趋化因子CCL18在过敏性疾病发病机理中的作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
本研究通过体外实验以及应用人体化的重症联合免疫缺陷(SCID)小鼠进行的体内实验对CCL18在过敏性疾病发病机理中的作用进行了研究,取得了以下学术进展:
    1.研究发现在过敏原的刺激下,过敏性哮喘患者外周血单个核细胞(PBMC)在转录及翻译水平表达CCL18基因增高。免疫酶联及免疫组化检测进一步发现,未经治疗的过敏性哮喘患者血清及支气管肺泡盥洗液中CCL18水平显著增高。发现CCL18在体外能够趋化Th2细胞和嗜碱性粒细胞。本研究的结果进一步提出了CCL18参与过敏性疾病发病的科学依据。
    2.柴油废气颗粒(DEP)能够通过IL-13在转录及翻译水平显著提高CCL18在非过敏者PBMC中的表达,从而导致Th2细胞的募集。更进一步表明CCL18可能在过敏性疾病产生的最初始时期发挥致病作用。
    3.通过对CCL5利用CCR5在SCID小鼠模型中募集炎症细胞的实验,证明了该模型应用于趋化因子研究中的可行性。利用人体化SCID小鼠模型研究的结果表明CCL18能够趋化单核/巨噬细胞、Th2细胞、原态和记忆T细胞。进一步通过体内实验证明了CCL18能通过募集Th2细胞为主的炎症细胞来促进过敏性疾病的发生发展。
    4.构建了人CCL18及连有人或小鼠IgG1 Fc片段的CCL18的真核表达载体。经扩增表达及克隆选择后获得在无血清条件下高效稳定表达的细胞克隆。为在真核系统中克隆CCL18受体奠定了基础。总之,本研究首次发现CCL18参与了过敏性哮喘的发病,并首次揭示了CCL18在过敏性哮喘中的细胞来源及在过敏性疾病发病机制中的重要作用。表明CCL18可能是治疗过敏性疾病的潜在分子靶点。
Chemokines are a group of small, mostly basic, structurally relatedmolecules that regulate cell trafficking of various types of leukocytes. Theinterreaction between chemokines and their related receptors controls the celltrafficking of immunocytes between circulatory system and tissues or organs,and play important roles in homeostasis or pathological conditions.Chemokines and cytokines together form an axis of mediating the trend ofimmune response induced by allergens.
    Numerous studies have shown that allergic diseases are Th2 lymphocyte-mediated inflammatory disorders. The involvement of some chemokines thatfavor Th2 cell recruitment leads to more Th2 cell trafficking to theinflammatory location and aggravates the imbalance of immune response.CCL18 is a chemokine recently cloned, preferentially expressed in lung tissueand induced by Th2-type cytokines like IL-4, IL-13 and IL-10, suggestingCCL18 may participate in the pathogenesis of allergic diseases especiallyallergic asthma. However, hereto few studies have been reported exceptseveral about allergic dermatitis. Therefore, we explored the roles of CCL18 in
    the pathogenesis of allergic diseases.Firstly, the roles of CCL18 in the pathogenesis of allergic asthma wereinvestigated. Under the stimulation of allergen, the production of CCL18 inperipheral blood mononuclear cells (PBMC) from allergic asthmatics waselevated at both transcriptional and translational levels. There are at least twopathways involved in allergen-induced CCL18 production: one throughmonocyte stimulation by T cell-derived Th2 cytokines including IL-4 andIL-13, and one through direct pDC stimulation. These data suggest that duringthe Th2 polarization induced by allergen, CCL18 expression inantigen-presenting cells is increased. In vitro CCL18 attracted Th2 cells andbasophils but not Th1 cells or eosinophils. The fact that CCL18 inducedF-actin polymerization in Th2 cells and histamine and intracellular calciumrelease from basophils confirmed its chemotactic activity on these two types ofcells. These data suggest that CCL18 may be involved in secondary responsesinvolving Th2 cells and not only in primary immune responses as first thoughtfrom its ability to recruit naive T cells. The CCL18 levels in thebronchoalveolar lavage and serum from untreated and treated asthmaticpatients and control subjects were assayed. The results showed increasedCCL18 in the bronchoalveolar lavage from untreated asthmatic patientscompared to control subjects and asthmatics treated by inhaled corticoids.Altogether, this study shows a new function for CCL18, which is thepreferential recruitment of Th2 cells and basophils in the context of allergicasthma. Its high expression in bronchi from asthmatics suggests a predominantrole in the pathophysiology of allergic diseases.Consequently, the effect of environmental factors, particularly dieselexposure, was analysed. Diesel activation of PBMC from nonatopic subjectsinduced a late increase in CCL18 expression. These data suggest that even for
    the nonatopic subjects, the expression of Th2 cytokines and CCL18 could stillbe elevated, furthermore the trend to allergy is exhibited, and CCL18 mayparticipate in the pathogenesis in the primary phase of allergic diseases.Studies to evaluate the in vivo role of CCL18 are limited due to theabsence of a murine homologue. Therefore, a model was established in SCID(Severe Combined ImmunoDeficient) mice grafted with human skin in orderto analyse the effect of chemokines and their receptors in vivo. We evaluatedthe role of CCR5 in the CCL5 (RANTES)-induced leukocyte recruitment inthis model. CCL5 induced a significant recruitment of memory T cells,monocytes/macrophages, eosinophils and IFN-γ+ cells by the use of CCR5.Thus, the feasibility of application of this model into in vivo studies ofchemokines was confirmed. We analysed the effect of CCL18 afterintradermal injection of SCID mice. Preliminary results showd a recruitmentof monocytes/macrophages, Th2 cells, na?ve and memory T cells, whichconfirmed the in vitro chemotactic activity of CCL18. These results suggestthat CCL18 could attract the inflammatory cells including Th2 cells toaggravate the development of allergic diseases.CCL18 receptor is still unknown. Our goal was to clone this receptorusing molecules produced in eukaryotic cells. CCL18 expressing vectors and asoluble fusion protein made of CCL18 fused to the Fc portion of human ormurine IgG1 were constructed. After amplification and clone selection, cloneswith stable and high productivity were obtained. These purified molecules willbe further used for the receptor cloning from a cDNA bank.Altogether, we firstly found that CCL18 participates in the pathogenesisof allergic asthma, and firstly disclosed the cell resource of CCL18 elevationin allergic asthma as well as its important roles in the pathogenesis of allergicdiseases. Thus, CCL18 could be a potential molecule target for the effective
    treatment of allergic diseases. These research results have been accepted forpublishing in the following SCI indexed journals: The Journal of Immunology(IF: 6.486), Journal of Allergy and Clinical Immunology (IF: 7.205) and TheJournal of Investigative Dermatology (IF: 4.238).
引文
1. Holgate ST, Lack G. Improving the management of atopic disease. Arch Dis Child, 2005, 90: 826–831.
    2. Gleich GJ. The eosinophil and bronchial asthma: current understanding. J Allergy Clin Immunol, 1990, 85: 422-36.
    3. A. B. Kay. Allergy and allergic diseases. N Engl J Med, 2001, 344(1): 30-37.
    4. Standards for the diagnosis and care of patients with chronic obstructive pulmonary disease (COPD) and asthma. This official statement of the American Thoracic Society was adopted by the ATS Board of Directors, November 1986. Am Rev Respir Dis, 1987, 136:225-44.
    5. Global Initiative for Asthma: Global strategy for Asthma Management and Prevention: NHLBI/WHO Workshop Report. March 1993. National Institutes of Health/NHLBI, 1995.
    6. Oettgen HC, Geha RS. IgE in asthma and atopy: cellular and molecular connections. J Clin Invest, 1999, 104(7): 829-35.
    7. Bacharier LB, Geha RS. Molecular mechanisms of IgE regulation. J Allergy Clin Immunol, 2000,
    105: S547-58.
    8. Gardby E, Chen XJ, Lycke NY. Impaired CD40-signalling in CD19-deficient mice selectively affects Th2-dependent isotype switching. Scand J Immunol, 2001, 53: 13-23.
    9. Vercelli D. Regulation of IgE synthesis in humans. J Biol Regul Homeost Agents, 1995, 9:1-6.
    10. Postma DS, Bleecker ER, Amelung PJ, et al. Genetic susceptibility to asthma-bronchial hyperresponsiveness coinherited with a major gene for atopy. N Engl J Med. 1995, 333: 894–900.
    11. Katoh N, Kraft S, Wessendorf JH, et al. The high-affinity IgE receptor (Fcepsilon RI) blocks apoptosis in normal human monocytes. J Clin Invest, 2000, 105:183-90.
    12. Katoh S, Matsumoto N, Fukushima K, et al. Elevated chemokine levels in bronchoalveolar lavage fluid of patients with eosinophilic pneumonia. J Allergy Clin Immunol, 2000, 106:730-6.
    13. Kraft S, Novak N, Katoh N, et al. Aggregation of the high-affinity IgE receptor Fc(epsilon)RI on human monocytes and dendritic cells induces NF-kappaB activation. J Invest Dermatol, 2002, 118:830-7.
    14. Metcalfe DD, Baram D, Mekori YA. Mast cells. Physiol Rev, 1997, 77:1033-79.
    15. Okumura S, Sagara H, Fukuda T, et al. FcepsilonRI-mediated amphiregulin production by human mast cells increases mucin gene expression in epithelial cells. J Allergy Clin Immunol, 2005, 115: 272-9.
    16. Suzukawa M, Hirai K, Iikura M, et al. IgE-and FcepsilonRI-mediated migration of human basophils. Int Immunol, 2005, 17(9):1249-55.
    17. Capron M, Jouault T, Prin L, et al. Functional study of a monoclonal antibody to IgE Fc receptor (Fc epsilon R2) of eosinophils, platelets, and macrophages. J Exp Med, 1986, 164: 72-89.
