Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
ACR Open Rheumatol ; 5(10): 536-546, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37710418

ABSTRACT

OBJECTIVE: To use cell-based gene signatures to identify patients with systemic lupus erythematous (SLE) in the phase II/III APRIL-SLE and phase IIb ADDRESS II trials most likely to respond to atacicept. METHODS: A published immune cell deconvolution algorithm based on Affymetrix gene array data was applied to whole blood gene expression from patients entering APRIL-SLE. Five distinct patient clusters were identified. Patient characteristics, biomarkers, and clinical response to atacicept were assessed per cluster. A modified immune cell deconvolution algorithm was developed based on RNA sequencing data and applied to ADDRESS II data to identify similar patient clusters and their responses. RESULTS: Patients in APRIL-SLE (N = 105) were segregated into the following five clusters (P1-5) characterized by dominant cell subset signatures: high neutrophils, T helper cells and natural killer (NK) cells (P1), high plasma cells and activated NK cells (P2), high B cells and neutrophils (P3), high B cells and low neutrophils (P4), or high activated dendritic cells, activated NK cells, and neutrophils (P5). Placebo- and atacicept-treated patients in clusters P2,4,5 had markedly higher British Isles Lupus Assessment Group (BILAG) A/B flare rates than those in clusters P1,3, with a greater treatment effect of atacicept on lowering flares in clusters P2,4,5. In ADDRESS II, placebo-treated patients from P2,4,5 were less likely to be SLE Responder Index (SRI)-4, SRI-6, and BILAG-Based Combined Lupus Assessment responders than those in P1,3; the response proportions again suggested lower placebo effect and a greater treatment differential for atacicept in P2,4,5. CONCLUSION: This exploratory analysis indicates larger differences between placebo- and atacicept-treated patients with SLE in a molecularly defined patient subset.

2.
Front Cell Dev Biol ; 8: 577662, 2020.
Article in English | MEDLINE | ID: mdl-33240880

ABSTRACT

B cell activation factor of the TNF family (BAFF/BLyS), an essential B cell survival factor of which circulating levels are elevated in several autoimmune disorders, is targeted in the clinic for the treatment of systemic lupus erythematosus (SLE). The soluble form of BAFF can exist as 3-mer, or as 60-mer that results from the ordered assembly of twenty 3-mers and that can be obtained from naturally cleaved membrane-bound BAFF or made as a recombinant protein. However, which forms of soluble BAFF exist and act in humans is unclear. In this study, BAFF 3-mer and 60-mer in biological fluids were characterized for size, activity and response to specific stimulators or inhibitors of BAFF. Human cerebrospinal fluids (CSF) from patients with multiple sclerosis and adult human sera contained exclusively BAFF 3-mer in these assays, also when BAFF concentrations were moderately SLE or highly (BAFFR-deficient individual) increased. Human sera, but not CSF, contained a high molecular weight, saturable activity that dissociated preformed recombinant BAFF 60-mer into 3-mer. This activity was lower in cord blood. Cord blood displayed BAFF levels 10-fold higher than in adults and consistently contained a fair proportion of active high molecular weight BAFF able to dissociate into 3-mer but not endowed with all properties of recombinant BAFF 60-mer. If BAFF 60-mer is produced in humans, it is dissociated, or at least attenuated in the circulation.

3.
Br J Pharmacol ; 176(20): 4019-4033, 2019 10.
Article in English | MEDLINE | ID: mdl-31355456

ABSTRACT

BACKGROUND AND PURPOSE: The TNF family ligands, B cell activating factor of the TNF family (BAFF, also known as B lymphocyte stimulator, BLyS) and a proliferation-inducing ligand (APRIL), share the transmembrane activator and calcium-modulator and cyclophilin ligand (CAML)-interactor (TACI) as one of their common receptors. Atacicept, a chimeric recombinant TACI/IgG1-Fc fusion protein, inhibits both ligands. TACI and APRIL also bind to proteoglycans and to heparin that is structurally related to proteoglycans. It is unknown whether the portion of TACI contained in atacicept can bind directly to proteoglycans, or indirectly via APRIL, and whether this could interfere with the anti-coagulant properties of heparin. EXPERIMENTAL APPROACH: Binding of atacicept and APRIL to proteoglycan-positive cells was measured by FACS. Activities of heparin and atacicept were measured with activated factor Xa inhibition and cell-based assays. Effects of heparin on circulating atacicept was monitored in mice. KEY RESULTS: Atacicept did not bind to proteoglycan-positive cells, but when complexed to APRIL could do so indirectly via APRIL. Multimers of atacicept obtained after exposure to cysteine or BAFF 60-mer bound directly to proteoglycans. Atacicept alone, or in complex with APRIL, or in a multimeric form did not interfere with heparin activity in vitro. Conversely, heparin did not influence inhibition of BAFF and APRIL by atacicept and did not change circulating levels of atacicept. CONCLUSIONS AND IMPLICATIONS: Lack of detectable interference of APRIL-bound or free atacicept on heparin activity makes it unlikely that atacicept at therapeutic doses will interfere with the function of heparin in vivo.


