Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Front Immunol ; 12: 629807, 2021.
Article in English | MEDLINE | ID: mdl-33936042

ABSTRACT

Autoantigen-directed tolerance can be induced by certain nucleosomal histone peptide epitope/s in nanomolar dosage leading to sustained remission of disease in mice with spontaneous SLE. By contrast, lupus is accelerated by administration of intact (whole) histones, or whole nucleosomes in microparticles from apoptotic cells, or by post-translationally acetylated histone-peptides. Low-dose therapy with the histone-peptide epitopes simultaneously induces TGFß and inhibits IL-6 production by DC in vivo, especially pDC, which then induce CD4+CD25+ Treg and CD8+ Treg cells that suppress pathogenic autoimmune response. Both types of induced Treg cells are FoxP3+ and act by producing TGFß at close cell-to-cell range. No anaphylactic adverse reactions, or generalized immunosuppression have been detected in mice injected with the peptides, because the epitopes are derived from evolutionarily conserved histones in the chromatin; and the peptides are expressed in the thymus during ontogeny, and their native sequences have not been altered. The peptide-induced Treg cells can block severe lupus on adoptive transfer reducing inflammatory cell reaction and infiltration in the kidney. In Humans, similar potent Treg cells are generated by the histone peptide epitopes in vitro in lupus patients' PBMC, inhibiting anti-dsDNA autoantibody and interferon production. Furthermore, the same types of Treg cells are generated in lupus patients who are in very long-term remission (2-8 years) after undergoing autologous hematopoietic stem cell transplantation. These Treg cells are not found in lupus patients treated conventionally into clinical remission (SLEDAI of 0); and consequently they still harbor pathogenic autoimmune cells, causing subclinical damage. Although antigen-specific therapy with pinpoint accuracy is suitable for straight-forward organ-specific autoimmune diseases, Systemic Lupus is much more complex. The histone peptide epitopes have unique tolerogenic properties for inhibiting Innate immune cells (DC), T cells and B cell populations that are both antigen-specifically and cross-reactively involved in the pathogenic autoimmune response in lupus. The histone peptide tolerance is a natural and non-toxic therapy suitable for treating early lupus, and also maintaining lupus patients after toxic drug therapy. The experimental steps, challenges and possible solutions for successful therapy with these peptide epitopes are discussed in this highly focused review on Systemic Lupus.


Subject(s)
Autoimmunity , Histones/pharmacology , Lupus Erythematosus, Systemic/immunology , Nucleosomes/chemistry , Peptide Fragments/pharmacology , Animals , DNA/immunology , Down-Regulation , Epitopes , Histones/immunology , Humans , Immune Tolerance , Interleukin-2/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Lymphocytes/immunology , Mice , T-Lymphocytes, Regulatory/immunology
3.
Bone Marrow Transplant ; 53(6): 692-700, 2018 06.
Article in English | MEDLINE | ID: mdl-29855561

ABSTRACT

Some patients with systemic lupus erythematosus (SLE) are refractory to traditional therapies, dependent on chronic corticosteroids, have organ damage, and are at high risk of mortality. In this group of patients, we report outcome at a median of five years after autologous hematopoietic stem cell transplant (HSCT) using two different non-myeloablative regimens. Four patients received a conditioning regimen of cyclophosphamide (200 mg/kg) and alemtuzumab (60 mg), while 26 patients underwent conditioning with cyclophosphamide (200 mg/kg), rATG (Thymoglobulin) (5.5 mg/kg), and rituximab 1000 mg. Unselected peripheral blood stem cells were infused on day 0. There were no treatment related deaths. Of the four patients treated with cyclophosphamide and alemtuzumab, none entered remission. For the 26 patients treated with cyclophosphamide, rATG, and rituximab, disease remission defined as no immune suppressive drugs except hydroxychloroquine and/or 10 mg or less of prednisone a day was 92% at 6 months, 92% at one year, 81% at 2 years, 71% at 3 years, and 62% at 4 and 5 years post-HSCT. Autologous HSCT outcome is dependent on the conditioning regimen but prior organ damage may cause lingering symptoms.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Lupus Erythematosus, Systemic/therapy , Peripheral Blood Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Adult , Chronic Disease , Female , Follow-Up Studies , Humans , Lupus Erythematosus, Systemic/pathology , Male , Treatment Outcome
4.
Clin Immunol ; 162: 9-26, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26521071

