Regulation of adaptive immune responses by innate cells expressing ...

1 downloads 55 Views 141KB Size Report
Streilein, Steven P. Balk, and Mark Exley for their helpful discussions on ... Medzhitov, R., Janeway, C. A., Jr. (1997) Innate immunity: The virtues of a nonclonal ..... Stein-Streilein, J., Lipscomb, M. F., Fisch, H., Whitney, P. L. (1987). Pulmonary ...
Regulation of adaptive immune responses by innate cells expressing NK markers and antigen-transporting macrophages J. Stein-Streilein,* K-H. Sonoda,* D. Faunce,* and J. Zhang-Hoover* *Schepens Eye Research Institute, Harvard Medical School, Boston; and †Pulmonary and Critical Care Division, Department of Medicine at Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts

Abstract: A continuing theme of work done in our laboratory involves regulation of adaptive immune response by innate cells, in general, and immuneregulation by natural killer (NK) and NKT cells, in particular. Studies include work with the lung and the eye. In addition to immune surveillance of tumor cells, the NK cell is often associated with secreting cytokines that contribute to the creation of microenvironments conducive to Th1 responses and with defense mechanisms that lessen the initial infecting viral load. Reported studies show that the NKT cells support both T helper cell responses (type 1 and 2), as well as their being absolutely central to the development of antigen-specific Tregulatory cells involved in peripheral tolerance. Because of the multifunctional capabilities of the NKT cell, we propose that yet another cell, such as the antigen-presenting cell (APC), may influence the effector pathway of the NKT cell. We postulate that the APC that transports the antigen from the entry environment provides both trafficking and activation signals for innate cells in the secondary lymphoid organs. Evidence is presented that macrophage-derived signals selectively recruit NKT cells and bias their cytokine synthesis. Data imply that, just as occurs in immune inflammation, a collection of innate and adaptive immune cells interact within the secondary lymphoid tissue to generate antigenspecific tolerance in the periphery. J. Leukoc. Biol. 67: 488–494; 2000. Key Words: inflammation · peripheral tolerance · pulmonary interstitial fibrosis · anterior chamber-associated immune deviation · influenza virus

INTRODUCTION Innate immune responses involve non-clonally derived defense cells that can respond immediately to danger [1, 2]. The defense mechanisms used by innate cells include regulatory mechanisms that contribute to the biasing of the clonally derived adaptive immune response. The relationship of the natural killer (NK) cell to the NKT cell is rarely studied, but the simple concept that the NK and NKT cell are in some sort of balancing 488

Journal of Leukocyte Biology Volume 67, April 2000

(yin-yang) relationship is sometimes implied [3]. It is well accepted that interferon-␥ (IFN-␥) promotes Th1 cellular immune responses [4–6], and the NK cell is an early and convenient source of this polarizing cytokine. Th2 cells appear to need interleukin-4 (IL-4) or IL-13 for their development, although the cellular source is unknown. Early studies suggested the NKT cell to be responsible for Th2 polarization, however, only a few experimental Th2 models depend on the NKT cell for IL-4 [7–9], because it was quickly revealed that several other models of Th2 responses still occurred in the absence of NKT cells [10–12]. At this time, the biological role for NKT cells is not fully understood. The role of the antigen-presenting cell (APC) is established in activation of immune inflammatory responses and adaptive immunity. However, the role of the APC in regulating other innate cell responses is less well understood. Evidence is presented that the antigen-transporting macrophages from local environments influence both innate and adaptive immune cells in the secondary lymphoid organs and influence whether inflammation or tolerance is the outcome. It has been known since the 1960s that regional spheres of immunological influence exist. At that time, the newly described IgA class of immunogloblin was shown to be particularly enriched in external bodily secretions such as tear, saliva, and intestinal contents. And, only 4 years later, it was shown that thoracic duct drainage contained recirculating lymphocytes passing through the lymph that were not homogeneous [13]. In fact, the IgA-bearing B lymphocytes were shown to preferentially localize in the gastrointestinal tract. Since that time, it was shown that antigen processing, and the subsequent immune responses in the gut and other mucosal tissues, was under the influence of the regional regulation and specialization [14, 15]. Studies in our laboratory support the notion of regional specialization in the lung [16]. Although the large airways are lined with mucosa and show some aspects of the mucosal immune system, the alveoli and air spaces are not part of the mucosal system and instead exhibit their own unique regulation to prevent immune responses and protect the lung’s physiological function of gas exchange.

Correspondence: Joan Stein-Streilein, Ph.D., Schepens Eye Research Institute, 20 Staniford St., Boston, MA 02114. E-mail: [email protected] Received November 4, 1999; revised January 19, 2000; accepted January 20, 2000.

http://www.jleukbio.org

Role of NK cells in viral infections The NK cell was first noticed for its ability to kill tumor cells without prior sensitization, ergo its name [17]. We and others have shown that NK cells are critical for protection against certain viral infections [18–23]. It is important to note that NK cells limit the expansion of the viral load while T effector cells differentiate in response to viral antigens. Then, effector T lymphocytes complete the job by eliminating virus-infected cells [24]. In particular, we reported that NK cells were necessary for controlling the influenzal viral load in mice and hamsters because, in the absence of NK cells (eliminated in vivo with specific antibodies to antigens on the NK cell), infected mice expressed more virus and succumbed faster to the virus [20, 23, 25]. Furthermore, we and others carefully showed that, when given before virus infection, R␣AsGM1 treatment removed NK cells but did not remove effector T cells or macrophages [23, 26, 27]. Moreover, because the NK cell is present locally in a variety of tissues and organs, including the lung, methods were available to selectively eliminate the NK cells in the lung, leaving blood-borne NK cells intact [20, 25, 28, 29]. Thus, these studies showed that the NK cells that were protective were within the local environment. The idea that NK cell defenses were regulated locally contributed to the notion that regional specialization is an important concept in immune defense in the organism as a whole [15]. The concept that the NK cell in the lung, and more recently, the NKT cell (see below), are part of regional specialization mechanisms is well supported by recent work on NK and NKT cells in the liver [30, 31].