Subject(s)
B-Lymphocytes/drug effects , Cytokines/antagonists & inhibitors , Heparin/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , B-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/metabolism , Dose-Response Relationship, Drug , Factor Xa/metabolism , Female , HEK293 Cells , Heparin/administration & dosage , Heparin/blood , Humans , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/blood , Structure-Activity Relationship
4.
Nat Commun ; 9(1): 1199, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572442

ABSTRACT

The B cell survival factor (TNFSF13B/BAFF) is often elevated in autoimmune diseases and is targeted in the clinic for the treatment of systemic lupus erythematosus. BAFF contains a loop region designated the flap, which is dispensable for receptor binding. Here we show that the flap of BAFF has two functions. In addition to facilitating the formation of a highly active BAFF 60-mer as shown previously, it also converts binding of BAFF to TNFRSF13C (BAFFR) into a signaling event via oligomerization of individual BAFF-BAFFR complexes. Binding and activation of BAFFR can therefore be targeted independently to inhibit or activate the function of BAFF. Moreover, structural analyses suggest that the flap of BAFF 60-mer temporarily prevents binding of an anti-BAFF antibody (belimumab) but not of a decoy receptor (atacicept). The observed differences in profiles of BAFF inhibition may confer distinct biological and clinical efficacies to these therapeutically relevant inhibitors.


Subject(s)
B-Cell Activating Factor/chemistry , B-Cell Activating Factor/physiology , B-Cell Activation Factor Receptor/chemistry , B-Lymphocytes/cytology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , B-Cell Activating Factor/genetics , Cell Differentiation , Cell Survival , Cross-Linking Reagents/chemistry , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Immunoglobulin Fragments/chemistry , Lymphopenia/metabolism , Male , Mice , Mice, Transgenic , Mutation , Protein Binding , Protein Domains , Recombinant Fusion Proteins/pharmacology
5.
Int Rev Immunol ; 36(1): 3-19, 2017 01 02.
Article in English | MEDLINE | ID: mdl-28215100

ABSTRACT

The B cell-stimulating molecules, BAFF (B cell activating factor) and APRIL (a proliferation-inducing ligand), are critical factors in the maintenance of the B cell pool and humoral immunity. In addition, BAFF and APRIL are involved in the pathogenesis of a number of human autoimmune diseases, with elevated levels of these cytokines detected in the sera of patients with systemic lupus erythematosus (SLE), IgA nephropathy, Sjögren's syndrome, and rheumatoid arthritis. As such, both molecules are rational targets for new therapies in B cell-driven autoimmune diseases, and several inhibitors of BAFF or BAFF and APRIL together have been investigated in clinical trials. These include the BAFF/APRIL dual inhibitor, atacicept, and the BAFF inhibitor, belimumab, which is approved as an add-on therapy for patients with active SLE. Post hoc analyses of these trials indicate that baseline serum levels of BAFF and BAFF/APRIL correlate with treatment response to belimumab and atacicept, respectively, suggesting a role for the two molecules as predictive biomarkers. It will, however, be important to refine future testing to identify active forms of BAFF and APRIL in the circulation, as well as to distinguish between homotrimer and heteromer configurations. In this review, we discuss the rationale for dual BAFF/APRIL inhibition versus single BAFF inhibition in autoimmune disease, by focusing on the similarities and differences between the physiological and pathogenic roles of the two molecules. A summary of the preclinical and clinical data currently available is also presented.