ABSTRACT

We studied effects of early and late apoptotic (necroptotic) cell products, related damage associated alarmins and TLR agonists, on hematopoietic stem and progenitor cells (HSPC). Surprisingly, normal HSPC themselves produced IL-17 and IL-21 after 1½days of stimulation, and the best stimulators were TLR 7/8 agonist; HMGB1-DNA; TLR 9 agonist, and necroptotic B cells. The stimulated HSPC expressed additional cytokines/mediators, directly causing rapid expansion of IL-17(+) memory CD4 T (Th17), and CD8 T (Tc17) cells, and antigen-experienced IL-17(+) T cells with "naïve" phenotype. In lupus marrow, HSPC were spontaneously pre-stimulated by endogenous signals to produce IL-17 and IL-21. In contrast to HSPC, megakaryocyte progenitors (MKP) did not produce IL-17, and unlike HSPC, they could process and present particulate apoptotic autoantigens to augment autoimmune memory Th17 response. Thus abnormally stimulated primitive hematopoietic progenitors augment expansion of IL-17 producing immune and autoimmune memory T cells in the bone marrow, which may affect central tolerance.


Subject(s)
Bone Marrow/immunology , Cytokines/immunology , Hematopoietic Stem Cells/immunology , Lupus Erythematosus, Systemic/immunology , Multipotent Stem Cells/immunology , Th17 Cells/immunology , Animals , Apoptosis/immunology , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Flow Cytometry , Humans , Interleukin-17/immunology , Interleukins/immunology , Lupus Erythematosus, Systemic/blood , Mice , Toll-Like Receptors
5.
Nat Commun ; 6: 7086, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25963626

ABSTRACT

Dendritic cells (DCs) are critical for immune homeostasis. To target DCs, we generated a mouse line with Flip deficiency in cells that express cre under the CD11c promoter (CD11c-Flip-KO). CD11c-Flip-KO mice spontaneously develop erosive, inflammatory arthritis, resembling rheumatoid arthritis, which is dramatically reduced when these mice are crossed with Rag(-/-) mice. The CD8α(+) DC subset is significantly reduced, along with alterations in NK cells and macrophages. Autoreactive CD4(+) T cells and autoantibodies specific for joint tissue are present, and arthritis severity correlates with the number of autoreactive CD4(+) T cells and plasmablasts in the joint-draining lymph nodes. Reduced T regulatory cells (Tregs) inversely correlate with arthritis severity, and the transfer of Tregs ameliorates arthritis. This KO line identifies a model that will permit in depth interrogation of the pathogenesis of rheumatoid arthritis, including the role of CD8α(+) DCs and other cells of the immune system.


Subject(s)
Arthritis/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CD11c Antigen/metabolism , Inflammation/metabolism , Animals , Arthritis/genetics , Arthritis/pathology , Autoantibodies , Autoimmune Diseases , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CD11c Antigen/genetics , CD4-Positive T-Lymphocytes , Dendritic Cells , Female , Inflammation/genetics , Male , Mice , Mice, Knockout , T-Lymphocytes, Regulatory
6.
Immunol Lett ; 163(1): 84-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25454068

ABSTRACT

Platelets, like stromal cells, present antigen only via MHC class I, but the immune potential of their progenitors has not been explored in humans. We derived CD34(+)CD117(+)CD41(+)CD151(+) megakaryocyte progenitors (MKp) in vitro from mobilized peripheral blood hematopoietic stem and progenitor cells (HSPC) of normal subjects using culture conditions akin to bone marrow niche, or organs that support extramedullary hematopoiesis. The MKp expressed MHC Class II in contrast to platelets and functioned as professional APC before they matured further. Moreover, MKp constitutively expressed mRNA encoding mediators for human Th17 expansion, including IL-1, IL-18, IL-6, TGFß, IL-23, BAFF, and COX2. MKp also expressed high levels of type I interferon and IRF5 mRNA. In contrast to platelets, MKp augmented the expansion of Th17, Th1, and potent Th17/Th1 double-positive cells in normal PBMC and CD4 line T cells from normal subjects or lupus patients. The Th cell augmentation involved pre-committed memory cells, and was significant although modest, because only non-cognate MKp-T cell interactions could be studied, under non-polarizing conditions. Importantly, the MKp-mediated expansion was observed in the presence or absence of direct MKp-T cell contact. Furthermore, MKp augmented Th17 responses against Candida albicans, a serious opportunistic pathogen. These results indicate an immunologic role of MKp in situations associated with extramedullary hematopoiesis and mobilization of HSPC.


