Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Exp Gerontol ; 38(6): 661-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12814801

ABSTRACT

IL-12 plays a crucial role in a variety of immune responses that are involved in immuno-senesence and autoimmune diseases. Experimental SLE can be induced in mice by the monoclonal anti-DNA autoantibody bearing the 16/6Id. Aging mice are less susceptible to disease induction. We evaluated the in vivo effects of IL-12 on several parameters related to aging and to experimental SLE. Young (2 months) and aging (10 months) mice that were or were not immunized with the 16/6Id were treated with IL-12. IL-12 treated unimmunized BALB/c and C3H.SW mice had significant elevated levels of anti-DNA antibodies. A high percentage of these mice had glomerular immune complex deposits (ICD). IL-12 treatment resulted in significant increase of IL-1 and IFN-gamma and a decrease of IL-10 production in unimmunized mice. The same IL-12 induced changes were observed in mice with experimental SLE but were less pronounced than in normal mice. In general, all the effects in SLE afflicted mice were more prominent in aging than in young mice. Treatment of mice with experimental SLE upregulated IL-1, IL-2, IFNgamma and downregulated IL-10. These changes were observed in the groups of young as well as old mice. In conclusion, administration of IL-12 to aging mice reversed their Th1/Th2 cytokine profile and thus rendered them vulnerable to the induction of experimental SLE.


Subject(s)
Aging/immunology , Interleukin-12/immunology , Lupus Erythematosus, Systemic/immunology , Animals , Antibodies, Antinuclear/biosynthesis , Antigen-Antibody Complex/metabolism , Cytokines/biosynthesis , DNA/immunology , Female , Kidney Glomerulus/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Th1 Cells/immunology , Th2 Cells/immunology
11.
Autoimmunity ; 35(3): 211-9, 2002 May.
Article in English | MEDLINE | ID: mdl-12389646

ABSTRACT

A peptide based on the complementarity determining region (CDR) 3 of a pathogenic anti-DNA monoclonal antibody that bears the 16/6 idiotype (Id) was shown previously to be a dominant T-cell epitope in experimental SLE, and to be capable of inhibiting SLE-associated responses. When injected, concomitant with active immunization with the pathogenic human anti-DNA, 16/6 Id+ mAb, pCDR3 inhibited the proliferation of LN-derived T cells stimulated in vitro with the 16/6 Id mAb. The inhibition of the specific proliferative responses could be reversed by the addition of exogenous IL-2 to the cultures. Analysis of secreted cytokine profile in supernatants of these cultures demonstrated that pCDR3 treatment reduced significantly the levels of both IL-2 and IFN-gamma that were elevated further in cells of the 16/6 Id-immunized mice. The CDR3-based peptide was shown here to immunomodulate in vivo experimental SLE, induced by the human anti-DNA 16/6 Id+ antibody. The beneficial effects of pCDR3 on the clinical manifestations of SLE were associated with downregulation of the Th1-type (IL-2, IFN-gamma) and proinflammatory (TNF-alpha) cytokines, whereas the immunosuppressive cytokine TGF-beta was up regulated.


Subject(s)
Antibodies, Antinuclear/therapeutic use , Complementarity Determining Regions/therapeutic use , DNA/immunology , Lupus Erythematosus, Systemic/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Cytokines/biosynthesis , Epitopes, T-Lymphocyte , Humans , Immunoglobulin Idiotypes/immunology , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Mice
12.
PLoS One ; 8(3): e60394, 2013.
Article in English | MEDLINE | ID: mdl-23555966