NKT cells NKT cells make up a subpopulation of nontraditional T cells that co-express NK markers and were first studied for their ability to initiate polarization of the T helper cell responses [32, 33]. Details of this cell are published elsewhere [31, 34–36]. The novel lymphoid lineage that gives rise to NKT cells produces lymphocytes that are distinct from other lymphoid cells, including T, B, and NK cells. The majority of NKT cells in the mouse co-express NK markers and a single invariant T cell receptor (TCR) encoded by the V␣14 and J␣281 gene segments [37–39] in association with a highly skewed set of V␤s [40–47]. Moreover, the generation of V␣14 J␣281 TCRNKT cells is exclusively dependent on the expression of the invariant V␣14 J␣281 TCR as evidenced by the fact that these cells do not develop in the invariant J␣281-deficient mice [48], and that the forced expression of the invariant V␣14 TCR led to expression of exclusively V␣14 NKT cells conventional T cell development [49, 50]. The invariant T cell receptor on a subpopulation of NKT cells binds CD1 with or without lipids [51]. A ligand for the invariant V␣14V␤ 8.2 TCR exclusively expressed on V␣14 NKT cells has been identified recently to be ␣ galactosylceramide (␣ GalCer) [52]. In addition, ␣ GalCer is presented by a monomorphic major histocompatibility gene complex (non-MHC) class I molecule, CD1.d, expressed on dendritic cells and other bone marrow-derived cells. The ligand/CD1 complex selectively stimulates V␣14 NKT cells, but not other lymphocytes, to proliferate [53, 54]. Thus the invariant V␣14V J␣281 TCRexpressing NKT cell subpopulation is CD1.d restricted. V␣14

J␣281 TCR is found on up to 85% of NKT cells [51, 55]. Unlike the human, which expresses CD1.a, b, c, and d, the mouse only expresses CD1.d. Moreover, human NKT cells that express the invariant V␣ 24J␣Q invariant TCR also bind to CD1.d [56]. Therefore, the CD1-restricted mouse models exploiting CD1/ NKT cell interactions are directly relevant to the function of human NKT cells [55].

Cytokine production in NK and NKT cells during regional immune responses Recent work in our laboratory analyzed cytokines produced by NK and NKT cells in several experimental animal models involving lung and eye immunology [57, 58]. In the lung we observed that NK cells within the alveolar spaces produced IFN-␥ during toxicity (bleomycin)-initiated inflammatory processes and immune-mediated inflammatory responses [hapten immune pulmonary interstitial fibrosis (HIPIF)] [59]. In contrast, NK cells were essentially absent in the bronchoalveolar lavage samples during the Th2-mediated inflammatory process found to be dominant in the mouse ovalbumin (OVA)-asthma lung model [57]. On the other hand, NKT cells were prominent in all three models of immune inflammation in the lung; albeit they displayed different intracellular cytokine profiles [57]. NKT cells containing IL-4 were increased in the OVA-asthma model, whereas NKT cells synthesizing IFN-␥ [60] were prominent in the inflammatory or Th1-initiated fibrotic animal models. Thus, as suggested by others, the NKT cell is quite pleotropic in its responses [48, 61–63]. Because the NKT cell responds to primary stimulation in a variety of ways, we reasoned that yet another cell might orchestrate the response seen in the NKT cell. We postulated that the antigen-transporting macrophage from the local environment of antigen entry may influence the subsequent innate and adaptive immune response. Evidence is published that supports the notion that antigen-transporting macrophages derived from the eye are important to the development of the immune deviation or peripheral tolerance that occurs during the development of anterior chamber-associated immune deviation (ACAID) [64, 65].

ACAID model of peripheral tolerance The model for ACAID is an established protocol for the study of mechanisms that induce peripheral tolerance in response to antigen injected into an immune-privileged site, such as the eye [66–68]. Published data show that eye-derived APCs identified by F4/80⫹ expression leave the eye by 3 days post-inoculation, travel through the blood as an ACAIDinducing signal, and settle in the spleen thereafter [69]. The F4/80⫹ cell, being influenced by the suppressive microenvironment of the anterior chamber, is specialized and produces transforming growth factor ␤ (TGF-␤), which induces autocrine production of TGF-␤ in the cells it binds [64, 65, 70]. Thus the antigen-transporting macrophage from the environment of antigen entry influences both the innate and adaptive immune response that occurs in the secondary lymphoid organ. During the ACAID induction process in the spleen, antigen is thought to be presented to CD8⫹ T cells that then differentiate into antigen-specific negative regulatory cells [70]. The signals that

Stein-Streilein et al. Innate cells promote immune responses and tolerance

489

influence the differentiation of CD8⫹ T cell into regulatory cells are not clearly defined. In part, the role of NKT cells in development of peripheral tolerance contributes to a better understanding of T lymphocyte regulatory cell development [58]. Initially, we observed that an accumulation of NKT cells in the spleens of anterior chamber-inoculated mice was associated with the development of CD8⫹ T regulatory cells during ACAID induction [58]. The detection of NKT cell accumulation in the spleen was reproducible and was observed in C57BL/6 (B6), BALB/c, and (B6 ⫻ 129) F1 strains of mice during ACAID induction (Table 1).Thus it is unlikely that NKT cell accumulation in the spleen during ACAID is dependent on a particular genetic background.

