биохимия атеросклероза
.pdf402 Stewart C. Whitman and Tanya A. Ramsamy
(α-GalCer), which is derived from marine sponges. In addition to being used as a way to distinguish NKT cells from NK cells, these four selection criteria have allowed NKT cells to be themselves partitioned into four broad categories as outlined in Fig. 18.1.
The ability of one unique subset of T lymphocytes to interact with CD1d results from the expression of a highly biased, evolutionarily conserved, TCR consisting of an invariant α-chain (a Vα14 segment joined to a Jα18 segment) that preferentially binds to one of three β-chains (Vβ8.2, Vβ-7, and Vβ-2) [70, 71]. As such, these T lymphocytes are known as invariant NKT cells [72] or Vα14 NKT cells [73]. In C57BL/6 mice, this unique subset of T lymphocytes generally, but not consistently, expresses the NK1.1 receptor to varying degrees [69, 74–76]. Although the physiological substrate for this subset of T lymphocytes is still unclear, invariant NKT cells have a strong response to and are selectively activated by the exogenous substrate, α- GalCer, a marine sponge-derived glycosphingolipid [77], which binds specifically to CD1d [74, 78]. Furthermore, invariant Vα14 T cells do not express CD8, a marker characteristic of cytotoxic T lymphocytes, but instead are either CD4+, a marker characteristic helper T lymphocytes, or double negatives (DN) [75, 76]. Although Vα14 NKT cells are by far the more abundant NKT cells present, other, less common NKT cells have been identified.
Like the Vα14 NKT cells, the non-Vα14 NKT cells are also CD1d-depend- ent and can either be CD4+ or DN [79–81]. This additional class of NKT cells, which expresses either the Vα3.2-Jα9 or Vα8 α-chain and preferentially binds the Vβ8 β-chain, is less common than the Vα14 NKT cells in the murine model [81]. Yet another class of NKT cells are those that are CD1dindependent and include NKT cells that express the NK1.1 receptor, have diverse TCR can be CD8+, CD4+, or DN [82, 83] or those that express CD49B, an antigen present on a majority of the murine NK cells [84].
Once activated, all NKT cell populations have the capacity to exert immunoregulatory functions by releasing large quantities of T helper (Th) 1 or Th2 cytokines. The Th1 cytokines release by NKT cells upon activation include the proatherogenic cytokines interleukin (IL)-12 [85, 86] and IFN-γ [87–90], whereas the antiatherogenic cytokines [86] and IL-10 [91–94] are associated with the Th2 response. The release of cytokines from NKT cells initiates a cascade of events and causes the bystander activation of adjacent NK cells, B lymphocytes, and dendritic cells [95], as well as the activation of conventional CD4+ and CD8+ T lymphocytes [71, 96]. Although the natural ligands for CD1d-dependent NKT cells, which includes the Vα14 NKT cells, still remain to be identified, α-GalCer has been shown to be a potent and specific activator of NKT cells [74]. As such, evidence exists to suggest that NKT cells can modulate antitumoral [97–99] and antimicrobial immunity [100, 101] as well as a number of autoimmune diseases, including experimental autoimmune encephalomyelitis [102] (a model for multiple sclerosis) and type 1 diabetes [103, 104], and as reviewed by van der Vliet et al. [105].
Chapter 18. The Role of the Immune System in Atherosclerosis |
403 |
Atherosclerosis is a chronic inflammatory disorder that is known to involve components of both the innate and adaptive immune systems [3, 5, 106, 107]. Given that NKT cells link the two arms of the immune system, it seems likely that they would also play a role in the pathogenesis of atherosclerosis. In fact, several studies have shown, using lipopolysaccharide or α-GalCer, that NKT cells are present in atherosclerotic lesions [15, 18, 108]. Furthermore, α-GalCer administration has recently been shown to specifically exacerbate atherosclerosis in Apoe–/– mice, a model predisposed to developing atherosclerotic lesions spontaneously, when compared to vehicle treated animals [16, 18], and those also deficient in CD1d [15]. Incidentally, CD1d has been detected in human atherosclerotic lesions [109] underscoring the probable involvement of NKT cells in the human disease process. The administration of α-GalCer to Apoe–/– mice has been shown to have dramatic effects on the local cytokine environment of the aortas with established atherosclerotic lesions by inducing the production of IFN-γ, IL-4, and IL-10 [16]. Others have observed an early burst of inflammatory cytokines, both Th1 (IFN-γ, tumor necrosis factor-α, IL-2) and Th2 (IL-4, IL-5) cytokines, in the sera of Apoe–/– [16]. The effect of α-GalCer on the development of atherosclerosis and the production of proatherogenic cytokines by NKT cells suggests that CD1d-dependent NKT cells might play a participatory role in the atherogenic process.