Subject(s)
B-Cell Activating Factor/metabolism , B-Lymphocytes/immunology , Immunotherapy/methods , Lupus Erythematosus, Systemic/drug therapy , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Autoantibodies/metabolism , Clinical Trials as Topic , Humans , Molecular Targeted Therapy , Recombinant Fusion Proteins/therapeutic use
6.
Clin Immunol ; 164: 65-77, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26821304

ABSTRACT

Bruton's tyrosine kinase (Btk) is expressed in a variety of immune cells and previous work has demonstrated that blocking Btk is a promising strategy for treating autoimmune diseases. Herein, we utilized a tool Btk inhibitor, M7583, to determine the therapeutic efficacy of Btk inhibition in two mouse lupus models driven by TLR7 activation and type I interferon. In BXSB-Yaa lupus mice, Btk inhibition reduced autoantibodies, nephritis, and mortality. In the pristane-induced DBA/1 lupus model, Btk inhibition suppressed arthritis, but autoantibodies and the IFN gene signature were not significantly affected; suggesting efficacy was mediated through inhibition of Fc receptors. In vitro studies using primary human macrophages revealed that Btk inhibition can block activation by immune complexes and TLR7 which contributes to tissue damage in SLE. Overall, our results provide translational insight into how Btk inhibition may provide benefit to a variety of SLE patients by affecting both BCR and FcR signaling.


Subject(s)
Lupus Erythematosus, Systemic/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase , Animals , Arthritis/drug therapy , Arthritis/pathology , Autoantibodies/blood , Disease Models, Animal , Female , Foot Joints/drug effects , Foot Joints/pathology , Humans , Immunosuppressive Agents , Interferon Type I/immunology , Kidney/drug effects , Kidney/pathology , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Nephritis/drug therapy , Nephritis/pathology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Proteinuria/drug therapy , Proteinuria/pathology , Terpenes , Toll-Like Receptor 7/immunology
7.
Front Immunol ; 5: 242, 2014.
Article in English | MEDLINE | ID: mdl-24904590

ABSTRACT

Human maternal autoantibodies can trigger autoimmune diseases such as congenital heart block (CHB) in the progeny of women with lupus or Sjogren's disease. The pathogenic effect of early autoantibody (autoAb) exposure has been investigated in a murine neonatal autoimmune ovarian disease (nAOD) model triggered by a unique ZP3 antibody. Although immune complexes (IC) are formed in adult and neonatal ovaries, ZP3 antibody triggers severe nAOD only in <7-day-old neonatal mice. Propensity to nAOD is due to the uniquely hyper-responsive neonatal natural killer (NK) cells that lack the inhibitory Ly49C/I receptors. In nAOD, the neonatal NK cells directly mediate ovarian inflammation and oocyte depletion while simultaneously promoting de novo pathogenic ovarian-specific T cell responses. Resistance to nAOD in older mice results from the emergence of the Ly49C/I(+) NK cells that regulate effector NK cells and from CD25(+) regulatory T cell control. In preliminary studies, FcγRIII(+) NK cells as well as the ovarian resident FcγRIII(+) macrophages and/or dendritic cells were found to be as indispensable players. Activated by ovarian IC, they migrate to lymphoid organs where NK cell priming occurs. Remarkably, the findings in nAOD are very similar to those reported for neonatal responses to a retrovirus and its cognate antibody that lead to long-lasting immunity. Studies on nAOD therefore provide insights into maternal autoAb-mediated neonatal autoimmunity, including CHB, while simultaneously uncovering new properties of the neonatal innate and adaptive responses, lethality of premature infant infection, and novel neonatal antiviral vaccine design.

8.
J Immunol ; 191(6): 2865-9, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23960238

ABSTRACT

NK cells are critical in immune responses against pathogens. However, their role in autoimmunity is still controversial. In this study, we demonstrate that neonatal NK cells render newborns more susceptible to neonatal autoimmunity induced by maternal autoantibodies (neonatal autoimmune ovarian disease); thus, neonatal but not adult NK cells are pathogenic after transfer into NK cell-deficient pups. The inhibitory receptors Ly49C/I are expressed in ∼5% of neonatal and ∼50% of adult NK cells. In this study, we show that the presence of Ly49C/I⁺ adult NK cells inhibits neonatal autoimmune ovarian disease induction. Thus, the ontogenetic regulation of Ly49C/I expression determines the propensity to autoantibody-induced autoimmunity. In summary, this study provides definitive evidence of a pathogenic role of NK cells in neonatal autoimmunity and also elucidates a novel mechanism by which neonatal NK cells render newborns more susceptible to autoantibody-induced autoimmunity.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily A/immunology , Ovarian Diseases/immunology , Animals , Animals, Newborn , Autoimmunity/immunology , Disease Models, Animal , Female , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pregnancy
9.
J Immunol ; 183(12): 7635-8, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19923458