Subject(s)
Antigen-Presenting Cells/immunology , Gene Expression Regulation , Histocompatibility Antigens Class II/immunology , Megakaryocyte Progenitor Cells/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Antigen-Presenting Cells/cytology , Antigens, CD/immunology , Blood Platelets/cytology , Blood Platelets/immunology , Cell Communication/immunology , Cytokines/immunology , Humans , Megakaryocyte Progenitor Cells/cytology , Th1 Cells/cytology , Th17 Cells/cytology
7.
Clin Immunol ; 149(3): 365-78, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24211843

ABSTRACT

Low-dose tolerance therapy with nucleosomal histone peptide epitopes blocks lupus disease in mouse models, but effect in humans is unknown. Herein, we found that CD4(+)CD25(high)FoxP3(+) or CD4(+)CD45RA(+)FoxP3(low) T-cells, and CD8(+)CD25(+)FoxP3(+) T-cells were all induced durably in PBMCs from inactive lupus patients and healthy subjects by the histone peptide/s themselves, but in active lupus, dexamethasone or hydroxychloroquine unmasked Treg-induction by the peptides. The peptide-induced Treg depended on TGFß/ALK-5/pSmad 2/3 signaling, and they expressed TGF-ß precursor LAP. Lupus patients' sera did not inhibit Treg induction. The peptide epitope-induced T cells markedly suppressed type I IFN related gene expression in lupus PBMC. Finally, the peptide epitopes suppressed pathogenic autoantibody production by PBMC from active lupus patients to baseline levels by additional mechanisms besides Treg induction, and as potently as anti-IL6 antibody. Thus, low-dose histone peptide epitopes block pathogenic autoimmune response in human lupus by multiple mechanisms to restore a stable immunoregulatory state.


Subject(s)
Autoimmunity/drug effects , CD8-Positive T-Lymphocytes/drug effects , Histones/immunology , Lupus Erythematosus, Systemic/immunology , Peptides/pharmacology , T-Lymphocytes, Regulatory/drug effects , Adult , Anti-Inflammatory Agents/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , Autoantibodies/biosynthesis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Epitopes , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Histones/chemistry , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation , Male , Middle Aged , Nucleosomes/chemistry , Nucleosomes/immunology , Peptides/immunology , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/immunology , Smad3 Protein/genetics , Smad3 Protein/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
8.
J Immunol ; 188(12): 5970-80, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22561152

ABSTRACT

In search of autoantigen-presenting cells that prime the pathogenic autoantibody-inducing Th cells of lupus, we found that CD41(+)CD151(+) cells among Lineage(-) (Lin(-)) CD117(+) (c-Kit(+)) CX3CR1(-) splenocytes depleted of known APCs were most proficient in presenting nuclear autoantigens from apoptotic cells to induce selectively an autoimmune Th17 response in different lupus-prone mouse strains. The new APCs have properties resembling megakaryocyte and/or bipotent megakaryocyte/erythroid progenitors of bone marrow, hence they are referred to as MM cells in this study. The MM cells produce requisite cytokines, but they require contact for optimal Th17 induction upon nucleosome feeding, and can induce Th17 only before undergoing differentiation to become c-Kit(-)CD41(+) cells. The MM cells expand up to 10-fold in peripheral blood of lupus patients and 49-fold in spleens of lupus mice preceding disease activity; they accelerate lupus in vivo and break tolerance in normal mice, inducing autoimmune Th17 cells. MM cells also cause Th17 skewing to foreign Ag in normal mice without Th17-polarizing culture conditions. Several molecules in MM cells are targets for blocking of autoimmunization. This study advances our understanding of lupus pathogenesis and Th17 differentiation biology by characterizing a novel category of APC.


Subject(s)
Antigen-Presenting Cells/immunology , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , Megakaryocyte Progenitor Cells/immunology , Th17 Cells/immunology , Adult , Animals , Antigen Presentation/immunology , Autoantibodies/immunology , Autoantigens/immunology , Cell Differentiation/immunology , Cell Separation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Humans , Mice , Mice, Mutant Strains , Middle Aged , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Th17 Cells/cytology
9.
J Clin Immunol ; 31(3): 379-94, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21287397

ABSTRACT

Tolerance therapy with nucleosomal histone peptides H4(71-94), H4(16-39), or H1'(22-42) controls disease in lupus-prone SNF1 mice. It would be clinically important to determine whether a cocktail of the above epitopes would be superior. Herein, we found that compared with cocktail peptides, H4(71-94) monotherapy more effectively delayed nephritis onset, prolonged lifespan, diminished immunoglobulin G autoantibody levels, reduced autoantigen-specific Th1 and Th17 responses and frequency of T(FH) cells in spleen and the helper ability of autoimmune T cells to B cells, by inducing potent CD8 Treg cells. H4(71-94) therapy was superior in "tolerance spreading," suppressing responses to other autoepitopes, nucleosomes, and ribonucleoprotein. We also developed an in vitro assay for therapeutic peptides (potentially in humans), which showed that H4(71-94), without exogenous transforming growth factor (TGF)-ß, was efficient in inducing stable CD4(+)CD25(+)Foxp3(+) T cells by decreasing interleukin 6 and increasing TGF-ß production by dendritic cells that induced ALK5-dependent Smad-3 phosphorylation (TGF-ß signal) in target autoimmune CD4(+) T cells.