    18. Tsicopoulos A, Joseph M. The role of CD23 in allergic disease. Clin Exp Allergy, 2000, 30: 602-5.
    19. Aubry JP, Pochon S, Graber P, et al. CD21 is a ligand for CD23 and regulates IgE production. Nature, 1992, 358: 505-7.
    20. Delespesse G, Sarfati M. An update on human CD23 (Fc epsilon RII). Fc epsilon RII and IgE-BFs (soluble CD23) play an essential role in the regulation of human IgE synthesis. Clin Exp Allergy, 1991, 21 Suppl 1:153-61.
    21. Armitage RJ, Goff LK, Beverley PC. Expression and functional role of CD23 on T cells. Eur J Immunol, 1989, 19:31-5.
    22. Gordon J, Cairns JA, Millsum MJ, et al. Interleukin 4 and soluble CD23 as progression factors for human B lymphocytes: analysis of their interactions with agonists of the phosphoinositide "dual pathway" of signalling. Eur J Immunol, 1988, 18:1561-5.
    23. Gosset P, Tillie-Leblond I, Oudin S, et al. Production of chemokines and proinflammatory and antiinflammatory cytokines by human alveolar macrophages activated by IgE receptors. J Allergy Clin Immunol, 1999, 103: 289-97.
    24. Yamaguchi, M, et al. IgE enhances mouse mast cell FceRI expression in vitro and in vivo. Evidence for a novel amplification mechanism for IgE-dependent reactions. J Exp Med, 1997, 185: 663–672.
    25. Kisselgof A.B, Oettgen H C. The expression of murine B cell CD23, in vivo, is regulated by its ligand, IgE. Int. Immunol, 1998, 10: 1377–1384.
    26. Lafaille JJ. The role of helper T cell subsets in autoimmune diseases. Cytokine Growth Factor Rev, 1998, 9:139-151.
    27. Moser M, Murphy KM. Dendritic cell regulation of TH1-TH2 development. Nat Immunol 2000, 1: 199-205.
    28. Zhai Y, Ghobrial RM, Busuttil RW, et al. Th1 and Th2 cytokines in organ transplantation: paradigm lost? Crit Rev Immunol, 1999, 19:155-72.
    29. Groux H, Sornasse T, Cottrez F, et al. Induction of human T helper cell type 1 differentiation results in loss of IFN-gamma receptor beta-chain expression. J Immunol, 1997, 158:5627-31.
    30. Pernis A, Gupta S, Gollob KJ, et al. Lack of interferon gamma receptor beta chain and the prevention of interferon gamma signaling in TH1 cells. Science, 1995, 269: 245-7.
    31. Annunziato F, Manetti R, Tomasevic I, et al. Expression and release of LAG-3-encoded protein by human CD4+ T cells are associated with IFN-gamma production. Faseb J, 1996, 10: 769-76.
    32. Del Prete G, Maggi E, Pizzolo G, et al. CD30, Th2 cytokines and HIV infection: a complex and fascinating link. Immunol Today, 1995, 16:76-80.
    33. Nagata K, Tanaka K, Ogawa K, et al. Selective expression of a novel surface molecule by human Th2 cells in vivo. J Immunol, 1999, 162:1278-86.
    34. Kaplan MH, Schindler U, Smiley ST, et al. Stat6 is required for mediating responses to IL-4 and for development of Th2 cells. Immunity, 1996, 4: 313-9.
    35. Kaplan MH, Sun YL, Hoey T, et al. Impaired IL-12 responses and enhanced development of Th2 cells in Stat4-deficient mice. Nature, 1996, 382: 174-7.
    36. Shimoda K, van Deursen J, Sangster MY, et al. Lack of IL-4-induced Th2 response and IgE class switching in mice with disrupted Stat6 gene. Nature, 1996, 380:630-3.
    37. Takeda K, Tanaka T, Shi W, et al. Essential role of Stat6 in IL-4 signalling. Nature, 1996, 380: 627-30.
    38. Thierfelder WE, van Deursen JM, Yamamoto K, et al. Requirement for Stat4 in interleukin-12-mediated responses of natural killer and T cells. Nature, 1996, 382: 171-4.
    39. Finkelman FD, Morris SC, Orekhova T, et al. Stat6 regulation of in vivo IL-4 responses. J Immunol, 2000, 164: 2303-10.
    40. Zhu J, Guo L, Watson CJ, et al. Stat6 is necessary and sufficient for IL-4's role in Th2 differentiation and cell expansion. J Immunol, 2001, 166: 7276-81.
    41. Zhang S, Lukacs NW, Lawless VA, et al. Cutting edge: differential expression of chemokines in Th1 and Th2 cells is dependent on Stat6 but not Stat4. J Immunol, 2000, 165: 10-4.
    42. Glimcher LH, Murphy KM. Lineage commitment in the immune system: the T helper lymphocyte grows up. Genes Dev, 2000, 14: 1693–1711.
    43. Lee GR, Fields PE, Flavell RA. Regulation of IL-4 gene expression by distal regulatory elements and GATA-3 at the chromatin level. Immunity, 2001, 14: 447-59.
    44. Szabo SJ, Sullivan BM, Stemmann C, et al. Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells. Science, 2002, 295: 338-42.
    45. Kidd P, Th1/Th2 Balance: the hypothesis, its limitations, and implications for health and disease. Alternative Medicine Review, 2003, 8: 223-46.
    46. Ying S, Humbert M, Barkans J, et al. Expression of IL-4 and IL-5 mRNA and protein product by CD4+ and CD8+ T cells, eosinophils, and mast cells in bronchial biopsies obtained from atopic and nonatopic (intrinsic) asthmatics. J Immunol, 1997, 158: 3539-44.
    47. Larché M, Robinson DS, Kay AB. The role of T lymphocytes in the pathogenesis of asthma. J Allergy Clin Immunol, 2003, 11(3): 450-463.
    48. Epstein MM. Targeting memory Th2 cells for the treatment of allergic asthma. Pharmacology & Therapeutics, 2006, 109: 107– 136.
    49. Neurath MF, Finotto S, Glimcher LH. The role of Th1/Th2 polarization in mucosal immunity. Nature Medicine, 2002, 8: 567-73.
    50. Finotto S, Neurath MF, Glickman JN, et al. Development of spontaneous airway changes consistent with human asthma in mice lacking T-bet. Science, 2002, 295: 336–338.
    51. Kim TS, DeKruyff RH, Rupper R, et al. An ovalbumin-IL-12 fusion protein is more effective than ovalbumin plus free recombinant IL-12 in inducing a T helper cell type 1-dominated immune response and inhibiting antigen-specific IgE production. J Immunol, 1997, 158: 4137-44.
    52. Hansen G, McIntire JJ, Yeung VP, et al. CD4(+) T helper cells engineered to produce latent TGF-beta1 reverse allergen-induced airway hyperreactivity and inflammation. J Clin Invest, 2000, 105: 61-70.
    53. Hansen G, Berry G, DeKruyff RH, et al. Allergen-specific Th1 cells fail to counterbalance Th2 cell-induced airway hyperreactivity but cause severe airway inflammation. J Clin Invest, 1999, 103: 175-83.
    54. Randolph DA, Carruthers CJ, Szabo SJ, et al. Modulation of airway inflammation by passive transfer of allergen-specific Th1 and Th2 cells in a mouse model of asthma. J Immunol, 1999, 162: 2375-83.
    55. Cui J, Pazdziorko S, Miyashiro JS, et al. TH1-mediated airway hyperresponsiveness independent of neutrophilic inflammation. J Allergy Clin Immunol 2005;115:309-15.
    56. Irifune K, Yokoyama A, Sakai K, et al. Adoptive transfer of T-helper cell type 1 clones attenuates an asthmatic phenotype in mice. Eur Respir J, 2005, 25: 653-9.
    57. Akdis M, Blaser K, Akdis CA. T regulatory cells in allergy: Novel concepts in the pathogenesis, prevention, and treatment of allergic diseases. J Allergy Clin Immunol, 2005, 116: 961-8.
    58. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor foxp3. Science, 2003, 299: 1057-61.
    59. Von Boehmer H. Mechanisms of suppression by suppressor T cells. Nat Immunol, 2005, 6: 338-44.
    60. Nakamura K, Kitani A, Strober W. Cell contact-dependent immunosuppression by CD4+CD25+ regulatory T cells is mediated by cell surface-bound transforming growth factor beta. J Exp Med, 2001, 194: 629-44.
    61. Seroogy CM, Gern JE. The role of T regulatory cells in asthma. J Allergy Clin Immunol, 2005, 116: 996-9.
    62. Hawrylowicz CM, Garra AO. Potential role of interleukin-10-secreting regulatory t cells in allergy and asthma. Nat Rev Immunol, 2005, 5: 271-283.
    63. Ling EM et al. Relation of CD4+CD25+ regulatory T-cell suppression of allergen-driven T-cell activation to atopic status and expression of allergic disease. Lancet, 2004, 363: 608–615.
    64. Lewkowich IP, Herman NS, Schleifer KW, et al. CD4+CD25+T cells protect against experimentally induced asthma and alter pulmonary dendritic cell phenotype and function. J Exp Med, 2005, 202: 1549-61.
    65. Suto, A. Nakajima H, Kagami S, et al. Role of CD4+ CD25+ regulatory T cells in T helper 2 cell-mediated allergic inflammation in the airways. Am J Respir Crit Care Med, 2001, 164: 680–687.
    66. Jaffar Z, Sivakuru T, Roberts K. CD4+CD25+ T cells regulate airway eosinophilic inflammation by modulating the TH2 cell phenotype. J Immunol, 2004, 172 : 3842–3849.
    67. Curotto de Lafaille, MA, et al. Hyper immunoglobulin E response in mice with monoclonal populations of B and T lymphocytes. J Exp Med, 2001, 194: 1349–1359.
    68. Ostroukhova M, et al. Tolerance induced by inhaled antigen involves CD4+ T cells expressing membrane-bound TGF-β and FOXP3. J Clin Invest, 2004, 114: 28–38.
    69. Akbari O, DeKruyff RH, Umetsu DT. Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen. Nature Immunol, 2001, 2: 725–731.
    70. Yssel H, Johnson KE, Schneider PV, et al. T cell activation-inducing epitopes of the house dust mite allergen Der p I. Proliferation and lymphokine production patterns by Der p I-specific CD4+ T cell clones. J Immunol, 1992, 148: 738-45.
    71. Ding L, Linsley PS, Huang LY, et al. IL-10 inhibits macrophage costimulatory activity by selectively inhibiting the up-regulation of B7 expression. J Immunol, 1993, 151: 1224-34.
    72. Kubin M, Chow JM, Trinchieri G. Differential regulation of interleukin-12 (IL-12), tumor necrosis factor alpha, and IL-1 beta production in human myeloid leukemia cell lines and peripheral blood mononuclear cells. Blood, 1994, 83: 1847-55.
    73. D'Andrea A, Aste-Amezaga M, Valiante NM, et al. Interleukin 10 (IL-10) inhibits human lymphocyte interferon gamma-production by suppressing natural killer cell stimulatory factor/IL-12 synthesis in accessory cells. J Exp Med, 1993, 178: 1041-8.
    74. Ralph P, Nakoinz I, Sampson-Johannes A, et al. IL-10, T lymphocyte inhibitor of human blood cell production of IL-1 and tumor necrosis factor. J Immunol, 1992, 148: 808-14.
    75. Kopydlowski KM, Salkowski CA, Cody MJ, et al. Regulation of macrophage chemokine expression by lipopolysaccharide in vitro and in vivo. J Immunol, 1999, 163: 1537-44.
    76. Macatonia SE, Doherty TM, Knight SC, et al. Differential effect of IL-10 on dendritic cell-induced T cell proliferation and IFN-gamma production. J Immunol ,1993, 150: 3755-65.
    77. Rissoan MC, Soumelis V, Kadowaki N, et al. Reciprocal control of T helper cell and dendritic cell differentiation. Science, 1999, 283: 1183-6.
    78. Burdin N, Van Kooten C, Galibert L, et al. Endogenous IL-6 and IL-10 contribute to the differentiation of CD40-activated human B lymphocytes. J Immunol, 1995, 154: 2533-44.
    79. Satoguina JS, Weyand E, Larbi J, et al. T regulatory-1 cells induce IgG4 production by B cells: role of IL-10. J Immunol, 2005, 174: 4718-26.
    80. Schandene L, Alonso-Vega C, Willems F, et al. B7/ CD28-dependent IL-5 production by human resting T cells is inhibited by IL-10. J Immunol, 1994, 152: 4368-74.
    81. Taga K, Cherney B, Tosato G. IL-10 inhibits apoptotic cell death in human T cells starved of IL-2. Int Immunol, 1993, 5: 1599-608.
    82. Makela MJ, Kanehiro A, Borish L, et al. IL-10 is necessary for the expression of airway hyperresponsiveness but not pulmonary inflammation after allergic sensitization. Proc Natl Acad Sci U S A, 2000, 97: 6007-12.
    83. Zuany-Amorim C, Haile S, Leduc D, et al. Interleukin-10 inhibits antigen-induced cellular recruitment into the airways of sensitized mice. J Clin Invest, 1995, 95: 2644-51.
    84. Quinn TJ, Taylor S, Wohlford-Lenane CL, et al. IL-10 reduces grain dust-induced airway inflammation and airway hyperreactivity. J Appl Physiol, 2000, 88: 173-9.
    85. Akbari O, et al. Antigen-specific regulatory T cells develop via the ICOS– ICOS-ligand pathway and inhibit allergeninduced airway hyperreactivity. Nature Med, 2002, 8: 1024–1032.
    86. Akdis, M. et al. Immune responses in healthy and allergic individuals are characterized by a fine balance between allergen-specific T regulatory 1 and T helper 2 cells. J Exp Med, 2004, 199, 1567–1575.
    87. Morgan DA, Ruscetti FW, Gallo R. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science, 1976, 193: 1007-8.
    88. Lenardo MJ. Interleukin-2 programs mouse alpha beta T lymphocytes for apoptosis. Nature, 1991, 353: 858-61.
    89. Granucci F, Vizzardelli C, Pavelka N, et al. Inducible IL-2 production by dendritic cells revealed by global gene expression analysis. Nat Immunol, 2001, 2: 882-8.
    90. Granucci F, Vizzardelli C, Virzi E, et al. Transcriptional reprogramming of dendritic cells by differentiation stimuli. Eur J Immunol, 2001, 31: 2539-46.
    91. Germann T, Gately MK, Schoenhaut DS, et al. Interleukin-12/T cell stimulating factor, a cytokine with multiple effects on T helper type 1 (Th1) but not on Th2 cells. Eur J Immunol, 1993, 23: 1762-70.
    92. Seder RA, Gazzinelli R, Sher A, et al. Interleukin 12 acts directly on CD4+ T cells to enhance priming for interferon gamma production and diminishes interleukin 4 inhibition of such priming. Proc Natl Acad Sci U S A , 1993, 90: 10188-92.
    93. Morris SC, Madden KB, Adamovicz JJ, et al. Effects of IL-12 on in vivo cytokine gene expression and Ig isotype selection. J Immunol, 1994, 152: 1047-56.
    94. Tang C, Rolland JM, Ward C, et al. Modulatory effects of alveolar macrophages on CD4+ T-cell IL-5 responses correlate with IL-1beta, IL-6, and IL-12 production. Eur Respir J, 1999, 14: 106-12.
    95. Gately MK, Renzetti LM, Magram J, et al. The interleukin-12 /interleukin-12-receptor system: role in normal and pathologic immune responses. Annu Rev Immunol, 1998, 16: 495-521.
    96. Kips JC, Brusselle GJ, Joos GF, et al. Interleukin-12 inhibits antigen-induced airway hyperresponsiveness in mice. Am J Respir Crit Care Med, 1996, 153: 535-9.
    97. Gavett SH, O'Hearn DJ, Li X, et al. Interleukin 12 inhibits antigen-induced airway hyperresponsiveness, inflammation, and Th2 cytokine expression in mice. J Exp Med, 1995, 182: 1527-36.
    98. Bryan SA, O'Connor BJ, Matti S, et al. Effects of recombinant human interleukin-12 on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet, 2000, 356: 2149-53.
    99. Bancroft GJ, Sheehan KC, Schreiber RD, et al. Tumor necrosis factor is involved in the T cell-independent pathway of macrophage activation in scid mice. J Immunol, 1989, 143: 127-30.
    100. Zlotnik A, Shimonkevitz RP, Gefter ML, et al. Characterization of the gamma-interferon-mediated induction of antigen-presenting ability in P388D1 cells. J Immunol,1983, 131: 2814-20.
    101. Dalton DK, Pitts-Meek S, Keshav S, et al. Multiple defects of immune cell function in mice with disrupted interferon-gamma genes. Science, 1993, 259: 1739-42.
    102. Pernis A, Gupta S, Gollob KJ, et al. Lack of interferon gamma receptor beta chain and the prevention of interferon gamma signaling in TH1 cells. Science, 1995, 269: 245-7.
    103. Elser B, Lohoff M, Kock S, et al. IFN-gamma represses IL-4 expression via IRF-1 and IRF-2. Immunity, 2002, 17: 703-12.
    104. Iwamoto I, Nakajima H, Endo H, et al. Interferon gamma regulates antigen-induced eosinophil recruitment into the mouse airways by inhibiting the infiltration of CD4+ T cells. J Exp Med, 1993, 177: 573-6.
    105. Coyle AJ, Tsuyuki S, Bertrand C, et al. Mice lacking the IFN-gamma receptor have impaired ability to resolve a lung eosinophilic inflammatory response associated with a prolonged capacity of T cells to exhibit a Th2 cytokine profile. J Immunol, 1996, 156: 2680-5.
    106. Xu D, Trajkovic V, Hunter D, et al. IL-18 induces the differentiation of Th1 or Th2 cells depending upon cytokine milieu and genetic background. Eur J Immunol, 2000, 30: 3147-56.
    107. Yoshimoto T, Min B, Sugimoto T, et al. Nonredundant roles for CD1d-restricted natural killer T cells and conventional CD4+ T cells in the induction of immunoglobulin E antibodies in response to interleukin 18 treatment of mice. J Exp Med, 2003, 197: 997-1005.
    108. Yoshimoto T, Okada K, Morishima N, et al. Induction of IgG2a class switching in B cells by IL-27. J Immunol, 2004, 173: 2479-85.
    109. Gurney AL. IL-22, a Th1 cytokine that targets the pancreas and select other peripheral tissues. Int Immunopharmacol, 2004, 4: 669-77.
    110. Howard M, Farrar J, Hilfiker M, et al. Identification of a T cell-derived b cell growth factor distinct from interleukin 2. J Exp Med. 1982, 155: 914-23.
    111. Lebman DA, Coffman RL. Interleukin 4 causes isotype switching to IgE in T cell-stimulated clonal B cell cultures. J Exp Med, 1988, 168: 853-62.
    112. Fernandez-Botran R, Sanders VM, Oliver KG, et al. Interleukin 4 mediates autocrine growth of helper T cells after antigenic stimulation. Proc Natl Acad Sci U S A, 1986, 83: 9689-93.
    113. Vella A, Teague TK, Ihle J, et al. Interleukin 4 (IL-4) or IL-7 prevents the death of resting T cells: stat6 is probably not required for the effect of IL-4. J Exp Med, 1997, 186: 325-30.
    114. Fiset PO, Soussi-Gounni A, Christodoulopoulos P, et al. Modulation of allergic response in nasal mucosa by antisense oligodeoxynucleotides for IL-4. J Allergy Clin Immunol, 2003, 111: 580-6.
    115. Zuany-Amorim C, Ruffie C, Haile S, et al. Requirement for gammadelta T cells in allergic airway inflammation. Science, 1998, 280: 1265-7.
    116. Xiao T, Fujita H, Saeki H, et al. Thymus and activation-regulated chemokine (TARC/CCL17) produced by mouse epidermal Langerhans cells is upregulated by TNF-alpha and IL-4 and downregulated by IFN-gamma. Cytokine, 2003, 23: 126-32.
    117. Yamashita U, Kuroda E. Regulation of macrophage-derived chemokine (MDC, CCL22) production. Crit Rev Immunol, 2002, 22: 105-14.
    118. Schleimer RP, Sterbinsky SA, Kaiser J, et al. IL-4 induces adherence of human eosinophils and basophils but not neutrophils to endothelium. Association with expression of VCAM-1. J Immunol, 1992, 148: 1086-92.
    119. Hsieh FH, Lam BK, Penrose JF, et al. T helper cell type 2 cytokines coordinately regulate immunoglobulin E-dependent cysteinyl leukotriene production by human cord blood-derived mast cells: profound induction of leukotriene C(4) synthase expression by interleukin 4. J Exp Med, 2001, 193: 123-33.
    120. Mosmann TR, Bond MW, Coffman RL, et al. T-cell and mast cell lines respond to B-cell stimulatory factor 1. Proc Natl Acad Sci U S A, 1986, 83: 5654-8.
    121. Hsieh CS, Heimberger AB, Gold JS, et al. Differential regulation of T helper phenotype development by interleukins 4 and 10 in an alpha beta T-cell-receptor transgenic system. Proc Natl Acad Sci U S A, 1992, 89: 6065-9.
    122. Dabbagh K, Takeyama K, Lee HM, et al. IL-4 induces mucin gene expression and goblet cell metaplasia in vitro and in vivo. J Immunol, 1999, 162: 6233-7.
    123. Robinson DS, Hamid Q, Ying S, et al. Predominant TH2-like bronchoalveolar T-lymphocyte population in atopic asthma. N Engl J Med, 1992, 326: 298-304.
    124. Kopf M, Le Gros G, Bachmann M, et al. Disruption of the murine IL-4 gene blocks Th2 cytokine responses. Nature, 1993, 362: 245-8.
    125. Henderson WR, Chi EY, Maliszewski CR. Soluble IL-4 receptor inhibits airway inflammation following allergen challenge in a mouse model of asthma. J Immunol, 2000, 164: 1086-95.
    126. Rankin JA, Picarella DE, Geba GP, et al. Phenotypic and physiologic characterization of transgenic mice expressing interleukin 4 in the lung: lymphocytic and eosinophilic inflammation without airway hyperreactivity. Proc Natl Acad Sci U S A, 1996, 93: 7821-5.
    127. Bradding P, Feather IH, Wilson S, et al. Immunolocalization of cytokines in the nasal mucosa of normal and perennial rhinitic subjects. The mast cell as a source of IL-4, IL-5, and IL-6 in human allergic mucosal inflammation. J Immunol, 1993, 151: 3853-65.
    128. Prussin C, Metcalfe DD. IgE, mast cells, basophils, and eosinophils. J Allergy Clin Immunol, 2003, 111: S486-94.
    129. Croft M, Swain SL. Recently activated naive CD4 T cells can help resting B cells, and can produce sufficient autocrine IL-4 to drive differentiation to secretion of T helper 2-type cytokines. J Immunol, 1995, 154: 4269-82.
    130. Wardlaw AJ. Molecular basis for selective eosinophil trafficking in asthma: A multistep paradigm. J Allergy Clin Immunol, 1999, 104: 917-26.
    131. Lopez AF, Sanderson CJ, Gamble JR, et al. Recombinant human interleukin 5 is a selective activator of human eosinophil function. J Exp Med, 1988, 167: 219-24.
    132. Kagami S, Nakajima H, Kumano K, et al. Both stat5a and stat5b are required for antigen-induced eosinophil and T-cell recruitment into the tissue. Blood, 2000, 95: 1370-7.
    133. Karras JG, McGraw K, McKay RA, et al. Inhibition of antigen-induced eosinophilia and late phase airway hyperresponsiveness by an IL-5 antisense oligonucleotide in mouse models of asthma. J Immunol, 2000, 164: 5409-15.
    134. Foster PS, Hogan SP, Ramsay AJ, et al. Interleukin 5 deficiency abolishes eosinophilia, airways hyperreactivity, and lung damage in a mouse asthma model. J Exp Med, 1996, 183: 195-201.
    135. Lee JJ, McGarry MP, Farmer SC, et al. Interleukin-5 expression in the lung epithelium of transgenic mice leads to pulmonary changes pathognomonic of asthma. J Exp Med, 1997, 185: 2143-56.
    136. Corry DB, Folkesson HG, Warnock ML, et al. Interleukin 4, but not interleukin 5 or eosinophils, is required in a murine model of acute airway hyperreactivity. J Exp Med, 1996, 183: 109-17.
    137. Leckie MJ, ten Brinke A, Khan J, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet, 2000, 356: 2144-8.
    138. de Waal Malefyt R, Abrams JS, Zurawski SM, et al. Differential regulation of IL-13 and IL-4 production by human CD8+ and CD4+ Th0, Th1 and Th2 T cell clones and EBV-transformed B cells. Int Immunol, 1995, 7: 1405-16.
    139. T cells: modulation by interleukin-4 and interleukin-12. Eur J Immunol, 1996, 26: 571-7.
    140. Akbari O, Stock P, Meyer E, et al. Essential role of NKT cells producing IL-4 and IL-13 in the development of allergen-induced airway hyperreactivity. Nat Med, 2003, 9: 582-8.
    141. Schmid-Grendelmeier P, Altznauer F, Fischer B, et al. Eosinophils express functional IL-13 in eosinophilic inflammatory diseases. J Immunol, 2002, 169: 1021-7.
    142. Grunstein MM, Hakonarson H, Leiter J, et al. IL-13-dependent autocrine signaling mediates altered responsiveness of IgE-sensitized airway smooth muscle. Am J Physiol Lung Cell Mol Physiol, 2002, 282: L520-8.
    143. Zurawski SM, Vega F Jr, Huyghe B, et al. Receptors for interleukin-13 and interleukin-4 are complex and share a novel component that functions in signal transduction. EMBO J, 1993, 12: 2663-70.
    144. Grunig G, Warnock M, Wakil AE, et al. Requirement for IL-13 independently of IL-4 in experimental asthma. Science, 1998, 282: 2261-3.
    145. Wills-Karp M, Luyimbazi J, Xu X, et al. Interleukin-13: central mediator of allergic asthma. Science, 1998, 282: 2258-61.
    146. Li L, Xia Y, Nguyen A, et al. Effects of Th2 cytokines on chemokine expression in the lung: IL-13 potently induces eotaxin expression by airway epithelial cells. J Immunol, 1999, 162: 2477-87.
    147. Zhu Z, Homer RJ, Wang Z, et al. Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J Clin Invest, 1999, 103: 779-88.
    148. Kuperman D, Schofield B, Wills-Karp M, et al. Signal transducer and activator of transcription factor 6 (Stat6)-deficient mice are protected from antigen-induced airway hyperresponsiveness and mucus production. J Exp Med, 1998, 187: 939-48.
    149. Tomkinson A, Duez C, Cieslewicz G, et al. A murine IL-4 receptor antagonist that inhibits IL-4-and IL-13-induced responses prevents antigen-induced airway eosinophilia and airway hyperresponsiveness. J Immunol, 2001, 166: 5792-800.
    150. Punnonen J, Yssel H, de Vries JE. The relative contribution of IL-4 and IL-13 to human IgE synthesis induced by activated CD4+ or CD8+ T cells. J Allergy Clin Immunol, 1997, 100: 792-801.
    151. Hesse M, Modolell M, La Flamme AC, et al. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of L-arginine metabolism. J Immunol, 2001, 167: 6533-44.
    152. Lee CG, Homer RJ, Zhu Z, et al. Interleukin-13 induces tissue fibrosis by selectively stimulating and activating transforming growth factor beta(1). J Exp Med, 2001, 194: 809-21.
    153. Wills-Karp M, Interleukin-13 in asthma pathogenesis. Immunol Rev, 2004, 202: 175-90.
    154. Ohshima Y, Yasutomi M, Omata N, et al. Dysregulation of IL-13 production by cord blood CD4+ T cells is associated with the subsequent development of atopic disease in infants. Pediatr Res, 2002, 51: 195-200.
    155. Renauld JC, Goethals A, Houssiau F, et al. Human P40 / IL-9. Expression in activated CD4+ T cells, genomic organization, and comparison with the mouse gene. J Immunol, 1990, 144: 4235-41.
    156. Hultner L, Druez C, Moeller J, et al. Mast cell growth-enhancing activity (MEA) is structurally related and functionally identical to the novel mouse T cell growth factor P40/TCGFIII (interleukin 9). Eur J Immunol, 1990, 20: 1413-6.
    157. Gounni AS, Gregory B, Nutku E, et al. Interleukin-9 enhances interleukin-5 receptor expression, differentiation, and survival of human eosinophils. Blood, 2000, 96: 2163-71.
    158. Tsicopoulos A, Shimbara A, de Nadai P, et al. Involvement of IL-9 in the bronchial phenotype of patients with nasal polyposis. J Allergy Clin Immunol, 2004, 113: 462-9.
    159. Fort MM, Cheung J, Yen D, et al. IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologies in vivo. Immunity, 2001, 15: 985-95.
    160. Liao SC, Cheng YC, Wang YC, et al. IL-19 induced Th2 cytokines and was up-regulated in asthma patients. J Immunol, 2004, 173: 6712-8.
    161. Gallagher G, Eskdale J, Jordan W, et al. Human interleukin-19 and its receptor: a potential role in the induction of Th2 responses. Int Immunopharmacol, 2004, 4: 615-26.
    162. Campbell JJ, Hedrick J, Zlotnik A, et al. Chemokines and the arrest of lymphocytes rolling under flow conditions. Science, 1998, 279: 381-4.
    163. Hoogewerf AJ, Kuschert GS, Proudfoot AE, et al. Glycosaminoglycans mediate cell surface oligomerization of chemokines. Biochemistry, 1997, 36: 13570-8.
    164. Cascieri MA, Springer MS. The chemokine/chemokine-receptor family: potential and progress for therapeutic intervention. Curr Opin Chem Biol, 2000, 4: 420-7.
    165. Yoshie O, Imai T, Nomiyama Chemokines in immunity. Adv Immunol, 2001, 78: 57-110.
    166. Panina-Bordignon P, Papi A, Mariani M, et al. The C-C chemokine receptors CCR4 and CCR8 identify airway T cells of allergen-challenged atopic asthmatics. J Clin Invest, 2001, 107: 1357-64.
    167. Pease JE, Weller CL, Williams TJ. Regulation of eosinophil trafficking in asthma and allergy. Ernst Schering Res Found Workshop, 2004, 45: 85-100.
    168. Sallusto F, Lanzavecchia A, Mackay CR. Chemokines and chemokine receptors in T-cell priming and Th1/Th2-mediated responses. Immunol Today, 1998, 19: 568-74.
    169. Oppenheim JJ, Zachariae CO, et al. Properties of the novel proinflammatory supergene "intercrine" cytokine family. Annu Rev Immunol, 1991, 9: 617-48.
    170. Rossi DL, Vicari AP, et al. Identification through bioinformatics of two new macrophage proinflammatory human chemokines: MIP-3alpha and MIP-3beta. J Immunol, 1997, 158: 1033-6.
    171. Bazan JF, Bacon KB, et al. A new class of membrane-bound chemokine with a CX3C motif. Nature, 1997, 385: 640-4.
    172. Proost P, Wuyts A, et al. Human monocyte chemotactic proteins-2 and -3: structural and functional comparison with MCP-1. J Leukoc Biol, 1996, 59: 67-74.
    173. Stellato C, Matsukura S, et al. Differential regulation of epithelial-derived C-C chemokine expression by IL-4 and the glucocorticoid budesonide. J Immunol, 1999, 163: 5624-32.
    174. Kayisli UA, Mahutte NG, et al. Uterine chemokines in reproductive physiology and pathology. Am J Reprod Immunol, 2002, 47: 213-21.
    175. Lilly, CM, Nakamura H, et al. Expression of eotaxin by human lung epithelial cells: induction by cytokines and inhibition by glucocorticoids. J Clin Invest, 1997, 99: 1767-73.
    176. Kaplan MH, Schindler U, et al. Stat6 is required for mediating responses to IL-4 and for development of Th2 cells. Immunity, 1996, 4: 313-9.
    177. Takeda K, Kamanaka M, et al. Impaired IL-13-mediated functions of macrophages in STAT6-deficient mice. J Immunol 1996, 157: 3220-2.
    178. Mattes J, Yang M, et al. IL-13 induces airways hyperreactivity independently of the IL-4R alpha chain in the allergic lung. J Immunol, 2001, 167: 1683-92.
    179. Akimoto T, Numata F, et al. Abrogation of bronchial eosinophilic inflammation and airway hyperreactivity in signal transducers and activators of transcription (STAT)6-deficient mice. J Exp Med, 1998,187: 1537-42.
    180. Zimmermann N, Hogan SP, et al. Murine eotaxin-2: a constitutive eosinophil chemokine induced by allergen challenge and IL-4 overexpression. J Immunol, 2000, 165: 5839-46.
    181. Keane-Myers A, Gause WC, et al. B7-CD28/CTLA-4 costimulatory pathways are required for the development of T helper cell 2-mediated allergic airway responses to inhaled antigens. J Immunol, 1997, 158: 2042-9.
    182. Oosterwegel MA, Mandelbrot DA, et al. The role of CTLA-4 in regulating Th2 differentiation. J Immunol, 1999, 163: 2634-9.
    183. Hidi R, Riches V, et al. Role of B7-CD28/CTLA-4 costimulation and NF-kappa B in allergen-induced T cell chemotaxis by IL-16 and RANTES. J Immunol, 2000, 164: 412-8.
    184. Padrid PA, Mathur M, et al. CTLA4Ig inhibits airway eosinophilia and hyperresponsiveness by regulating the development of Th1/Th2 subsets in a murine model of asthma. Am J Respir Cell Mol Biol, 1998, 18(4): 453-62.
    185. Murphy PM. The molecular biology of leukocyte chemoattractant receptors. Annu Rev Immunol, 1994, 12: 593-633.
    186. Bokoch GM. Chemoattractant signaling and leukocyte activation. Blood, 1995, 86: 1649-60.
    187. Bacon KB, Szabo MC, et al. RANTES induces tyrosine kinase activity of stably complexed p125FAK and ZAP-70 in human T cells. J Exp Med, 1996, 184: 873-82.
    188. Mellado M, Rodriguez-Frade JM, et al. The chemokine monocyte chemotactic protein 1 triggers Janus kinase 2 activation and tyrosine phosphorylation of the CCR2B receptor. J Immunol, 1998, 161: 805-13.
    189. Kampen, GT, Stafford S, et al. Eotaxin induces degranulation and chemotaxis of eosinophils through the activation of ERK2 and p38 mitogen-activated protein kinases. Blood, 2000, 95: 1911-7.
    190. Uguccioni M, D'Apuzzo M, et al. Actions of the chemotactic cytokines MCP-1, MCP-2, MCP-3, RANTES, MIP-1 alpha and MIP-1 beta on human monocytes. Eur J Immunol, 1995, 25: 64-8.
    191. Loetscher P, Pellegrino A, Gong JH, et al. The ligands of CXC chemokine receptor 3, I-TAC, Mig, and IP10, are natural antagonists for CCR3. J Biol Chem, 2001, 276: 2986-91.
    192. Weng Y, Siciliano SJ, Waldburger KE, et al. Binding and functional properties of recombinant and endogenous CXCR3 chemokine receptors. J Biol Chem, 1998, 273: 18288-91.
    193. Ogilvie P, Bardi G, Clark-Lewis I, et al. Eotaxin is a natural antagonist for CCR2 and an agonist for CCR5. Blood, 2001, 97: 1920-1924.
    194. Ogilvie P, Paoletti S, Clark-Lewis I, et al. Eotaxin-3 is a natural antagonist for CCR2 and exerts a repulsive effect on human monocytes. Blood, 2003, 102: 789-94.
    195. Kaplan AP. Chemokines, chemokine receptors and allergy. Int Arch Allergy Immunol 2001;124:423-31.
    196. D'Ambrosio D, Iellem A, et al. Selective up-regulation of chemokine receptors CCR4 and CCR8 upon activation of polarized human type 2 Th cells. J Immunol, 1998, 161: 5111-5.
    197. Sallusto F, Palermo B, et al. Distinct patterns and kinetics of chemokine production regulate dendritic cell function. Eur J Immunol, 1999, 29: 1617-25.
    198. Power CA, Meyer A, et al. Molecular cloning and functional expression of a novel CC chemokine receptor cDNA from a human basophilic cell line. J Biol Chem, 1995, 270: 19495-500.
    199. Sallusto F, Mackay CR, et al. Selective expression of the eotaxin receptor CCR3 by human T helper 2 cells. Science, 1997, 277: 2005-7.
    200. Hieshima K, Imai T, Baba M, et al. A novel human CC chemokine PARC that is most homologous to macrophage-inflammatory protein-1 alpha/LD78 alpha and chemotactic for T lymphocytes, but not for monocytes. J Immunol, 1997, 159: 1140-9.
    201. Adema GJ, Hartgers F, Verstraten R, et al. A dendritic-cell-derived C-C chemokine that preferentially attracts naive T cells. Nature, 1997, 387: 713-7.
    202. Kodelja V, Muller C, Politz O, et al. Alternative macrophage activation-associated CC-chemokine-1, a novel structural homologue of macrophage inflammatory protein-1 alpha with a Th2-associated expression pattern. J Immunol, 1998, 160: 1411-18.
    203. Guan P, Burghes AH, Cunningham A, et al. Genomic organization and biological characterization of the novel human CC chemokine DC-CK-1/PARC/MIP-4/SCYA18. Genomics, 1999, 56: 296-302.
    204. Wells TN, Peitsch MC. The chemokine information source: identification and characterization of novel chemokines using the WorldWideWeb and expressed sequence tag databases. J Leukoc Biol, 1997, 61: 545-50.
    205. Politz O, Kodelja V, Guillot P, et al. Pseudoexons and regulatory elements in the genomic sequence of the beta-chemokine, alternative macrophage activation-associated CC-chemokine (AMAC)-1. Cytokine, 2000, 12: 120-6.
    206. Tasaki Y, Fukuda S, Iio M, et al. Chemokine PARC gene (SCYA18) generated by fusion of two MIP-1alpha/LD78alpha-like genes. Genomics, 1999, 55: 353-7.
    207. Struyf S, Schutyser E, Gouwy M, et al. PARC/CCL18 is a plasma CC chemokine with increased levels in childhood acute lymphoblastic leukemia. Am J Pathol, 2003, 163: 2065-75.
    208. Schutyser E, Richmond A, Van Damme J. Involvement of CC chemokine ligand 18 (CCL18) in normal and pathological processes. J Leukoc Biol, 2005, 78: 14-26.
    209. Lindhout E, Vissers JLM, Hartgers FC. The dendritic cell-specific CC-chemokine DC-CK1 is expressed by germinal center dendritic cells and attracts CD38-negative mantle zone B lymphocytes. J Immunol, 2001, 166, 3284–89.
    210. Schraufstatter I, Takamori H, Sikora L, et al. Eosinophils and monocytes produce pulmonary and activation-regulated chemokine, which activates cultured monocytes/macrophages. Am J Physiol Lung Cell Mol Physiol, 2004, 286: L494-501.
    211. Pivarcsi A, Gombert M, Dieu-Nosjean MC, et al. CC chemokine ligand 18, an atopic dermatitisassociated and dendritic cell-derived chemokine, is regulated by staphylococcal products and allergen exposure. J Immunol, 2004, 173: 5810-17.
    212. Song E, Ouyang N, Horbelt M, et al. Influence of alternatively and classically activated macrophages on fibrogenic activities of human fibroblasts. Cell. Immunol, 2000, 204, 19–28.
    213. Schutyser E, Struyf S, Wuyts A, et al. Selective induction of CCL18/PARC by staphylococcal enterotoxins in mononuclear cells and enhanced levels in septic and rheumatoid arthritis. Eur J Immunol, 2001, 31, 3755–62.
    214. Struyf S, Proost P, Van Damme J. Regulation of the immune response by the interaction of chemokines and proteases. Adv. Immunol, 2003, 81: 1–44.
    215. Standiford TJ, Kunkel SL, Liebler JM, et al. Gene expression of macrophage inflammatory protein-1_ from human blood monocytes and alveolar macrophages is inhibited by interleukin-4. Am J Respir Cell Mol Biol, 1993, 9: 192–198.
    216. Berkman N, John M, Roesems G, et al. Inhibition of macrophage inflammatory protein-1α expression by IL-10. Differential sensitivities in human blood monocytes and alveolar macrophages. J Immunol, 1995, 155: 4412–18.
    217. Radstake TR, van der Voort R, Ten Brummelhuis M, et al. Increased expression of CCL18, CCL19, and CCL17 by dendritic cells from patients with rheumatoid arthritis (RA) and regulation by Fcγ receptors. Ann Rheum Dis, 2004, 64, 359–67.
    218. Vulcano M, Struyf S, Scapini P, et al. Unique regulation of CCL18 production by maturing dendritic cells. J Immunol, 2003, 170: 3843-49.
    219. De Ceuninck F, Dassencourt L, Anract P. The inflammatory side of human chondrocytes unveiled by antibody microarrays. Biochem Biophys Res Commun, 2004, 323: 960–69.
    220. Gunther C, Bello-Fernandez C, Kopp T, et al. CCL18 is expressed in atopic dermatitis and mediates skin homing of human memory T cells. J Immunol, 2005, 174: 1723-8.
    221. Atamas SP, Luzina IG, Choi J, et al. Pulmonary and activation-regulated chemokine stimulates collagen production in lung fibroblasts. Am J Respir Cell Mol Biol, 2003, 29: 743-9.
    222. Broxmeyer HE, Kim CH. Regulation of hematopoiesis in a sea of chemokine family members with a plethora of redundant activities. Exp Hematol, 1999, 27: 1113-23.
    223. Nibbs RJ, Salcedo TW, Campbell JD, et al. C-C chemokine receptor 3 antagonism by the beta-chemokine macrophage inflammatory protein 4, a property strongly enhanced by an amino-terminal alanine-methionine swap. J Immunol, 2000, 164: 1488-1497.
    224. Wan Y, Jakway JP, Qiu H, et al. Identification of full, partial and inverse CC chemokine receptor 3 agonists using [35S]GTP_S binding. Eur J Pharmacol, 2002, 456: 1–10.
    225. Schraufstatter I, Takamori H, Sikora L,et al. Eosinophils and monocytes produce pulmonary and activationregulated chemokine, which activates cultured monocytes/macrophages. Am J Physiol Lung Cell Mol Physiol, 2004, 286: L494–L501.
    226. Bruna-Romero O, Schmieg J, Del Val M, et al. The dendritic cell-specific chemokine, dendritic cell-derived CC chemokine 1, enhances protective cell-mediated immunity to murine malaria. J Immunol, 2003, 170: 3195–3203.
    227. Jonuleit H, Schmitt E, Schuler G, et al. Induction of interleukin 10-producing, nonproliferating CD4(+) T cells with regulatory properties by repetitive stimulation with allogeneic immature human dendritic cells. J Exp Med, 2000, 192: 1213–22.
    228. Roncarolo MG., Levings MK, et al. Differentiation of T regulatory cells by immature dendritic cells. J Exp Med, 2001, 193: F5–F9.
    229. Cyster JG. Chemokines and cell migration in secondary lymphoid organs. Science, 1999, 286: 2098–2102.
    230. Yoshie O, Imai T, Nomiyama H. Chemokines in immunity. Adv Immunol, 2001, 78: 57–110.
    231. Pardo A, Smith KM, Abrams J, et al. CCL18/DC-CK-1/PARC up-regulation in hypersensitivity pneumonitis. J Leukoc Biol, 2001, 70: 610-6.
    232. Luzina IG, Atamas SP, Wise R, et al. Gene expression in bronchoalveolar lavage cells from scleroderma patients. Am J Respir Cell Mol Biol, 2002, 26: 549–57.
    233. Zou J, Young S, Zhu F, et al. Microarray profile of differentially expressed genes in a monkey model of allergic asthma. Genome Biol, 2002, 3: research0020.
    234. Schutyser E, Struyf S, Proost P, et al. Identification of biologically active chemokine isoforms from ascitic fluid and elevated levels of CCL18/pulmonary and activation-regulated chemokine in ovarian carcinoma. J Biol Chem, 2002, 277: 24584-93.
    235. Leung SY, Yuen ST, Chu KM, et al. Expression profiling identifies chemokine (C-C motif) ligand 18 as an independent prognostic indicator in gastric cancer. Gastroenterology, 2004, 127: 457-69.
    236. Loercher AE, Nash MA, Kavanagh JJ, et al. Identification of an IL-10-producing HLA-DR-negative monocyte subset in the malignant ascites of patients with ovarian carcinoma that inhibits cytokine protein expression and proliferation of autologous T cells. J Immunol, 1999, 163: 6251–60.
    237. Sica A, Saccani A, Bottazzi B, et al. Autocrine production of IL-10 mediates defective IL-12 production and NF-kB activation in tumor-associated macrophages. J Immunol, 2000, 164 : 762–67.
    238. Goebeler M, Trautmann A, et al. Differential and sequential expression of multiple chemokines during elicitation of allergic contact hypersensitivity. Am J Pathol, 2001,158: 431-40.
    239. Nomura I, Gao B, Boguniewicz M, et al. Distinct patterns of gene expression in the skin lesions of atopic dermatitis and psoriasis: a gene microarray analysis. J Allergy Clin Immunol, 2003, 112: 1195-1202.
    240. Boot RG, Verhoek M, De Fost M, et al. Marked elevation of the chemokine CCL18/PARC in Gaucher disease: a novel surrogate marker for assessing therapeutic intervention. Blood, 2004, 103: 33–39.
    241. Moran MT, Schofield JP, Hayman AR, et al. Pathologic gene expression in Gaucher disease: up-regulation of cysteine proteinases including osteoclastic cathepsin K. Blood, 2000, 96: 1969–78.
    242. Xanthou G, Polihronis M, Tzioufas AG, et al. Lymphoid chemokine messenger RNA expression by epithelial cells in the chronic inflammatory lesion of the salivary glands of Sjogren's syndrome patients: possible participation in lymphoid structure formation. Arthritis Rheum, 2001, 44: 408–18.
    243. Kusano F, Tanaka Y, Marumo F, et al. Expression of C-C chemokines is associated with portal and periportal inflammation in the liver of patients with chronic hepatitis C. Lab Invest, 2000, 80: 415–22.
    244. Krupa WM, Dewan M, Jeon MS, et al. Trapping of misdirected dendritic cells in the granulomatous lesions of giant cell arteritis. Am J Pathol, 2002, 161, 1815–23.
    245. Reape TJ, Rayner K, Manning CD, et al. Expression and cellular localization of the CC chemokines PARC and ELC in human atherosclerotic plaques. Am J Pathol, 1999, 154: 365-74.
    246. El-Asrar AM, Struyf S, Al-Kharashi S, et al. Expression of T lymphocyte chemoattractants and activation markers in vernal keratoconjunctivitis. Br J Ophthalmol, 2002, 86: 1175–80.
    267. Jamieson SE, Miller EN, Black GF, et al. Evidence for a cluster of genes on chromosome 17q11–q21 controlling susceptibility to tuberculosis and leprosy in Brazilians. Genes Immun, 2004, 5: 46–57.
    268. Bosma GC, Custer RP, Bosma MJ. A severe combined immunodeficiency mutation in the mouse. Nature, 1983, 301:527-30.
    269. Blunt T, Gell D, Fox M, et al. Identification of a nonsense mutation in the carboxyl-terminal region of DNA-dependent protein kinase catalytic subunit in the scid mouse. Proc Natl Acad Sci U S A, 1996, 93: 10285-90.
    270. Kirchgessner CU, Patil CK, Evans JW, et al. DNA-dependent kinase (p350) as a candidate gene for the murine SCID defect. Science,1995, 267: 1178-83.
    271. Murray AG, Petzelbauer P, Hughes CC, et al. Human T-cell-mediated destruction of allogeneic dermal microvessels in a severe combined immunodeficient mouse. Proc Natl Acad Sci U S A, 1994, 91: 9146-50.
