 up-regulated Foxp3-expressing CD8(+)CD28(-) Tregs in association with clinical amelioration of lupus manifestations. Furthermore, the in vivo depletion of the latter cells diminished the clinical improvement and the inhibitory effects of hCDR1 on the secretion of IFN-gamma and resulted in the up-regulation of IL-10. However, the stimulatory effect of hCDR1 on the secretion of TGF-beta was not affected by the CD8 Tregs. In the absence of CD8 Tregs, CD4(+)CD25(+) Tregs were unable to expand in the hCDR1-treated mice, and the expression of Foxp3 was reduced, thereby interfering further with the suppressive function of CD4(+)CD25(+) Tregs as determined in the in vitro assays. However, CD8 cells from hCDR1-treated mice that were adoptively transferred into SLE-afflicted mice led to up-regulation of CD4(+)CD25(+) cells with intensified Foxp3 expression in the recipient mice. Thus, a functional link between two subsets of Tregs is demonstrated in which CD8(+)CD28(-) Tregs are required for both the optimal expansion and function of lupus ameliorating hCDR1-induced CD4(+)CD25(+) Tregs.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors , Immune Tolerance , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Nerve Tissue Proteins/immunology , Adoptive Transfer , Animals , Autoantigens/pharmacology , Autoantigens/therapeutic use , CD8-Positive T-Lymphocytes/transplantation , Cell Proliferation , Disease Models, Animal , Female , Humans , Mice , Nerve Tissue Proteins/pharmacology , Nerve Tissue Proteins/therapeutic use , T-Lymphocytes, Regulatory/cytology
10.
Immunology ; 128(1 Suppl): e395-405, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19040426

ABSTRACT

Systemic lupus erythematosus (SLE) is characterized in its early stages by the expansion of autoreactive T cells that trigger B-cell activation with subsequent multi-organ injury. Dendritic cells (DCs) in lupus were found to display an aberrant phenotype with higher expression of the maturation markers major histocompatibility complex (MHC) class II, CD80 and CD86, as well as higher production of proinflammatory cytokines including interleukin-12 (IL-12), resulting in an increased ability to activate T cells. A peptide (hCDR1) based on the complementarity determining region-1 of an anti-DNA antibody ameliorated SLE in both induced and spontaneous lupus models by downregulating T-cell functions. Our objectives were to determine whether DCs play a role in promoting the beneficial effects of hCDR1. We showed here that treatment with hCDR1 lowered the expression levels of MHC class II, CD80 and CD86 on DCs. The latter effect was associated with downregulation of messenger RNA expression and secretion of IL-12, a cytokine that upregulated T-cell proliferation and interferon-gamma (IFN-gamma) secretion. Moreover, DCs derived from hCDR1-treated mice downregulated proliferation and IFN-gamma secretion by T cells from untreated mice. Upregulation of transforming growth factor-beta (TGF-beta) secretion by T cells, following treatment with hCDR1, resulted in downregulation of IFN-gamma production and contributed to the phenotypic changes and magnitude of IL-12 secretion by DCs. The ameliorating effects of hCDR1 are therefore mediated at least partially by the upregulated secretion of TGF-beta by T cells that contribute to the induction of DCs with immature phenotype and suppressed functions. The resulting DCs further downregulate autoreactive T-cell functions.


Subject(s)
Autoantigens/therapeutic use , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Lupus Erythematosus, Systemic/drug therapy , Nerve Tissue Proteins/therapeutic use , Animals , Autoantigens/pharmacology , B7-1 Antigen/drug effects , B7-1 Antigen/immunology , B7-2 Antigen/drug effects , B7-2 Antigen/immunology , Dendritic Cells/drug effects , Down-Regulation , Female , Histocompatibility Antigens Class II/drug effects , Humans , Interferon-gamma/agonists , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12/antagonists & inhibitors , Interleukin-12/immunology , Interleukin-12/pharmacology , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/pharmacology , Peptides/pharmacology , Peptides/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transforming Growth Factor beta/pharmacology , Up-Regulation
11.
Immunology ; 128(2): 245-52, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19740381