Subject(s)
Autoimmunity/drug effects , Histones/pharmacology , Immune Tolerance/drug effects , Immunoassay , Immunologic Factors/pharmacology , Lupus Nephritis/drug therapy , Peptides/pharmacology , Animals , Autoantibodies/immunology , Autoantigens/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Drug Combinations , Epitopes/immunology , Female , Flow Cytometry , Histones/chemical synthesis , Histones/immunology , Immune Tolerance/immunology , Immunologic Factors/chemical synthesis , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Nucleosomes/immunology , Nucleosomes/metabolism , Peptides/chemical synthesis , Peptides/immunology
10.
J Immunol ; 183(10): 6346-58, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19841178

ABSTRACT

Compared with conventional drug therapy, autologous hemopoietic stem cell transplantation (HSCT) can induce very-long-term remission in refractory lupus patients. Herein, we show that in posttransplant patients, both CD4(+)CD25(high)FoxP3(+) and an unusual CD8(+)FoxP3(+) Treg subset return to levels seen in normal subjects; accompanied by almost complete inhibition of pathogenic T cell response to critical peptide autoepitopes from histones in nucleosomes, the major lupus autoantigen from apoptotic cells. In addition to a stably sustained elevation of FoxP3, posttransplant CD8 T cells also maintained markedly higher expression levels of latency-associated peptide (LAP), CD103, PD-1, PD-L1, and CTLA-4, as compared with pretransplant CD8 T cells that were identically treated by a one-time activation and rest in short-term culture. The posttransplant CD8 regulatory T cells (Treg) have autoantigen-specific and nonspecific suppressive activity, which is contact independent and predominantly TGF-beta dependent. By contrast, the pretransplant CD8 T cells have helper activity, which is cell contact dependent. Although CD4(+)CD25(high) Treg cells return during clinical remission of conventional drug-treated lupus, the posttransplant patient's CD8 Treg cells are considerably more potent, and they are absent in drug-treated patients in whom CD4 T cell autoreactivity to nucleosomal epitopes persists even during clinical remission. Therefore, unlike conventional drug therapy, hemopoietic stem cell transplantation generates a newly differentiated population of LAP(high)CD103(high) CD8(TGF-beta) Treg cells, which repairs the Treg deficiency in human lupus to maintain patients in true immunological remission.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation , Lupus Erythematosus, Systemic/surgery , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Antigens, CD/immunology , Antigens, CD/metabolism , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , B7-H1 Antigen , CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen , Female , Humans , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-13/immunology , Interleukin-13/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Male , Middle Aged , Nucleosomes/immunology , Nucleosomes/metabolism , Programmed Cell Death 1 Receptor , Remission, Spontaneous , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Young Adult
11.
Arthritis Res Ther ; 11(2): R59, 2009.
Article in English | MEDLINE | ID: mdl-19405952