CD1d Null Mice
Animal models that combine genetic risks for atherosclerosis, such as the Apoe–/– or the Ldl-r−/− [90] mouse, with an altered immune system have been invaluable in demonstrating a link between atherosclerosis and immunity [3, 5]. Using these models, several groups have provided evidence to suggest that NKT cells are potentially proatherogenic [15–19]. The most commonly used model of NKT cells deficiency is the CD1d–/– mouse, which was first created by Mendiratta et al. [110]. In this model, the CD1d–/– mice lack the molecule necessary for normal development of the CD1d-dependent NKT cell and are therefore deficient in functional CD1d-dependent NKT cells. Recent studies have shown that deficiency of CD1d significantly slows the development of atherosclerosis in wild-type mice on an atherogenic diet [18] or in Apoe–/– mice whether they were fed a chow diet [15] or an atherosclerotic diet, which is a diet enriched in both cholesterol and fat [16]. The reduction in atherosclerotic lesion size ranged from 25% in double knockouts on a chow diet to 70% on an atherosclerotic diet. Similar observations were shown when CD1d–/– was put onto an Ldl-r–/– genetic background. These mice had 50% less lesions than the CD1d+/+ controls when on an atherosclerotic diet for 4 weeks [17]. However, the authors did note that this difference was lost when both groups were compared at 8 and 12 weeks.
Using the technique of bone marrow transplantation following lethal irradiation, Nakai et al. [18] showed that Ldl-r–/– mice reconstituted with CD1d–/–
404 Stewart C. Whitman and Tanya A. Ramsamy
bone marrow had significantly less atherosclerosis compared to lethally irradiated Ldl-r–/– mice reconstituted with wild-type bone marrow. These data suggest that replenishing the hematopoietic stem cells with bone marrow from mice that have CD1d-dependent NKT cells is sufficient to stimulate the development of early stage atherosclerotic lesions. These results, combined with those obtained using CD1d null mice, suggest that the absence of CD1dreactive NKT cells attenuates lesion formation in mice during the early fatty streak formation.
Jα18 Null Mice
The exclusive expression of the invariant Vα14/Vβ receptor on Vα14 NKT cells and the essential requirement of Vα14 expression for the development of Vα14 NKT cells was demonstrated in Vα14 NKT cell-deficient (Jα18–/–) mice [111]. Here, Cui et al. [111] showed that targeted deletion of the Jα18 gene caused a complete failure of the mice to develop Vα14 NKT cells, leaving other lymphoid lineages intact. This observation strongly suggests that the invariant Vα14 segment is indispensable for the generation of Vα14 NKT cells. Targeted deletion of the Jα18 gene resulting in the selective depletion of CD1d-dependent, Vα14 NKT cells without affecting the population of other NKT (including the other CD1d-dependent NKT cells), NK, and conventional T lymphocytes [73, 111]. By superimposing the Jα18 deficiency onto mice that are already susceptible to atherosclerosis by virtue of being Ldl-r–/–, Whitman et al. have shown that the loss of functionally active Vα14 NKT cells significantly slows the formation of lesions by 20% in both genders of mice [19]. As such, this study shows that a single population of NKT cells participates in the process of early-stage atherosclerotic lesion formation. Upon stimulation, Vα14 NKT cells have the capacity to exert immunoregulatory functions by releasing large amounts of inflammatory cytokines, including the proatherogenic cytokine IFN-γ [87–90]. This release of cytokines will, in turn, cause the activation of adjacent NK cells, B cells, CD4+ and CD8+ T lymphocytes [71, 112], as well as adjacent antigen-presenting cells [95]. This hierarchical form of activation may be one of the key mechanisms by which NKT cells promote atherosclerosis at the level of the vessel wall.
