1051
|
Montanez E, Piwko-Czuchra A, Bauer M, Li S, Yurchenco P, Fässler R. Analysis of integrin functions in peri-implantation embryos, hematopoietic system, and skin. Methods Enzymol 2007; 426:239-89. [PMID: 17697888 DOI: 10.1016/s0076-6879(07)26012-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Integrins mediate cell adhesion, permit traction forces important for cell migration, and cross-talk with growth factor receptors to regulate cell proliferation, cell survival, and cell differentiation. The plethora of functions explains their central role for development and disease. The progress in mouse genetics and the ease with which the mouse genome can be manipulated enormously contributed to our understanding of how integrins exert their functions at the molecular level. In the present chapter, we describe tests that are routinely used in our laboratory to investigate embryos, organs, and cells (peri-implantation embryos, hematopoietic system, epidermis, and hair follicles) that lack the expression of integrins or integrin-associated proteins.
Collapse
Affiliation(s)
- Eloi Montanez
- Max Planck Institute of Biochemistry, Department of Molecular Medicine, Martinsried, Germany
| | | | | | | | | | | |
Collapse
|
1052
|
Nakae S, Suto H, Berry GJ, Galli SJ. Mast cell-derived TNF can promote Th17 cell-dependent neutrophil recruitment in ovalbumin-challenged OTII mice. Blood 2006; 109:3640-8. [PMID: 17197430 PMCID: PMC1874568 DOI: 10.1182/blood-2006-09-046128] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Both mast cells and IL-17 can contribute to host defense and pathology in part by orchestrating neutrophil recruitment, but the possible role of mast cells in IL-17-induced inflammation remains to be defined. We found that mast cells and IL-17, but neither IFN-gamma nor FcRgamma signaling, contributed significantly to the antigen (Ag)-dependent airway neutrophilia elicited in ovalbumin-specific T-cell receptor (TCR)-expressing C57BL/6-OTII mice, and that IFN-gamma significantly suppressed IL-17-dependent airway neutrophilia in this setting. IL-18, IL-1beta, and TNF each contributed significantly to the development of Ag- and T helper 17 (Th17 cell)-mediated airway neutrophilia. Moreover, IL-17 enhanced mast cell TNF production in vitro, and mast cell-associated TNF contributed significantly to Ag- and Th17 cell-mediated airway neutrophilia in vivo. By contrast, we detected no significant role for the candidate mediators histamine, PGD(2), LTB(4), CXCL10, or IL-16, each of which can be produced by mast cells and other cell types, in the neutrophil infiltration elicited in this model. These findings establish that mast cells and mast cell-derived TNF can significantly enhance, by FcRgamma-independent mechanisms, the Ag- and Th17 cell-dependent development of a neutrophil-rich inflammatory response at a site of Ag challenge.
Collapse
Affiliation(s)
- Susumu Nakae
- Department of Pathology L-235, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
1053
|
Smith CM, Rosa GTL, May JS, Bennett NJ, Mount AM, Belz GT, Stevenson PG. CD4+ T cells specific for a model latency-associated antigen fail to control a gammaherpesvirus in vivo. Eur J Immunol 2006; 36:3186-97. [PMID: 17109468 DOI: 10.1002/eji.200636164] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CD4(+) T cells play a major role in containing herpesvirus infections. However, their cellular targets remain poorly defined. In vitro CD4(+) T cells have been reported to kill B cells that harbor a latent gammaherpesvirus. We used the B cell-tropic murine gammaherpesvirus-68 (MHV-68) to test whether this also occurred in vivo. MHV-68 that expressed cytoplasmic ovalbumin (OVA) in tandem with its episome maintenance protein, ORF73, stimulated CD8(+) T cells specific for the H2-K(b)-restricted OVA epitope SIINFEKL and was rapidly eliminated from C57BL/6 (H2(b)) mice. However, the same virus failed to stimulate CD4(+) T cells specific for the I-A(d)/I-A(b)-restricted OVA(323-339) epitope. We overcame any barrier to the MHC class II-restricted presentation of an endogenous epitope by substituting OVA(323-339) for the CLIP peptide of the invariant chain (ORF73-IRES-Ii-OVA), again expressed in tandem with ORF73. This virus presented OVA(323-339) but showed little or no latency deficit in either BALB/c (H2(d)) or C57BL/6 mice. Latent antigen-specific CD4(+) T cells therefore either failed to recognize key virus-infected cell populations in vivo or lacked the effector functions required to control them.