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease mediated by T and B cells. It is characterized by a variety of autoantibodies and systemic clinical manifestations. A tolerogenic peptide, designated hCDR1, ameliorated the serological and clinical manifestations of SLE in both spontaneous and induced models of lupus. In the present study, we evaluated the status of mature B cells in the bone marrow (BM) of SLE-afflicted mice, and determined the effect of treatment with the tolerogenic peptide hCDR1 on these cells. We demonstrate herein that mature B cells of the BM of SLE-afflicted (New Zealand Black x New Zealand White)F(1) mice were largely expanded, and that treatment with hCDR1 down-regulated this population. Moreover, treatment with hCDR1 inhibited the expression of the pathogenic cytokines [interferon-gamma and interleukin (IL)-10], whereas it up-regulated the expression of transforming growth factor-beta in the BM. Treatment with hCDR1 up-regulated the rates of apoptosis of mature B cells. The latter was associated with inhibited expression of the survival Bcl-xL gene and of IL-7 by BM cells. Furthermore, the addition of recombinant IL-7 abrogated the suppressive effects of hCDR1 on Bcl-xL in the BM cells and resulted in elevated levels of apoptosis. Hence, the down-regulated production of IL-7 contributes to the hCDR1-mediated apoptosis of mature B cells in the BM of SLE-afflicted mice.


Subject(s)
Antibodies, Monoclonal/immunology , B-Lymphocyte Subsets/immunology , Bone Marrow Cells/immunology , Interleukin-7/antagonists & inhibitors , Lupus Erythematosus, Systemic/immunology , Peptide Fragments/immunology , Animals , Apoptosis/immunology , Disease Models, Animal , Down-Regulation/immunology , Female , Immune Tolerance/immunology , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-7/immunology , Mice , Mice, Inbred Strains , Reverse Transcriptase Polymerase Chain Reaction/methods , Spleen/immunology , Transforming Growth Factor beta/biosynthesis , Up-Regulation/immunology
12.
Clin Immunol ; 131(2): 223-32, 2009 May.
Article in English | MEDLINE | ID: mdl-19188092

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by dysregulated immune responses mediated by T and B cells. A tolerogenic peptide, designated hCDR1, ameliorated the serological and clinical manifestations of SLE in mouse models of lupus. We investigated the role of B-cell activating factor (BAFF) in the beneficial effects of hCDR1. BAFF production was reduced in hCDR1-treated mice in association with diminished production of dsDNA-specific autoantibodies and proteinuria levels. In addition, IFN-gamma and IL-10, which induce BAFF secretion, were down-regulated in hCDR1-treated mice. The reduced levels of BAFF correlated with a lower rate of maturation and differentiation of B cells, and with a decrease in integrin expression and anti-apoptotic gene expression by B cells. Moreover, BAFF signaling through the NF-kB pathways was inhibited in hCDR1-treated mice. Thus, down-regulation of BAFF plays a role in the mechanism of action by which hCDR1 ameliorates lupus manifestations.


Subject(s)
Autoantigens/pharmacology , B-Cell Activating Factor/metabolism , Lupus Erythematosus, Systemic/physiopathology , Nerve Tissue Proteins/pharmacology , Peptides/pharmacology , Animals , Autoantigens/genetics , B-Cell Activating Factor/blood , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Cell Differentiation/drug effects , Down-Regulation/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Integrins/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lupus Erythematosus, Systemic/drug therapy , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/genetics , Peptides/genetics , Peptides/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Spleen/drug effects , Spleen/metabolism , Survival Analysis
13.
Clin Immunol ; 133(1): 61-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19631585

ABSTRACT

Interferon-gamma (IFN-gamma) plays a pathogenic role in systemic lupus erythematosus (SLE). Uncontrolled IFN-gamma signaling may result from a deficiency in the negative regulator, namely, suppressor of cytokine signaling-1 (SOCS-1). We investigated the activation status of IFN-gamma signaling pathway in SLE-afflicted (New-Zealand-BlackxNew-Zealand-White)F1 mice and determined its responsiveness when treating with a tolerogenic peptide, hCDR1, which ameliorates SLE. SOCS-1 was suppressed and pSTAT1 was enhanced in spleen-derived cells from SLE-affected mice as compared with healthy controls. Treatment with hCDR1 reversed the expression of these two molecules in association with clinical amelioration. In vitro stimulation with IFN-gamma resulted in elevated levels of SOCS-1 in cells from both vehicle and hCDR1-treated mice but this effect reached significance only in cells of the latter group, which also exhibited reduced levels of pSTAT1. Thus, SOCS-1 is diminished in SLE-affected mice, and treatment with hCDR1 results in its up-regulation thereby restoring control of IFN-gamma signaling pathway.