ABSTRACT

BACKGROUND: The tolerogenic peptide, hCDR1, ameliorated manifestations of systemic lupus erythematosus (SLE) via the immunomodulation of pro-inflammatory and immunosuppressive cytokines and the induction of regulatory T cells. Because type I interferon (IFN-α) has been implicated to play a role in SLE pathogenesis, we investigated the effects of hCDR1 on IFN-α in a murine model of SLE and in human lupus. METHODOLOGY PRINCIPAL FINDINGS: (NZBxNZW)F1 mice with established SLE were treated with hCDR1 (10 weekly injections). Splenocytes were obtained for gene expression studies by real-time RT-PCR. hCDR1 down-regulated significantly IFN-α gene expression (73% inhibition compared to vehicle treated mice, p = 0.002) in association with diminished clinical manifestations. Further, hCDR1 reduced, in vitro, IFN-α gene expression in peripheral blood mononuclear cells (PBMC) of 10 lupus patients (74% inhibition compared to medium, p = 0.002) but had no significant effects on the expression levels of IFN-α in PBMC of primary anti-phospholipid syndrome patients or of healthy controls. Lupus patients were treated for 24 weeks with hCDR1 (5) or placebo (4) by weekly subcutaneous injections. Blood samples collected, before and after treatment, were frozen until mRNA isolation. A significant reduction in IFN-α was determined in hCDR1 treated patients (64.4% inhibition compared to pretreatment expression levels, p = 0.015). No inhibition was observed in the placebo treated patients. In agreement, treatment with hCDR1 resulted in a significant decrease of disease activity. IFN-α appears to play a role in the mechanism of action of hCDR1 since recombinant IFN-α diminished the immunomodulating effects of hCDR1 on IL-1ß, TGFß and FoxP3 gene expression. CONCLUSIONS SIGNIFICANCE: We reported previously that hCDR1 affected various cell types and immune pathways in correlation to disease amelioration. The present studies demonstrate that hCDR1 is also capable of down-regulating significantly (and specifically to lupus) IFN-α gene expression. Thus, hCDR1 has a potential role as a novel, disease specific treatment for lupus.


Subject(s)
Autoantigens/therapeutic use , Down-Regulation/drug effects , Immunologic Factors/therapeutic use , Interferon-alpha/genetics , Lupus Erythematosus, Systemic/drug therapy , Nerve Tissue Proteins/therapeutic use , Peptides/therapeutic use , Amino Acid Sequence , Animals , Autoantigens/chemistry , Cells, Cultured , Female , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/genetics , Male , Mice , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Peptides/chemistry
13.
J Neuroimmunol ; 232(1-2): 151-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21129786

ABSTRACT

To determine the effect of the tolerogenic peptide hCDR1 on hippocampal neurogenesis, we treated SLE-afflicted (NZBxNZW)F1 mice with hCDR1 (once a week for 10weeks). The treatment resulted in the up-regulation of neurogenesis in the dentate gyrus and restored the NeuN immunoreactivity in brain hippocampi of the mice in association with increased gene expression of IGF-1, NGF and BDNF. Furthermore, hCDR1 treatment significantly up-regulated p-ERK and p-Akt that are suggested to be key components in mediating growth factor-induced neurogenesis. The observed effects of hCDR1 on hippocampal-neurogenesis and on associated signaling pathways suggest a potential role for hCDR1 in CNS lupus.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hippocampus/drug effects , Lupus Vasculitis, Central Nervous System/drug therapy , Neurogenesis/drug effects , Peptide Fragments/therapeutic use , Animals , Blotting, Western , Disease Models, Animal , Enzyme Activation/drug effects , Female , Gene Expression/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunohistochemistry , Lupus Vasculitis, Central Nervous System/metabolism , Lupus Vasculitis, Central Nervous System/pathology , Mice , Nerve Growth Factors/drug effects , Protein Kinases/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
14.
PLoS One ; 4(12): e8447, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20041142

ABSTRACT

BACKGROUND: Regulatory T cells (Tregs) were shown to be central in maintaining immunological homeostasis and preventing the development of autoimmune diseases. Several subsets of Tregs have been identified to date; however, the dynamics of the interactions between these subsets, and their implications on their regulatory functions are yet to be elucidated. METHODOLOGY/PRINCIPAL FINDINGS: We employed a combination of mathematical modeling and frequent in vivo measurements of several T cell subsets. Healthy BALB/c mice received a single injection of either hCDR1--a tolerogenic peptide previously shown to induce Tregs, a control peptide or vehicle alone, and were monitored for 16 days. During this period, splenocytes from the treated mice were analyzed for the levels of CD4, CD25, CD8, CD28 and Foxp3. The collected data were then fitted to mathematical models, in order to test competing hypotheses regarding the interactions between the followed T cell subsets. In all 3 treatment groups, a significant, lasting, non-random perturbation of the immune system could be observed. Our analysis predicted the emergence of functional CD4 Tregs based on inverse oscillations of the latter and CD4(+)CD25(-) cells. Furthermore, CD4 Tregs seemed to require a sufficiently high level of CD8 Tregs in order to become functional, while conversion was unlikely to be their major source. Our results indicated in addition that Foxp3 is not a sufficient marker for regulatory activity. CONCLUSIONS/SIGNIFICANCE: In this work, we unraveled the dynamics of the interplay between CD4, CD8 Tregs and effector T cells, using, for the first time, a mathematical-mechanistic perspective in the analysis of Treg kinetics. Furthermore, the results obtained from this interdisciplinary approach supported the notion that CD4 Tregs need to interact with CD8 Tregs in order to become functional. Finally, we generated predictions regarding the time-dependent function of Tregs, which can be further tested empirically in future work.