ABSTRACT

Natural CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg) effectively prevent autoimmune disease development, but their role in maintaining physiological tolerance against self-Ag of internal organs is not yet defined. In this study, we quantified disease-specific Treg (DS-Treg) as Treg that preferentially suppress one autoimmune disease over another in day 3 thymectomized recipients. A striking difference was found among individual lymph nodes (LN) of normal mice; Treg from draining LN were 15-50 times more efficient than those of nondraining LN at suppressing autoimmune diseases of ovary, prostate, and lacrimal glands. The difference disappeared upon auto-Ag ablation and returned upon auto-Ag re-expression. In contrast, the CD4(+)CD25(-) effector T cells from different individual LN induced multiorgan inflammation with comparable organ distribution. We propose that peripheral tolerance for internal organs relies on the control of autoreactive effector T cells by strategic enrichment of Ag-specific Treg in the regional LN.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Epitopes, T-Lymphocyte/biosynthesis , Lymph Nodes/immunology , Lymph Nodes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmune Diseases/pathology , CD4 Antigens/biosynthesis , Cell Movement/immunology , Cell Proliferation , Female , Interleukin-2 Receptor alpha Subunit/biosynthesis , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/immunology , Ovarian Diseases/immunology , Ovarian Diseases/pathology , Ovarian Diseases/prevention & control , Self Tolerance/immunology , T-Lymphocytes, Regulatory/pathology
10.
J Autoimmun ; 31(2): 123-30, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18650065

ABSTRACT

We sought to determine if the histone deacetylase inhibitor (HDI), trichostatin A (TSA), would alter systemic lupus erythematosus (SLE) in NZB/W mice. Fourteen to sixteen-week-old female NZB/W F1 mice were given TSA (1.0mg/kg body weight (BW)) intraperitonealy (i.p.) daily, TSA (1.0mg/kg BW) i.p.+anti-CD25 (250mg/mouse) i.p. every third day, only anti-CD25 (250mg/mouse) i.p., DMSO or isotype IgG. Disease progression was assessed as they aged. Mice were sacrificed at 26 or 38 weeks of age, tissues collected and evaluated. At 36 weeks, TSA-treated animals had decreased anti-double stranded DNA (dsDNA) autoantibodies and decreased protein excretion compared to controls. Spleen size and the percentage of CD4+CD69+ cells were decreased, with an increase in CD4+CD25+ T cells in the TSA-treated mice. Real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis of T cells showed a decrease in IL-6 production but an increase in TGF-beta1 and Foxp3 in the TSA-treated animals. Kidney analysis showed a decrease in IgG and C3 deposition, decrease in pathologic glomerular disease and renal MCP-1, MMP-9, and IL-6 mRNA expression. Anti-CD25-treated mice euthanized at 26 weeks of age showed decreased Foxp3+CD4+CD25+ T cells compared to TSA-treated mice. These data suggest TSA administration modulates lupus-like disease, in part, by increasing T regulatory cells.


Subject(s)
Autoimmunity , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Antinuclear/blood , Autoimmunity/drug effects , CD4 Antigens/metabolism , Disease Models, Animal , Disease Progression , Female , Flow Cytometry , Immunologic Factors/pharmacology , Interleukin-2 Receptor alpha Subunit/metabolism , Kidney/pathology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/drug therapy , Mice , Mice, Inbred NZB , Organ Size , T-Lymphocytes, Regulatory/drug effects , Up-Regulation/drug effects , Up-Regulation/immunology
11.
J Immunol ; 180(7): 4366-70, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18354156

ABSTRACT

Female B6AF1 mice thymectomized on day 3 (d3tx) develop autoimmune ovarian disease (AOD) and dacryoadenitis. It has been hypothesized that d3tx breaks tolerance by depleting late ontogeny regulatory T cells (Treg). We now report that Treg greatly expand over effector T cells in d3tx mice and adoptively suppress autoimmune disease in d3tx recipients. In the d3tx donors, Treg from ovarian lymph nodes (LN) preferentially suppress AOD and Treg from lacrimal gland LN preferentially suppress dacryoadenitis, suggesting they are strategically positioned for disease control. Indeed, the autologous disease in d3tx mice is dramatically enhanced by in vivo depletion of endogenous Treg. Moreover, normal 3-day-old mice possess Treg that suppress AOD and autoimmune gastritis as efficiently as adult cells. Thus, d3tx mice possess disease-relevant Treg of presumed neonatal origin. They accumulate in the regional LN and actively inhibit concurrent autoimmune disease; however, they cannot fully prevent autoimmune disease development.