Requirement for CD1/NKT cell interaction in ACAID-induced peripheral tolerance To test the role of NKT cells in the development of ACAID, we used CD1 knockout (KO) mice as hosts for the model. The CD1 KO mice used (developed by S. Balk and colleagues at Beth Israel-Deaconess Medical Center, Boston, MA) lack both genes for CD1 and, it is important to note, lack NKT cells because the CD1.d that is expressed by cortical thymocytes is needed for positive selection of the major subpopulation of NKT cells [71, 72]. Indeed, CD1 KO mice were unable to develop T regulatory cells specific for the antigen ovalbumin after anterior chamber inoculation unless the mice were first reconstituted with wild-type NKT cells [58]. The model of ACAID can be recreated in part or in full in vitro [73–75]. Partial in vitro ACAID requires that APCs [in this case, thioglycolate-induced peritoneal exudate cells (PECs)] be pulsed with antigen in the presence of TGF-␤ before being inoculated intravenously into a naive mouse to induce peripheral tolerance. Previously it was noted that TGF-␤-treated PECs down-regulate CD40 and IL-12 expression [76]. We observed that OVA-pulsed PECs that were treated with TGF-␤ responded, in part, with increased expression of their CD1 molecules (Fig. 1). Therefore, we predict that eye-derived APCs in vivo also express more CD1 molecules per cell because of their exposure to TGF-␤ in the aqueous humor. We presume that when NKT cells bind to CD1 on the APC, the NKT cells are stimulated to produce immune-suppressive cytokines (TGF-␤ or IL-10) that in turn contribute to the differentiation of the CD8⫹ precursor T cells. Because there is no intracytoplasmic portion of the CD1 molecule, the CD1expressing cell is thought to not receive signals when the CD1 molecule is engaged [77]. Published results show that when TABLE 1.

Number of NKT Cells in the Spleen After Anterior Chamber Inoculation of Ovalbumina,b Mouse strain

Treatment

AC-HBSS AC-OVA

C57BL/6

BALB/c

17,589.8 ⫾ 1652.6 34,528.8 ⫾ 3542.1c

14,480.8 ⫾ 313.1 23,150 ⫾ 2340.7c

a Data are represented as mean number of NKT cells per 1 ⫻ 106 spleen cells ⫾ SEM. b NKT cell counts were performed 7 days after anterior chamber inoculation. c Significant from anterior chamber-HBSS-treated group when P ⬍ 0.05 as determined by ANOVA.

490

Journal of Leukocyte Biology Volume 67, April 2000

Fig. 1. Flow cytomertry analysis of CD1.d expression in TGF-␤-treated PECs. PECs were obtained from peritoneal washes of mice 3 days after they received an intraperitoneal inoculation of 2.5 mL of 3% aged-thioglycolate solution (Sigma Chemical, St. Louis, MO). After counting, PECs (2 ⫻ 106) were cultured with OVA (5 mg/mL), with and without porcine TGF-␤2 (5 ng/mL, R & D Systems, Minneapolis, MN) in a 24-well culture plate in serum-free medium [RPMI 1640 medium, 10 mM HEPES, 0.1 mM nonessential amino acids, 1 mM sodium pyruvate, 100 U/mL penicillin, 100 µg/mL streptomycin (Biowhitaker, Walkersville, MD), and supplemented with 0.1% bovine serum albumin (Sigma), ITS⫹ culture supplement (1 µg/mL iron-free transferrin, 10 ng/mL linoleic acid, 0.3 ng/mL Na2Se, and 0.2 µg/mL Fe (NO3)3, Collaborative Biomedical Products, Bedford, MA)]. Nonadherent cells were removed form the cultures after 24 h by washing and the remaining adherent cells were collected by vigorous pipetting with cold phosphate-buffered saline (4°C) before washing with HBSS to remove free OVA and TGF-␤2. TGF-␤2-treated or nontreated PECs were double stained with phycoerythrin-conjugated Mac-1 (M1/70.15, Caltag, South San Francisco, CA), biotin-conjugated anti-CD1.d (1B1, PharMingen, San Diego, CA) counterstained by SA-fluorescein isothiocyanate (PharMingen). Stained cells were analyzed on an EPICS XL flow cytometer (Beckman Coulter, Miami, FL). Total Mac-1-positive cells (8 ⫻ 103) were gated, and surface CD1.d expression was evaluated.

CD1 interaction with NKT cells is blocked in vivo with CD1-specific antibodies, CD8⫹ T regulatory cells do not develop. Of course, we do not know whether the CD1 on the eye-derived APC is the only CD1 molecule to which the TCR on the NKT cell binds. NKT cells could bind CD1 on CD8⫹ T cells, B cells, as well as stromal cells. The novel idea of a cluster of cells being required for the induction of peripheral tolerance is being explored.

Chemokines, NKT cells, and tolerance Chemokines make up a large family of small proteins that are known to play a critical role in immune and inflammatory reactions and in viral infections and are reviewed elsewhere [78–82]. Chemokines are made predominantly by myeloid-derived cells. Usually the chemokines share receptors, and many have overlapping functions. In addition to recruitment, several recent reports now attribute a variety of nontrafficking biological functions to these small cytokine molecules. Both the role of chemokines in the development or maintenance of tolerance and the effects of chemokines on NKT cells are unexplored areas. Recently, we turned our attention to the question of how NKT cells accumulate in the spleen during ACAID. They could http://www.jleukbio.org

Fig. 2. Kinetics of MIP-2 production and NKT cell accumulation in blood and spleen after anterior chamber OVA. Accumulation of NKT cells in the spleen after anterior chamber inoculation of OVA correlates with the kinetics of F4/80 and MIP-2 mRNA expression in the spleen (solid areas). Total cellular RNA was isolated from blood monocytes and splenic adherent cells (macrophages) at various times after anterior chamber inoculation of OVA. MIP-2 mRNA expression was examined with a multiprobe RNase protection assay and quantified by phosphorimaging and densitometry. The frequencies of splenic F4/80⫹ and NK1.1/TCR␤⫹ (NKT) cells were determined by flow cytometry. Note that shortly after anterior chamber OVA, MIP-2 mRNA can be detected among blood monocytes, but disappears from the blood and then appears in the splenic macrophage population at 1 week after anterior chamber OVA, suggesting that MIP-2-producing cells traffic via the blood to the spleen. The trafficking of MIP-2-producing cells from the blood to the spleen resembles that of the F4/80⫹ cell (hatched bar) and NKT cell (checked bar) accumulation. The up-regulation of MIP-2 and appearance of F4/80⫹ cells and NKT cells is unique to the induction of peripheral tolerance in the spleen after antigen inoculation in the eye.