Collapse
Affiliation(s)
- Christopher M Smith
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | | |
Collapse
|
1054
|
Brännström J, Hässler S, Peltonen L, Herrmann B, Winqvist O. Defect internalization and tyrosine kinase activation in Aire deficient antigen presenting cells exposed to Candida albicans antigens. Clin Immunol 2006; 121:265-73. [PMID: 16982213 DOI: 10.1016/j.clim.2006.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2006] [Revised: 07/10/2006] [Accepted: 08/08/2006] [Indexed: 01/12/2023]
Abstract
Patients with Autoimmune polyendocrine syndrome type I (APS I) present with multiple endocrine failures due to organ-specific autoimmune disease, thought to be T-cell-mediated. Paradoxically, APS I patients suffer from chronic mucocutaneous candidiasis. The mutated gene has been identified as the Autoimmune regulator (AIRE). Aire is expressed in medullary epithelial cells of the thymus and in antigen presenting cells in the periphery. T cells from Aire deficient mice and men displayed an enhanced proliferative response against Candida antigen in vitro, suggesting that Aire deficient T cells are competent in recognizing Candida albicans. In contrast, monocytes from APS I patients displayed a decreased and delayed internalization of zymosan. Furthermore, Candida antigen activated monocytes from APS I patients show decreased and altered phoshotyrosine kinase activation. In conclusion, Aire deficient APCs have a defect receptor mediated internalization of Candida which affects kinase activation, likely altering the innate Candida immune response.
Collapse
Affiliation(s)
- Johan Brännström
- Karolinska Institutet, Department of Medicine, Unit of Clinical Allergy Research L2:04, Karolinska Hospital, SE-171 76 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
1055
|
Matthews KE, Karabeg A, Roberts JM, Saeland S, Dekan G, Epstein MM, Ronchese F. Long-term deposition of inhaled antigen in lung resident CD11b-CD11c+ cells. Am J Respir Cell Mol Biol 2006; 36:435-41. [PMID: 17122367 DOI: 10.1165/rcmb.2006-0330oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In this study we report the characterization of a population of lung resident CD11b(-)CD11c(+) cells that are able to take up inhaled antigen and retain it for extended periods of time. Ovalbumin conjugated to fluorescein-isothiocyanate (FITC-OVA) administered intranasally to mice was taken up by two main populations of cells in the lung, a migratory CD11c(+)CD11b(+) population consisting of dendritic cells (DC), which rapidly transported antigen to the draining lymph node (LN), and a resident CD11b(-)CD11c(+) population that retained engulfed antigen without apparently degrading it for up to 8 wk after administration. The FITC(+)CD11b(-)CD11c(+) cells did not migrate to draining LN at a detectable rate, and did not up-regulate expression of costimulatory molecules in response to LPS treatment. FITC(+)CD11b(-)CD11c(+) cells were found in the lung and bronchoalveolar lavage fluid, and their distribution was compatible with macrophages. Although FITC(+)CD11b(-)CD11c(+) cells expressed the DC marker DEC205 and other molecules associated with antigen-presenting cell function, they did not induce proliferation of antigen-specific CD4(+) T cells in vitro or acute cytokine production by activated CD4(+) T cells in vivo. Thus, FITC(+)CD11b(-)CD11c(+) cells appear to represent an intermediate cell type sharing properties with DC and macrophages. These cells may have a role in modulating the responses of lung resident T cells to inhaled antigens.