Subject(s)
Autoantigens/therapeutic use , Immunosuppression Therapy , Interferon-gamma/immunology , Lupus Erythematosus, Systemic/therapy , Nerve Tissue Proteins/therapeutic use , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Disease Models, Animal , Down-Regulation/genetics , Down-Regulation/immunology , Female , Immune Tolerance/drug effects , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/immunology , Mice , Peptides/therapeutic use , STAT1 Transcription Factor/agonists , Signal Transduction/drug effects , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/agonists , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Up-Regulation/genetics , Up-Regulation/immunology
14.
J Autoimmun ; 33(1): 58-67, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19329278

ABSTRACT

In primary adrenocortical failure (Addison's disease) caused by autoimmunity, autoantibodies to the steroidogenic cytochrome P450 enzyme 21-hydroxylase (21OH) are detected in the majority of patients. It is currently uncertain whether the autoantibodies themselves participate in the pathogenesis, or if they merely reflect an on-going T cell mediated response. The identification of T cells reactive with 21OH, if any, has been hampered by the lack of a high-quality antigen. In the current study recombinant human 21OH has been expressed in Spodoptera frugiperda insect cells using a baculovirus expression system. Recombinant enzymatically active 21OH was purified to apparent homogeneity by immobilized metal ion affinity chromatography. The purified enzyme was highly immunogenic in immunized SJL/J mice, and immune responses to 21OH-derived peptides assayed as T cell proliferation and interferon gamma production could be invoked after priming with the recombinant protein. Furthermore, purified 21OH was recognized by sera from patients with autoimmune Addison's disease, and it could block the binding of radiolabeled in vitro translated 21OH in a sensitive fluid-phase radioimmunoassay. We conclude that the recombinant preparation of 21OH presented here is of sufficient purity and quality to be used for studies of cellular and humoral immunity in autoimmune Addison's disease.


Subject(s)
Addison Disease/immunology , Autoantigens/metabolism , Peptides/metabolism , Steroid 21-Hydroxylase/metabolism , T-Lymphocytes/metabolism , Addison Disease/blood , Addison Disease/diagnosis , Addison Disease/pathology , Animals , Autoantibodies/blood , Autoantigens/chemistry , Autoantigens/genetics , Autoantigens/immunology , Baculoviridae/genetics , Cell Line , Cell Proliferation , Chromatography, Affinity , Cytochrome P-450 Enzyme System/metabolism , Humans , Immune Sera , Immunization , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Peptides/chemistry , Peptides/genetics , Peptides/immunology , Spodoptera/enzymology , Spodoptera/genetics , Steroid 21-Hydroxylase/chemistry , Steroid 21-Hydroxylase/genetics , Steroid 21-Hydroxylase/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transgenes/immunology
15.
J Autoimmun ; 33(1): 77-82, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19346102

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by dysregulation of cytokines, apoptosis, and B- and T-cell functions. The tolerogenic peptide, hCDR1 (Edratide), ameliorated the clinical manifestations of murine lupus via down-regulation of pro-inflammatory cytokines and apoptosis, up-regulation of the immunosuppressive cytokine TGF-beta, and the induction of regulatory T-cells. In the present study, gene expression was determined in peripheral blood mononuclear cells of 9 lupus patients that were treated for 26 weeks with either hCDR1 (five patients), or placebo (four patients). Disease activity was assessed by SLEDAI-2K and the BILAG scores. Treatment with hCDR1 significantly down-regulated the mRNA expression of the pathogenic cytokines IL-1beta, TNF-alpha, IFN-gamma, and IL-10, of BLyS (B-lymphocyte stimulator) and of the pro-apoptotic molecules caspase-3 and caspase-8. In contrast, the treatment up-regulated in vivo gene expression of both TGF-beta and FoxP3. Furthermore, hCDR1 treatment resulted in a significant decrease in SLEDAI-2K (from 8.0+/-2.45 to 4.4+/-1.67; P=0.02) and BILAG (from 8.2+/-2.7 to 3.6+/-2.9; P=0.03) scores. Thus, the tolerogenic peptide hCDR1, immunomodulates, in vivo, the expression of genes that play a role in SLE, consequently restoring the global immune dysregulation of lupus patients. Hence, hCDR1 has a potential role as a novel disease-specific treatment for lupus patients.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Forkhead Transcription Factors/metabolism , Immunotherapy , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/immunology , Peptide Fragments/administration & dosage , Adult , Apoptosis/drug effects , Caspases/genetics , Caspases/immunology , Caspases/metabolism , Complementarity Determining Regions/immunology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Progression , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/physiopathology , Lupus Erythematosus, Systemic/therapy , Male , Middle Aged , Self Tolerance/immunology , Severity of Illness Index , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Treatment Outcome
16.
J Rheumatol ; 45(3): 310-319, 2018 03.
Article in English | MEDLINE | ID: mdl-29335340