Subject(s)
Autoimmune Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Thymectomy , Animals , Animals, Newborn , Autoimmune Diseases/pathology , Female , Forkhead Transcription Factors/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Lymph Nodes/immunology , Male , Mice , Time Factors
12.
J Immunol ; 179(9): 5644-8, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17947634

ABSTRACT

Sphingosine kinase (Sphk) phosphorylates sphingosine into sphingosine-1-phosphate (S1P), but its recently identified isoform Sphk2 has been suggested to have distinct subcellular localization and substrate specificity. We demonstrate here that, surprisingly, Sphk2(-/-) CD4(+) T cells exhibit a hyperactivated phenotype with significantly enhanced proliferation and cytokine secretion in response to IL-2 as well as reduced sensitivity to regulatory T cell-mediated suppression in vitro, apparently independent of effects upon S1P. Such findings appear to reflect a requirement for Sphk2 to suppress IL-2 signaling because, in Sphk2(-/-) CD4(+) T cells, IL-2 induced abnormally accentuated STAT5 phosphorylation and small interfering RNA knockdown of STAT5 abrogated their hyperactive phenotype. This pathway physiologically modulates autoinflammatory responses, because Sphk2(-/-) T cells induced more rapid and robust inflammatory bowel disease in scid recipients. Thus, Sphk2 regulates IL-2 pathways in T cells, and the modulation of Sphk2 activity may be of therapeutic utility in inflammatory and/or infectious diseases.


Subject(s)
Autoimmunity/immunology , Interleukin-2/immunology , Interleukin-2/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction/immunology , Animals , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Lysophospholipids/metabolism , Mice , Mice, Knockout , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/immunology
13.
Immunol Rev ; 212: 170-84, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16903914

ABSTRACT

The CD4+ CD25+ regulatory T cells (Tregs) are efficient regulators of autoimmunity, but the mechanism remains elusive. We summarize recent data for the conclusion that disease-specific Tregs respond to tissue antigens to maintain physiological tolerance and prevent autoimmunity. First, polyclonal Tregs from antigen-positive donors suppress autoimmune ovarian disease (AOD) or experimental autoimmune prostatitis in day 3 thymectomized (d3tx) mice more efficiently than Tregs from antigen-negative donors. Second, Tregs of antigen-negative adult mice respond to cognate antigen in vivo and rapidly gain disease-specific Treg function. Third, in d3tx female recipients devoid of neonatal ovarian antigens, only female Tregs suppressed AOD; the male Tregs gain AOD-suppressing function by responding to the ovarian antigen in the recipients and mask the supremacy of female Tregs in AOD suppression. Fourth, when Tregs completely suppress AOD, the ovary-draining lymph node is the only location with evidence of profound and persistent (but reversible) host T-cell suppression. Fifth, from these nodes, highly potent AOD-suppressing Tregs are retrievable. We conclude that self-tolerance involves the continuous priming of Tregs by autoantigens, and in autoimmune disease suppression, the effector T-cell response is continuously negated by potent disease-specific Tregs that accumulate at the site of autoantigen presentation.


Subject(s)
Autoantigens/physiology , Self Tolerance , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Immunosuppression Therapy , Mice , T-Lymphocytes, Regulatory/transplantation
14.
Blood ; 107(3): 1056-62, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16223778

