Abstract
The characterization of functional CD8+ inhibitory or regulatory T cells and their gene regulation remains a critical challenge in the field of tolerance and autoimmunity. Investigating the genes induced in regulatory cells and the regulatory networks and pathways that underlie mechanisms of immune resistance and prevent apoptosis in the CD8+ T cell compartment are crucial to understanding tolerance mechanisms in systemic autoimmunity. Little is currently known about the genetic control that governs the ability of CD8+ Ti or regulatory cells to suppress anti-DNA Ab production in B cells. Silencing genes with siRNA or shRNA and overexpression of genes with lentiviral cDNA transduction are established approaches to identifying and understanding the function of candidate genes in tolerance and immunity. Elucidation of interactions between genes and proteins, and their synergistic effects in establishing cell-cell cross talk, including receptor modulation/antagonism, are essential for delineating the roles of these cells. In this review, we will examine recent reports which describe the modulation of cells from lupus prone mice or lupus patients to confer anti-inflammatory and protective gene expression and novel associated phenotypes. We will highlight recent findings on the role of selected genes induced by peptide tolerance in CD8+ Ti.
Keywords: Autoimmunity, Systemic Lupus Erythematosus, CD8+ Treg, Genes and Tolerance
1. Introduction
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by widespread inflammation, autoantibody production, and immune complex deposition. While SLE affects multiple major organ systems in the body, lupus nephritis is the leading cause of death. Approximately two million people suffer from SLE with the majority of cases being women of childbearing age. Lupus is a gender-biased disease with a female to male ratio of 9:1. African American women are three times more likely to get lupus (SLE) than white women. Lupus is also more common in Hispanic, Asian, and Native American women. In SLE, immune homeostasis is impaired, and both T and B cells acquire abnormal characteristics in terms of activation, cytokine production, and proliferative potential. The modulation of abnormal immune regulation is the subject of intense investigations in several experimental autoimmune diseases. Significant therapeutic goals include limiting the number of abnormal pathogenic cells and autoantibodies, and restoration of immune system self-tolerance by the administration of peptides that induce regulatory/inhibitory T cells (Ti).
Decreased numbers, and or impaired function, of CD4+ regulatory T cells (Treg) have emerged as an important pathogenic mechanism in SLE [1–9]. Targeting of regulatory CD4+ T cells and CD8+Ti in SLE for therapeutic purposes through use of self-peptides to alter the functional properties of these cells and to halt disease progression has been attempted [10–19]. We have shown previously that BWF1 lupus mice are protected from autoimmune disease after i.v. injection of high doses of pConsensus (pCons), a synthetic peptide based on sequences of murine anti-dsDNA antibodies that are presented by both MHC class I and II molecules [11]. Tolerance induction by pCons peptide treatment enhances the numbers of both CD8+Ti and CD4+ Treg. Critically, both of these cell populations suppress the proliferation of effector CD4+CD25− CD4+ T cells and B cells [8, 10, 16, 17, 19]. We also have evidence that pCons peptide induces Treg in SLE patient cells in vitro and these cells suppress the proliferation of autologous CD4+CD25− effector cells. Furthermore, we found an inverse correlation between the expression levels of the Foxp3 gene in Treg and SLE disease activity (SLEDAI) [20]. In this review, we will discuss some of our recent findings and highlight the work of others in the field.
2. Potential contributions of CD8+ regulatory T cells to immune tolerance in Lupus
The role of CD8+ Ti as Treg has only recently begun to be examined as a novel approach in the field of immune tolerance [21–24]. Clues to the regulatory function of CD8+T cells have emerged from studies in autoimmune diseases such as experimental autoimmune encephalomyelitis [25–28], myasthenia gravis [29], and SLE [21, 30–33]. Recent studies have provided evidence that both CD4+ Treg and CD8+ suppressor T cells play crucial roles in the prevention of autoimmunity [6, 8, 10, 16, 17, 34–36]. Via and colleagues recently ascribed to donor CD8+T cells a role in the prevention of lupus in a murine model of graft vs host disease, by inhibition of effector T cells that cause the disease [37–39]. Fan and Singh reported that therapeutically induced CD8+CTL kill autoantibody-producing B cells and inhibit murine lupus [40]. By administration of nucleosomal histone peptides to (SWRXNZB) F1 (SNF1) mice, Datta and colleagues induced CD4+ and CD8+ TGFβ+ Treg that subsequently delayed B cell activation and nephritis [13, 41]. This group also reported that TGFβ-producing human CD8+ Treg are associated with immunological remission of lupus following autologous hematopoietic stem cell transplantation in SLE patients [32]. Kumar and colleagues showed that Qa-1 restricted CD8αα+ TCRαβ+ T cells regulate immunity [23, 42, 43]. Using the BWF1 SLE mouse model, Mozes’ group studied induced Treg in mice treated with a tolerogenic peptide based on the light chain complementarity-determining region 1 (hCDR1) of human anti-dsDNA antibodies [15, 44]. Tolerization of mice with hCDR1 induced CD4+CD25high and CD8+CD28 Treg, which suppressed lymphocyte proliferation and autoantibody production [45]. We found, in our similar model of tolerance induced by pCons, that inhibitory cells were present in both CD8+CD28+ and CD8+CD28− subsets. However, the expression of Foxp3 and TGFβ mRNAs was higher and lasted longer in the CD28− subsets [17]. Recently, the Cantor group described a population of Qa-1 restricted CD8+ T cells that inhibit lupus-like disease and target autoreactive CD4+T follicular helper cells (TFH) [22, 46]. These CD8+ Ti cells maintain self-tolerance by recognition of Qa-1 peptide ligands expressed at the surface of follicular helper T cells. Recently, we have shown that pCons-induced CD8+Ti suppress autoimmunity in a murine model of SLE in a manner dependent on Foxp3 expression [10, 16, 17]. Following pCons administration, CD8+ Ti display a unique genetic profile, with upregulated genes including Foxp3, Trp53, Bcl2, CCR7, IFNAR1, and IFI202b and downregulated genes including regulator of G protein signaling proteins (RGS2, RGS16, and RGS17), glutamic pyruvate transaminase (GPT2), BAX, programmed cell death-1 (PD1), growth arrest and DNA damage inducible 45 beta (GADD45β), and phosphodiesterase 3b (PDE3b) [47]. CD8+Ti in our tolerance model expressed low levels of PD1, CTLA4 and CD122, and a partial characterization of the genetic basis of their suppressive capacity indicates some dependence on the expression of FoxP3, PD1, and IFI202b [10, 16, 17, 19]. However, as yet not all the critical genetic elements required for the full spectrum and function of suppressive activity in this cell population has been elucidated.