    272. Yan HC, Delisser HM, Pilewski JM, et al. Leukocyte recruitment into human skin transplanted onto severe combined immunodeficient mice induced by TNF-alpha is dependent on E-selectin. J Immunol, 1994, 152: 3053-63.
    273. Kunstfeld R, Lechleitner S, Wolff K, et al. MCP-1 and MIP-1alpha are most efficient in recruiting T cells into the skin in vivo. J Invest Dermatol, 1998, 111: 1040-4.
    274. Oka M, Berking C, Nesbit M, et al. Interleukin-8 overexpression is present in pyoderma gangrenosum ulcers and leads to ulcer formation in human skin xenografts. Lab Invest, 2000, 80:595-604.
    275. Biedermann T, Schwarzler C, Lametschwandtner G, et al. Targeting CLA/E-selectin interactions prevents CCR4-mediated recruitment of human Th2 memory cells to human skin in vivo. Eur J Immunol, 2002, 32: 3171-80.
    276. Carballido JM, Biedermann T, Schwarzler C, et al. The SCID-hu Skin mouse as a model to investigate selective chemokine mediated homing of human T-lymphocytes to the skin in vivo. J Immunol Methods, 2003, 273: 125-35.
    277. Fahy O, Porte H, Senechal S, et al. Chemokine-induced cutaneous inflammatory cell infiltration in a model of Hu-PBMC-SCID mice grafted with human skin. Am J Pathol, 2001, 158: 1053-1063.
    278. Senechal S, Fahy O, Gentina T, et al. CCR3-blocking antibody inhibits allergen-induced eosinophil recruitment in human skin xenografts from allergic patients. Lab Invest, 2002, 82: 929-39.
    279. Asselin S, Conjeaud H, Minty A, et al. Stable polarization of peripheral blood T cells towards type 1 or type 2 phenotype after polyclonal activation. Eur J Immunol, 1998, 28: 532-9.
    280. Imai T, Nagira M, Takagi S, et al. Selective recruitment of CCR4-bearing Th2 cells toward antigen-presenting cells by the CC chemokines thymus and activation-regulated chemokine and macrophage-derived chemokine. Int Immunol, 1999, 11: 81-8.
    281. Kurimoto Y, de Weck AL, Dahinden CA. Interleukin 3-dependent mediator release in basophils triggered by C5a. J Exp Med, 1989, 170: 467-479.
    282. Jahnsen FL, Lund-Johansen F, Dunne J F, et al. Experimentally induced recruitment of plasmacytoid (CD123high) dendritic cells in human nasal allergy. J. Immunol, 2000, 165:4062-68.
    283. de Heer HJ, Hammad H, Soullie T, et al. Essential role of lung plasmacytoid dendritic cells in preventing asthmatic reactions to harmless inhaled antigen. J Exp Med, 2004, 200: 89-98.
    284. Charbonnier AS, Hammad H, Gosset P, et al. Der p 1-pulsed myeloid and plasmacytoid dendritic cells from house dust mite-sensitized allergic patients dysregulate the T cell response. J Leukoc Biol, 2003, 73: 91-99.
    285. Farkas L, Kvale EO, Johansen FE, et al. Plasmacytoid dendritic cells activate allergen-specific TH2 memory cells: modulation by CpG oligodeoxynucleotides. J Allergy Clin Immunol, 2004, 114: 436-43.
    286. Moller GM, Overbeek SE, Van Helden-Meeuwsen CG, et al. Increased numbers of dendritic cells in the bronchial mucosa of atopic asthmatic patients: downregulation by inhaled corticosteroids. Clin Exp Allergy, 1996, 26: 517-24.
    287. Campbell JJ, Haraldsen G, et al. The chemokine receptor CCR4 in vascular recognition by cutaneous but not intestinal memory T cells. Nature. 1999, 400: 776-80.
    288. Diaz-Sanchez D, Proietti L, Polosa R. Diesel fumes and the rising prevalence of atopy: an urban legend? Curr Allergy Asthma Rep, 2003, 3: 146-52.Farber, JM. Mig and IP-10: CXC chemokines that target lymphocytes. J Leukoc Biol, 1997, 61: 246-57.
    289. Yu CP, Xu GB. Predictive models for deposition of inhaled diesel exhaust particles in humans and laboratory species. Res Rep Health Eff Inst, 1987, 10: 3-22.
    290. Diaz-Sanchez D. The role of diesel exhaust particles and their associated polyaromatic hydrocarbons in the induction of allergic airway disease. Allergy, 1997, 52: 52-6;discussion 57-8.
    291. Diaz-Sanchez D, Tsien A, Fleming J,et al. Combined diesel exhaust particulate and ragweed allergen challenge markedly enhances human in vivo nasal ragweed-specific IgE and skews cytokine production to a T helper cell 2-type pattern. J Immunol, 1997, 158: 2406-13.
    292. Fujieda S, Diaz-Sanchez D, Saxon A.Combined nasal challenge with diesel exhaust particles and allergen induces In vivo IgE isotype switching. Am J Respir Cell Mol Biol, 1998, 19: 507-12.
    293. Fahy O, Hammad H, Senechal S, et al. Synergistic effect of diesel organic extracts and allergen Der p 1 on the release of chemokines by peripheral blood mononuclear cells from allergic subjects: involvement of the map kinase pathway. Am J Respir Cell Mol Biol, 2000, 23: 247-54.
    294. Fahy O, Senechal S, Pene J, et al. Diesel exposure favors Th2 cell recruitment by mononuclear cells and alveolar macrophages from allergic patients by differentially regulating macrophage-derived chemokine and IFN-gamma-induced protein-10 production. J Immunol, 2002, 168: 5912-9.
    295. Senechal S, de Nadai P, Ralainirina N, et al. Effect of diesel on chemokines and chemokine receptors involved in helper T cell type 1/type 2 recruitment in patients with asthma. Am J Respir Crit Care Med, 2003, 168: 215-21.
    296. Salvi SS, Nordenhall C, Blomberg A, et al. Acute exposure to diesel exhaust increases IL-8 and GRO-alpha production in healthy human airways. Am J Respir Crit Care Med, 2000, 161(2 Pt 1):550-7.
    297. Steerenberg PA, Zonnenberg JA, Dormans JA, et al. Diesel exhaust particles induced release of interleukin 6 and 8 by (primed) human bronchial epithelial cells (BEAS 2B) in vitro. Exp Lung Res 1998;24: 85-100.
    298. Ohtoshi T, Takizawa H, Okazaki H, Kawasaki S, Takeuchi N, Ohta K, et al. Diesel exhaust particles stimulate human airway epithelial cells to produce cytokines relevant to airway inflammation in vitro. J Allergy Clin Immunol, 1998, 101: 778-85.
    299. Jimenez LA, Drost EM, Gilmour PS, Rahman I, Antonicelli F, Ritchie H, et al. PM(10)-exposed macrophages stimulate a proinflammatory response in lung epithelial cells via TNF-alpha. Am J Physiol Lung Cell Mol Physiol, 2002, 282: L237-48.
    300. Fahy O, Tsicopoulos A, Hammad H, et al. Effects of diesel organic extracts on chemokine production by peripheral blood mononuclear cells. J Allergy Clin Immunol, 1999, 103: 1115-24.
    301. Farber, JM. Mig and IP-10: CXC chemokines that target lymphocytes. J Leukoc Biol, 1997, 61: 246-57.
    302. Flier J, Boorsma DM, van Beek PJ, et al. Differential expression of CXCR3 targeting chemokines CXCL10, CXCL9, and CXCL11 in different types of skin inflammation. J Pathol, 2001, 94: 398-405.
    303. Gasperini S, Marchi M, Calzetti F, et al. Gene expression and production of the monokine induced by IFN-gamma (MIG), IFN-inducible T cell alpha chemoattractant (I-TAC), and IFN-gamma-inducible protein-10 (IP-10) chemokines by human neutrophils. J Immunol, 1999, 62: 4928-37.
    304. Piali L, Weber C, LaRosa G, et al. The chemokine receptor CXCR3 mediates rapid and shear-resistant adhesion-induction of effector T lymphocytes by the chemokines IP10 and Mig. Eur J Immunol, 1998, 28: 961-72.
    305. Boorsma DM, Flier J, Sampat S, et al. Chemokine IP-10 expression in cultured human keratinocytes. Arch Dermatol Res, 1998, 290: 335-41.
    306. Sauty A, Dziejman M, Taha RA, et al. The T cell-specific CXC chemokines IP-10, Mig, and I-TAC are expressed by activated human bronchial epithelial cells. J Immunol 1999;162: 3549-58.
    307. Jinquan T, Jing C, Jacobi HH, et al. CXCR3 expression and activation of eosinophils: role of IFN-gamma-inducible protein-10 and monokine induced by IFN-gamma. J Immunol, 2000, 165: 1548-56.
    308. Gangur V, Simons FE, HayGlass KT. IP-10 mediated reinforcement of human type 1 cytokine synthesis to environmental allergens among non-atopic subjects. Int Arch Allergy Immunol, 1999, 118: 387-90.
    309. Fahy O, Senechal S, Pene J, et al. Diesel exposure favors Th2 cell recruitment by mononuclear cells and alveolar macrophages from allergic patients by differentially regulating macrophage-derived chemokine and IFN-gamma-induced protein-10 production. J Immunol, 2002, 168: 5912-9.
    310. Nilsen AM, Hagemann R, Eikas H, et al. Reduction of IL-12 p40 production in activated monocytes after exposure to diesel exhaust particles. Int Arch Allergy Immunol, 2003, 131: 201-8.
    311. Ohtani T, Nakagawa S, Kurosawa M, et al. Cellular basis of the role of diesel exhaust particles in inducing Th2-dominant response. J Immunol, 2005, 174: 2412-9.
    312. Chvatchko Y, Hoogewerf AJ, Meyer A, et al. A key role for CC chemokine receptor 4 in lipopolysaccharide-induced endotoxic shock. J Exp Med 2000, 191: 1755-64.
    313. Iellem A, Colantonio L, et al. Inhibition by IL-12 and IFN-alpha of I-309 and macrophage-derived chemokine production upon TCR triggering of human Th1 cells. Eur J Immunol, 2000, 30: 1030-9.
    314. Inngjerdingen M, Damaj B, et al. Human NK cells express CC chemokine receptors 4 and 8 and respond to thymus and activation-regulated chemokine, macrophage-derived chemokine, and I-309. J Immunol, 2000, 164: 4048-54.
    315. Haque NS, Fallon JT, et al. The chemokine receptor CCR8 mediates human endothelial cell chemotaxis induced by I-309 and Kaposi sarcoma herpesvirus-encoded vMIP-I and by lipoprotein(a)-stimulated endothelial cell conditioned medium. Blood, 2001, 97: 39-45.
    316. Andrew DP, Chang MS, McNinch J, et al. STCP-1 (MDC) CC chemokine acts specifically on chronically activated Th2 lymphocytes and is produced by monocytes on stimulation with Th2 cytokines IL-4 and IL-13. J Immunol, 1998, 161: 5027-38.
    317. Zingoni A, Soto H, Hedrick JA, et al. The chemokine receptor CCR8 is preferentially expressed in Th2 but not Th1 cells. J Immunol, 1998, 161:547-51.
    318. Bochner BS, Hudson SA, Xiao HQ, et al. Release of both CCR4-active and CXCR3-active chemokines during human allergic pulmonary late-phase reactions. J Allergy Clin Immunol, 2003, 112: 930-4.
    319. Devouassoux G, Saxon A, Metcalfe DD, et al. Chemical constituents of diesel exhaust particles induce IL-4 production and histamine release by human basophils. J Allergy Clin Immunol, 2002, 109(5): 847-53.
    320. Stenfors N, Nordenhall C, Salvi SS, et al. Different airway inflammatory responses in asthmatic and healthy humans exposed to diesel. Eur Respir J, 2004, 23: 82-6.
    321. Diaz-Sanchez D, Tsien A, Casillas A, et al. Enhanced nasal cytokine production in human beings after in vivo challenge with diesel exhaust particles. J Allergy Clin Immunol, 1996, 98: 114-23.
    322. Pourazar J, Frew AJ, Blomberg A, et al. Diesel exhaust exposure enhances the expression of IL-13 in the bronchial epithelium of healthy subjects. Respir Med, 2004, 98: 821-5.
    323. McKenzie GJ, Emson CL, Bell SE, et al. Impaired development of Th2 cells in IL-13-deficient mice. Immunity, 1998, 9(3): 423-32.
    324. Yan HC, Juhasz I, Pilewski J, et al. Human/severe combined immunodeficient mouse chimeras. An experimental in vivo model system to study the regulation of human endothelial cell-leukocyte adhesion molecules. J Clin Invest, 1993, 91: 986.
    325. Berger EA, Murphy PM, Farber JM. Chemokine receptors as HIV-1 coreceptors: roles in viral entry, tropism, and disease. Annu Rev Immunol, 1999, 17: 657-700.
    326. Beck LA, Dalke S, Leiferman KM, et al. Cutaneous injection of RANTES causes eosinophil recruitment: comparison of nonallergic and allergic human subjects. J Immunol, 1997, 159: 2962-72.
    327. Oliveira SH, Lira S, Martinez-A C, et al. Increased responsiveness of murine eosinophils to MIP-1beta (CCL4) and TCA-3 (CCL1) is mediated by their specific receptors, CCR5 and CCR8. J Leukoc Biol, 2002, 71: 1019-25.
    328. Proudfoot AE, Handel TM, Johnson Z, et al. Glycosaminoglycan binding and oligomerization are essential for the in vivo activity of certain chemokines. Proc Natl Acad Sci U S A, 2003, 100: 1885-90.
    329. D'Amico G, Frascaroli G, Bianchi G, et al. Uncoupling of inflammatory chemokine receptors by IL-10: generation of functional decoys. Nat Immunol, 2000, 1: 387-391.
    330. Lloyd CM, Delaney T, Nguyen T, et al. CC chemokine receptor (CCR)3/eotaxin is followed by CCR4/monocyte-derived chemokine in mediating pulmonary T helper lymphocyte type 2 recruitment after serial antigen challenge in vivo. J Exp Med, 2000, 191: 265-274.
    331. Proudfoot AE. Chemokine receptors: multifaceted therapeutic targets, Nat Rev Immunol, 2002, 2: 106-115.
    332. Liang M, Rosser M, Ng HP, et al. Species selectivity of a small molecule antagonist for the CCR1 chemokine receptor. Eur J Pharmacol, 2000, 389: 41-49.
    333. Schall TJ, Bacon K, Toy KJ, et al. Selective attraction of monocytes and T lymphocytes of the memory phenotype by cytokine RANTES. Nature, 1990, 347: 669-71.
    334. Bonecchi R, Bianchi G, Bordignon PP, et al. Differential expression of chemokine receptors and chemotactic responsiveness of type 1 T helper cells (Th1s) and Th2s. J Exp Med, 1998, 187: 129-134.
    335. Blanpain C, Libert F, Vassart G, et al. CCR5 and HIV infection. Receptors Channels, 2002, 8: 19-31.
    336. He J, Chen Y, Farzan M, et al. CCR3 and CCR5 are co-receptors for HIV-1 infection of microglia. Nature, 1997, 385: 645-49.
    337. Suzuki N, Nakajima A, Yoshino S, et al. Selective accumulation of CCR5+ T lymphocytes into inflamed joints of rheumatoid arthritis. Int Immunol, 1999, 11: 553-59.
    338. Oki M, Ohtani H, Kinouchi Y, et al. Accumulation of CCR5+ T cells around RANTES+ granulomas in Crohn's disease: a pivotal site of Th1-shifted immune response? Lab Invest, 2005, 85: 137-45.
    339. Zang YC, Samanta AK, Halder JB, et al. Aberrant T cell migration toward RANTES and MIP-1 alpha in patients with multiple sclerosis. Overexpression of chemokine receptor CCR5. Brain, 2000, 123 (Pt 9): 1874-82.
    340. Panzer U, Reinking RR, Steinmetz OM, et al. CXCR3 and CCR5 positive T-cell recruitment in acute human renal allograft rejection. Transplantation, 2004, 78: 1341-50.
    341. Hellier S, Frodsham AJ, Hennig BJ, et al. Association of genetic variants of the chemokine receptor CCR5 and its ligands, RANTES and MCP-2, with outcome of HCV infection. Hepatology, 2003, 38: 1468-76.
    342. Zhou Y, Huang D, Farver C, et al. Relative importance of CCR5 and antineutrophil cytoplasmic antibodies in patients with Wegener's granulomatosis. J Rheumatol, 2003, 30: 1541-47.
    343. Schuh JM, Blease K, Hogaboam CM. The role of CC chemokine receptor 5 (CCR5) and RANTES/CCL5 during chronic fungal asthma in mice. Faseb J, 2002, 16: 228-30.
    344. Amano H, Bickerstaff A, Orosz CG, et al. Absence of recipient CCR5 promotes early and increased allospecific antibody responses to cardiac allografts. J Immunol, 2005, 174: 6499-6508.
    345. Luckow B, Joergensen J, Chilla S, et al. Reduced intragraft mRNA expression of matrix metalloproteinases Mmp3, Mmp12, Mmp13 and Adam8, and diminished transplant arteriosclerosis in Ccr5-deficient mice. Eur J Immunol, 2004, 34: 2568-78.
    346. Algood HM, Flynn JL. CCR5-deficient mice control Mycobacterium tuberculosis infection despite increased pulmonary lymphocytic infiltration. J Immunol, 2004, 173: 3287-96.
    347. Field J, Marshall AC, J Hertzog P, et al. Chemokine receptor CCR5 is not required for development of experimental autoimmune gastritis. Clin Immunol, 2003, 109: 238-47.
    348. Zhong MX, Kuziel WA, Pamer EG, et al. Chemokine receptor 5 is dispensable for innate and adaptive immune responses to Listeria monocytogenes infection. Infect Immun, 2004, 72: 1057-64.
    349. Chen Z, Yu S, Bakhiet M, et al. The chemokine receptor CCR5 is not a necessary inflammatory mediator in kainic acid-induced hippocampal injury: evidence for a compensatory effect by increased CCR2 and CCR3. J Neurochem, 2003, 86: 61-68.
    350. Lee B, Sharron M, Blanpain C, et al. Epitope mapping of CCR5 reveals multiple conformational states and distinct but overlapping structures involved in chemokine and coreceptor function. J Biol Chem,1999, 274: 9617-26.
    351. Blanpain C, Buser R, Power CA, et al. A chimeric MIP-1alpha/RANTES protein demonstrates the use of different regions of the RANTES protein to bind and activate its receptors. J Leukoc Biol, 2001, 69: 977-985.
    352. Ying S, Meng Q, Zeibecoglou K, et al. Eosinophil chemotactic chemokines (eotaxin, eotaxin-2, RANTES, monocyte chemoattractant protein-3 (MCP-3), and MCP-4), and C-C chemokine receptor 3 expression in bronchial biopsies from atopic and nonatopic (Intrinsic) asthmatics. J Immunol, 1999, 163: 6321-29.
    353. Weber C, Belge KU, von Hundelshausen P, et al. Differential chemokine receptor expression and function in human monocyte subpopulations. J Leukoc Biol, 2000, 67: 699-704.
    354. Oravecz T, Pall M, Roderiquez G, et al. Regulation of the receptor specificity and function of the chemokine RANTES (regulated on activation, normal T cell expressed and secreted) by dipeptidyl peptidase IV (CD26)-mediated cleavage. J Exp Med, 1997, 86: 1865-72.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700