proliferate or be recruited. We postulated that NKT cells were recruited by a select group of chemokines. Recent evidence showed that macrophage inflammatory protein-2 (MIP-2; mouse IL-8 analog) and IP-10 were selectively up-regulated in both in vitro-treated ACAID-conferring-macrophages and the mono-

cytes/macrophages in the peripheral blood of mice 3 days post anterior chamber inoculation [83]. It was interesting to us that the kinetics of the MIP-2 expression from the blood monocytes to splenic adherent cells mimicked the path of the F4/80⫹ cell, noted previously by Wilbanks and Streilein [64, 65] to be the ACAID-inducing signal (Fig. 2). Boyden chamber studies showed that MIP-2 was a chemoattractant for NKT cells and not for other lymphocytes tested [83]. As expected, NKT cells expressed the CXCR2 receptor (data not shown). Moreover, MIP-2-neutralizing mAb, given systemically, effectively blocked the recruitment of the NKT cells to the spleen and the subsequent differentiation of the CD8⫹ T regulatory cells and therefore prevented the induction of peripheral tolerance [83]. We now postulate that the involvement of MIP-2 in tolerance induction is not a response unique to antigens injected into the eye, but will be a general mechanism used in other immuneprivileged sites as well as organs and tissues when CD8⫹ T regulatory cells are generated to suppress expression of immune responses.

DISCUSSION In summary, reports by our laboratory showed that NK cells provided cytokines (IFN-␥) that helped to clear influenza virus (and other viruses) and contributed to the biasing of the immune response toward a Th1 response [22, 84, 85]. During pulmonary conditions in which the Th1 response was chronic or the toxicity of the chemical led to the release of inflammatory cytokines, the NK cell contributed to the pathophysiological state of the organ by promoting an inflammatory response through its IFN-␥ release. Others reported that, under other conditions, the NK cell can make Th2-type cytokines in the absence of IFN-␥ [15]. On the other hand, in our animal models, the NKT cell was prominent in both Th1 and Th2 responses, as well as being critical to the development of peripheral tolerance to the eye-inoculated antigens. In both mouse and human a major

Fig. 3. Model for role of NKT cells and antigentransporting macrophage in spleen during ACAID. The model shows that APCs (derived from the eye) selectively up-regulate chemokines capable of attracting NKT cells. Then, NKT cells brought to the spleen by MIP-2, bind CD1 expressed on APCs. TCR binding to CD1 may induce NKT cells to synthesize and secrete chemokines that recruit CD8⫹ T cells to the site of tolerance induction. The cluster of cells participating may include APCs, CD8⫹ T cells, B cells, and NKT cells. The cells could be held together by CD1 and other unknown adhesion molecules. The NKT cell, stimulated through its TCR by CD1 ligation, produces regulatory cytokines (IL-10, TGF-␤) that in turn influence the differentiation of CD8⫹ T cells into negative regulatory cells for delayed-type hypersensitivity (DTH) and peripheral tolerance induced via the eye.

Stein-Streilein et al. Innate cells promote immune responses and tolerance

491

subpopulation of NKT lymphocytes responds to signals provided ligation of its TCR to CD1 molecules [51, 56]. It is possible that the CD1-induced signals in NKT cells may depend on the character of the antigen-transporting APC that expresses it (i.e., TGF-␤ versus IL-12 secretion). In addition, the transporting APC may express critical co-receptors (or cytokines) for activation of NKT cell secretion of inflammatory cytokine (IFN-␥ that are different or absent from the antigentransporting APCs that signal the NKT cell to secrete downmodulatory cytokines and support the development of Tregulatory cells. Others showed that the cytokine microenvironment that is present during the binding of the NKT cell to the CD1 molecule will influence the type of cytokine produced by the NKT cell [48, 61, 63]. Understanding the subtle differences among the signals received by NKT cells through CD1 ligation on antigen-transporting macrophages might lead to new knowledge and therapies in conditions where there is a breakdown in tolerance (autoimmunity). Recent studies [83] showed a unique profile of chemokines that was responsible for recruitment of NKT cells during peripheral tolerance induction in the spleen after the antigen was inoculated into an immune-privileged site. Recruitment of the appropriate cells to the secondary lymphoid organ for tolerance supports the concept of required cellular interactions dependent on chemokines and ligands different from those that encourage the promotion of cell clusters required for activation and induction of inflammation (Fig. 3). The model for immuneprivileged site-induced tolerance is equally applicable as a model of self-tolerance in tissues and organs. Generation of negative regulatory T lymphocytes may be defective in certain autoimmune diseases. Reports of an association of defective or deficient NKT cells in a variety of autoimmune diseases in both mouse and humans have been published [86–90]. Moreover, diabetic-prone NOD mice were protected from the onset of diabetes with the adoptive transfer of NKT cells [90, 91]. The phenotype of such NKT cells is not well described, but it is assumed that the protective NKT cells must prevent the Th1-destructive environment and would imply, therefore, that there may be a role for NKT cell-induced antigen-specific CD8⫹ T regulatory cells in the prevention of diabetes and other autoimmune diseases.