3. Cellular, molecular, and functional basis for gene selection in tolerance model
While SLE and related autoimmune diseases are known to be complex and polygenic, not all the susceptibility genes are either sufficient or required for disease progression. Furthermore, some genes regarded as susceptibility genes when expressed in effector T or B cells, e.g., upregulated anti-apoptotic (bcl2) and downregulated apoptotic genes (Bax, GADD45β), may be advantageous in the activation and suppressive actions of CD8+Ti when expressed in these cells. Presumably, certain genes involved in important T cell functions, such as those mediating chemokine/cytokine receptor trafficking or signaling, (e.g., IFNAR, Ifi202b, CCR7) could prove to be crucial for the suppressive effects of CD8+Ti. Our recent data highlight multiple genes whose products appear to be advantageous in these cells. We will next discuss further the salient genes we have identified in microarray analyses [47], offering insights into their possible roles in immune tolerance and autoimmunity. Validation of our results by RT-PCR and protein expression studies has substantiated the likelihood that these genes play roles in autoimmunity.
IFI202b
IFI202b is an interferon inducible gene. We have found significant increased expression of ifi202b in tolerized CD8+Ti cells [19]. This increased expression lasted at least four weeks after pCons tolerance. Upregulation of the IFN-inducible gene Ifi202b, is significant because: a) in B cells, Ifi202 upregulation protects them from apoptosis and predisposes NZB mice to SLE [48], b) we and others have demonstrated that the “profile” of peripheral blood cells from SLE patients exhibits multiple upregulated genes under the control of interferons [49, 50], and c) recent experiments show that deficiency of IFNγRII (surface receptor for type II IFN) in MRL/lpr/lpr mice prevents SLE, whereas knockout of IFNγRI (type I IFN receptor) accelerates the disease [51]. Another gene in the IFI200 cluster, Ifi202a, has recently been shown to be dispensable for B cell function [52]. A recent study has found that female and male sex hormones differentially regulate the expression of Ifi202 within the Nba2 interval of C57BL/6 mice [53]. Female sex hormone 17-β estradiol upregulates, and male sex hormone androgen decreases, Ifi202 expression. The Ifi202 gene product has also been reported to bind to p53 and prevent its effects on the expression of pro-apoptosis genes [54]. Thus, the role IFI202b of is complex and more studies are needed before its role can be elucidated.
Bcl-2 (B cell lymphoma 2)
Bcl-2 is an important apoptosis regulatory gene. Bcl-2 has been implicated in many cancers [55, 56]. Association between Bcl2 polymorphism and SLE has been reported [57]. We previously demonstrated that apoptosis was significantly decreased in tolerized CD8+Ti cells [10]. We have found increased expression of Bcl-2 in tolerized CD8+ Ti cells in BWF1 mice [58]. Similarly, others have found increased expression of Bcl-2 and Bcl-XL in a model of immune tolerance in SLE [59, 60]. Our work and that of others reinforce the concept that Bcl-2 plays an important role in regulating and shaping the T cell repertoire and immune tolerance by inhibiting programmed cell death in specific T cell subsets including CD8+Ti [61]. This probably allows the suppressive cells to survive long enough to produce clinically significant suppression of effector T and B cells.
p53
p53, a tumor suppressor gene, is a DNA-binding transcription factor that acts to regulate the cell cycle and plays an important role in control of cancer. Mutations at the p53 gene lead to cancer [62]. P53 has been linked to SMAD/TGFβ interactions that promote cell senescence [63], and has multiple effects, both positive and negative, on cell cycle arrest and apoptosis [63, 64]. The p53 protein has recently been shown to play major roles in infectious diseases and immunity [65], and its gene is located in a human gene region linked to SLE [66]. Its levels are increased in lymphocytes from SLE patients compared to controls [67] and in one ethnic group a polymorphism of p53 is associated with SLE [68]. A recent study identifies the major role of p53 in the increased formation of exosomes, a non-classical protein secretion pathway, by upregulated expression of the target gene TSAP6 [65, 69]. We detected increased expression of p53 in CD8+Ti [58]. Conceivably, p53 may play a novel role in support of CD8+Ti-mediated tolerance by promoting the exosome-mediated delivery of TGFβ or other key growth factors.
Foxp3
Foxp3 is a master regulator and it plays a significant role in the development and function of Treg. It is a marker for both natural and induced Treg. In humans, mutation in the Foxp3 gene leads to immunodysregulation, polyendocrinopathy, and enteropathy, X-linked (IPEX) syndrome [70]. In mice, Foxp3 deficiency (The Scurfy mouse) is associated with lymphoproliferation (ref). Whereas Foxp3 is known to be critical to the function of Treg [71], not all of the important signaling pathways are yet understood [72]. Indeed, Foxp3 either down- (as in most cases) or upregulated [73] by virtue of its high-affinity binding to a specific consensus binding site, ATAAACAA, in its target genes. Upregulation of Foxp3 in rodent CD8+CD28− Ti may induce CD4+ Treg [74]. TGFβ upregulation of Foxp3 expression [75] identified a new role for this growth factor in immunosuppression. Foxp3 expression CD4+ regulatory T cells) was decreased in the absence of CD8+ Treg, demonstrating a functional link between the two subsets of Treg in which, CD8+CD28− Treg are required for the optimal expansion and function of peptide hCDR1 induced CD4+ Treg in murine lupus [45, 76, 77]. We have found increased expression of Foxp3 in CD8+ Ti cells, and silencing of Foxp3 with siRNA abrogates the suppression of anti-DNA Ab induced by CD8+ Ti cells [10, 17]. These data indicate that Foxp3 plays an important role in the suppression of anti- DNA Ab in murine SLE.
Selected downregulated genes in our model of tolerance will be discussed here due to their known roles in autoimmunity [78, 79].
Regulator of G protein signaling (RGS) genes
RGS proteins are potent GTPase-activating proteins (GAP) for heterotrimeric G protein (Gq, Gi, and Go family) alpha subunits, acting as multifunctional inhibitors of signal transduction in many cells [80, 81]. They are also capable of “fine-tuning” GPCR signaling in lymphocytes [82]. In particular, RGS2, a growth-inhibitory protein, plays a role in leukemogenesis [83]. Semplicini et al reported that reduction of RGS2 signaling increases Ca2+ mobilization and ERK1/2 activation in response to GPCR stimulation [84]. We found that multiple RGS proteins are downregulated in tolerized CD8+Ti cells [58].
GPT2
GPT2, is an alanine transaminase, that has a role in gluconeogenesis and amino acid homeostasis, and is an enzyme/biomarker [85] that is upregulated in disease states. GPT2 is implicated in exacerbating autoimmune disease [86], and its levels were shown to be increased in NZB/NZW F1 mice [87]. Increased serum aminotransferases have been reported to be associated with anti-mitochondrial antibodies in SLE patients with autoimmune liver disease [88]. We have found decreased GPT2 mRNA and protein expression in CD8+Ti from tolerized BWF1 mice [58].