ABSTRACT

Studies on CD4+ CD25+ regulatory T cells (Tregs) with transgenic T-cell receptors indicate that Tregs may receive continuous antigen (Ag) stimulation in the periphery. However, the consequence of this Ag encounter and its relevance to physiologic polyclonal Treg function are not established. In autoimmune prostatitis (EAP) of the day-3 thymectomized (d3tx) mice, male Tregs suppressed EAP 3 times better than Tregs from female mice or male mice without prostates. Importantly, the superior EAP-suppressing function was acquired after a 6-day exposure to prostate Ag in the periphery, unaffected by sex hormones. Thus, a brief exposure of physiologic prostate Ag capacitates peripheral polyclonal Tregs to suppress EAP. In striking contrast, autoimmune ovarian disease (AOD) was suppressed equally by male and female Tregs. We now provide evidence that the ovarian Ag develops at birth, 14 days earlier than prostate Ag, and that male Tregs respond to neonatal ovarian Ag in the Treg recipients to gain AOD-suppressing capacity. When d3tx female recipients were deprived of ovarian Ag in the neonatal period, AOD was suppressed by female but not by male Tregs, whereas dacryoadenitis was suppressed by both. We conclude that the physiologic autoAg quickly and continuously enhances disease-specific polyclonal Treg function to maintain self-tolerance.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/immunology , Immune Tolerance , Prostatitis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/pathology , Dacryocystitis/immunology , Dacryocystitis/pathology , Female , Gonadal Steroid Hormones/immunology , Male , Mice , Oophoritis/immunology , Oophoritis/pathology , Prostatitis/pathology , T-Lymphocytes, Regulatory/pathology
15.
J Exp Med ; 202(6): 771-81, 2005 Sep 19.
Article in English | MEDLINE | ID: mdl-16172257

ABSTRACT

This study investigated the unresolved issue of antigen-dependency and antigen-specificity of autoimmune disease suppression by CD4+CD25+ T cells (T regs). Based on autoimmune ovarian disease (AOD) in day 3 thymectomized (d3tx) mice and polyclonal T regs expressing the Thy1.1 marker, we determined: (a) the location of recipient T cell suppression, (b) the distribution of AOD-suppressing T regs, and (c) the relative efficacy of male versus female T regs. Expansion of recipient CD4+ T cells, activation/memory marker expression, and IFN-gamma production were inhibited persistently in the ovary-draining LNs but not elsewhere. The cellular changes were reversed upon Thy1.1+ T reg depletion, with emergence of potent pathogenic T cells and severe AOD. Similar changes were detected in the regional LNs during autoimmune dacryoadenitis and autoimmune prostatitis suppression. Although the infused Thy1.1+ T regs proliferated and were disseminated in peripheral lymphoid organs, only those retrieved from ovary-draining LNs adoptively suppressed AOD at a suboptimal cell dose. By depriving d3tx recipients of ovarian antigens, we unmasked the supremacy of ovarian antigen-exposed female over male T regs in AOD suppression. Thus, disease suppression by polyclonal T regs depends on endogenous antigen stimulation; this occurs in a location where potent antigen-specific T regs accumulate and continuously negate pathogenic T cell response.


Subject(s)
Autoimmune Diseases/immunology , Epitopes, T-Lymphocyte/physiology , Lymph Nodes/immunology , Ovarian Diseases/immunology , Receptors, Interleukin-2/biosynthesis , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Clone Cells , Female , Lymph Nodes/cytology , Lymph Nodes/metabolism , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Ovarian Diseases/pathology , Receptors, Interleukin-2/metabolism , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation
16.
Int Rev Immunol ; 24(3-4): 227-45, 2005.
Article in English | MEDLINE | ID: mdl-16036376

ABSTRACT

Although previous studies have emphasized the tolerogenic property of murine neonatal immune system, recent studies indicate that neonatal mice are prone to autoimmune disease. This chapter will summarize the evidence for neonatal propensity to autoimmune ovarian disease (AOD) and describe the new finding that autoantibody can trigger a T cell-dependent autoimmune disease in neonatal but not adult mice. Based on depletion or addition of the CD4+ CD25+ T cells, disease resistance of older mice is explicable by the emergence of CD4+ CD25+ regulatory T-cell function after day 5, whereas disease susceptibility is associated with resistance to regulation by CD4+ CD25+ T cells.


Subject(s)
Autoimmune Diseases/immunology , CD4 Antigens/immunology , Infant, Newborn, Diseases/immunology , Ovarian Diseases/immunology , Receptors, Interleukin-2/immunology , T-Lymphocytes/immunology , Autoimmune Diseases/genetics , Female , Humans , Infant, Newborn , Infant, Newborn, Diseases/genetics , Ovarian Diseases/genetics
17.
Biochim Biophys Acta ; 1627(1): 26-38, 2003 May 13.
Article in English | MEDLINE | ID: mdl-12759189