ACKNOWLEDGMENTS Dr. Douglas Faunce is a recipient of the National Eye Institute National Research Service Award 1 F32 EY07021-01; and Dr. Jie Zhang-Hoover is a recipient of the National Heart Lung and Blood Institute National Research Service Award 1 F32 HL10148-01A1. This work was supported in part by National Heart Lung and Blood Institute Grant R01 HL 33709-09, National Eye Institute Grant R01 EY1 EY11983-01A2, and The Schepens Eye Research Institute. We thank Drs. J. Wayne Streilein, Steven P. Balk, and Mark Exley for their helpful discussions on ideas and concepts presented in this review. We also appreciate Ms. Gayle Barry’s efforts in preparing the manuscript. 492

Journal of Leukocyte Biology Volume 67, April 2000

REFERENCES 1. Medzhitov, R., Janeway, C. A., Jr. (1997) Innate immunity: The virtues of a nonclonal system of recognition. Cell 91, 295–298. 2. Matzinger, P. (1996) An innate sense of danger. Semin. Immunol. 10, 399–415. 3. Carnaud, C., Lee, D., Donnars, O., Park, S-H., Beavis, A., Koezuka, Y., Bendelac, A. (1999) Cutting edge: Cross-talk between cells of the innate immune system. NKT cells rapidly activate NK cells. J. Immunol. 163, 4647–4650. 4. Mandelboim, O., Kent, S., Davis, D. M., Wilson, S. B., Okazaki, T., Jackson, R., Hafler, D., Strominger, J. L. (1998) Natural killer activating receptors trigger interferon gamma secretion from T cells and natural killer cells, Proc. Natl. Acad. Sci. USA 95, 3798–3803. 5. Arase, H., Arase, N., Saito, T. (1996) Interferon-␥ production by natural killer (NK) cells and NK1.1⫹ T cells upon NKR-P1 cross-linking. J. Exp. Med. 183, 2391–2396. 6. Young, H. A., Hardy, K. J. (1995) Role of interferon-gamma in immune cell regulation. J. Leukoc. Biol. 58, 373–381. 7. Yoshimoto, T., Bendelac, A., Watson, C., Hu-Li, J., Paul, W. E. (1995) Role of NK1.1⫹ T cells in a TH2 response and in immunoglobulin E production. Science 270, 1845–1847. 8. Chen, Y. H., Chiu, N. M., Mandal, M., Wang, N., Wang, C. R. (1997) Impaired NK1⫹ T cell development and early IL-4 production in CD1-deficient mice. Immunity 6, 459–467. 9. Mendiratta, S. K., Martin, W. D., Hong, S., Boesteanu, A., Joyce, S., Van Kaer, L. (1997) CD1d1 mutant mice are deficient in natural T cells that promptly produce IL-4. Immunity 6, 469–477. 10. Smiley, S. T., Kaplan, M. H., Grusby, M. J. (1997) Immunolglobulin E production in the absence of interleukin-4-secreting CD1-dependent cells. Science 275, 977–979. 11. Brown, D. R., Fowell, D. J., Corry, D. B., Wynn, T. A., Moskowitz, N. H., Cheever, A. W., Locksley, R. M., Reiner, S. L. (1996) ␤-2-microglobulindependent NK1.1⫹ T cells are not essential for T helper cell 2 immune responses. J. Exp. Med. 184, 1295–1304. 12. Zhang, Y., Rogers, K. H., Lewis, D. B. (1996) Beta 2-microglobulindependent T cells are dispensable for allergen-induced T helper 2 responses. J. Exp. Med. 184, 1507–1512. 13. Gowans, J. L., Knight, E. J. (1964) The route of re-circulation of lymphocytes in the rat. Proc. R. Soc. Lond. B. Biol. Sci. 159, 257–282. 14. Guy-Grand, D., Griscelli, C., Vassalli, P. (1974) The gut-associated lymphoid system: Nature and properties of the large dividing cells. Eur. J. Immunol. 4, 435–443. 15. Streilein, J. W., Stein-Streilein, J., Head, J. (1986) Regional specialization in antigen presentaion. In The Reticuloendothelial System. New York: Plenum, 37–93. 16. Zhang-Hoover, J., Sutton, A., van Rooijen, N., Stein-Streilein, J. (1999) Alveolar macrophages determine if a fibrogenic immune response occurs or not in the lung. In Society for Leukocyte Biology 15th International Congress with the European Macrophage Study Group, September 22–26, 1999, Cambridge, UK, Bethesda, MD: Society for Leukocyte Biology, 15. 17. Trinchieri, G. (1989) Biology of natural killer cells [Review] [1182 refs.]. Adv. Immunol. 47, 187–376. 18. Bukowski, J. F., Woda, B. A., Habu, S., Okumura, K., Welsh, R. A. (1983) Natural killer cell depletion enhances virus synthesis and virus-induced hepatitis in vivo. J. Immunol. 131, 1531–1538. 19. Bukoswki, J. F., Warner, J. F., Dennert, G., Welsh, R. M. (1985) Adoptive transfer studies demonstrating the antiviral effect of natural killer cells in vivo. J. Exp. Med. 161, 40–52. 20. Stein-Streilein, J., Bennett, M., Mann, D., Kumar, V. (1983) Natural killer cells in mouse lung: surface phenotype, target preference, and response to local influenza virus infection. J. Immunol. 131, 2699–2704. 21. Bukowski, J. F., Welsh, R. M. (1985) Inability of interferon to protect virus-infected cells against lysis by natural killer (NK) cells correlates with NK cell-mediated antiviral effects in vivo. J. Immunol. 135, 3537–3541. 22. Bukowski, J. F., Welsh, R. M. (1986) The role of natural killer cells and interferon in resistance to acute infection of mice with herpes simplex virus type 1. J. Immunol. 136, 3481–3485. 23. Stein-Streilein, J., Guffee, J. (1986) In vivo treatment of mice and hamsters with antibodies to Asialo GM1 increases morbidity and mortality to pulmonary influenza infection. J. Immunol. 136, 1435–1441. 24. Kos, F. J., Engleman, E. G. (1996) Role of natural killer cells in the generation of influenza virus-specific cytotoxic T cells. Cell. Immunol. 173, 1–6. 25. Stein-Streilein, J., Guffee, J., Fan, W. (1988) Locally and systemically derived natural killer cells participate in defense against intranasally inoculated influenza virus. Reg. Immunol. 1, 100–105.