PDE3b
PDE3b was downregulated in CD8+ Ti. Foxp3 binding to its conserved site within the first intron of the PDE3b gene potently represses expression of this negative signaling regulatory enzyme. PDE3b is involved in controlling the abundance of cyclic adenosine monophosphate (cAMP) and plays an important role in the suppression of T cell function [89]. The downregulation of PDE3b has been associated with enhanced insulin secretion, suggesting that secretion of other factors could also be positively modulated. Consistent with the notion that reversal of its cAMP-degrading activity is important for maintenance of CD8+Ti, PDE3b is one of the most down-regulated genes in Treg [89].
Bax
The Bcl-2–associated X protein (Bax), is a protein of the Bcl-2 family. It promotes apoptosis by competing with Bcl-2. The expression of BAX is upregulated by p53 and Bax has been shown to be involved in p53-mediated apoptosis. The Bax gene has also been implicated in lupus nephritis [78]. Decreased expression of Bax has been demonstrated in active SLE patient PBLs compared to inactive or normal controls [90]. However, recent reports showing increased apoptosis and higher expression of Bax and caspase-3 in kidney cells in human lupus nephritis (LN) suggests that apoptosis might be induced through these protein pathways, possibly with resultant tissue damage that plays a role in lupus nephritis progression. [91]. The Bax gene has also been linked to cancer and aging [92, 93]. Altered Bax expression and decreased apoptosis in bone marrow cells of lupus-susceptible NZB/W mice has been reported [94]. We have found decreased expression of the Bax gene both at the mRNA and protein level in CD8+ Ti [58]. The Bax protein forms heterodimers with bcl-2 protein, which bind to the voltage-dependent anion channels (VDAC) in mitochondria and induce loss of membrane potential and release of cytochrome c to the cytoplasm. Our data showing upregulation of Bcl2 and downregulation of Bax in CD8+ Ti may explain the resistance of these induced suppressor cells to apoptosis.
PD1
Programmed cell death 1 (PD-1) is an inhibitory receptor known to play an important role in the regulation of autoimmunity and in the maintenance of peripheral tolerance [16, 95, 96]. PD-1 is a member of the CD28/B7 super family of costimulatory molecules that regulates T cell tolerance and is expressed on activated CD4+T cells, CD8+T cells, and antigen presenting cells (APCs) [95, 97]. PD1 has two ligands, PD ligand 1 (PD-L1) and PD-L2, both of which are type I transmembrane proteins, yet their expression patterns differ [98–100]. PD-L1 is constitutively expressed on murine T cells, B cells, macrophages, dendritic cells (DCs), mesenchymal stem cells, and bone marrow-derived mast cells, and is further upregulated in a number of these cell types upon activation [101]. Expression of PD-L2 is restricted to macrophages, DCs, bone marrow-derived mast cells, and peritoneal B1 cells [95, 97, 101–103].
PD-1-deficient mice develop spontaneous autoimmune diseases including SLE (nephritis and autoantibodies in C57BL/6 mice and cardiomyopathy in Balb/c mice), indicating an essential function of PD-1 in immune tolerance mechanisms [96, 104, 105]. Studies have suggested that PD-1/PD-L1 interaction plays a key role in downregulating the activation of T cells [106]. Blockade of PD-1 has been shown to lead to a worsening of symptoms in murine models of graft-verus-host disease [107] and Experimental Autoimmune Encephomyelitis (EAE) [108]. In contrast, we have shown that blockade of PD-1 protects lupus-prone mice from disease[109]. PD-1 mediated peripheral tolerance involves CD4+CD25+ T regulatory cells (Treg), which have been shown to highly express both PD-1 and PD-L1 [110]. We have shown that there is reduced expression of PD-1 in inhibitory CD8+T cells (CD8+Ti) compared to non-inhibitory CD8+ T cells following tolerization with the pCons peptide [16]. However, inhibiting expression of PD-1 even more with specific siRNA abrogates the ability of the cells to suppress. Thus, expression of a small quantity of PD-1 is required for the cells to be inhibitory, but expression of high amounts is associated with inability to suppress[16]. We also found that blocking of PD-1 decreased FoxP3 expression in CD8+Ti cells from tolerized mice indicating cross regulation[16]. PD-1 is also important in inducing Treg as well as in the mechanisms of action of other suppressive/inhibitory T cell populations [111–113].
4. Conclusions
Peptide tolerance can induce specific CD8+ Ti or regulatory cells with a unique gene signature. Silencing of selected genes abrogates the suppression induced by regulatory CD8+T cells. Interferon and apoptosis gene signatures are characteristic of a protective phenotype in regulatory CD8+ Ti cells. Peptide tolerance induces distinct gene signature including both up and down regulation of selected genes in our tolerance model. Some of the gene signature changes may play a significant role in lupus pathology and could have the potential to aid in the development of future therapeutics.
Take-home messages.
Regulatoy CD8+T cells are heterogeneous in mode of induction, phenotype, and function. Their regulation depends on tolerance mechanisms and treatment regimens.
Peptide tolerance, corticosteroids and various epigenetic agents can alter the gene signature in CD8+ Ti cells.
Interferon- and apoptosis-related genes can be induced in lupus cells with peptide tolerance and other epigenetic agents such as histones and vitamin A metabolites.
Both up and down regulated genes are equally important in “resetting” immune responses in SLE.