ABSTRACT

INTRODUCTION: Lupus patients need alternatives to steroids and cytotoxic drugs. We recently found that apigenin, a non-mutagenic dietary flavonoid, can sensitize recurrently activated, normal human T cells to apoptosis by inhibiting nuclear factor-kappa-B (NF-kappaB)-regulated Bcl-xL, cyclooxygenase 2 (COX-2), and cellular FLICE-like inhibitory protein (c-FLIP) expression. Because sustained immune activation and hyperexpression of COX-2 and c-FLIP contribute to lupus, we treated SNF1 mice that spontaneously develop human lupus-like disease with apigenin. METHODS: SNF1 mice with established lupus-like disease were injected with 20 mg/kg of apigenin daily and then monitored for development of severe nephritis. Histopathologic changes in kidneys, IgG autoantibodies to nuclear autoantigens in serum and in cultures of splenocytes, along with nucleosome-specific T helper 1 (Th1) and Th17 responses, COX-2 expression, and apoptosis of lupus immune cells were analyzed after apigenin treatment. RESULTS: Apigenin in culture suppressed responses of Th1 and Th17 cells to major lupus autoantigen (nucleosomes) up to 98% and 92%, respectively, and inhibited the ability of lupus B cells to produce IgG class-switched anti-nuclear autoantibodies helped by these Th cells in presence of nucleosomes by up to 82%. Apigenin therapy of SNF1 mice with established lupus suppressed serum levels of pathogenic autoantibodies to nuclear antigens up to 97% and markedly delayed development of severe glomerulonephritis. Apigenin downregulated COX-2 expression in lupus T cells, B cells, and antigen-presenting cells (APCs) and caused their apoptosis. Autoantigen presentation and Th17-inducing cytokine production by dendritic cells were more sensitive to the inhibitory effect of apigenin in culture, as evident at 0.3 to 3 muM, compared with concentrations (10 to 100 microM) required for inducing apoptosis. CONCLUSIONS: Apigenin inhibits autoantigen-presenting and stimulatory functions of APCs necessary for the activation and expansion of autoreactive Th1 and Th17 cells and B cells in lupus. Apigenin also causes apoptosis of hyperactive lupus APCs and T and B cells, probably by inhibiting expression of NF-kappaB-regulated anti-apoptotic molecules, especially COX-2 and c-FLIP, which are persistently hyperexpressed by lupus immune cells. Increasing the bioavailability of dietary plant-derived COX-2 and NF-kappaB inhibitors, such as apigenin, could be valuable for suppressing inflammation in lupus and other Th17-mediated diseases like rheumatoid arthritis, Crohn disease, and psoriasis and in prevention of inflammation-based tumors overexpressing COX-2 (colon, breast).


Subject(s)
Antigen Presentation/drug effects , Apigenin/pharmacology , Autoantigens/immunology , Lupus Erythematosus, Systemic/immunology , T-Lymphocyte Subsets/drug effects , Th1 Cells/drug effects , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Apoptosis/drug effects , Autoantibodies/biosynthesis , Autoantibodies/blood , Autoantibodies/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cyclooxygenase 2 , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/drug effects , Interferon-gamma/biosynthesis , Interferon-gamma/drug effects , Interleukin-17/immunology , Lupus Nephritis/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred NZB , Nucleosomes/immunology , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology
13.
Immunol Lett ; 121(1): 74-83, 2008 Nov 16.
Article in English | MEDLINE | ID: mdl-18812189

ABSTRACT

Resistance of T cells to activation-induced cell death (AICD) is associated with autoimmunity and lymphoproliferation. We found that apigenin (4',5,7-trihydroxyflavone), a non-mutagenic dietary flavonoid, augmented both extrinsic and intrinsic pathways of apoptosis in recurrently activated, but not in primarily stimulated, human blood CD4+ T cells. Apigenin potentiated AICD by inhibiting NF-kappaB activation and suppressing NF-kappaB-regulated anti-apoptotic molecules, cFLIP, Bcl-x(L), Mcl-1, XIAP and IAP, but not Bcl-2. Apigenin suppressed NF-kappaB translocation to nucleus and inhibited IkappaBalpha phosphorylation and degradation in response to TCR stimulation in reactivated peripheral blood CD4 T cells, as well as in leukemic Jurkat T cell lines. Among the pathways that lead to NF-kappaB activation upon TCR stimulation, apigenin selectively inhibited PI3K-PKB/Akt, but not PKC-theta activation in the human T cells, and synergized with a PI3K inhibitor to markedly augment AICD. Apigenin also suppressed expression of anti-apoptotic cyclooxygenase 2 (COX-2) protein in activated human T cells, but it did not affect activation of Erk MAPKinase. Thus, in chronically activated human T cells, relatively non-toxic apigenin can suppress anti-apoptotic pathways involving NF-kappaB activation, and especially cFLIP and COX-2 expression that are important for functioning and maintenance of immune cells in inflammation, autoimmunity and lymphoproliferation.