http://www.jleukbio.org

26. Stitz, L., Althage, A., Hengartner, H., Zinkernagel, R. (1985) Natural killer cells vs. cytotoxic T cells in the peripheral blood of virus-infected mice. J. Immunol. 134, 598–602. 27. Stitz, L., Baenziger, J., Pircher, H., Hengartner, H., Zinkernagel, R. M. (1986) Effect of rabbit anti-asialo GM1 treatment in vivo or with anti-asialo GM1 plus complement in vitro on cytotoxic T cell activities. J. Immunol. 136, 4674–4680. 28. Stein-Streilein, J. (1988) Immunobiology of lymphocytes in the lung. Reg. Immunol. 1, 128–136. 29. Stein-Streilein, J., Witte, P. L., Streilein, J., Guffee, J. (1985) Local cellular defenses in influenza-infected lungs. Cell. Immunol. 95, 234–246. 30. MacDonald, H. R. (1995) NK1.1⫹ T cell receptor-␣/␤⫹ cells: new clues to their origin, specificity, and function. J. Exp. Med. 182, 633–638. 31. Watanabe, H., Miyaji, C., Kawachi, Y., Liai, T., Ohtsuka, K., Iwanage, T., Takahashi-Iwanaga, H., Abo, T. (1995) Relationships between intermediate TCR cells and NK1.1⫹ T cells in various immune organs: NK1.1⫹ T cells are present within a population of intermediate TCR cells. J. Immunol. 155, 2972–2983. 32. Yoshimoto, T., Paul, W. E. (1994) CD4⫹, NK1.1⫹ T cells promptly produce interleukin 4 in response to in vivo challenge with anti-CD3. J. Exp. Med. 179, 1285–1295. 33. Kawano, T., Cui, J., Koezuka, Y., Toura, I., Kaneko, Y., Sato, H., Kondo, E., Harada, M., Koseki, H., Nakayama, T., Tanaka, Y., Taniguchi, M. (1998) Natural killer-like nonspecific tumor cell lysis mediated by specific ligand-activated Valpha14 NKT cells. Proc. Natl. Acad. Sci. USA 95, 5690–5693. 34. Kariv, I., Hardy, R. R., Hayakawa, K. (1993) Selective enrichment of major histocompatibility complex class II-specific autoreactive cells in the thymic Thy0 subset. J. Exp. Med. 177, 1429–1437. 35. Gombert, J. M., Herbelin, A., Tancrede-Bohin, E., Dy, M., Carnaud, M., Bach, J. P. (1998) Early quantitative and functional deficiency of NK1⫹-like thymocytes in NOD mouse. Eur. J. Immunol. 26, 2989–2998. 36. Asea, A., Stein-Streilein, J. (1998) Signalling through NK1.1 triggers NK cells to die but induces NKT cells to produce IL-4. Immunology 93, 296–305. 37. Imai, K., Kanno, M., Kimoto, H., Shigemoto, K., Yamamoto, S., Taniguchi, M. (1986) Sequence and expression of transcripts of the T-cell antigen receptor alpha-chain gene in a functional, antigen-specific suppressor-Tcell hybridoma, Proc. Natl. Acad. Sci. USA 83, 8708–8712. 38. Lantz, O., Bendelac, A. (1994) An invariant T cell receptor alpha chain is used by a unique subset of major histocompatibility complex class I-specific CD4⫹ and CD4⫺8⫺ T cells in mice and humans. J. Exp. Med. 180, 1097–1106. 39. Makino, Y., Kanno, R., Ito, T., Higashino, K., Taniguchi, M. (1995) Predominant expression of invariant V alpha 14⫹ TCR alpha chain in NK1.1⫹ T cell populations. Int. Immunol. 7, 1157–1161. 40. Fowlkes, B. J., Kruisbeek, A. M., Ton-That, H., Weston, M. A., Coligan, J. E., Schwartz, R. H., Pardoll, D. M. (1987) A novel population of T-cell receptor alpha beta-bearing thymocytes which predominantly expresses a single V beta gene family. Nature 329, 251–254. 41. Budd, R. C., Miescher, G. C., Howe, R. C., Lees, R. K., Bron, C., MacDonald, H. R. (1987) Developmentally regulated expression of T cell receptor beta chain variable domains in immature thymocytes. J. Exp. Med. 166, 577–582. 42. Ballas, Z. K., Rasmussen, W. (1990) NK1.1⫹ thymocytes adult murine CD4⫺, CD8⫺ thymocytes contain an NK1.1⫹, CD3⫹, CD5hi, CD44hi, TCR-V beta 8⫹ subset. J. Immunol. 145, 1039–1045. 43. Sykes, M. (1990) Unusual T cell populations in adult murine bone marrow prevalence of CD3⫹CD4⫺CD8⫺ and alpha beta TCR⫹NK1.1⫹ cells. J. Immunol. 145, 3209–3215. 44. Levitsky, H. I., Golumbek, P. T., Pardoll, D. M. (1991) The fate of CD4-8⫺ T cell receptor-alpha beta⫹ thymocytes. J. Immunol. 146, 1113–1117. 45. Takahama, Y., Kosugi, A., Singer, A. (1991) Phenotype, ontogeny, and repertoire of CD4⫺CD8⫺ T cell receptor alpha beta⫹ thymocytes variable influence of self-antigens on T cell receptor V beta usage. J. Immunol. 146, 1134–1141. 46. Masuda, K., Makino, Y., Cui, J., Ito, T., Tokuhisa, T., Takahama, Y., Koseki, H., Tsuchida, K., Koike, T., Moriya, H., Amano, M., Taniguchi, M. (1997) Phenotypes and invariant alpha beta TCR expression of peripheral V alpha 14⫹ NK T cells. J. Immunol. 158, 2076–2082. 47. Bendelac, A., Killeen, N., Littman, D. R., Schwartz, R. H. (1994) A subset of CD4⫹ thymocytes selected by MHC class I molecule. Science 263, 1774–1778. 48. Cui, J., Shin, T., Kawano, T., Sato, H., Kondo, E., Toura, I., Kaneko, Y., Koseki, H., Kanno, M., Taniguchi, M. (1997) Requirement for V␣14 NKT cells in IL-12-mediated rejection of tumors. Science 278, 1623–1625. 49. Taniguchi, M., Koseki, H., Tokuhisa, T., Masuda, K., Sato, H., Kondo, E., Kawano, T., Cui, J., Perkes, A., Koyasu, S., Makino, Y. (1996) Essential

50.