ABSTRACT

OBJECTIVE: Anticitrullinated protein antibodies (ACPA) have major diagnostic significance in rheumatoid arthritis (RA). ACPA are directed against different citrullinated antigens, including filaggrin, fibrinogen, vimentin, and collagen. The presence of ACPA is associated with joint damage and extraarticular manifestations, suggesting that ACPA may have a significant role in the pathogenesis of RA. METHODS: To verify the effect of ACPA on RA-immune cells, peripheral blood mononuclear cells (PBMC) from cyclic citrullinated peptide (CCP)-positive patients with RA and healthy controls were cocultured in vitro with ACPA. ACPA-positive stained cells were analyzed by flow cytometry and the effect of ACPA on mRNA expression levels was evaluated by real-time PCR. We tested whether the stimulatory effects induced by ACPA could be inhibited by the addition of a new multiepitope citrullinated peptide (Cit-ME). RESULTS: We found that ACPA bind specifically to PBMC from CCP-positive patients with RA through the Fab portion. ACPA induce upregulation of pathogenic cytokine expression (4- to 13-fold increase) in PBMC derived from CCP-positive patients with RA. Moreover, ACPA upregulated IL-1ß and IL-6 mRNA expression levels by 10- and 6-fold, respectively, compared to control IgG. Cit-ME, a genuine ligand of ACPA, inhibited the ACPA-induced upregulation of IL-1ß and IL-6 by 30%. CONCLUSION: ACPA bind to a limited percentage of PBMC and upregulate inflammatory cytokine expression, suggesting that ACPA is involved in RA pathogenesis. Targeting ACPA to decrease their pathogenic effects might provide a novel direction in developing therapeutic strategies for RA.


Subject(s)
Arthritis, Rheumatoid/blood , Autoantibodies/blood , Citrulline/immunology , Leukocytes, Mononuclear/metabolism , Peptides, Cyclic/metabolism , Transcriptome/immunology , Antibody Affinity/immunology , Citrulline/chemical synthesis , Filaggrin Proteins , Humans , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fc Fragments/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
17.
Endocrinology ; 147(5): 2411-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16497795

ABSTRACT

The steroidogenic enzyme 21-hydroxylase (21OH) is the main autoantigen in autoimmune primary adrenal failure (Addison's disease). Autoantibodies against 21OH are immunological markers of an ongoing autoimmune process but are not directly involved in the tissue destruction. Autoreactive T cells are thought to mediate tissue damage, but the T cell antigen(s) has not been identified. To find out whether 21OH contains important immunodominant epitopes for T cells, we first immunized BALB/c and SJL inbred mouse strains with recombinant 21OH and showed that lymph node cells proliferated effectively following in vitro stimulation with recombinant 21OH (stimulation indices (SI) 20-40). We further synthesized a series of peptides based on 21OH with amino acid sequences with propensity to bind to major histocompatibility complex class II molecules. Only a few peptides could trigger lymphocytes of 21OH-primed mice to proliferate. One of these, 21OH (342-361), stimulated effectively 21OH-primed lymph node cells of SJL mice (SI = 4-8) and also, although to a lesser extent, of BALB/c mice (SI = 2.5). When SJL mice were immunized with 21OH (342-361), the immunizing peptide as well as peptide 21OH (346-361) triggered a significant proliferative response (SI = 24). A peptide from another part of 21OH, namely 21OH (191-202), did not stimulate the 21OH (342-361)-primed cells. Moreover, stimulation of lymph node cells of mice immunized with 21OH (342-361) with 21OH resulted in a significant proliferative response. We conclude that 21OH (342-361) is an immunodominant determinant for T cells in SJL and probably BALB/c mice. 21OH (342-361) corresponds to the substrate binding site of the enzyme. The p342-361 region may be involved in the pathogenesis of autoimmune adrenal failure in humans.


Subject(s)
Addison Disease/immunology , Autoimmune Diseases/immunology , Epitopes, T-Lymphocyte/chemistry , Steroid 21-Hydroxylase/chemistry , Animals , Antibodies, Monoclonal/chemistry , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Proliferation , Dose-Response Relationship, Drug , Epitopes/chemistry , Female , Humans , Immunodominant Epitopes , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Substrate Specificity , T-Lymphocytes/metabolism
18.
J Neuroimmunol ; 177(1-2): 63-75, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16757035

ABSTRACT

A dual altered peptide ligand (APL) composed of the tandemly arranged two single amino acid analogs of two myasthenogenic peptides, p195-212 and p259-271 was demonstrated to down-regulate in vitro and in vivo myasthenia gravis (MG) associated autoreactive responses. In this study, we demonstrate the suppressive properties of the dual APL following immunization with the whole Torpedo AChR (TAChR) and in mice with established experimental autoimmune MG (EAMG). The dual APL acts by up-regulating CD4+ CD25+ cells expressing characteristic regulatory markers along with an associated increase in levels of IL-10 and TGF-beta. The latter cytokine plays a key role in the ameliorating effects of the dual APL.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Immunosuppression Therapy/methods , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/therapy , Peptides/pharmacology , Receptors, Nicotinic/immunology , Animals , Autoimmunity/drug effects , Autoimmunity/immunology , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/immunology , Female , Interleukin-10/immunology , Interleukin-10/metabolism , Ligands , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myasthenia Gravis, Autoimmune, Experimental/physiopathology , Receptors, Interleukin-2/biosynthesis , Subcellular Fractions , Torpedo , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/immunology
19.
Eur Cytokine Netw ; 17(2): 109-16, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16840029

ABSTRACT

IL-1 is one of the most pleiotropic pro-inflammatory and immunostimulatory cytokines. Overproduction of IL-1 has been shown to be involved in the pathogenicity of various autoimmune inflammatory diseases, including systemic lupus erythematosus (SLE). However, the different contributions that the IL-1 agonistic molecules make in their in vivo native milieu, IL-1beta which is mainly secreted against IL-1alpha which is mainly cell-associated, have not been established. Experimental SLE can be induced in mice by injection with monoclonal anti-DNA antibodies bearing a major idiotype designated, 16/6Id. In the present study, experimental SLE was induced in mice deficient in specific IL-1 molecules, i.e. IL-1alpha(-/-), IL-1beta(-/-), IL-1alpha/beta(-/-) (double KO) and in control BALB/c mice. Mice deficient in IL-1beta , i.e. IL-1beta(-/-) and IL-1alpha/beta(-/-) mice, developed lower levels of anti-dsDNA antibodies after immunization with 16/6Id, as compared to IL-1alpha(-/-) or control BALB/c mice. Disease manifestations were milder in mice deficient in IL-1beta expression. The representative cytokine cascade that is characteristic of overt experimental SLE was also shown to be reduced in groups of mice that lacked IL-1beta as compared to mice deficient in IL-1alpha, which is mainly cell-associated. Altogether, our results point to the importance of secretable IL-1beta, rather than cell-associated IL-1alpha, in the immunostimulatory and inflammatory phenomena that mediate the pathogenesis of experimental SLE.


Subject(s)
Disease Models, Animal , Interleukin-1/genetics , Lupus Erythematosus, Systemic/genetics , Animals , Antibodies/blood , Female , Genetic Predisposition to Disease , Immunity, Innate/genetics , Interleukin-1/deficiency , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout
20.
Am J Med Sci ; 331(1): 4-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16415656

ABSTRACT

OBJECTIVE: The objective of this study was to define the prevalence of systemic lupus erythematosus (SLE) in patients with myasthenia gravis (MG). METHODS: Seventy-eight MG patients recruited unselectively from Israeli MG database were evaluated by medical history, physical examination and serology (ANA at 1:100 and anti-ds-DNA at 1:10 dilution) for the presence of SLE, which was defined by the presence of four or more American College of Rheumatology diagnostic criteria. RESULTS: Thirty-one (40%) of our patients were males and 47 (60%) were females. Their mean age at time of the study was 51.5 +/- 14.5 years. Forty patients (51%) had an early-onset disease (<40 years); 90% had generalized and 10% had limited ophthalmic MG. Significant titers of ANA and ds-DNA autoantibodies were observed in 38.5% and 19.2% of the patients. In six (7.7%), a definitive diagnosis of SLE was established (MG was first diagnosed; there was no association with previous thymectomy), three of them revealed lupus-related neurologic manifestations. All six patients were females with an early onset generalized MG. CONCLUSION: High prevalence of SLE and lupus-related autoantibodies exist in female MG patients. Thus, MG patients should be evaluated for the coexistence of SLE, and assessment for MG is suggested in lupus patients with unexplained muscular weakness.


Subject(s)
Lupus Erythematosus, Systemic/epidemiology , Myasthenia Gravis/complications , Adult , Aged , Antibodies, Antinuclear/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lupus Erythematosus, Systemic/complications , Male , Middle Aged , Prevalence
SELECTION OF CITATIONS
SEARCH DETAIL