Subject(s)
Apigenin/administration & dosage , Apoptosis Regulatory Proteins/immunology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Proliferation/drug effects , Chromones/pharmacology , Cyclooxygenase 2/metabolism , Diet , Humans , I-kappa B Kinase/metabolism , Morpholines/pharmacology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
14.
Arthritis Rheum ; 56(12): 4132-41, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18050205

ABSTRACT

OBJECTIVE: To investigate the role of cyclooxygenase 2 (COX-2) in the functioning of different cell types involved in the lupus autoimmune response, and to examine the therapeutic effect of COX-2 inhibitors in mice prone to spontaneously develop systemic lupus erythematosus (SLE). METHODS: Lupus-prone (SWR x NZB)F(1) mice were fed with a diet containing different doses of the COX-2-specific inhibitor celecoxib or the nonspecific inhibitor aspirin, or a combination of both, and the effects of the therapy on autoantibody production, development of lupus nephritis, and mortality were determined. Expression of COX-2 by different cells of the lupus immune system and the effect of COX-2 inhibitors on the function of these cells in vitro and in vivo were assessed. RESULTS: The immune cells of mice with SLE spontaneously hyperexpressed COX-2, and COX-2 inhibitors could cause cell apoptosis. Treatment with COX-2 inhibitors resulted in decreased autoantibody production and inhibition of the T cell response to the major lupus autoantigen, nucleosome, and its presentation by antigen-presenting cells. Surprisingly, a significant increase in survival occurred only in mice receiving intermittent therapy with the lowest dose of celecoxib (500 parts per million), approximating <100 mg of celecoxib/day in humans. A continuous diet, but not intermittent feeding, with the combination of celecoxib and aspirin delayed development of nephritis temporarily, but failed to prolong survival. Indeed, treatment with aspirin alone increased mortality. CONCLUSION: The contributions of the major players in the pathogenic autoimmune response, namely, T cells, B cells, dendritic cells, and macrophages that are abnormally hyperactive in lupus, depend on the increased expression and activity of COX-2, similar to inflammatory cells in target organs. Intermittent pulse therapy with low doses of select COX-2 inhibitors would be of value in the treatment of lupus.


Subject(s)
Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase 2/metabolism , Immune System/enzymology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/metabolism , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Administration, Oral , Animals , Antibodies, Antinuclear/blood , Apoptosis/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , B-Lymphocytes/pathology , Celecoxib , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/enzymology , Dendritic Cells/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Glomerulonephritis/prevention & control , Lupus Erythematosus, Systemic/pathology , Macrophages/drug effects , Macrophages/enzymology , Macrophages/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred NZB , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , T-Lymphocytes/pathology
15.
J Immunol ; 178(12): 7849-58, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17548623

ABSTRACT

Subnanomolar doses of an unaltered, naturally occurring nucleosomal histone peptide epitope, H4(71-94), when injected s.c. into lupus-prone mice, markedly prolong lifespan by generating CD4+25+ and CD8+ regulatory T cells (Treg) producing TGF-beta. The induced Treg cells suppress nuclear autoantigen-specific Th and B cells and block renal inflammation. Splenic dendritic cells (DC) captured the s.c.-injected H4(71-94) peptide rapidly and expressed a tolerogenic phenotype. The DC of the tolerized animal, especially plasmacytoid DC, produced increased amounts of TGF-beta, but diminished IL-6 on stimulation via the TLR-9 pathway by nucleosome autoantigen and other ligands; and those plasmacytoid DC blocked lupus autoimmune disease by simultaneously inducing autoantigen-specific Treg and suppressing inflammatory Th17 cells that infiltrated the kidneys of untreated lupus mice. Low-dose tolerance with H4(71-94) was effective even though the lupus immune system is spontaneously preprimed to react to the autoepitope. Thus, H4(71-94) peptide tolerance therapy that preferentially targets pathogenic autoimmune cells could spare lupus patients from chronically receiving toxic agents or global immunosuppressants and maintain remission by restoring autoantigen-specific Treg cells.


Subject(s)
Autoimmune Diseases/drug therapy , Dendritic Cells/immunology , Immunosuppression Therapy , Lupus Erythematosus, Systemic/drug therapy , Peptide Fragments/therapeutic use , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Autoantibodies/immunology , Autoantigens/immunology , B-Lymphocytes/immunology , CD4 Antigens/analysis , CD8 Antigens/analysis , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/metabolism , Histones/chemistry , Histones/immunology , Immune Tolerance , Immunoglobulin G/immunology , Interleukin-17/metabolism , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-6/metabolism , Mice , Nucleosomes/immunology , Peptide Fragments/administration & dosage , Peptides/administration & dosage , Peptides/therapeutic use , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/metabolism , Transforming Growth Factor beta/metabolism
16.
Int Rev Immunol ; 25(1-2): 5-25, 2006.
Article in English | MEDLINE | ID: mdl-16531276