Acknowledgments
RPS is supported by NIH grants AR54034, AI 083894, AI65645, UCLA Clinical Translational Seed Grant (CTSG) and American Autoimmune Related Disease Association grant (AARDA). BHH is supported by an award from TEVA Pharmaceuticals. BHH and RPS are also supported by UCLA Senate Core Grant.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Crispin JC, Alcocer-Varela J, de Pablo P, Martinez A, Richaud-Patin Y, Alarcon-Segovia D. Immunoregulatory defects in patients with systemic lupus erythematosus in clinical remission. Lupus. 2003;12:386–93. doi: 10.1191/0961203303lu368oa. [DOI] [PubMed] [Google Scholar]
- 2.Crispin JC, Martinez A, Alcocer-Varela J. Quantification of regulatory T cells in patients with systemic lupus erythematosus. J Autoimmun. 2003;21:273–6. doi: 10.1016/s0896-8411(03)00121-5. [DOI] [PubMed] [Google Scholar]
- 3.Liu MF, Wang CR, Fung LL, Wu CR. Decreased CD4+CD25+ T cells in peripheral blood of patients with systemic lupus erythematosus. Scand J Immunol. 2004;59:198–202. doi: 10.1111/j.0300-9475.2004.01370.x. [DOI] [PubMed] [Google Scholar]
- 4.Miyara M, Amoura Z, Parizot C, Badoual C, Dorgham K, Trad S, et al. Global natural regulatory T cell depletion in active systemic lupus erythematosus. J Immunol. 2005;175:8392–400. doi: 10.4049/jimmunol.175.12.8392. [DOI] [PubMed] [Google Scholar]
- 5.Valencia X, Yarboro C, Illei G, Lipsky PE. Deficient CD4+CD25high T Regulatory Cell Function in Patients with Active Systemic Lupus Erythematosus. J Immunol. 2007;178:2579–88. doi: 10.4049/jimmunol.178.4.2579. [DOI] [PubMed] [Google Scholar]
- 6.Scalapino KJ, Tang Q, Bluestone JA, Bonyhadi ML, Daikh DI. Suppression of disease in New Zealand Black/New Zealand White lupus-prone mice by adoptive transfer of ex vivo expanded regulatory T cells. J Immunol. 2006;177:1451–9. doi: 10.4049/jimmunol.177.3.1451. [DOI] [PubMed] [Google Scholar]
- 7.Horwitz DA, Gray JD, Zheng SG. The potential of human regulatory T cells generated ex vivo as a treatment for lupus and other chronic inflammatory diseases. Arthritis Res. 2002;4:241–6. doi: 10.1186/ar414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.La Cava A, Ebling FM, Hahn BH. Ig-reactive CD4+CD25+ T cells from tolerized (New Zealand Black × New Zealand White)F1 mice suppress in vitro production of antibodies to DNA. J Immunol. 2004;173:3542–8. doi: 10.4049/jimmunol.173.5.3542. [DOI] [PubMed] [Google Scholar]
- 9.Horwitz DA, Zheng SG, Gray JD, Wang JH, Ohtsuka K, Yamagiwa S. Regulatory T cells generated ex vivo as an approach for the therapy of autoimmune disease. Semin Immunol. 2004;16:135–43. doi: 10.1016/j.smim.2003.12.009. [DOI] [PubMed] [Google Scholar]
- 10.Hahn BH, Singh RP, La Cava A, Ebling FM. Tolerogenic treatment of lupus mice with consensus peptide induces Foxp3-expressing, apoptosis-resistant, TGFbeta-secreting CD8+ T cell suppressors. J Immunol. 2005;175:7728–37. doi: 10.4049/jimmunol.175.11.7728. [DOI] [PubMed] [Google Scholar]
- 11.Hahn BH, Singh RR, Wong WK, Tsao BP, Bulpitt K, Ebling FM. Treatment with a consensus peptide based on amino acid sequences in autoantibodies prevents T cell activation by autoantigens and delays disease onset in murine lupus. Arthritis Rheum. 2001;44:432–41. doi: 10.1002/1529-0131(200102)44:2<432::AID-ANR62>3.0.CO;2-S. [DOI] [PubMed] [Google Scholar]
- 12.Kang HK, Liu M, Datta SK. Low-dose Peptide tolerance therapy of lupus generates plasmacytoid dendritic cells that cause expansion of autoantigen-specific regulatory T cells and contraction of inflammatory th17 cells. J Immunol. 2007;178:7849–58. doi: 10.4049/jimmunol.178.12.7849. [DOI] [PubMed] [Google Scholar]
- 13.Kang HK, Michaels MA, Berner BR, Datta SK. Very low-dose tolerance with nucleosomal peptides controls lupus and induces potent regulatory T cell subsets. J Immunol. 2005;174:3247–55. doi: 10.4049/jimmunol.174.6.3247. [DOI] [PubMed] [Google Scholar]
- 14.Riemekasten G, Langnickel D, Enghard P, Undeutsch R, Humrich J, Ebling FM, et al. Intravenous injection of a D1 protein of the Smith proteins postpones murine lupus and induces type 1 regulatory T cells. J Immunol. 2004;173:5835–42. doi: 10.4049/jimmunol.173.9.5835. [DOI] [PubMed] [Google Scholar]
- 15.Sharabi A, Haviv A, Zinger H, Dayan M, Mozes E. Amelioration of murine lupus by a peptide, based on the complementarity determining region-1 of an autoantibody as compared to dexamethasone: different effects on cytokines and apoptosis. Clin Immunol. 2006;119:146–55. doi: 10.1016/j.clim.2006.01.007. [DOI] [PubMed] [Google Scholar]
- 16.Singh RP, La Cava A, Hahn BH. pConsensus Peptide Induces Tolerogenic CD8+ T Cells in Lupus-Prone (NZB × NZW)F1 Mice by Differentially Regulating Foxp3 and PD1 Molecules. J Immunol. 2008;180:2069–80. doi: 10.4049/jimmunol.180.4.2069. [DOI] [PubMed] [Google Scholar]
- 17.Singh RP, La Cava A, Wong M, Ebling F, Hahn BH. CD8+ T Cell-Mediated Suppression of Autoimmunity in a Murine Lupus Model of Peptide-Induced Immune Tolerance Depends on Foxp3 Expression. J Immunol. 2007;178:7649–57. doi: 10.4049/jimmunol.178.12.7649. [DOI] [PubMed] [Google Scholar]
- 18.Singh RR, Ebling FM, Albuquerque DA, Saxena V, Kumar V, Giannini EH, et al. Induction of autoantibody production is limited in nonautoimmune mice. J Immunol. 2002;169:587–94. doi: 10.4049/jimmunol.169.1.587. [DOI] [PubMed] [Google Scholar]
- 19.Dinesh R, Hahn BH, La Cava A, Singh RP. Interferon-inducible gene 202b controls CD8(+) T cell-mediated suppression in anti-DNA Ig peptide-treated (NZB × NZW) F1 lupus mice. Genes Immun. 2011:12. doi: 10.1038/gene.2011.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hahn BH, Anderson M, Le E, La Cava A. Anti-DNA Ig peptides promote Treg cell activity in systemic lupus erythematosus patients. Arthritis Rheum. 2008;58:2488–97. doi: 10.1002/art.23609. [DOI] [PubMed] [Google Scholar]