Subject(s)
CD4 Antigens/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Communication , Interleukin-2 Receptor alpha Subunit/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , CD28 Antigens/metabolism , CD8 Antigens/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Kinetics , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Mice , Mice, Inbred BALB C , Models, Immunological , Spleen/cytology
15.
Hum Immunol ; 70(3): 139-45, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19280712

ABSTRACT

A tolerogenic peptide, hCDR1, ameliorated murine lupus via the upregulation of functional regulatory cells and by immunomodulating cytokine production. In the present study we analyzed the ability of hCDR1 to similarly affect gene expression and regulatory T cells when incubated with peripheral blood mononuclear cells (PBMC) of lupus patients. To this end, peripheral blood mononuclear cells (PBMC) of 11 lupus patients and five gender- and age-matched healthy controls were cultured with hCDR1 or a control peptide. Gene expression and regulatory T-cells were assessed. hCDR1 significantly downregulated interleukin (IL)-1beta, interferon (IFN)-gamma, and IL-10 gene expression. Furthermore, hCDR1 upregulated the expression of the anti-apoptotic Bcl-xL molecule and downregulated the pro-apoptotic caspase-3, resulting in reduced rates of apoptosis. hCDR1 increased the expression of transforming growth factor (TGF)-beta, FoxP3 and the negative regulators Foxj1 and Foxo3a. No significant effects were observed using a control peptide or when PBMC of healthy donors were incubated with hCDR1. The elevated gene expression of FoxP3 was due to hCDR1-induced upregulation of TGF-beta, resulting in an increase of CD4+CD25+FoxP3+ functional, regulatory cells. The ability of the regulatory cells to diminish IFN-gamma expression and to upregulate TGF-beta was abrogated after the addition of a neutralizing anti-CD25 antibody, confirming their role in the beneficial effects of hCDR1.


Subject(s)
Complementarity Determining Regions/immunology , Cytokines/metabolism , Leukocytes, Mononuclear/immunology , Lupus Erythematosus, Systemic/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Apoptosis/immunology , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/pharmacology , Cytokines/antagonists & inhibitors , Down-Regulation/drug effects , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Immunosuppression Therapy , Interleukin-2 Receptor alpha Subunit/biosynthesis , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Lupus Erythematosus, Systemic/blood , Lymphocyte Activation/drug effects , Male , Middle Aged , Peptides/chemistry , Peptides/immunology , Peptides/pharmacology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Up-Regulation/drug effects
16.
Arthritis Rheum ; 56(7): 2371-81, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17599765

ABSTRACT

OBJECTIVE: To identify genes that are differently expressed in (NZB x NZW)F(1) mice with established lupus compared with healthy controls, and to determine how gene expression is affected by treatment with hCDR1 (Edratide), a peptide synthesized on the basis of the sequence of the first complementarity-determining region (CDR1) of an autoantibody. METHODS: RNA was extracted from spleen cells of young, disease-free mice and of older mice with systemic lupus erythematosus (SLE) that were treated with hCDR1 or with vehicle alone. Gene expression was assessed using the DNA microarray technique and verified by real-time reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS: In mice with SLE, numerous genes showed increased or decreased expression relative to that in the disease-free controls. Treatment with hCDR1 restored the expression of many of these genes to control levels. Real-time RT-PCR verified that in diseased mice RNA transcripts of Tnfsf4, Il5ra, Zbtb20, and Nid1 were up-regulated, while transcripts of Tfpi and S100a8 were down-regulated, and confirmed the effects of hCDR1 on the expression of those genes. Kidney immunostaining demonstrated that the up-regulated expression of OX40 ligand, which is a protein product of the gene tumor necrosis factor (ligand) superfamily member 4, in diseased mice was reduced by hCDR1. CONCLUSION: Expression of numerous genes in mice with SLE differs from that in young, disease-free control mice. Treatment with hCDR1 restores the expression of 22% of these genes to levels similar to those in controls. Thus, one of the mechanisms by which hCDR1 exerts its beneficial effects on the clinical symptoms of SLE is through regulation of gene expression.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Gene Expression Regulation/drug effects , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/genetics , Peptide Fragments/therapeutic use , Amino Acid Sequence , Animals , DNA Primers , Disease Models, Animal , Female , Mice , Mice, Inbred Strains , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Peptide Fragments/chemistry , RNA/isolation & purification , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Spleen/pathology
17.
Clin Immunol ; 119(2): 146-55, 2006 May.
Article in English | MEDLINE | ID: mdl-16503419