ABSTRACT

The RET protooncogene encodes for a transmembrane receptor tyrosine kinase and plays a crucial role in nephrogenesis and the enteric nervous system (ENS) development. Alternative splicing at the 3' end of the RET gene generates 3' splicing variants that encode RET 9, RET 51 and RET 43 isoforms. It has been hypothesized that these isoforms perform distinct functions and that their expressions are differentially regulated during mammalian development. To gain an insight into the expression patterns of various ret isoforms during embryogenesis, we investigate the temporal and spatial expressions of ret gene in mouse embryos and in adult mice. We characterized the 3' end of the mouse ret gene and localized the alternatively spliced exons. Using 3' rapid amplification of cDNA ends (3' RACE) and reverse transcription-polymerase chain reaction (RT-PCR), ret 9 and ret 51 transcripts were identified in both mouse embryos and adult mouse tissues. However, the ret 43 transcript was not. Using in situ hybridization, we showed that ret 9 was the dominant ret encoding transcript in mouse embryos. Transcripts of ret 9 were detected in all cranial ganglia; in the sensory and autonomic ganglia of the trunk; in a subset of neurons of the dorsal root ganglion (DRG); in the motor neurons of the spinal cord; in the developing lung and excretory systems; in the enteric neuroblasts of the ENS; and in the thyroid lobes. In contrast, ret 51 expression was weak and restricted to the motor column of the spinal cord, the DRG, the enteric neuroblasts, the lung bud and the kidney. In adult mice, ret 9 expression was relatively widespread in many organs while that of ret 51 was rather restricted. Our data indicated that ret isoforms are temporally and spatially regulated in mouse embryos and adult mouse.


Subject(s)
Alternative Splicing , Protein Isoforms/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Amino Acid Sequence , Animals , Base Sequence , In Situ Hybridization , Mice , Molecular Sequence Data , Organ Specificity , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
J Immunol ; 170(9): 4656-64, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12707344

ABSTRACT

Although human maternal autoantibodies may transfer transient manifestation of autoimmune disease to their progeny, some neonatal autoimmune diseases can progress, leading to the loss of tissue structure and function. In this study we document that murine maternal autoantibody transmitted to progeny can trigger de novo neonatal pathogenic autoreactive T cell response and T cell-mediated organ-specific autoimmune disease. Autoantibody to a zona pellucida 3 (ZP3) epitope was found to induce autoimmune ovarian disease (AOD) and premature ovarian failure in neonatal, but not adult, mice. Neonatal AOD did not occur in T cell-deficient pups, and the ovarian pathology was transferable by CD4(+) T cells from diseased donors. Interestingly, neonatal AOD occurred only in pups exposed to ZP3 autoantibody from neonatal days 1-5, but not from day 7 or day 9. The disease susceptibility neonatal time window was not related to a propensity of neonatal ovaries to autoimmune inflammation, and it was not affected by infusion of functional adult CD4(+)CD25(+) T cells. However, resistance to neonatal AOD in 9-day-old mice was abrogated by CD4(+)CD25(+) T cell depletion. Finally, neonatal AOD was blocked by Ab to IgG-FcR, and interestingly, the disease was not elicited by autoantibody to a second, independent native ZP3 B cell epitope. Therefore, a new mechanism of neonatal autoimmunity is presented in which epitope-specific autoantibody stimulates de novo autoimmune pathogenic CD4(+) T cell response.


Subject(s)
Animals, Newborn/immunology , Autoantibodies/physiology , Autoimmune Diseases/immunology , Maternal-Fetal Exchange/immunology , Ovarian Diseases/immunology , Receptors, Cell Surface , T-Lymphocyte Subsets/immunology , Zona Pellucida/immunology , Administration, Oral , Adoptive Transfer , Aging/immunology , Amino Acid Sequence , Animals , Autoantibodies/administration & dosage , Autoantigens/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Disease Progression , Disease Susceptibility/immunology , Dose-Response Relationship, Immunologic , Egg Proteins/immunology , Epitopes, B-Lymphocyte/administration & dosage , Epitopes, B-Lymphocyte/immunology , Female , Immunity, Innate , Immunohistochemistry , Immunophenotyping , Lymphocyte Depletion , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred A , Mice, Inbred C57BL , Milk/immunology , Molecular Sequence Data , Organ Specificity/immunology , Ovarian Diseases/pathology , Ovarian Diseases/prevention & control , Peptide Fragments/immunology , Pregnancy , Receptors, IgG/physiology , Receptors, Interleukin-2/biosynthesis , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Zona Pellucida Glycoproteins
19.
Dev Biol ; 256(1): 73-88, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12654293

ABSTRACT

Selected for its high relative abundance, a protein spot of MW approximately 75 kDa, pI 5.5 was cored from a Coomassie-stained two-dimensional gel of proteins from 2850 zona-free metaphase II mouse eggs and analyzed by tandem mass spectrometry (TMS), and novel microsequences were identified that indicated a previously uncharacterized egg protein. A 2.4-kb cDNA was then amplified from a mouse ovarian adapter-ligated cDNA library by RACE-PCR, and a unique 2043-bp open reading frame was defined encoding a 681-amino-acid protein. Comparison of the deduced amino acid sequence with the nonredundant database demonstrated that the protein was approximately 40% identical to the calcium-dependent peptidylarginine deiminase (PAD) enzyme family. Northern blotting, RT-PCR, and in situ hybridization analyses indicated that the protein was abundantly expressed in the ovary, weakly expressed in the testis, and absent from other tissues. Based on the homology with PADs and its oocyte-abundant expression pattern, the protein was designated ePAD, for egg and embryo-abundant peptidylarginine deiminase-like protein. Anti-recombinant ePAD monospecific antibodies localized the molecule to the cytoplasm of oocytes in primordial, primary, secondary, and Graafian follicles in ovarian sections, while no other ovarian cell type was stained. ePAD was also expressed in the immature oocyte, mature egg, and through the blastocyst stage of embryonic development, where expression levels began to decrease. Immunoelectron microscopy localized ePAD to egg cytoplasmic sheets, a unique keratin-containing intermediate filament structure found only in mammalian eggs and in early embryos, and known to undergo reorganization at critical stages of development. Previous reports that PAD-mediated deimination of epithelial cell keratin results in cytoskeletal remodeling suggest a possible role for ePAD in cytoskeletal reorganization in the egg and early embryo.


Subject(s)
Cleavage Stage, Ovum/enzymology , Hydrolases/metabolism , Oocytes/enzymology , Amino Acid Sequence , Animals , Antibody Specificity , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Female , Gene Expression Regulation, Developmental , Hydrolases/genetics , Hydrolases/immunology , In Situ Hybridization , Male , Mice , Mice, Inbred ICR , Microscopy, Immunoelectron , Molecular Sequence Data , Oocytes/ultrastructure , Ovary/enzymology , Pregnancy , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Solubility
20.
J Immunol ; 170(4): 1667-74, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574329

ABSTRACT

The blockade of CD40 ligand (CD40L) is effective in autoimmune disease prevention. Recently, a brief period of CD40L mAb treatment was reported to induce tolerance and enhancement of CD4(+)CD25(+) regulatory T cell activity. We therefore determined the efficacy of CD40L mAb treatment in autoimmunity that resulted from CD4(+)CD25(+) regulatory T cell deficiency. Autoimmune ovarian disease (AOD) and oocyte autoantibody response of day 3-thymectomized (d3tx) mice were inhibited by continuous CD40L mAb treatment from day 3, or from days 10-14, whereas CD40L mAb treatment confined to the neonatal week was ineffective. The enhanced expression of memory markers (CD44 and CD62L(low)) on CD4(+) T cells of the d3tx mice was unaffected by CD40L mAb treatment. In contrast, their increased T cell activation markers (CD69 and CD25) were eliminated by CD40L mAb treatment. Moreover, ex vivo activated T cells of d3tx mice expressed elevated intracellular IFN-gamma, and this was also blocked by CD40L mAb. The memory T cells, although nonpathogenic in CD40L mAb-positive environment, transferred severe AOD to CD40L mAb(-) neonatal recipients. Most importantly, CD40L mAb treatment inhibited AOD in recipients of T cells from d3tx donors with severe AOD and led to regression of AOD in d3tx mice documented at 4 wk. Therefore, 1) the continuous presence of CD40L mAb both prevents and causes regression of AOD in the d3tx mice; and 2) the multiple steps of the d3tx autoimmune disease, including T cell activation, cytokine production, T cell-mediated inflammation, and tissue injury, are CD40L dependent.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , CD40 Ligand/immunology , CD40 Ligand/physiology , Ovarian Diseases/immunology , Ovarian Diseases/prevention & control , Thymectomy , Adoptive Transfer , Animals , Animals, Newborn , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/therapeutic use , Antibodies, Monoclonal/administration & dosage , Autoimmune Diseases/pathology , Female , Injections, Intraperitoneal , Interferon-gamma/biosynthesis , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphopenia/immunology , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Ovarian Diseases/pathology , Spleen/cytology , Spleen/immunology , Spleen/transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...