Concluding Remarks
Despite the ever-growing list of immune cells now identified within the developing atherosclerotic lesion of both humans and mice, this chapter has only focused on two unique populations of lymphocytes, namely NK cells and NKT cells. As noted in the introduction, macrophages and conventional T lymphocytes constitute the major cell types involved in the earlyand latter stages of this disease. However, despite the apparent underrepresentation of both cell types, NK cells and NKT cells have now been shown to have a
Chapter 18. The Role of the Immune System in Atherosclerosis |
405 |
significant participatory role alongside these other more “prominent” classes of lymphocytes. As highlighted above, recent experimental evidence from our laboratory [13, 14, 19] and others [15–18] suggest that neither NK cells nor NKT cells cause the disease, but rather these immune cells play an important role in accelerating lesion development by modulating, via the elaboration of IFN-γ, the function of other more prominent immune cells (conventional T lymphocytes and macrophages) found within the developing atherosclerotic lesion. In this context, research from a number of laboratories strongly suggests that the proinflammatory cytokines IL-12 [85, 86] and IL-18 [113, 114] play a significant role in promoting atherosclerosis via the activation of NKT and NK cells, respectively.
Current in vivo models that combine genetic risks for atherosclerosis with an altered immune system have revealed to us the important modulatory role played by both NK and NKT cells. These studies have also highlighted the fact that the transgenic mouse models of atherosclerosis, although not a perfect substitute for human lesion pathology, are currently our best all-around model that affords us a significant degree of flexibility by allowing us access to a vast array of different transgenic and gene-targeted mice. As new advancements occur in mouse gene manipulation techniques, the use of the mouse will increase our ability to define these complicated mechanistic pathways, leaving us with the easier task of having to only confirm such pathways in our human atherosclerotic specimens.
Acknowledgments: S. C. Whitman is the recipient of a Great-West Life & London Life New Investigator Award from the Heart and Stroke Foundation of Canada and is supported by the Heart and Stroke Foundation of Ontario Grant NA-5086 and Canadian Institutes of Health Research Grants MOP53344 and GHS-60663. T. A. Ramsamy is the recipient of a Heart and Stroke Foundation of Canada Post-Doctoral Fellowship awarded to the Coronary Disease and Heart Failure Group at the University of Ottawa Heart Institute. The authors apologize to those investigators whose excellent contributions to the field of atherosclerosis and immunity research, which could not be included in this review due to space limitations.
References
1.McMillan GC: Historical review of research on atherosclerosis. Adv Exp Med Biol 369: 1–6, 1995.
2.Ross R: Atherosclerosis—an inflammatory disease. N Engl J Med 340: 115–126, 1999.
3.Hansson GK, Libby P, Schonbeck U, Yan ZQ: Innate and adaptive immunity in the pathogenesis of atherosclerosis. Circ Res 91: 281–291, 2002.
4.VanderLaan PA, Reardon CA: Thematic review series: the immune system and atherogenesis. The unusual suspects: an overview of the minor leukocyte populations in atherosclerosis. J Lipid Res 46: 829–838, 2005.
406Stewart C. Whitman and Tanya A. Ramsamy
5.Hansson GK: Immune mechanisms in atherosclerosis. Arterioscler Thromb Vasc Biol 21: 1876–1890, 2001.
6.Daugherty A, Rateri DL: T lymphocytes in atherosclerosis: the yin-yang of Th1 and Th2 influence on lesion formation. Circ Res 90: 1039–1040, 2002.
7.Roselaar SE, Kakkanathu PX, Daugherty A: Lymphocyte populations in atherosclerotic lesions of apoE –/– and LDL receptor –/– mice. Decreasing density with disease progression. Arterioscler Thromb Vasc Biol 16: 1013–1018, 1996.
8.Dansky HM, Charlton SA, Harper MM, Smith JD: T and B lymphocytes play a minor role in atherosclerotic plaque formation in the apolipoprotein E-defi- cient mouse. Proc Natl Acad Sci U S A 94: 4642–4646, 1997.
9.Daugherty A, Pure E, Delfel-Butteiger D, Chen S, Leferovich J, Roselaar SE, Rader DJ: The effects of total lymphocyte deficiency on the extent of atherosclerosis in apolipoprotein E–/– mice. J Clin Invest 100: 1575–1580, 1997.
10.Song L, Leung C, Schindler C: Lymphocytes are important in early atherosclerosis. J Clin Invest 108: 251–259, 2001.
11.Smith JD, Trogan E, Ginsberg M, Grigaux C, Tian J, Miyata M: Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc Natl Acad Sci U S A 92: 8264–8268, 1995.
12.de Villiers WJ, Smith JD, Miyata M, Dansky HM, Darley E, Gordon S: Macrophage phenotype in mice deficient in both macrophage-colony-stimulat- ing factor (op) and apolipoprotein E. Arterioscler Thromb Vasc Biol 18: 631–640, 1998.
13.Whitman SC, Rateri DL, Szilvassy SJ, Yokoyama W, Daugherty A: Depletion of natural killer cell function decreases atherosclerosis in low-density lipoprotein receptor null mice. Arterioscler Thromb Vasc Biol 24: 992–994, 2004.
14.Ramsamy TA, Rogers L, Hasu M, Siddiqui MM, Huynh T, Tam NL, Yokoyama WM, Whitman SC: Natural killer cell deficiency impairs early-stage lesion development in apolipoprotein E null mice. Arterioscler Thromb Vasc Biol 25: E-82, 2005.
15.Tupin E, Nicoletti A, Elhage R, Rudling M, Ljunggren HG, Hansson GK, Berne GP: CD1d-dependent activation of NKT cells aggravates atherosclerosis. J Exp Med 199: 417–422, 2004.
16.Major AS, Wilson MT, McCaleb JL, Ru SY, Stanic AK, Joyce S, Van Kaer L, Fazio S, Linton MF: Quantitative and qualitative differences in proatherogenic NKT cells in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 24: 2351–2357, 2004.
17.Aslanian AM, Chapman HA, Charo IF: Transient role for CD1d-restricted natural killer T cells in the formation of atherosclerotic lesions. Arterioscler Thromb Vasc Biol 25: 628–632, 2005.
18.Nakai Y, Iwabuchi K, Fujii S, Ishimori N, Dashtsoodol N, Watano K, Mishima T, Iwabuchi C, Tanaka S, Bezbradica JS, Nakayama T, Taniguchi M, Miyake S, Yamamura T, Kitabatake A, Joyce S, Van Kaer L, Onoe K: Natural killer T cells accelerate atherogenesis in mice. Blood 104: 2051–2059, 2004.
19.Purdy L, Hasu M, Whitman SC: Depletion of invariant natural killer T cells decreases atherosclerosis in LDL receptor null mice. Arterioscler Thromb Vasc Biol 24: E57, 2004.
20.Sohma Y, Sasano H, Shiga R, Saeki S, Suzuki T, Nagura H, Nose M, Yamamoto T: Accumulation of plasma cells in atherosclerotic lesions of Watanabe heritable hyperlipidemic rabbits. Proc Natl Acad Sci U S A 92: 4937–4941, 1995.
Chapter 18. The Role of the Immune System in Atherosclerosis |
407 |
21.Zhou X, Hansson GK: Detection of B cells and proinflammatory cytokines in atherosclerotic plaques of hypercholesterolaemic apolipoprotein E knockout mice. Scand J Immunol 50: 25–30, 1999.
22.Major AS, Fazio S, Linton MF: B-lymphocyte deficiency increases atherosclerosis in LDL receptor-null mice. Arterioscler Thromb Vasc Biol 22: 1892–1898, 2002.
23.Kaartinen M, Penttila A, Kovanen PT: Accumulation of activated mast cells in the shoulder region of human coronary atheroma, the predilection site of atheromatous rupture. Circulation 90: 1669–1678, 1994.
24.Bobryshev YV, Taksir T, Lord RS, Freeman MW: Evidence that dendritic cells infiltrate atherosclerotic lesions in apolipoprotein E-deficient mice. Histol Histopathol 16: 801–808, 2001.
25.Strong JP, Malcom GT, Oalmann MC, Wissler RW: The PDAY study: natural history, risk factors, and pathobiology. Pathobiological determinants of atherosclerosis in youth. Ann N Y Acad Sci 811: 226–235, 1997.
26.Millonig G, Malcom GT, Wick G: Early inflammatory–immunological lesions in juvenile atherosclerosis from the Pathobiological Determinants of Atherosclerosis in Youth (PDAY)-study. Atherosclerosis 160: 441–448, 2002.
27.Millonig G, Schwentner C, Mueller P, Mayerl C, Wick G: The vascular-associ- ated lymphoid tissue: a new site of local immunity. Curr Opin Lipidol 12: 547–553, 2001.
28.Piedrahita JA, Zhang SH, Hagaman JR, Oliver PM, Maeda N: Generation of mice carrying a mutant apolipoprotein E gene inactivated by gene targeting in embryonic stem cells. Proc Natl Acad Sci U S A 89: 4471–4475, 1992.
29.Plump AS, Smith JD, Hayek T, Aalto-Setala K, Walsh A, Verstuyft JG, Rubin EM, Breslow JL: Severe hypercholesterolemia and atherosclerosis in apolipoprotein E-deficient mice created by homologous recombination in ES cells. Cell 71: 343–353, 1992.
30.Nakashima Y, Plump AS, Raines EW, Breslow JL, Ross R: ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree. Arterioscler Thromb 14: 133–140, 1994.
31.Ishibashi S, Goldstein JL, Brown MS, Herz J, Burns DK: Massive xanthomatosis and atherosclerosis in cholesterol-fed low density lipoprotein receptor-nega- tive mice. J Clin Invest 93: 1885–1893, 1994.
32.Stary HC: Macrophages, macrophage foam cells, and eccentric intimal thickening in the coronary arteries of young children. Atherosclerosis 64: 91–108, 1987.
33.Stary HC: The sequence of cell and matrix changes in atherosclerotic lesions of coronary arteries in the first forty years of life. Eur Heart J 11 (Suppl E): 3–19, 1990.
34.Stary HC: Composition and classification of human atherosclerotic lesions. Virchows Arch A Pathol Anat Histopathol 421: 277–290, 1992.
35.Kaartinen M, Penttila A, Kovanen PT: Mast cells in rupture-prone areas of human coronary atheromas produce and store TNF-alpha. Circulation 94: 2787–2792, 1996.
36.Getz GS: The involvement of lipoproteins in atherogenesis. Evolving concepts. Ann N Y Acad Sci 598: 17–28, 1990.
37.Schwenke DC, Carew TE: Initiation of atherosclerotic lesions in cholesterol-fed rabbits. I. Focal increases in arterial LDL concentration precede development of fatty streak lesions. Arteriosclerosis 9: 895–907, 1989.
408Stewart C. Whitman and Tanya A. Ramsamy
38.Schwenke DC, Carew TE: Initiation of atherosclerotic lesions in cholesterol-fed rabbits. II. Selective retention of LDL vs. selective increases in LDL permeability in susceptible sites of arteries. Arteriosclerosis 9: 908–918, 1989.
39.Fry DL: Mass transport, atherogenesis, and risk. Arteriosclerosis 7: 88–100, 1987.
40.Steinberg D, Witztum JL: Lipoproteins and atherogenesis. Current concepts. JAMA 264: 3047–3052, 1990.
41.Quinn MT, Parthasarathy S, Fong LG, Steinberg D: Oxidatively modified low density lipoproteins: a potential role in recruitment and retention of monocyte/macrophages during atherogenesis. Proc Natl Acad Sci U S A 84: 2995–2998, 1987.
42.Cybulsky MI, Iiyama K, Li H, Zhu S, Chen M, Iiyama M, Davis V, GutierrezRamos JC, Connelly PW, Milstone DS: A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J Clin Invest 107: 1255–1262, 2001.
43.Boullier A, Bird DA, Chang MK, Dennis EA, Friedman P, Gillotre-Taylor K, Horkko S, Palinski W, Quehenberger O, Shaw P, Steinberg D, Terpstra V, Witztum JL: Scavenger receptors, oxidized LDL, and atherosclerosis. Ann N Y Acad Sci 947: 214–222, 2001.
44.Jonasson L, Holm J, Skalli O, Bondjers G, Hansson GK: Regional accumulations of T cells, macrophages, and smooth muscle cells in the human atherosclerotic plaque. Arteriosclerosis 6: 131–138, 1986.
45.Jeziorska M, McCollum C, Woolley DE: Mast cell distribution, activation, and phenotype in atherosclerotic lesions of human carotid arteries. J Pathol 182: 115–122, 1997.
46.Bobryshev YV, Lord RS: Identification of natural killer cells in human atherosclerotic plaque. Atherosclerosis 180: 423–427, 2005.
47.Bobryshev YV, Lord RS: Co-accumulation of dendritic cells and natural killer T cells within rupture-prone regions in human atherosclerotic plaques. J Histochem Cytochem 53: 781–785, 2005.
48.Rosenfeld ME, Polinsky P, Virmani R, Kauser K, Rubanyi G, Schwartz SM: Advanced atherosclerotic lesions in the innominate artery of the ApoE knockout mouse. Arterioscler Thromb Vasc Biol 20: 2587–2592, 2000.
49.Calara F, Silvestre M, Casanada F, Yuan N, Napoli C, Palinski W: Spontaneous plaque rupture and secondary thrombosis in apolipoprotein E-deficient and LDL receptor-deficient mice. J Pathol 195: 257–263, 2001.
50.Williams H, Johnson JL, Carson KG, Jackson CL: Characteristics of intact and ruptured atherosclerotic plaques in brachiocephalic arteries of apolipoprotein E knockout mice. Arterioscler Thromb Vasc Biol 22: 788–792, 2002.
51.Zhou J, Moller J, Danielsen CC, Bentzon J, Ravn HB, Austin RC, Falk E: Dietary supplementation with methionine and homocysteine promotes early atherosclerosis but not plaque rupture in ApoE-deficient mice. Arterioscler Thromb Vasc Biol 21: 1470–1476, 2001.
52.Lanier LL: NK cell receptors. Annu Rev Immunol 16: 359–393, 1998.
53.Trinchieri G: Biology of natural killer cells. Adv Immunol 47: 187–376, 1989.
54.Whiteside TL, Herberman RB: Role of human natural killer cells in health and disease. Clin Diagn Lab Immunol 1: 125–133, 1994.
55.Cerwenka A, Lanier LL: Natural killer cells, viruses and cancer. Nat Rev Immunol 1: 41–49, 2001.
56.Yokoyama WM, Kim S, French AR: The dynamic life of natural killer cells. Annu Rev Immunol 22: 405–429, 2004.
Chapter 18. The Role of the Immune System in Atherosclerosis |
409 |
57.Schiller NK, Boisvert WA, Curtiss LK: Inflammation in atherosclerosis: lesion formation in LDL receptor-deficient mice with perforin and Lyst(beige) mutations. Arterioscler Thromb Vasc Biol 22: 1341–1346, 2002.
58.Hansson GK: Immune mechanisms in atherosclerosis. Arterioscler Thromb Vasc Biol 21: 1876–1890, 2001.
59.Roder J, Duwe A: The beige mutation in the mouse selectively impairs natural killer cell function. Nature 278: 451–453, 1979.
60.Roder JC: The beige mutation in the mouse. I. A stem cell predetermined impairment in natural killer cell function. J Immunol 123: 2168–2173, 1979.
61.Roder JC, Lohmann-Matthes ML, Domzig W, Wigzell H: The beige mutation in the mouse. II. Selectivity of the natural killer (NK) cell defect. J Immunol 123: 2174–2181, 1979.
62.Paigen B, Holmes PA, Novak EK, Swank RT: Analysis of atherosclerosis susceptibility in mice with genetic defects in platelet function. Arteriosclerosis 10: 648–652, 1990.
63.Kim S, Iizuka K, Aguila HL, Weissman IL, Yokoyama WM: In vivo natural killer cell activities revealed by natural killer cell-deficient mice. Proc Natl Acad Sci U S A 97: 2731–2736, 2000.
64.Budd RC, Miescher GC, Howe RC, Lees RK, Bron C, MacDonald HR: Developmentally regulated expression of T cell receptor beta chain variable domains in immature thymocytes. J Exp Med 166: 577–582, 1987.
65.Fowlkes BJ, Kruisbeek AM, Ton-That H, Weston MA, Coligan JE, Schwartz RH, Pardoll DM: A novel population of T-cell receptor alpha beta-bearing thymocytes which predominantly expresses a single V beta gene family. Nature 329: 251–254, 1987.
66.Ceredig R, Lynch F, Newman P: Phenotypic properties, interleukin 2 production, and developmental origin of a “mature” subpopulation of Lyt-2-. Proc Natl Acad Sci U S A 84: 8578–8582, 1987.
67.Makino Y, Kanno R, Ito T, Higashino K, Taniguchi M: Predominant expression of invariant V alpha 14+ TCR alpha chain in NK1.1+ T cell populations. Int Immunol 7: 1157–1161, 1995.
68.Reichlin A, Yokoyama WM: Natural killer cell proliferation induced by antiNK1.1 and IL-2. Immunol Cell Biol 76: 143–152, 1998.
69.Hammond KJ, Pellicci DG, Poulton LD, Naidenko OV, Scalzo AA, Baxter AG, Godfrey DI: CD1d-restricted NKT cells: an interstrain comparison. J Immunol
167:1164–1173, 2001.
70.Bendelac A, Bonneville M, Kearney JF: Autoreactivity by design: innate B and T lymphocytes. Nat Rev Immunol 1: 177–186, 2001.
71.Wilson SB, Delovitch TL: Janus-like role of regulatory iNKT cells in autoimmune disease and tumour immunity. Nat Rev Immunol 3: 211–222, 2003.
72.Kronenberg M, Gapin L: The unconventional lifestyle of NKT cells. Nat Rev Immunol 2: 557–568, 2002.
73.Taniguchi M, Harada M, Kojo S, Nakayama T, Wakao H: The regulatory role of Valpha14 NKT cells in innate and acquired immune response. Annu Rev Immunol 21: 483–513, 2003.
74.Kawano T, Cui J, Koezuka Y, Toura I, Kaneko Y, Motoki K, Ueno H, Nakagawa R, Sato H, Kondo E, Koseki H, Taniguchi M: CD1d-restricted and TCR-mediated activation of valpha14 NKT cells by glycosylceramides. Science
278:1626–1629, 1997.
410Stewart C. Whitman and Tanya A. Ramsamy
75.Benlagha K, Weiss A, Beavis A, Teyton L, Bendelac A: In vivo identification of glycolipid antigen-specific T cells using fluorescent CD1d tetramers. J Exp Med
191:1895–1903, 2000.
76.Matsuda JL, Naidenko OV, Gapin L, Nakayama T, Taniguchi M, Wang CR, Koezuka Y, Kronenberg M: Tracking the response of natural killer T cells to a glycolipid antigen using CD1d tetramers. J Exp Med 192: 741–754, 2000.
77.Wilson MT, Singh AK, Van Kaer L: Immunotherapy with ligands of natural killer T cells. Trends Mol Med 8: 225–231, 2002.
78.Singh N, Hong S, Scherer DC, Serizawa I, Burdin N, Kronenberg M, Koezuka Y, Van Kaer L: Cutting edge: activation of NK T cells by CD1d and alphagalactosylceramide directs conventional T cells to the acquisition of a Th2 phenotype. J Immunol 163: 2373–2377, 1999.
79.Cardell S, Tangri S, Chan S, Kronenberg M, Benoist C, Mathis D: CD1-restricted CD4+ T cells in major histocompatibility complex class II-deficient mice. J Exp Med 182: 993–1004, 1995.
80.Behar SM, Dascher CC, Grusby MJ, Wang CR, Brenner MB: Susceptibility of mice deficient in CD1D or TAP1 to infection with Mycobacterium tuberculosis. J Exp Med 189: 1973–1980, 1999.
81.Park SH, Weiss A, Benlagha K, Kyin T, Teyton L, Bendelac A: The mouse CD1d-restricted repertoire is dominated by a few autoreactive T cell receptor families. J Exp Med 193: 893–904, 2001.
82.Eberl G, Lees R, Smiley ST, Taniguchi M, Grusby MJ, MacDonald HR: Tissuespecific segregation of CD1d-dependent and CD1d-independent NK T cells. J Immunol 162: 6410–6419, 1999.
83.Hammond KJ, Pelikan SB, Crowe NY, Randle-Barrett E, Nakayama T, Taniguchi M, Smyth MJ, van Driel IR, Scollay R, Baxter AG, Godfrey DI: NKT cells are phenotypically and functionally diverse. Eur J Immunol 29: 3768–3781, 1999.
84.Arase H, Saito T, Phillips JH, Lanier LL: Cutting edge: the mouse NK cellassociated antigen recognized by DX5 monoclonal antibody is CD49b (alpha 2 integrin, very late antigen-2). J Immunol 167: 1141–1144, 2001.
85.Lee TS, Yen HC, Pan CC, Chau LY: The role of interleukin 12 in the development of atherosclerosis in ApoE-deficient mice. Arterioscler Thromb Vasc Biol
19:734–742, 1999.
86.Davenport P, Tipping PG: The role of interleukin-4 and interleukin-12 in the progression of atherosclerosis in apolipoprotein E-deficient mice. Am J Pathol
163:1117–1125, 2003.
87.Gupta S, Pablo AM, Jiang X, Wang N, Tall AR, Schindler C: IFN-gamma potentiates atherosclerosis in ApoE knock-out mice. J Clin Invest 99: 2752–2761, 1997.
88.Whitman SC, Ravisankar P, Daugherty A: IFN-gamma deficiency exerts gen- der-specific effects on atherogenesis in apolipoprotein E–/– mice. J Interferon Cytokine Res 22: 661–670, 2002.
89.Whitman SC, Ravisankar P, Elam H, Daugherty A: Exogenous interferongamma enhances atherosclerosis in apolipoprotein E–/– mice. Am J Pathol 157: 1819–1824, 2000.
90.Buono C, Come CE, Stavrakis G, Maguire GF, Connelly PW, Lichtman AH: Influence of interferon-gamma on the extent and phenotype of diet-induced atherosclerosis in the LDLR-deficient mouse. Arterioscler Thromb Vasc Biol 23: 454–460, 2003.
Chapter 18. The Role of the Immune System in Atherosclerosis |
411 |
91.Caligiuri G, Rudling M, Ollivier V, Jacob MP, Michel JB, Hansson GK, Nicoletti A: Interleukin-10 deficiency increases atherosclerosis, thrombosis, and low-density lipoproteins in apolipoprotein E knockout mice. Mol Med 9: 10–17, 2003.
92.Mallat Z, Besnard S, Duriez M, Deleuze V, Emmanuel F, Bureau MF, Soubrier F, Esposito B, Duez H, Fievet C, Staels B, Duverger N, Scherman D, Tedgui A: Protective role of interleukin-10 in atherosclerosis. Circ Res 85: e17–e24, 1999.
93.Pinderski LJ, Fischbein MP, Subbanagounder G, Fishbein MC, Kubo N, Cheroutre H, Curtiss LK, Berliner JA, Boisvert WA: Overexpression of inter- leukin-10 by activated T lymphocytes inhibits atherosclerosis in LDL receptordeficient mice by altering lymphocyte and macrophage phenotypes. Circ Res 90: 1064–1071, 2002.
94.Pinderski Oslund LJ, Hedrick CC, Olvera T, Hagenbaugh A, Territo M, Berliner JA, Fyfe AI: Interleukin-10 blocks atherosclerotic events in vitro and in vivo. Arterioscler Thromb Vasc Biol 19: 2847–2853, 1999.
95.Fujii S, Shimizu K, Smith C, Bonifaz L, Steinman RM: Activation of natural killer T cells by alpha-galactosylceramide rapidly induces the full maturation of dendritic cells in vivo and thereby acts as an adjuvant for combined CD4 and CD8 T cell immunity to a coadministered protein. J Exp Med 198: 267–279, 2003.
96.Godfrey DI, Hammond KJ, Poulton LD, Smyth MJ, Baxter AG: NKT cells: facts, functions and fallacies. Immunol Today 21: 573–583, 2000.
97.Kobayashi E, Motoki K, Uchida T, Fukushima H, Koezuka Y: KRN7000, a novel immunomodulator, and its antitumor activities. Oncol Res 7: 529–534, 1995.
98.Kawano T, Cui J, Koezuka Y, Toura I, Kaneko Y, Sato H, Kondo E, Harada M, Koseki H, Nakayama T, Tanaka Y, Taniguchi M: Natural killer-like nonspecific tumor cell lysis mediated by specific ligand-activated Valpha14 NKT cells. Proc Natl Acad Sci U S A 95: 5690–5693, 1998.
99.Nakagawa R, Motoki K, Ueno H, Iijima R, Nakamura H, Kobayashi E, Shimosaka A, Koezuka Y: Treatment of hepatic metastasis of the colon26 adenocarcinoma with an alpha-galactosylceramide, KRN7000. Cancer Res 58: 1202–1207, 1998.
100.Exley MA, Bigley NJ, Cheng O, Tahir SM, Smiley ST, Carter QL, Stills HF, Grusby MJ, Koezuka Y, Taniguchi M, Balk SP: CD1d-reactive T-cell activation leads to amelioration of disease caused by diabetogenic encephalomyocarditis virus. J Leukoc Biol 69: 713–718, 2001.
101.Gonzalez-Aseguinolaza G, de Oliveira C, Tomaska M, Hong S, Bruna-Romero O, Nakayama T, Taniguchi M, Bendelac A, Van Kaer L, Koezuka Y, Tsuji M: Alpha-galactosylceramide-activated Valpha 14 natural killer T cells mediate protection against murine malaria. Proc Natl Acad Sci U S A 97: 8461–8466, 2000.
102.Miyamoto K, Miyake S, Yamamura T: A synthetic glycolipid prevents autoimmune encephalomyelitis by inducing TH2 bias of natural killer T cells. Nature 413: 531–534, 2001.
103.Sharif S, Arreaza GA, Zucker P, Mi QS, Sondhi J, Naidenko OV, Kronenberg M, Koezuka Y, Delovitch TL, Gombert JM, Leite-De-Moraes M, Gouarin C, Zhu R, Hameg A, Nakayama T, Taniguchi M, Lepault F, Lehuen A, Bach JF, Herbelin A: Activation of natural killer T cells by alpha-galactosylceramide treatment prevents the onset and recurrence of autoimmune Type 1 diabetes. Nat Med 7: 1057–1062, 2001.