51.

52.

53.

54.

55.

56.

57. 58.

59.

60.

61.

62. 63.

64.

65.

66. 67. 68. 69. 70.

requirement of an invariant V alpha 14 T cell antigen receptor expression in the development of natural killer T cells. Proc. Natl. Acad. Sci. USA 93, 11025–11028. Porcelli, S., Yockey, C. E., Brenner, M. B., Balk, S. P. (1993) Analysis of T cell antigen receptor (TCR) expression by human peripheral blood CD4⫺8⫺ alpha/beta T cells demonstrates preferential use of several V beta genes and an invariant TCR alpha chain. J. Exp. Med. 178, 1–16. Bendelac, A., Lantz, O., Quimby, M. E., Yewdell, J. W., Bennink, J. R., Brutkiewicz, R. R. (1995) CD1 recognition by mouse NK1⫹ T lymphocytes. Science 268, 863–865. Kawano, T., Dui, J., Koezuka, Y., Toura, I., Kaneko, Y., Motoki, K., Ueno, H., Nakagawa, R., Sato, H., Kondo, E., Koseki, H., Taniguchi, M. (1997) CD1d-restricted and TCR-mediated activation of Va14 NKT cells by glycosylceramides. Science 278, 1626–1629. Burdin, N., Brossay, L., Kronenberg, M. (1999) Immunization with alpha-galactosylceramide polarizes CD1-reactive NK T cells towards Th2 cytokine synthesis. Eur. J. Immunol. 29, 2014–2025. Kitamura, H., Iwakabe, K., Yahata, T., Nishimura, S., Ohta, A., Ohmi, Y., Sato, M., Takeda, K., Okumura, K., Van Kaer, L., Kawano, T., Taniguchi, M., Nishimura, T. (1999) The natural killer T (NKT) cell ligand alphagalactosylceramide demonstrates its immunopotentiating effect by inducing interleukin (IL)-12 production by dendritic cells and IL-12 receptor expression on NKT cells. J. Exp. Med. 189, 1121–1128. Bendelac, A., Rivera, M. N., Parks, H., Roark, J. H. (1996) Mouse CD1-specific NK1 T cells: Development, specificity and function. Annu. Rev. Immunol. 15, 535–562. Exley, M., Garcia, J., Balk, S. P., Porcelli, S. (1997) Requirements for CD1d recognition by human invariant Valpha24⫹ CD4⫺CD8⫺ T cells. J. Exp. Med. 186, 109–120. Stein-Streilein, J., Asea, A., Montalvo, J., Maniaci, V., Wanner, A. (1996) The role of NK1.1⫹ cells in immune regulation. Period. Biol. 98, 427–432. Sonoda, K-H., Exley, M., Snapper, S., Balk, S., Stein-Streilein, J. (1999) CD1 reactive NKT cells are required for development of systemic tolerance through an immune privileged site. J. Exp. Med. 190, 1215–1225. Stein-Streilein, J., Lipscomb, M. F., Fisch, H., Whitney, P. L. (1987) Pulmonary interstitial fibrosis induced in hapten immune hamsters. Am. Rev. Respir. Dis. 136, 119–123. Leite-De-Moraes, M. C., Moreau, G., Arnould, A., Machavoine, F., Garcia, C., Papiernik, M., Dy, M. (1998) IL-4-producing NK T cells are biased towards IFN-gamma production by IL-12. Influence of the microenvironment on the functional capacities of NK T cells. Eur. J. Immunol. 28, 1507–1515. Takahashi, M., Ogasawara, K., Takeda, K., Hashimoto, W., Sakihara, H., Kumagai, K., Anzai, R., Satoh, M., Seki, S. (1996) LPS induces NK1.1⫹ ␣␤ T cells with potent cytotoxicity in the liver of mice via production of IL-12 from kupffer cells. J. Immunol. 156, 2436–2442. Chen, H., Paul, W. E. (1997) Cultured NK1.1⫹ CD4⫹ T cells produce large amounts of IL-4 and IFN-␥ upon activation by anti-CD3 or CD1. J. Immunol. 159, 2240–2249. Tomura, M., Yu, W. G., Ahn, H. J., Yamashita, M., Yang, Y. F., Ono, S., Hamaoka, T., Kawano, T., Taniguchi, M., Koezuka, Y., Fujiwara, H. (1999) A novel function of Valpha14⫹CD4⫹NKT cells: stimulation of IL-12 production by antigen-presenting cells in the innate immune system. J. Immunol. 163, 93–101. Wilbanks, G. A., Mammolenti, M., Streilein, J. W. (1991) Studies on the induction of anterior chamber-associated immune deviation (ACAID) II. Eye-derived cells participate in generating blood-borne signals that induce ACAID. J. Immunol. 146, 3018–3024. Wilbanks, G. A., Mammolenti, M., Streilein, J. W. (1992) Studies on the induction of anterior chamber-associated immune deviation (ACAID) III. Induction of ACAID depends upon intraocular transforming growth factor-beta. Eur. J. Immunol. 22, 165–173. Streilein, J. W. (1990) Anterior chamber associated immune deviation: the privilege of immunity in the eye. Surv. Ophthal. 35, 67–73. Streilein, J. W., Takeuchi, M., Taylor, A. W. (1997) Immune privilege, T-cell tolerance, and tissue-restricted autoimmunity [Review] [29 refs.]. Hum. Immunol. 52, 138–143. Kaplan, H. J., Streilein, J. W. (1977) Immune response to immunization via the anterior chamber of the eye. I.F. lymphocyte-induced immune deviation. J. Immunol. 118, 809–814. Streilein, J. W., Niederkorn, J. Y. (1981) Induction of anterior chamberassociated immune deviation requires an intact, functional spleen. J. Exp. Immunol. 153, 1058–1067. Wilbanks, G. A., Streilein, J. W. (1996) Characterization of suppressor cells in anterior chamber-associated immune deviation (ACAID) induced by soluble antigen. Evidence of two functionally and phenotypically distinct T-suppressor cell populations. Immunology 71, 383–389.