ABSTRACT

Naturally occurring, CD4+ CD25+ regulatory T cells that are exported from the thymus early in life play an important role in controlling organ-specific autoimmune diseases, but they may not be critical for suppressing systemic autoimmunity in lupus. On the other hand, lupus-prone subjects appear to be deficient in generation of adaptive T-regulatory cells that can be induced by various means. We review autoantigen-specific therapeutic approaches that induce such regulatory T cells. Of particular interest are TGF-ss producing CD4+ CD25+ and CD8+ regulatory T cells that are induced by low dose tolerance therapy of lupus-prone mice with nucleosomal histone peptide epitopes, administered subcutaneously in subnanomolar doses. These regulatory T cells are not only efficient in suppressing autoantigen recognition and autoantibody production, but they also inhibit migration/accumulation of pathogenic autoimmune cells in the target organ, such as the kidneys of mice prone to develop lupus nephritis. We discuss why and under what conditions such therapeutic approaches would be beneficial in lupus patients and lupus-prone subjects.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Autoantibodies/immunology , Autoantigens/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Mice , Models, Immunological
17.
J Immunol ; 175(9): 5857-65, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16237078

ABSTRACT

To study central tolerance to the major product of ongoing apoptosis in the thymus, we made new lines of transgenic (Tg) mice expressing TCR of a pathogenic autoantibody-inducing Th cell that was specific for nucleosomes and its histone peptide H4(71-94). In the lupus-prone (SWR x NZB)F1 (SNF1) thymus, introduction of the lupus TCR transgene caused no deletion, but marked down-regulation of the Tg TCR and up-regulation of endogenous TCRs. Paradoxically, autoimmune disease was suppressed in the alphabetaTCR Tg SNF1 mice with induction of highly potent regulatory T cells in the periphery. By contrast, in the MHC-matched, normal (SWR x B10. D2)F1 (SBF1), or in the normal SWR backgrounds, marked deletion of transgenic thymocytes occurred. Thymic lymphoid cells of the normal or lupus-prone mice were equally susceptible to deletion by anti-CD3 Ab or irradiation. However, in the steady state, spontaneous presentation of naturally processed peptides related to the nucleosomal autoepitope was markedly greater by thymic dendritic cells (DC) from normal mice than that from lupus mice. Unmanipulated thymic DC of SNF1 mice expressed lesser amounts of MHC class II and costimulatory molecules than their normal counterparts. These results indicate that apoptotic nucleosomal autoepitopes are naturally processed and presented to developing thymocytes, and a relative deficiency in the natural display of nucleosomal autoepitopes by thymic DC occurs in lupus-prone SNF1 mice.


Subject(s)
Autoimmunity , Dendritic Cells/physiology , Lupus Vulgaris/immunology , Nucleosomes/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Apoptosis , Histones/pharmacology , Mice , Mice, Inbred NZB , Mice, Transgenic , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes, Regulatory/physiology
18.
J Immunol ; 174(6): 3247-55, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15749855

ABSTRACT

We induced very low-dose tolerance by injecting lupus prone (SWR x NZB)F1 (SNF1) mice with 1 mug nucleosomal histone peptide autoepitopes s.c. every 2 wk. The subnanomolar peptide therapy diminished autoantibody levels and prolonged life span by delaying nephritis, especially by reducing inflammatory cell reaction and infiltration in kidneys. H4(71-94) was the most effective autoepitope. Low-dose tolerance therapy induced CD8+, as well as CD4+ CD25+ regulatory T (Treg) cell subsets containing autoantigen-specific cells. These adaptive Treg cells suppressed IFN-gamma responses of pathogenic lupus T cells to nucleosomal epitopes at up to a 1:100 ratio and reduced autoantibody production up to 90-100% by inhibiting nucleosome-stimulated T cell help to nuclear autoantigen-specific B cells. Both CD4+ CD25+ and CD8+ Treg cells produced and required TGF-beta1 for immunosuppression, and were effective in suppressing lupus autoimmunity upon adoptive transfer in vivo. The CD4+ CD25+ T cells were partially cell contact dependent, but CD8+ T cells were contact independent. Thus, low-dose tolerance with highly conserved histone autoepitopes repairs a regulatory defect in systemic lupus erythematosus by generating long-lasting, TGF-beta-producing Treg cells, without causing allergic/anaphylactic reactions or generalized immunosuppression.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/therapy , Nuclear Proteins/immunology , Nucleosomes/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Antibodies, Antinuclear/blood , Autoantigens/administration & dosage , Autoantigens/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes/administration & dosage , Epitopes/genetics , Female , Immune Tolerance , Immunosuppression Therapy , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Lupus Nephritis/prevention & control , Lupus Nephritis/therapy , Mice , Mice, Inbred NZB , Molecular Sequence Data , Nuclear Proteins/genetics , Peptides/immunology , Receptors, Interleukin-2/metabolism , Transforming Growth Factor beta/biosynthesis
19.
Arthritis Rheum ; 52(2): 659-67, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15693014