Collapse
Affiliation(s)
- Kate E Matthews
- Malaghan Institute of Medical Research, PO Box 7060, Wellington South, New Zealand
| | | | | | | | | | | | | |
Collapse
|
1056
|
Mirenda V, Jarmin SJ, David R, Dyson J, Scott D, Gu Y, Lechler RI, Okkenhaug K, Marelli-Berg FM. Physiologic and aberrant regulation of memory T-cell trafficking by the costimulatory molecule CD28. Blood 2006; 109:2968-77. [PMID: 17119120 DOI: 10.1182/blood-2006-10-050724] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
Productive T-cell immunity requires both the activation and the migration of specific T cells to the antigenic tissue. The costimulatory molecule CD28 plays an essential role in the initiation of T-cell–mediated immunity. We investigated the possibility that CD28 may also regulate migration of primed T cells to target tissue. In vitro, CD28-mediated signals enhanced T-cell transendothelial migration, integrin clustering, and integrin-mediated migration. In vivo, T cells bearing a mutation in the CD28 cytoplasmic domain, which abrogates PI3K activation, displayed normal clonal expansion but defective localization to antigenic sites following antigenic rechallenge. Importantly, antibody-mediated CD28 stimulation led to unregulated memory T-cell migration to extra-lymphoid tissue, which occurred independently of T-cell receptor (TCR)–derived signals and homing-receptor expression. Finally, we provide evidence that CD28- and CTLA-4–mediated signals exert opposite effects on T-cell trafficking in vivo. These findings highlight a novel physiologic function of CD28 that has crucial implications for the therapeutic manipulation of this and other costimulatory molecules.
Collapse
Affiliation(s)
- Vincenzo Mirenda
- Department of Immunology, Division of Medicine, Imperial College London, Hammersmith Campus, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
1057
|
Dorn T, Kuhn U, Bungartz G, Stiller S, Bauer M, Ellwart J, Peters T, Scharffetter-Kochanek K, Semmrich M, Laschinger M, Holzmann B, Klinkert WEF, Straten PT, Køllgaard T, Sixt M, Brakebusch C. RhoH is important for positive thymocyte selection and T-cell receptor signaling. Blood 2006; 109:2346-55. [PMID: 17119112 DOI: 10.1182/blood-2006-04-019034] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
RhoH is a small GTPase expressed only in the hematopoietic system. With the use of mice with targeted disruption of the RhoH gene, we demonstrated that RhoH is crucial for thymocyte maturation during DN3 to DN4 transition and during positive selection. Furthermore, the differentiation and expansion of DN3 and DN4 thymocytes in vitro were severely impaired. These defects corresponded to defective TCR signaling. Although RhoH is not required for TCR-induced activation of ZAP70 and ZAP70-mediated activation of p38, it is crucial for the tyrosine phosphorylation of LAT, PLCγ1, and Vav1 and for the activation of Erk and calcium influx. These data suggest that RhoH is important for pre–TCR and TCR signaling because it allows the efficient interaction of ZAP70 with the LAT signalosome, thus regulating thymocyte development.
Collapse
Affiliation(s)
- Tatjana Dorn
- Max Planck Institute of Biochemistry, Heisenberg Group Regulation of Cytoskeletal Organization, Martinsried, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1058
|
Ainge GD, Parlane NA, Denis M, Hayman CM, Larsen DS, Painter GF. Phosphatidylinositol mannosides: Synthesis and adjuvant properties of phosphatidylinositol di- and tetramannosides. Bioorg Med Chem 2006; 14:7615-24. [PMID: 16876422 DOI: 10.1016/j.bmc.2006.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2006] [Revised: 06/26/2006] [Accepted: 07/01/2006] [Indexed: 10/24/2022]
Abstract
Phosphatidylinositol mannosides (PIMs) isolated from mycobacteria have been identified as an important class of glycolipids with significant immune modulating properties. We present here the syntheses of phosphatidylinositol dimannoside (PIM2, 1) and phosphatidylinositol tetramannoside (PIM4, 2) and evaluate their adjuvant properties in a transgenic mouse model. The key step in the synthetic methodology for the synthesis of 2 relies on the selective glycosylation of diol 3 with mannosyl donor 11. Both synthetic PIMs were effective at enhancing IFN-gamma when given as adjuvants with a model antigen, with PIM2 being the more active. These data suggest that in this assay the PIM core structure is responsible for the observed biological activity.