Stein-Streilein et al. Innate cells promote immune responses and tolerance

493

71. Ballas, Z. K., Rasmussen, W. L., Alber, C. A., Sandor, M. (1997) Ontogeny of thymic NK1.1⫹ cells. J. Immunol. 159, 1174–1181. 72. Bendelac, A. (1995) Positive selection of mouse NK1⫹ T cells by CD1-expressing cortical thymocytes. J. Exp. Med. 182, 2091–2096. 73. D’Orazio, T. J., Niederkorn, J. Y. (1996) A novel role for TGF-beta and IL-10 in the induction of immune privilege. J. Immunol. 160, 2089–2098. 74. Hara, Y., Caspi, R. R., Wiggert, B., Dorf, M., Streilein, J. W. (1992) Analysis of an in vitro-generated signal that induces systemic immune deviation similar to that elicited by antigen injected into the anterior chamber of the eye. J. Immunol. 149, 1531–1538. 75. Hara, Y., Okamoto, S., Rouse, B., Streilein, J. W. (1993) Evidence that peritoneal exudate cells cultured with eye-derived fluids are the proximate antigen-presenting cells in immune deviation of the ocular type. J. Immunol. 151, 5162–5171. 76. Takeuchi, M., Alard, P., Streilein, J. W. (1998) TGF-beta promotes immune deviation by altering accessory signals of antigen-presenting cells. J. Immunol. 160, 1589–1597. 77. Porcelli, S. A., Modlin, R. L. (1999) The CD1 system: Antigen-presenting molecules for T cell recognition of lipids and glycolipids. Annu. Rev. Immunol. 17, 297–329. 78. Mantovani, A. (1999) The chemokine system: redundancy for robust outputs. Immunol. Today 20, 254–257. 79. Sallusto, F., Lanzavecchia, A., Mackay, C. R. (1998) Chemokines and chemokine receptors in T-cell priming and Th1/Th2-mediated responses. Immunol. Today 19, 568–574. 80. Baggiolini, M. (1998) Chemokines and leukocyte traffic. Nature 392, 565–568. 81. Hedrick, J. A., Zlotnik, A. (1996) Chemokines and lymphocyte biology. Curr. Opin. Immunol. 8, 343–347. 82. Rollins, B. J. (1997) Chemokines. Blood 90, 909–928. 83. Faunce, D., Sonoda, K.-H., Stein-Streilein, J. (1999) MIP-2 mediated recruitment of NKT cells is required for sytemic tolerance induced via the eye. In Society for Leukocyte Biology 15th International Congress with the

494

Journal of Leukocyte Biology Volume 67, April 2000

84. 85. 86.

87.

88.

89.

90. 91.

European Macrophage Study Group; September 22–26, 1999, Cambridge, UK. Bethesda, MD: Society for Leukocyte Biology, p. 9 [abstract]. Welsh, R. M. (1981) Natural cell-mediated immunity during viral infections. In Natural Resistance to Tumors and Viruses. Berlin: Springer-Verlag, 83–105. Welsh, R. M. (1986) Regulation of virus infections by natural killer cells. A review. Nat. Immun. Cell Growth Reg. 5, 169–199. Sumida, T., Sakamoto, A., Murata, H., Makino, Y., Takahashi, H., Yoshida, S., Nishioka, K., Iwamoto, I., Taniguchi, M. (1995) Selective reduction of T cells bearing invariant V alpha 24J alpha Q antigen receptor in patients with systemic sclerosis. J. Exp. Med. 182, 1163–1168. Wilson, S. B., Kent, S. C., Patton, K. T., Orban, T., Jackson, R. A., Exley, M., Porcelli, S., Schatz, D. A., Atkinson, M. A., Balk, S. P., Strominger, J. L., Hafler, D. A. (1998) Extreme Th1 bias of invariant Valpha24JalphaQ T cells in type 1 diabetes. Nature 391, 177–181 [published erratum appears in Nature (1999) 399, 84]. Mieza, M. A., Itoh, T., Cui, J. Q., Makino, Y., Kawano, T., Tsuchida, K., Koike, T., Shirai, T., Yagita, H., Matsuzawa, A., Koseki, H., Taniguchi, M. (1996) Selective reduction of V alpha 14⫹ NK T cells associated with disease development in autoimmune-prone mice. J. Immunol. 156, 4035– 4040. Yoshimoto, T., Bendelac, A., Hu-Li, J., Paul, W. E. (1995) Defective IgE production by SJL mice is linked to the absence of CD4⫹, NK1.1⫹ T cells that promptly produce interleukin 4. Proc. Natl. Acad. Sci. USA 92, 11931–11934. Baxter, A. G., Kinder, S. J., Hammond, K. J., Scollay, R., Godfrey, D. I. (1997) Association between alphabetaTCR⫹CD4⫺CD8⫺ T-cell deficiency and IDDM in NOD/Lt mice. Diabetes 46, 572–582. Hammond, K. J. L., Poulton, L. D., Palmisano, L. J., Silveira, P. A., Godfrey, D. I., Baxter, A. G. (1998) Alpha/beta-T cell receptor (TCR)⫹CD4⫺CD8⫺ (NKT) thymocytes prevent insulin-dependent diabetes mellitus in nonobese diabetic (NOD)/Lt mice by the influence of interleukin (IL)-4 and/or IL-10. J. Exp. Med. 187, 1047–1056.

http://www.jleukbio.org