ABSTRACT

A peptide (hCDR1) based on the sequence of the complementarity-determining region-1 of an anti-DNA autoantibody ameliorates clinical manifestations of lupus. We analyzed the beneficial effects of hCDR1 when given alone or in combination with dexamethasone, while comparing the mechanisms of action of the latter. Treatment with either hCDR1 or dexamethasone, or a combination of the latter significantly reduced titers of dsDNA-specific autoantibodies, levels of proteinuria, and intensity of glomerular immune complex deposits. Both drugs down-regulated the secretion and expression of IFN-gamma and IL-10, but only treatment with hCDR1 up-regulated TGF-beta. While both drugs reduced the expression of Fas ligand (FasL) and caspase 8, treatment with hCDR1 resulted in reduced whereas dexamethasone administration resulted in increased rate of apoptosis. Furthermore, down-regulation of FasL appeared to play a role in cytokine modulation. We conclude that specific treatment with hCDR1 ameliorates murine lupus via distinct mechanisms of action than those of dexamethasone.


Subject(s)
Apoptosis/immunology , Autoantibodies/physiology , Complementarity Determining Regions/physiology , Cytokines/biosynthesis , Dexamethasone/pharmacology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Peptide Fragments/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cytokines/metabolism , Fas Ligand Protein , Female , Humans , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred NZB , Tumor Necrosis Factors/metabolism
18.
Proc Natl Acad Sci U S A ; 103(23): 8810-5, 2006 Jun 06.
Article in English | MEDLINE | ID: mdl-16735466

ABSTRACT

Systemic lupus erythematosus is an autoimmune disease characterized by autoantibodies and systemic clinical manifestations. A peptide, designated hCDR1, based on the complementarity-determining region (CDR) 1 of an autoantibody, ameliorated the serological and clinical manifestations of lupus in both spontaneous and induced murine models of lupus. The objectives of the present study were to determine the mechanism(s) underlying the beneficial effects induced by hCDR1. Adoptive transfer of hCDR1-treated cells to systemic lupus erythematosus-afflicted (NZBxNZW)F1 female mice down-regulated all disease manifestations. hCDR1 treatment up-regulated (by 30-40%) CD4+CD25+ cells in association with CD45RBlow, cytotoxic T lymphocyte antigen 4, and Foxp3 expression. Depletion of the CD25+ cells diminished significantly the therapeutic effects of hCDR1, whereas administration of the enriched CD4+CD25+ cell population was beneficial to the diseased mice. Amelioration of disease manifestations was associated with down-regulation of the pathogenic cytokines (e.g., IFN-gamma and IL-10) and up-regulation of the immunosuppressive cytokine TGF-beta, which substantially contributed to the suppressed autoreactivity. TGF-beta was secreted by CD4+ cells that were affected by hCDR1-induced immunoregulatory cells. The hCDR1-induced CD4+CD25+ cells suppressed autoreactive CD4+ cells, resulting in reduced rates of activation-induced apoptosis. Thus, hCDR1 ameliorates lupus through the induction of CD4+CD25+ cells that suppress activation of the autoreactive cells and trigger the up-regulation of TGF-beta.