ABSTRACT

OBJECTIVE: (SWR x NZB)F(1) (or SNF(1)) hybrid mice succumb to lupus nephritis. A previous analysis of SNF(1) x NZB backcross mice revealed the existence of 4 SWR loci (H2 on chromosome 17, Swrl-1 on chromosome 1, Swrl-2 on chromosome 14, and Swrl-3 on chromosome 18) and 2 NZB loci (Nba1 and Lbw2/Sbw2, both on chromosome 4). A second study focusing on SNF(1) x SWR backcross offspring uncovered 5 suggestive loci for antinuclear antibody formation, consisting of 3 dominant NZB contributions (Nba4 on chromosome 5, Lbw4 on chromosome 6, and Nba5 on chromosome 7) and 2 recessive SWR contributions (Swrl-1 on chromosome 1 and Swrl-4 on chromosome 10). The present intercross study was executed to replicate the earlier findings, using an independent panel of (SWR x NZB)F(2) offspring. METHODS: A panel of (NZB x SWR)F(2) hybrids were phenotyped (for renal disease, early mortality, and a variety of autoantibodies) and genotyped (using 95 microsatellite primers positioned across all 19 autosomes and the X chromosome). Linkage analysis was conducted using the derived phenotype and genotype data, with the interval-mapping program MapManager. RESULTS: Four suggestive loci were mapped: Swrl-5 on chromosome 1 (peak at 106 cM), linked to hypergammaglobulinemia; an NZB locus on chromosome 5 (Nba4; peak at 15 cM), linked to IgG anti-single-stranded DNA (anti-ssDNA) antibodies, IgG anti-doubled-stranded DNA (anti-dsDNA) antibodies, and glomerulonephritis; an NZB locus on chromosome 13 (Nba6; peak at 28 cM), linked to IgG anti-dsDNA antibodies; and an SWR locus on chromosome 14 (Swrl-2; peak at 30 cM), linked to IgG anti-ssDNA antibodies. Eight additional loci revealed linkage at P < 0.01, of which 7 co-mapped with lupus susceptibility loci previously identified in other models. CONCLUSION: Considering all 3 mapping studies together, lupus in SWR/NZB hybrids appears to be the epistatic end product of several distinct loci, of which 3 SWR-derived loci (Swrl-1, Swrl-2, and Swrl-3) and 5 NZB-derived loci (Nba1, Nba3, Nba4, Nba5, and Lbw4) have been independently confirmed. The immunologic functions and molecular identities of these loci await elucidation.


Subject(s)
Lupus Nephritis/genetics , Animals , Antibodies, Antinuclear/blood , Chimera , Chromosome Mapping , Crosses, Genetic , DNA/immunology , DNA, Single-Stranded/immunology , Genetic Linkage , Genotype , Immunoglobulin G/analysis , Mice , Mice, Inbred NZB
20.
J Mol Med (Berl) ; 83(4): 267-78, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15630591

ABSTRACT

Special populations of T helper cells drive B cells to produce IgG class switched, pathogenic autoantibodies in lupus. The major source of antigenic determinants (epitopes) that trigger interactions between lupus T and B cells is nucleosomes of apoptotic cells. These epitopes can be used for antigen-specific therapy of lupus. Secondly, the autoimmune T cells of lupus are sustained because they resist anergy and activation-induced programmed cell death by markedly upregulating cyclooxygenase (COX) 2 along with the antiapoptotic molecule c-FLIP. Only certain COX-2 inhibitors block pathogenic anti-DNA autoantibody production in lupus by causing death of autoimmune T helper cells. Hence COX-2 inhibitors may work independently of their ability to block the enzymatic function of COX-2, and structural peculiarities of these select inhibitors may lead to better drug discovery and design.


Subject(s)
Autoantigens/immunology , Lupus Vulgaris/immunology , T-Lymphocytes, Helper-Inducer/pathology , Animals , Apoptosis/immunology , Apoptosis/physiology , B-Lymphocytes/immunology , CASP8 and FADD-Like Apoptosis Regulating Protein , CD40 Ligand/metabolism , Clonal Anergy , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Lupus Vulgaris/pathology , Lupus Vulgaris/therapy , Membrane Proteins , Nucleosomes/immunology , Prostaglandin-Endoperoxide Synthases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-cbl , T-Lymphocytes, Helper-Inducer/immunology , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...