- 21.Dinesh RK, Skaggs BJ, La Cava A, Hahn BH, Singh RP. CD8+ Tregs in lupus, autoimmunity, and beyond. Autoimmun Rev. 2010;9:560–8. doi: 10.1016/j.autrev.2010.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kim HJ, Verbinnen B, Tang X, Lu L, Cantor H. Inhibition of follicular T-helper cells by CD8(+) regulatory T cells is essential for self tolerance. Nature. 2010;467:328–32. doi: 10.1038/nature09370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Smith TR, Kumar V. Revival of CD8+ Treg-mediated suppression. Trends Immunol. 2008;29:337–42. doi: 10.1016/j.it.2008.04.002. [DOI] [PubMed] [Google Scholar]
- 24.Wang YM, Alexander SI. CD8 regulatory T cells: what’s old is now new. Immunol Cell Biol. 2009;87:192–3. doi: 10.1038/icb.2009.8. [DOI] [PubMed] [Google Scholar]
- 25.Baughman EJ, Mendoza JP, Ortega SB, Ayers CL, Greenberg BM, Frohman EM, et al. Neuroantigen-specific CD8+ regulatory T-cell function is deficient during acute exacerbation of multiple sclerosis. J Autoimmun. 2011;36:115–24. doi: 10.1016/j.jaut.2010.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kumar V, Sercarz E. An integrative model of regulation centered on recognition of TCR peptide/MHC complexes. Immunol Rev. 2001;182:113–21. doi: 10.1034/j.1600-065x.2001.1820109.x. [DOI] [PubMed] [Google Scholar]
- 27.Najafian N, Chitnis T, Salama AD, Zhu B, Benou C, Yuan X, et al. Regulatory functions of CD8+CD28− T cells in an autoimmune disease model. J Clin Invest. 2003;112:1037–48. doi: 10.1172/JCI17935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.York NR, Mendoza JP, Ortega SB, Benagh A, Tyler AF, Firan M, et al. Immune regulatory CNS-reactive CD8+T cells in experimental autoimmune encephalomyelitis. J Autoimmun. 2010;35:33–44. doi: 10.1016/j.jaut.2010.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ben-David H, Sharabi A, Dayan M, Sela M, Mozes E. The role of CD8+CD28 regulatory cells in suppressing myasthenia gravis-associated responses by a dual altered peptide ligand. Proc Natl Acad Sci U S A. 2007;104:17459–64. doi: 10.1073/pnas.0708577104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Filaci G, Bacilieri S, Fravega M, Monetti M, Contini P, Ghio M, et al. Impairment of CD8+ T suppressor cell function in patients with active systemic lupus erythematosus. J Immunol. 2001;166:6452–7. doi: 10.4049/jimmunol.166.10.6452. [DOI] [PubMed] [Google Scholar]
- 31.Zheng SG, Wang JH, Koss MN, Quismorio F, Jr, Gray JD, Horwitz DA. CD4+ and CD8+ regulatory T cells generated ex vivo with IL-2 and TGF-beta suppress a stimulatory graft-versus-host disease with a lupus-like syndrome. J Immunol. 2004;172:1531–9. doi: 10.4049/jimmunol.172.3.1531. [DOI] [PubMed] [Google Scholar]
- 32.Zhang L, Bertucci AM, Ramsey-Goldman R, Burt RK, Datta SK. Regulatory T cell (Treg) subsets return in patients with refractory lupus following stem cell transplantation, and TGF-beta-producing CD8+ Treg cells are associated with immunological remission of lupus. J Immunol. 2009;183:6346–58. doi: 10.4049/jimmunol.0901773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kim HJ, Wang X, Radfar S, Sproule TJ, Roopenian DC, Cantor H. CD8+ T regulatory cells express the Ly49 Class I MHC receptor and are defective in autoimmune prone B6-Yaa mice. Proc Natl Acad Sci U S A. 2011;108:2010–5. doi: 10.1073/pnas.1018974108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kohm AP, Carpentier PA, Anger HA, Miller SD. Cutting edge: CD4+CD25+ regulatory T cells suppress antigen-specific autoreactive immune responses and central nervous system inflammation during active experimental autoimmune encephalomyelitis. J Immunol. 2002;169:4712–6. doi: 10.4049/jimmunol.169.9.4712. [DOI] [PubMed] [Google Scholar]
- 35.Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S. Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med. 2002;196:389–99. doi: 10.1084/jem.20020399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Malek TR, Yu A, Vincek V, Scibelli P, Kong L. CD4 regulatory T cells prevent lethal autoimmunity in IL-2Rbeta-deficient mice. Implications for the nonredundant function of IL-2. Immunity. 2002;17:167–78. doi: 10.1016/s1074-7613(02)00367-9. [DOI] [PubMed] [Google Scholar]
- 37.Puliaev R, Puliaeva I, Welniak LA, Ryan AE, Haas M, Murphy WJ, et al. CTL-promoting effects of CD40 stimulation outweigh B cell-stimulatory effects resulting in B cell elimination and disease improvement in a murine model of lupus. J Immunol. 2008;181:47–61. doi: 10.4049/jimmunol.181.1.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Puliaeva I, Puliaev R, Via CS. Therapeutic potential of CD8+ cytotoxic T lymphocytes in SLE. Autoimmun Rev. 2009;8:219–23. doi: 10.1016/j.autrev.2008.07.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Via CS, Sharrow SO, Shearer GM. Role of cytotoxic T lymphocytes in the prevention of lupus-like disease occurring in a murine model of graft-vs-host disease. J Immunol. 1987;139:1840–9. [PubMed] [Google Scholar]
- 40.Fan GC, Singh RR. Vaccination with minigenes encoding V(H)-derived major histocompatibility complex class I-binding epitopes activates cytotoxic T cells that ablate autoantibody-producing B cells and inhibit lupus. J Exp Med. 2002;196:731–41. doi: 10.1084/jem.20020223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kang SM, Tang Q, Bluestone JA. CD4(+)CD25(+) Regulatory T Cells in Transplantation: Progress, Challenges and Prospects. Am J Transplant. 2007;7:1457–63. doi: 10.1111/j.1600-6143.2007.01829.x. [DOI] [PubMed] [Google Scholar]
- 42.Tang X, Maricic I, Kumar V. Anti-TCR antibody treatment activates a novel population of nonintestinal CD8 alpha alpha+ TCR alpha beta+ regulatory T cells and prevents experimental autoimmune encephalomyelitis. J Immunol. 2007;178:6043–50. doi: 10.4049/jimmunol.178.10.6043. [DOI] [PubMed] [Google Scholar]
- 43.Tang X, Maricic I, Purohit N, Bakamjian B, Reed-Loisel LM, Beeston T, et al. Regulation of immunity by a novel population of Qa-1-restricted CD8alphaalpha+TCRalphabeta+ T cells. J Immunol. 2006;177:7645–55. doi: 10.4049/jimmunol.177.11.7645. [DOI] [PubMed] [Google Scholar]
- 44.Eilat E, Dayan M, Zinger H, Mozes E. The mechanism by which a peptide based on complementarity-determining region-1 of a pathogenic anti-DNA auto-Ab ameliorates experimental systemic lupus erythematosus. Proc Natl Acad Sci U S A. 2001;98:1148–53. doi: 10.1073/pnas.98.3.1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Sharabi A, Mozes E. The suppression of murine lupus by a tolerogenic peptide involves foxp3-expressing CD8 cells that are required for the optimal induction and function of foxp3-expressing CD4 cells. J Immunol. 2008;181:3243–51. doi: 10.4049/jimmunol.181.5.3243. [DOI] [PubMed] [Google Scholar]
- 46.Lu L, Cantor H. Generation and regulation of CD8(+) regulatory T cells. Cell Mol Immunol. 2008;5:401–6. doi: 10.1038/cmi.2008.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Singh RP, Dinesh R, Elashoff D, de Vos S, Rooney RJ, Patel D, et al. Distinct gene signature revealed in white blood cells, CD4(+) and CD8(+) T cells in (NZBx NZW) F1 lupus mice after tolerization with anti-DNA Ig peptide. Genes Immun. 2010 doi: 10.1038/gene.2010.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Rozzo SJ, Allard JD, Choubey D, Vyse TJ, Izui S, Peltz G, et al. Evidence for an interferon-inducible gene, Ifi202, in the susceptibility to systemic lupus. Immunity. 2001;15:435–43. doi: 10.1016/s1074-7613(01)00196-0. [DOI] [PubMed] [Google Scholar]
- 49.Ye S, Pang H, Gu YY, Hua J, Chen XG, Bao CD, et al. Protein interaction for an interferon-inducible systemic lupus associated gene, IFIT1. Rheumatology (Oxford) 2003;42:1155–63. doi: 10.1093/rheumatology/keg315. [DOI] [PubMed] [Google Scholar]
- 50.Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, et al. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A. 2003;100:2610–5. doi: 10.1073/pnas.0337679100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hron JD, Peng SL. Type I IFN protects against murine lupus. J Immunol. 2004;173:2134–42. doi: 10.4049/jimmunol.173.3.2134. [DOI] [PubMed] [Google Scholar]
- 52.Gubbels Bupp MR, Li M, Pashine A, Aud D, Peng SL. The candidate lupus susceptibility gene Ifi202a is largely dispensable for B-cell function. Rheumatology (Oxford) 2008;47:103–4. doi: 10.1093/rheumatology/kem259. [DOI] [PubMed] [Google Scholar]
- 53.Panchanathan R, Shen H, Bupp MG, Gould KA, Choubey D. Female and male sex hormones differentially regulate expression of Ifi202, an interferon-inducible lupus susceptibility gene within the Nba2 interval. J Immunol. 2009;183:7031–8. doi: 10.4049/jimmunol.0802665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Xin H, D’Souza S, Jorgensen TN, Vaughan AT, Lengyel P, Kotzin BL, et al. Increased expression of Ifi202, an IFN-activatable gene, in B6.Nba2 lupus susceptible mice inhibits p53-mediated apoptosis. J Immunol. 2006;176:5863–70. doi: 10.4049/jimmunol.176.10.5863. [DOI] [PubMed] [Google Scholar]
- 55.Cleary ML, Smith SD, Sklar J. Cloning and structural analysis of cDNAs for bcl-2 and a hybrid bcl-2/immunoglobulin transcript resulting from the t(14;18) translocation. Cell. 1986;47:19–28. doi: 10.1016/0092-8674(86)90362-4. [DOI] [PubMed] [Google Scholar]
- 56.Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature. 1988;335:440–2. doi: 10.1038/335440a0. [DOI] [PubMed] [Google Scholar]
- 57.Wu H, Shen N, Gu Y, Qian J, Feng X, Wang Y, et al. Association between Bcl-2 gene polymorphism with systemic lupus erythematosus. Zhonghua Yi Xue Za Zhi. 2002;82:515–8. [PubMed] [Google Scholar]
- 58.Singh RP, Dinesh R, Elashoff D, de Vos S, Rooney RJ, Patel D, et al. Distinct gene signature revealed in white blood cells, CD4(+) and CD8(+) T cells in (NZBx NZW) F1 lupus mice after tolerization with anti-DNA Ig peptide. Genes Immun. 2010;11:294–309. doi: 10.1038/gene.2010.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Sharabi A, Dayan M, Zinger H, Mozes E. A new model of induced experimental systemic lupus erythematosus (SLE) in pigs and its amelioration by treatment with a tolerogenic peptide. J Clin Immunol. 2010;30:34–44. doi: 10.1007/s10875-009-9326-4. [DOI] [PubMed] [Google Scholar]
- 60.Sharabi A, Luger D, Ben-David H, Dayan M, Zinger H, Mozes E. The role of apoptosis in the ameliorating effects of a CDR1-based peptide on lupus manifestations in a mouse model. J Immunol. 2007;179:4979–87. doi: 10.4049/jimmunol.179.8.4979. [DOI] [PubMed] [Google Scholar]
- 61.Wiegers GJ, Kaufmann M, Tischner D, Villunger A. Shaping the T-cell repertoire: a matter of life and death. Immunol Cell Biol. 2011;89:33–9. doi: 10.1038/icb.2010.127. [DOI] [PubMed] [Google Scholar]
- 62.Harris CC. p53: at the crossroads of molecular carcinogenesis and risk assessment. Science. 1993;262:1980–1. doi: 10.1126/science.8266092. [DOI] [PubMed] [Google Scholar]
- 63.Kim KH, Park GT, Lim YB, Rue SW, Jung JC, Sonn JK, et al. Expression of connective tissue growth factor, a biomarker in senescence of human diploid fibroblasts, is up-regulated by a transforming growth factor-beta-mediated signaling pathway. Biochem Biophys Res Commun. 2004;318:819–25. doi: 10.1016/j.bbrc.2004.04.108. [DOI] [PubMed] [Google Scholar]
- 64.Itoh T, Cado D, Kamide R, Linn S. DDB2 gene disruption leads to skin tumors and resistance to apoptosis after exposure to ultraviolet light but not a chemical carcinogen. Proc Natl Acad Sci U S A. 2004;101:2052–7. doi: 10.1073/pnas.0306551101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Levine AJ, Hu W, Feng Z. The P53 pathway: what questions remain to be explored? Cell Death Differ. 2006;13:1027–36. doi: 10.1038/sj.cdd.4401910. [DOI] [PubMed] [Google Scholar]
- 66.Nath SK, Kelly JA, Namjou B, Lam T, Bruner GR, Scofield RH, et al. Evidence for a susceptibility gene, SLEV1, on chromosome 17p13 in families with vitiligo-related systemic lupus erythematosus. Am J Hum Genet. 2001;69:1401–6. doi: 10.1086/324470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Miret C, Molina R, Filella X, Garcia-Carrasco M, Claver G, Ingelmo M, et al. Relationship of p53 with other oncogenes, cytokines and systemic lupus erythematosus activity. Tumour Biol. 2003;24:185–8. doi: 10.1159/000074428. [DOI] [PubMed] [Google Scholar]
- 68.Lee YH, Rho YH, Choi SJ, Ji JD, Song GG. The functional p53 codon 72 polymorphism is associated with systemic lupus erythematosus. Lupus. 2005;14:842–5. doi: 10.1191/0961203305lu2224oa. [DOI] [PubMed] [Google Scholar]
- 69.Lespagnol A, Duflaut D, Beekman C, Blanc L, Fiucci G, Marine JC, et al. Exosome secretion, including the DNA damage-induced p53-dependent secretory pathway, is severely compromised in TSAP6/Steap3-null mice. Cell Death Differ. 2008;15:1723–33. doi: 10.1038/cdd.2008.104. [DOI] [PubMed] [Google Scholar]
- 70.Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–1. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
- 71.Hori S, Takahashi T, Sakaguchi S. Control of autoimmunity by naturally arising regulatory CD4+ T cells. Adv Immunol. 2003;81:331–71. doi: 10.1016/s0065-2776(03)81008-8. [DOI] [PubMed] [Google Scholar]
- 72.Peng SL. Forkhead transcription factors in chronic inflammation. Int J Biochem Cell Biol. 42:482–5. doi: 10.1016/j.biocel.2009.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zheng Y, Josefowicz SZ, Kas A, Chu TT, Gavin MA, Rudensky AY. Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature. 2007;445:936–40. doi: 10.1038/nature05563. [DOI] [PubMed] [Google Scholar]
- 74.Xystrakis E, Dejean AS, Bernard I, Druet P, Liblau R, Gonzalez-Dunia D, et al. Identification of a novel natural regulatory CD8 T-cell subset and analysis of its mechanism of regulation. Blood. 2004 doi: 10.1182/blood-2004-03-1214. [DOI] [PubMed] [Google Scholar]
- 75.Schramm C, Huber S, Protschka M, Czochra P, Burg J, Schmitt E, et al. TGFbeta regulates the CD4+CD25+ T-cell pool and the expression of Foxp3 in vivo. Int Immunol. 2004;16:1241–9. doi: 10.1093/intimm/dxh126. [DOI] [PubMed] [Google Scholar]
- 76.Arazi A, Sharabi A, Zinger H, Mozes E, Neumann AU. In vivo dynamical interactions between CD4 Tregs, CD8 Tregs and CD4+ CD25− cells in mice. PLoS One. 2009;4:e8447. doi: 10.1371/journal.pone.0008447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Mozes E, Sela U, Sharabi A. A novel synthetic peptide for the specific treatment of lupus: clinical effects and mechanism of action. Isr Med Assoc J. 2008;10:40–2. [PubMed] [Google Scholar]
- 78.Badillo-Almaraz I, Daza L, Avalos-Diaz E, Herrera-Esparza R. Glomerular expression of Fas ligand and Bax mRNA in lupus nephritis. Autoimmunity. 2001;34:283–9. doi: 10.3109/08916930109014697. [DOI] [PubMed] [Google Scholar]
- 79.Liu L, Tran E, Zhao Y, Huang Y, Flavell R, Lu B. Gadd45 beta and Gadd45 gamma are critical for regulating autoimmunity. J Exp Med. 2005;202:1341–7. doi: 10.1084/jem.20051359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Druey KM, Blumer KJ, Kang VH, Kehrl JH. Inhibition of G-protein-mediated MAP kinase activation by a new mammalian gene family. Nature. 1996;379:742–6. doi: 10.1038/379742a0. [DOI] [PubMed] [Google Scholar]
- 81.Heximer SP, Blumer KJ. RGS proteins: Swiss army knives in seven-transmembrane domain receptor signaling networks. Sci STKE. 2007;2007:pe2. doi: 10.1126/stke.3702007pe2. [DOI] [PubMed] [Google Scholar]
- 82.Kehrl JH. G-protein-coupled receptor signaling, RGS proteins, and lymphocyte function. Crit Rev Immunol. 2004;24:409–23. doi: 10.1615/critrevimmunol.v24.i6.20. [DOI] [PubMed] [Google Scholar]
- 83.Schwable J, Choudhary C, Thiede C, Tickenbrock L, Sargin B, Steur C, et al. RGS2 is an important target gene of Flt3-ITD mutations in AML and functions in myeloid differentiation and leukemic transformation. Blood. 2005;105:2107–14. doi: 10.1182/blood-2004-03-0940. [DOI] [PubMed] [Google Scholar]
- 84.Semplicini A, Lenzini L, Sartori M, Papparella I, Calo LA, Pagnin E, et al. Reduced expression of regulator of G-protein signaling 2 (RGS2) in hypertensive patients increases calcium mobilization and ERK1/2 phosphorylation induced by angiotensin II. J Hypertens. 2006;24:1115–24. doi: 10.1097/01.hjh.0000226202.80689.8f. [DOI] [PubMed] [Google Scholar]
- 85.Yang RZ, Blaileanu G, Hansen BC, Shuldiner AR, Gong DW. cDNA cloning, genomic structure, chromosomal mapping, and functional expression of a novel human alanine aminotransferase. Genomics. 2002;79:445–50. doi: 10.1006/geno.2002.6722. [DOI] [PubMed] [Google Scholar]
- 86.Sundrud MS, Koralov SB, Feuerer M, Calado DP, Kozhaya AE, Rhule-Smith A, et al. Halofuginone inhibits TH17 cell differentiation by activating the amino acid starvation response. Science. 2009;324:1334–8. doi: 10.1126/science.1172638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hsu TC, Huang CY, Chiang SY, Lai WX, Tsai CH, Tzang BS. Transglutaminase inhibitor cystamine alleviates the abnormality in liver from NZB/W F1 mice. Eur J Pharmacol. 2008;579:382–9. doi: 10.1016/j.ejphar.2007.10.059. [DOI] [PubMed] [Google Scholar]
- 88.Li CH, Xu PS, Wang CY, Zhang Y, Zou GL. The presence of anti-mitochondrial antibodies in Chinese patients with liver involvement in systemic lupus erythematosus. Rheumatol Int. 2006;26:697–703. doi: 10.1007/s00296-005-0034-y. [DOI] [PubMed] [Google Scholar]
- 89.Gavin MA, Rasmussen JP, Fontenot JD, Vasta V, Manganiello VC, Beavo JA, et al. Foxp3-dependent programme of regulatory T-cell differentiation. Nature. 2007;445:771–5. doi: 10.1038/nature05543. [DOI] [PubMed] [Google Scholar]
- 90.Ohsako S, Hara M, Harigai M, Fukasawa C, Krajewski S, Reed JC, Kashiwazaki S. The Bcl-2/Bax ratio of lymphocytes from human systemic lupus erythematosus patients. Modern Rheumatology. 1997;7:305–13. [Google Scholar]
- 91.Cui JH, Qiao Q, Guo Y, Zhang YQ, Cheng H, He FR, et al. Increased apoptosis and expression of FasL, Bax and caspase-3 in human lupus nephritis class II and IV. J Nephrol. 2011:0. doi: 10.5301/JN.2011.8451. [DOI] [PubMed] [Google Scholar]
- 92.Pistilli EE, Siu PM, Alway SE. Molecular regulation of apoptosis in fast plantaris muscles of aged rats. J Gerontol A Biol Sci Med Sci. 2006;61:245–55. doi: 10.1093/gerona/61.3.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Vekrellis K, McCarthy MJ, Watson A, Whitfield J, Rubin LL, Ham J. Bax promotes neuronal cell death and is downregulated during the development of the nervous system. Development. 1997;124:1239–49. doi: 10.1242/dev.124.6.1239. [DOI] [PubMed] [Google Scholar]
- 94.Nazareth M, Fanti P, Schwach C, Poppenberg K, Janis K, Aronica SM. Altered Bax expression and decreased apoptosis in bone marrow cells of lupus-susceptible NZB/W mice. Lupus. 2001;10:785–93. doi: 10.1177/096120330101001105. [DOI] [PubMed] [Google Scholar]
- 95.Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704. doi: 10.1146/annurev.immunol.26.021607.090331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Nishimura H, Minato N, Nakano T, Honjo T. Immunological studies on PD-1 deficient mice: implication of PD-1 as a negative regulator for B cell responses. Int Immunol. 1998;10:1563–72. doi: 10.1093/intimm/10.10.1563. [DOI] [PubMed] [Google Scholar]
- 97.Keir ME, Francisco LM, Sharpe AH. PD-1 and its ligands in T-cell immunity. Curr Opin Immunol. 2007;19:309–14. doi: 10.1016/j.coi.2007.04.012. [DOI] [PubMed] [Google Scholar]
- 98.Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:3887–95. doi: 10.1002/j.1460-2075.1992.tb05481.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Nishimura H, Agata Y, Kawasaki A, Sato M, Imamura S, Minato N, et al. Developmentally regulated expression of the PD-1 protein on the surface of double-negative (CD4−CD8−) thymocytes. Int Immunol. 1996;8:773–80. doi: 10.1093/intimm/8.5.773. [DOI] [PubMed] [Google Scholar]
- 100.Okazaki T, Honjo T. The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 2006;27:195–201. doi: 10.1016/j.it.2006.02.001. [DOI] [PubMed] [Google Scholar]
- 101.Yamazaki T, Akiba H, Iwai H, Matsuda H, Aoki M, Tanno Y, et al. Expression of programmed death 1 ligands by murine T cells and APC. J Immunol. 2002;169:5538–45. doi: 10.4049/jimmunol.169.10.5538. [DOI] [PubMed] [Google Scholar]
- 102.Ishida M, Iwai Y, Tanaka Y, Okazaki T, Freeman GJ, Minato N, et al. Differential expression of PD-L1 and PD-L2, ligands for an inhibitory receptor PD-1, in the cells of lymphohematopoietic tissues. Immunol Lett. 2002;84:57–62. doi: 10.1016/s0165-2478(02)00142-6. [DOI] [PubMed] [Google Scholar]
- 103.Zhong X, Tumang JR, Gao W, Bai C, Rothstein TL. PD-L2 expression extends beyond dendritic cells/macrophages to B1 cells enriched for V(H)11/V(H)12 and phosphatidylcholine binding. Eur J Immunol. 2007;37:2405–10. doi: 10.1002/eji.200737461. [DOI] [PubMed] [Google Scholar]
- 104.Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11:141–51. doi: 10.1016/s1074-7613(00)80089-8. [DOI] [PubMed] [Google Scholar]
- 105.Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 2001;291:319–22. doi: 10.1126/science.291.5502.319. [DOI] [PubMed] [Google Scholar]
- 106.Mazanet MM, Hughes CC. B7-H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. J Immunol. 2002;169:3581–8. doi: 10.4049/jimmunol.169.7.3581. [DOI] [PubMed] [Google Scholar]
- 107.Blazar BR, Carreno BM, Panoskaltsis-Mortari A, Carter L, Iwai Y, Yagita H, et al. Blockade of programmed death-1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma-dependent mechanism. J Immunol. 2003;171:1272–7. doi: 10.4049/jimmunol.171.3.1272. [DOI] [PubMed] [Google Scholar]
- 108.Ortler S, Leder C, Mittelbronn M, Zozulya AL, Knolle PA, Chen L, et al. B7-H1 restricts neuroantigen-specific T cell responses and confines inflammatory CNS damage: implications for the lesion pathogenesis of multiple sclerosis. Eur J Immunol. 2008;38:1734–44. doi: 10.1002/eji.200738071. [DOI] [PubMed] [Google Scholar]
- 109.Wong M, La Cava A, Singh RP, Hahn BH. Blockade of programmed death-1 in young (New Zealand black × New Zealand white)F1 mice promotes the activity of suppressive CD8+ T cells that protect from lupus-like disease. J Immunol. 2010;185:6563–71. doi: 10.4049/jimmunol.0903401. [DOI] [PubMed] [Google Scholar]
- 110.Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA. CD4+CD25+ regulatory cells from human peripheral blood express very high levels of CD25 ex vivo. Novartis Found Symp. 2003;252:67–88. doi: 10.1002/0470871628.ch6. discussion -91, 106–14. [DOI] [PubMed] [Google Scholar]
- 111.Krupnick AS, Gelman AE, Barchet W, Richardson S, Kreisel FH, Turka LA, et al. Murine vascular endothelium activates and induces the generation of allogeneic CD4+25+Foxp3+ regulatory T cells. J Immunol. 2005;175:6265–70. doi: 10.4049/jimmunol.175.10.6265. [DOI] [PubMed] [Google Scholar]
- 112.Totsuka T, Kanai T, Makita S, Fujii R, Nemoto Y, Oshima S, et al. Regulation of murine chronic colitis by CD4+CD25− programmed death-1+ T cells. Eur J Immunol. 2005;35:1773–85. doi: 10.1002/eji.200425109. [DOI] [PubMed] [Google Scholar]
- 113.Hornung M, Farkas SA, Sattler C, Schlitt HJ, Geissler EK. DX5+ NKT cells induce the death of colitis-associated cells: involvement of programmed death ligand-1. Eur J Immunol. 2006;36:1210–21. doi: 10.1002/eji.200535332. [DOI] [PubMed] [Google Scholar]