Subject(s)
Autoantibodies/immunology , CD4-Positive T-Lymphocytes/immunology , Complementarity Determining Regions/immunology , Lupus Vulgaris/immunology , Peptides/immunology , Receptors, Interleukin-2/immunology , Transforming Growth Factor beta/genetics , Animals , Autoantibodies/chemistry , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Death , Complementarity Determining Regions/chemistry , Female , Humans , Immunotherapy, Adoptive , Lupus Vulgaris/drug therapy , Lymphocyte Activation/immunology , Mice , Peptides/chemistry , Spleen/cytology , Transforming Growth Factor beta/metabolism , Up-Regulation
19.
Clin Immunol ; 117(3): 262-70, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16257268

ABSTRACT

Treatment with peptides based on the complementarity determining regions (CDR) of murine and human monoclonal anti-DNA antibodies that bear the common idiotype, 16/6 Id, ameliorates disease manifestations of mice with either induced or spontaneous SLE. Aberrant expression and function of the p21Ras/MAP kinase pathway are associated with active SLE. Therefore, we examined the effect of treatment with a CDR1-based peptide of a human autoantibody (hCDR1) on the p21Ras pathway and SLE manifestations of SLE-prone (NZBxNZW)F1 mice. Untreated SLE-afflicted mice demonstrated increased expression of p21Ras and the phosphorylated active form of its down-stream element JNK kinase in conjunction with reduced hSOS and unchanged p120GAP, as compared to healthy controls. Amelioration of SLE manifestations following treatment with hCDR1 was associated with a diminished expression of phosphorylated JNK kinase, mainly in the T cell population that also exhibited reduced rates of apoptosis. Thus, hCDR1 therapy ameliorates SLE, at least in part, via down-regulation of the activity of the pro-apoptotic JNK kinase.


Subject(s)
Autoantibodies/drug effects , Autoimmune Diseases/immunology , Complementarity Determining Regions/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , MAP Kinase Kinase 4/metabolism , Peptide Fragments/therapeutic use , Animals , Apoptosis/immunology , Autoantibodies/immunology , Autoimmune Diseases/genetics , Crosses, Genetic , Disease Models, Animal , Down-Regulation , Female , Immunization , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred NZB , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology
20.
J Immunol ; 175(11): 7255-63, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16301630

ABSTRACT

Systemic lupus erythematosus (SLE), which is characterized by the increased production of autoantibodies and defective T cell responses, can be induced in mice by immunization with a human anti-DNA mAb that expresses a major Id, designated 16/6Id. A peptide based on the sequence of the CDR1 of the 16/6Id (human CDR1 (hCDR1)) ameliorated the clinical manifestations of SLE and down-regulated, ex vivo, the 16/6Id-induced T cell proliferation. In this study, we examined the mechanism responsible for the hCDR1-induced modulation of T cell functions related to the pathogenesis of SLE. We found that injection of hCDR1 into BALB/c mice concomitant with their immunization with 16/6Id resulted in a marked elevation of TGF-beta secretion 10 days later. Addition of TGF-beta suppressed the 16/6Id-stimulated T cell proliferation similarly to hCDR1. In addition, we provide evidence that one possible mechanism underlying the hCDR1- and TGFbeta-induced inhibition of T cell proliferation is by down-regulating the expression, and therefore the functions, of a pair of key cell adhesion receptors, LFA-1 (alphaLbeta2) and CD44, which operate as accessory molecules in mediating APC-T cell interactions. Indeed, T cells of mice treated with hCDR1 showed a TGF-beta-induced suppression of adhesion to the LFA-1 and CD44 ligands, hyaluronic acid and ICAM-1, respectively, induced by stromal cell-derived factor-1alpha and PMA. The latter suppression is through the inhibition of ERK phosphorylation. Thus, the down-regulation of SLE-associated responses by hCDR1 treatment may be due to the effect of the up-regulated TGF-beta on the expression and function of T cell adhesion receptors and, consequently, on T cell stimulation, adhesion, and proliferation.


Subject(s)
Antibodies, Antinuclear/immunology , Autoimmunity/immunology , Hyaluronan Receptors/immunology , Lymphocyte Function-Associated Antigen-1/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Adhesion/immunology , Cell Communication/immunology , Cell Proliferation , Complementarity Determining Regions/immunology , Disease Models, Animal , Flow Cytometry , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Immunoglobulin Idiotypes , Intercellular Adhesion Molecule-1/metabolism , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred BALB C , Peptides/immunology , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL