Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
2.
Front Immunol ; 13: 809247, 2022.
Article in English | MEDLINE | ID: mdl-35693780

ABSTRACT

Continuous exposure of tissue antigen (Ag) to the autoantigen-specific regulatory T cells (Treg) is required to maintain Treg-dependent systemic tolerance. Thus, testis autoantigens, previously considered as sequestered, may not be protected by systemic tolerance. We now document that the complete testis antigen sequestration is not valid. The haploid sperm Ag lactate dehydrogenase 3 (LDH3) is continuously exposed and not sequestered. It enters the residual body (RB) to egress from the seminiferous tubules and interact with circulating antibody (Ab). Some LDH3 also remains inside the sperm cytoplasmic droplets (CD). Treg-depletion in the DEREG mice that express diphtheria toxin receptor on the Foxp3 promoter results in spontaneous experimental autoimmune orchitis (EAO) and Ab to LDH3. Unlike the wild-type male mice, mice deficient in LDH3 (wild-type female or LDH3 NULL males) respond vigorously to LDH3 immunization. However, partial Treg depletion elevated the wild-type male LDH3 responses to the level of normal females. In contrast to LDH3, zonadhesin (ZAN) in the sperm acrosome displays properties of a sequestered Ag. However, when ZAN and other sperm Ag are exposed by vasectomy, they rapidly induce testis Ag-specific tolerance, which is terminated by partial Treg-depletion, leading to bilateral EAO and ZAN Ab response. We conclude that some testis/sperm Ag are normally exposed because of the unique testicular anatomy and physiology. The exposed Ag: 1) maintain normal Treg-dependent systemic tolerance, and 2) are pathogenic and serve as target Ag to initiate EAO. Unexpectedly, the sequestered Ags, normally non-tolerogenic, can orchestrate de novo Treg-dependent, systemic tolerance when exposed in vasectomy.


Subject(s)
Orchitis , Vasectomy , Animals , Autoantigens , Female , Humans , Immune Tolerance , Male , Mice , T-Lymphocytes, Regulatory
3.
Clin Immunol ; 224: 108675, 2021 03.
Article in English | MEDLINE | ID: mdl-33482358

ABSTRACT

Cgnz1 on chromosome 1 mapped into a 1.34 Mb region of chromosome 1 in NZM2328 confers the progression of immune complex (IC)-mediated glomerulonephritis (GN) from acute GN (aGN) to chronic GN (cGN) with severe proteinuria and end stage renal disease in female mice. This genetic locus mediates podocyte susceptibility to IC-mediated damage. Taking advantage of the published observation that Cgnz1 is derived from NZW and that NZW is susceptible to orchitis, epididymitis and vasitis while C57L/J is resistant to these diseases, the possibility that this genetic region also confers germ cells susceptible to damage with aspermatogenesis and sterility in an active experimental autoimmune orchitis (EAO) model was investigated. Male mice from multiple intrachromosome (chromosome 1) recombinant strains were subjected to immunization with a sperm homogenate in CFA with concomitant administration of Bordetella pertussis toxin. There was concordance of the progression from aGN to cGN, severe proteinuria and end stage renal disease with susceptibility of EAO in NZM2328 and its congenic strains with various chromosome 1 genetic intervals introgressed from C57L/J to NZM2328. Both resistant and susceptible strains made comparable anti-testis and anti-sperm Abs. Thus the genetic interval that determines susceptibility to EAO is identical to that of Cgnz1 and mapped to the 1.34 Mb region in chromosone 1. This region likely confers germ cells in the male gonad susceptible to damage by immunologically mediated inflammation. This region has been tentatively renamed Cgnz1/Eaoz1. These observations further emphasize the importance of end organ susceptibility to damage in the pathogenesis of both systemic and organ specific autoimmune diseases.


Subject(s)
Autoimmune Diseases/immunology , Genetic Predisposition to Disease , Glomerulonephritis/immunology , Kidney Failure, Chronic/immunology , Orchitis/immunology , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/genetics , Female , Gene Expression Regulation/immunology , Glomerulonephritis/complications , Glomerulonephritis/genetics , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/genetics , Male , Mice , Orchitis/etiology , Orchitis/genetics
4.
Hum Reprod Update ; 24(4): 416-441, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29648649

ABSTRACT

BACKGROUND: Infection and inflammation of the reproductive tract are significant causes of male factor infertility. Ascending infections caused by sexually transmitted bacteria or urinary tract pathogens represent the most frequent aetiology of epididymo-orchitis, but viral, haematogenous dissemination is also a contributory factor. Limitations in adequate diagnosis and therapy reflect an obvious need for further understanding of human epididymal and testicular immunopathologies and their contribution to infertility. A major obstacle for advancing our knowledge is the limited access to suitable tissue samples. Similarly, the key events in the inflammatory or autoimmune pathologies affecting human male fertility are poorly amenable to close examination. Moreover, the disease processes generally have occurred long before the patient attends the clinic for fertility assessment. In this regard, data obtained from experimental animal models and respective comparative analyses have shown promise to overcome these restrictions in humans. OBJECTIVE AND RATIONALE: This narrative review will focus on male fertility disturbances caused by infection and inflammation, and the usefulness of the most frequently applied animal models to study these conditions. SEARCH METHODS: An extensive search in Medline database was performed without restrictions until January 2018 using the following search terms: 'infection' and/or 'inflammation' and 'testis' and/or 'epididymis', 'infection' and/or 'inflammation' and 'male genital tract', 'male infertility', 'orchitis', 'epididymitis', 'experimental autoimmune' and 'orchitis' or 'epididymitis' or 'epididymo-orchitis', antisperm antibodies', 'vasectomy'. In addition to that, reference lists of primary and review articles were reviewed for additional publications independently by each author. Selected articles were verified by each two separate authors and discrepancies discussed within the team. OUTCOMES: There is clear evidence that models mimicking testicular and/or epididymal inflammation and infection have been instructive in a better understanding of the mechanisms of disease initiation and progression. In this regard, rodent models of acute bacterial epididymitis best reflect the clinical situation in terms of mimicking the infection pathway, pathogens selected and the damage, such as fibrotic transformation, observed. Similarly, animal models of acute testicular and epididymal inflammation using lipopolysaccharides show impairment of reproduction, endocrine function and histological tissue architecture, also seen in men. Autoimmune responses can be studied in models of experimental autoimmune orchitis (EAO) and vasectomy. In particular, the early stages of EAO development showing inflammatory responses in the form of peritubular lymphocytic infiltrates, thickening of the lamina propria of affected tubules, production of autoantibodies against testicular antigens or secretion of pro-inflammatory mediators, replicate observations in testicular sperm extraction samples of patients with 'mixed atrophy' of spermatogenesis. Vasectomy, in the form of sperm antibodies and chronic inflammation, can also be studied in animal models, providing valuable insights into the human response. WIDER IMPLICATIONS: This is the first comprehensive review of rodent models of both infectious and autoimmune disease of testis/epididymis, and their clinical implications, i.e. their importance in understanding male infertility related to infectious and non-infectious/autoimmune disease of the reproductive organs.


Subject(s)
Autoimmune Diseases/complications , Infections/complications , Infertility, Male/etiology , Infertility, Male/pathology , Inflammation/complications , Animals , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Disease Models, Animal , Humans , Infections/diagnosis , Infections/pathology , Infections/therapy , Infertility, Male/diagnosis , Infertility, Male/therapy , Inflammation/diagnosis , Inflammation/immunology , Inflammation/therapy , Male , Orchitis/complications , Orchitis/diagnosis , Orchitis/pathology , Orchitis/therapy , Rodentia
5.
J Clin Invest ; 127(3): 1046-1060, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28218625

ABSTRACT

Autoimmune responses to meiotic germ cell antigens (MGCA) that are expressed on sperm and testis occur in human infertility and after vasectomy. Many MGCA are also expressed as cancer/testis antigens (CTA) in human cancers, but the tolerance status of MGCA has not been investigated. MGCA are considered to be uniformly immunogenic and nontolerogenic, and the prevailing view posits that MGCA are sequestered behind the Sertoli cell barrier in seminiferous tubules. Here, we have shown that only some murine MGCA are sequestered. Nonsequestered MCGA (NS-MGCA) egressed from normal tubules, as evidenced by their ability to interact with systemically injected antibodies and form localized immune complexes outside the Sertoli cell barrier. NS-MGCA derived from cell fragments that were discarded by spermatids during spermiation. They egressed as cargo in residual bodies and maintained Treg-dependent physiological tolerance. In contrast, sequestered MGCA (S-MGCA) were undetectable in residual bodies and were nontolerogenic. Unlike postvasectomy autoantibodies, which have been shown to mainly target S-MGCA, autoantibodies produced by normal mice with transient Treg depletion that developed autoimmune orchitis exclusively targeted NS-MGCA. We conclude that spermiation, a physiological checkpoint in spermatogenesis, determines the egress and tolerogenicity of MGCA. Our findings will affect target antigen selection in testis and sperm autoimmunity and the immune responses to CTA in male cancer patients.


Subject(s)
Autoantigens/immunology , Immune Tolerance , Seminiferous Tubules/immunology , Spermatogenesis/immunology , Spermatozoa/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Male , Mice , Mice, Inbred BALB C , Sertoli Cells/immunology
6.
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.

7.
Arthritis Rheum ; 64(8): 2518-28, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22488218

ABSTRACT

OBJECTIVE: Male rats transgenic for HLA-B27 and human ß(2) -microglobulin (hß(2) m) spontaneously develop epididymoorchitis (EO) preceding the development of spondylarthritis (SpA). In the specific B27/hß(2) m-transgenic rat cross-strain (21-3 × 382-2)F(1) , only the males develop SpA, and neither sex develops gut inflammation. This study was undertaken to determine whether EO and SpA in male (21-3 × 382-2)F(1) rats are causally related. In addition, the primary characteristics of EO in this rat arthritis model were assessed. METHODS: Male B27/hß(2) m-transgenic (21-3 × 382-2)F(1) rats underwent bilateral, unilateral, or sham epididymoorchiectomy between ages 36 and 125 days. The castrated rats were given testosterone replacement. Alternatively, the 21-3 and 283-2 transgene loci were crossed with a transgene inducing aspermatogenesis. Rats were observed for the development of EO, arthritis, and spondylitis. RESULTS: In unmanipulated transgenic rats, inflammation was first evident in the ductuli efferentes (DE; ducts linking the rete testis to epididymis) as early as age 30 days. The inflammation was initially neutrophilic, and later became granulomatous. Antisperm and anti-testis cell antibodies appeared in the rat serum after age 70 days. Cells infiltrating the testes were predominantly CD4+ T cells and CD68+ or CD163+ macrophages. Quantitative polymerase chain reaction of the DE, epididymis, and testis showed elevations in the levels of interferon-γ, interleukin-10 (IL-10), and IL-17A. In addition, levels of IL-12A, IL-22, IL-23A, and IL-23 receptor were found to be elevated in the DE. Remarkably, castration of the rats before age 91 days completely prevented the subsequent onset of arthritis and spondylitis, as did transgene-induced azospermia. CONCLUSION: Autoimmune EO develops spontaneously in HLA-B27/hß(2) m-transgenic (21-3 × 283-2)F(1) rats at age 30 days, the age when antigen-positive meiotic germ cells first exit the testis. Persistent testicular inflammation and/or antigenic stimulation are essential prerequisites for the subsequent development of SpA. Thus, dysregulated innate immunity at immune-privileged sites may be an essential mechanism triggering the onset of SpA.


Subject(s)
Autoimmune Diseases/complications , Epididymitis/complications , HLA-B27 Antigen/genetics , Orchitis/complications , Sex Characteristics , Spondylarthritis/etiology , Spondylarthritis/genetics , Animals , Autoimmune Diseases/immunology , Cytokines/metabolism , Disease Models, Animal , Epididymis/metabolism , Epididymis/surgery , Epididymitis/immunology , Female , Immunity, Innate/immunology , Male , Orchitis/immunology , Phenotype , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Spondylarthritis/immunology , Testis/metabolism , Testis/surgery , Transgenes/genetics
8.
PLoS Genet ; 8(12): e1003140, 2012.
Article in English | MEDLINE | ID: mdl-23300462

ABSTRACT

Experimental autoimmune orchitis (EAO), the principal model of non-infectious testicular inflammatory disease, can be induced in susceptible mouse strains by immunization with autologous testicular homogenate and appropriate adjuvants. As previously established, the genome of DBA/2J mice encodes genes that are capable of conferring dominant resistance to EAO, while the genome of BALB/cByJ mice does not and they are therefore susceptible to EAO. In a genome scan, we previously identified Orch3 as the major quantitative trait locus controlling dominant resistance to EAO and mapped it to chromosome 11. Here, by utilizing a forward genetic approach, we identified kinesin family member 1C (Kif1c) as a positional candidate for Orch3 and, using a transgenic approach, demonstrated that Kif1c is Orch3. Mechanistically, we showed that the resistant Kif1c(D2) allele leads to a reduced antigen-specific T cell proliferative response as a consequence of decreased MHC class II expression by antigen presenting cells, and that the L(578) → P(578) and S(1027) → P(1027) polymorphisms distinguishing the BALB/cByJ and DBA/2J alleles, respectively, can play a role in transcriptional regulation. These findings may provide mechanistic insight into how polymorphism in other kinesins such as KIF21B and KIF5A influence susceptibility and resistance to human autoimmune diseases.


Subject(s)
Disease Resistance/genetics , Genes, Dominant , Kinesins/genetics , Orchitis , Alleles , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Gene Expression , Genes, MHC Class II , Genetic Predisposition to Disease , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Orchitis/genetics , Orchitis/immunology , Quantitative Trait Loci/genetics , Testis/immunology
9.
Proc Natl Acad Sci U S A ; 108(18): 7511-6, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502500

ABSTRACT

Vasectomy is a well accepted global contraceptive approach frequently associated with epididymal granuloma and sperm autoantibody formation. To understand the long-term sequelae of vasectomy, we investigated the early immune response in vasectomized mice. Vasectomy leads to rapid epithelial cell apoptosis and necrosis, persistent inflammation, and sperm granuloma formation in the epididymis. Vasectomized B6AF1 mice did not mount autoimmune response but instead developed sperm antigen-specific tolerance, documented as resistance to immunization-induced experimental autoimmune orchitis (EAO) but not experimental autoimmune encephalomyelitis. Strikingly, tolerance switches over to pathologic autoimmune state following concomitant CD4(+)CD25(+)Foxp3(+) regulatory T cell (Treg) depletion: unilaterally vasectomized mice produce dominant autoantibodies to an orchitogenic antigen (zonadhesin), and develop CD4 T-cell- and antibody-dependent bilateral autoimmune orchitis. Therefore, (i) Treg normally prevents spontaneous organ-specific autoimmunity induction by persistent endogenous danger signal, and (ii) autoantigenic stimulation with sterile autoinflammation can lead to tolerance. Finally, postvasectomy tolerance occurs in B6AF1, C57BL/6, and A/J strains. However, C57BL/6 mice resisted EAO after 60% Treg depletion, but developed EAO after 97% Treg reduction. Therefore, variance in intrinsic Treg function--a possible genetic trait--can influence the divergent tolerogenic versus autoimmune response to vasectomy.


Subject(s)
Autoimmunity/immunology , Immune Tolerance/immunology , Spermatozoa/immunology , T-Lymphocytes, Regulatory/immunology , Vasectomy , Animals , Autoantibodies/immunology , Blotting, Western , Cell Proliferation , Electrophoresis, Polyacrylamide Gel , Male , Membrane Proteins/immunology , Mice , Mice, Mutant Strains , Statistics, Nonparametric
10.
J Immunol ; 186(1): 382-9, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21135167

ABSTRACT

Day 3 thymectomy (D3Tx) results in a loss of peripheral tolerance mediated by natural regulatory T cells (nTregs) and development of autoimmune ovarian dysgenesis (AOD) and autoimmune dacryoadenitis (ADA) in A/J and (C57BL/6J × A/J) F(1) hybrids (B6A), but not in C57BL/6J (B6) mice. Previously, using quantitative trait locus (QTL) linkage analysis, we showed that D3Tx-AOD is controlled by five unlinked QTL (Aod1-Aod5) and H2. In this study, using D3Tx B6-Chr(A/J)/NaJ chromosome (Chr) substitution strains, we confirm that QTL on Chr16 (Aod1a/Aod1b), Chr3 (Aod2), Chr1 (Aod3), Chr2 (Aod4), Chr7 (Aod5), and Chr17 (H2) control D3Tx-AOD susceptibility. In addition, we also present data mapping QTL controlling D3Tx-ADA to Chr17 (Ada1/H2), Chr1 (Ada2), and Chr3 (Ada3). Importantly, B6-ChrX(A/J) mice were as resistant to D3Tx-AOD and D3Tx-ADA as B6 mice, thereby excluding Foxp3 as a susceptibility gene in these models. Moreover, we report quantitative differences in the frequency of nTregs in the lymph nodes (LNs), but not spleen or thymus, of AOD/ADA-resistant B6 and AOD/ADA-susceptible A/J, B6A, and B6-Chr17(A/J) mice. Similar results correlating with experimental allergic encephalomyelitis and orchitis susceptibility were seen with B10.S and SJL/J mice. Using H2-congenic mice, we show that the observed difference in frequency of LN nTregs is controlled by Ada1/H2. These data support the existence of an LN-specific, H2-controlled mechanism regulating the prevalence of nTregs in autoimmune disease susceptibility.


Subject(s)
Autoimmune Diseases/immunology , H-2 Antigens/physiology , Lymph Nodes/cytology , Lymph Nodes/immunology , Oophoritis/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Thymectomy , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/surgery , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Chromosomes/genetics , Dacryocystitis/genetics , Dacryocystitis/immunology , Disease Susceptibility/immunology , Female , Genetic Linkage/immunology , Lymph Nodes/metabolism , Lymphocyte Count , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Transgenic , Oophoritis/genetics , Quantitative Trait Loci/immunology
11.
Cell Immunol ; 260(2): 119-27, 2010.
Article in English | MEDLINE | ID: mdl-19914609

ABSTRACT

The postnatal maternal environment is known to increase susceptibility to a number of autoimmune diseases. Here we asked whether the postnatal maternal environment could influence autoimmune disease development to day 3 thymectomy (d3tx)-induced autoimmune ovarian disease (AOD) and experimental allergic encephalomyelitis (EAE) in cross-fostered A/J and B6 mice. A/J pups foster-nursed by B6 mothers exhibit an increase in autoimmune disease development while cross-fostering B6 pups on A/J mothers did not alter their susceptibility. The increase in AOD incidence seen in foster-nursed d3tx A/J mice correlated with a decrease in the total number of CD4(+) T cells in the lymph nodes of these animals. Analysis of the cellular composition in the milk revealed that B6 mice shed significantly more maternally derived lymphocytes into their milk compared to A/J mothers. These data suggest that there are maternally derived postnatal factors that influence the development of autoimmune disease in A/J mice.


Subject(s)
Animals, Newborn/immunology , Animals, Suckling/immunology , Autoimmune Diseases/immunology , Disease Susceptibility/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Chemokine CXCL1/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunity, Maternally-Acquired/immunology , Interleukin-13/metabolism , Interleukin-9/metabolism , Lactation/immunology , Lactation/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Ovarian Diseases/immunology , Thymectomy
12.
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
13.
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
14.
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
15.
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
16.
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
17.
J Biol Chem ; 280(44): 36865-72, 2005 Nov 04.
Article in English | MEDLINE | ID: mdl-16093248

ABSTRACT

Immunotherapeutic drugs that mimic sphingosine 1-phosphate (S1P) disrupt lymphocyte trafficking and cause T helper and T effector cells to be retained in secondary lymphoid tissue and away from sites of inflammation. The prototypical therapeutic agent, 2-alkyl-2-amino-1,3-propanediol (FTY720), stimulates S1P signaling pathways only after it is phosphorylated by one or more unknown kinases. We generated sphingosine kinase 2 (SPHK2) null mice to demonstrate that this kinase is responsible for FTY720 phosphorylation and thereby its subsequent actions on the immune system. Both systemic and lymphocyte-localized sources of SPHK2 contributed to FTY720 induced lymphopenia. Although FTY720 was selectively activated in vivo by SPHK2, other S1P pro-drugs can be phosphorylated to cause lymphopenia through the action of additional sphingosine kinases. Our results emphasize the importance of SPHK2 expression in both lymphocytes and other tissues for immune modulation and drug metabolism.


Subject(s)
Immunosuppressive Agents/toxicity , Lymphocytes/drug effects , Lymphocytes/enzymology , Lymphopenia , Phosphotransferases (Alcohol Group Acceptor)/physiology , Propylene Glycols/toxicity , Animals , Fingolimod Hydrochloride , Flow Cytometry , Homozygote , Immunosuppressive Agents/metabolism , Lymphopenia/chemically induced , Lymphopenia/enzymology , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prodrugs/metabolism , Prodrugs/toxicity , Propylene Glycols/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/analogs & derivatives , Sphingosine/metabolism
18.
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
19.
J Immunol ; 175(2): 944-50, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16002693

ABSTRACT

The development of organ-specific autoimmune diseases in mice thymectomized on day 3 of life (d3tx mice) can be prevented by transferring CD4(+)CD25(+) T cells from syngeneic, normal adult mice. Using a d3tx model, we asked whether CD4(+)CD25(+) T cell deficiency contributes to glomerulonephritis (GN) in lupus-prone mice. New Zealand Mixed 2328 (NZM2328) mice spontaneously develop autoantibodies to dsDNA and female-dominant, fatal GN. After d3tx, both male and female NZM2328 mice developed 1) accelerated dsDNA autoantibody response, 2) early onset and severe proliferative GN with massive mesangial immune complexes, and 3) autoimmune disease of the thyroid, lacrimal gland, and salivary gland. The d3tx male mice also developed autoimmune prostatitis. The transfer of CD25(+) cells from 6-wk-old asymptomatic NZM2328 donors effectively suppressed dsDNA autoantibody and the development of autoimmune diseases, with the exception of proliferative lupus GN and sialoadenitis. This finding indicates that NZM2328 lupus mice have a selective deficiency in T cells that regulates the development of lupus GN and sialoadenitis. After d3tx, the proliferative GN of female mice progressed to fatal GN, but largely regressed in the male, thereby revealing a checkpoint in lupus GN progression that depends on gender.


Subject(s)
Genetic Predisposition to Disease , Glomerulonephritis, Membranoproliferative/immunology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Receptors, Interleukin-2/biosynthesis , Sialadenitis/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Age Factors , Animals , Antibodies, Antinuclear/biosynthesis , Antibodies, Antinuclear/blood , Antigen-Antibody Complex/blood , Antigen-Antibody Complex/metabolism , Chronic Disease , DNA/immunology , Disease Progression , Female , Glomerulonephritis, Membranoproliferative/physiopathology , Glomerulonephritis, Membranoproliferative/prevention & control , Kidney Glomerulus/immunology , Kidney Glomerulus/metabolism , Lacrimal Apparatus Diseases/prevention & control , Lupus Nephritis/physiopathology , Male , Mice , Prostatitis/prevention & control , Sex Factors , Sialadenitis/physiopathology , Sialadenitis/prevention & control , T-Lymphocytes, Regulatory/metabolism , Thymectomy , Thyroiditis, Autoimmune/prevention & control
20.
J Immunol ; 173(2): 1051-8, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15240693

ABSTRACT

A model of neonatal autoimmune disease has been described recently in which an epitope-specific autoantibody to murine zona pellucida 3 induces severe ovarian disease in neonatal, but not adult, mice (neonatal AOD). The autoantibody forms immune complex with endogenous ovarian zona pellucida 3, and a pathogenic CD4(+) T cell response is triggered. The basis for the predominant neonatal susceptibility has not been clarified. In this study innate immunity, including neonatal NK cells, in neonatal AOD was investigated. Neonatal spleen contained readily detectable NK1.1(+)TCRVbeta(-), but not NK1.1(+)TCRVbeta(+), cells. Ab depletion of NK1.1(+)TCRVbeta(-) cells inhibited neonatal AOD development. Moreover, in adoptive transfer of neonatal AOD, recipient disease was ameliorated when either donor or recipient NK cells were depleted. Thus, NK cells operate in both induction and effector phases of the disease. IFN-gamma was produced by neonatal NK cells in vivo, and it may be important in neonatal AOD. Indeed, ovaries with neonatal AOD expressed high levels of IFN-gamma and TNF-alpha which correlated with disease severity, and the disease was inhibited by IFN-gamma or TNF-alpha Ab. Importantly, disease was enhanced by recombinant IFN-gamma, and treatment of T cell donors with IFN-gamma Ab also significantly reduced adoptive transfer of neonatal AOD. Finally, neonatal AOD was ameliorated in mice deficient in FcgammaRIII and was enhanced in FcgammaRIIB-deficient mice. We conclude that neonatal NK cells promote pathogenic T cell response at multiple stages during neonatal autoimmune disease pathogenesis. Also operative in neonatal AOD are other mediators of the innate system, including proinflammatory cytokines and FcgammaRIII signaling.


Subject(s)
Autoimmunity/immunology , Cytokines/metabolism , Immune Complex Diseases/immunology , Killer Cells, Natural/immunology , Ovarian Diseases/immunology , Animals , Female , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Mice , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...