Collapse
Affiliation(s)
- Gary D Ainge
- Carbohydrate Chemistry Team, Industrial Research Limited, Lower Hutt, New Zealand
| | | | | | | | | | | |
Collapse
|
1059
|
Nagaishi T, Pao L, Lin SH, Iijima H, Kaser A, Qiao SW, Chen Z, Glickman J, Najjar SM, Nakajima A, Neel BG, Blumberg RS. SHP1 Phosphatase-Dependent T Cell Inhibition by CEACAM1 Adhesion Molecule Isoforms. Immunity 2006; 25:769-81. [PMID: 17081782 DOI: 10.1016/j.immuni.2006.08.026] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2005] [Revised: 06/26/2006] [Accepted: 08/30/2006] [Indexed: 11/20/2022]
Abstract
T cell activation through the T cell receptor (TCR) is subsequently modified by secondary signals that are either stimulatory or inhibitory. We show that CEACAM1 adhesion molecule isoforms containing a long cytoplasmic domain inhibited multiple T cell functions as a consequence of TCR ligation. Overexpression of CEACAM1 resulted in decreased proliferation, allogeneic reactivity, and cytokine production in vitro and delayed type hypersensitivity and inflammatory bowel disease in mouse models in vivo. Conditioned deletion of CEACAM1 in T cells caused increased TCR-CD3 complex signaling. This T cell regulation was dependent upon the presence of immunoreceptor tyrosine-based inhibition motifs (ITIM) within the cytoplasmic domain of CEACAM1 and the Src homology 2 domain-containing protein tyrosine-phosphatase 1 (SHP1) in the T cell. Thus, CEACAM1 overexpression or deletion in T cells resulted in T cell inhibition or activation, respectively, revealing a role for CEACAM1 as a class of inhibitory receptors potentially amenable to therapeutic manipulation.
Collapse
Affiliation(s)
- Takashi Nagaishi
- Gastroenterology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1060
|
Krishnamoorthy G, Lassmann H, Wekerle H, Holz A. Spontaneous opticospinal encephalomyelitis in a double-transgenic mouse model of autoimmune T cell/B cell cooperation. J Clin Invest 2006. [PMID: 16955140 DOI: 10.1172/jci28330.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We describe a double-transgenic mouse strain (opticospinal EAE [OSE] mouse) that spontaneously develops an EAE-like neurological syndrome closely resembling a human variant of multiple sclerosis, Devic disease (also called neuromyelitis optica). Like in Devic disease, the inflammatory, demyelinating lesions were located in the optic nerve and spinal cord, sparing brain and cerebellum, and the murine lesions showed histological similarity with their human correlates. OSE mice have recombination-competent immune cells expressing a TCR-alphabeta specific for myelin oligodendrocyte glycoprotein (MOG) aa 35-55 peptide in the context of I-Ab along with an Ig J region replaced by the recombined heavy chain of a monoclonal antibody binding to a conformational epitope on MOG. OSE mouse B cells bound even high dilutions of recombinant MOG, but not MOG peptide, and processed and presented it to autologous T cells. In addition, in OSE mice, but not in single-transgenic parental mice, anti-MOG antibodies were switched from IgM to IgG1.
Collapse
|
1061
|
Krishnamoorthy G, Lassmann H, Wekerle H, Holz A. Spontaneous opticospinal encephalomyelitis in a double-transgenic mouse model of autoimmune T cell/B cell cooperation. J Clin Invest 2006; 116:2385-92. [PMID: 16955140 PMCID: PMC1555668 DOI: 10.1172/jci28330] [Citation(s) in RCA: 253] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2006] [Accepted: 06/13/2006] [Indexed: 01/22/2023] Open
Abstract
We describe a double-transgenic mouse strain (opticospinal EAE [OSE] mouse) that spontaneously develops an EAE-like neurological syndrome closely resembling a human variant of multiple sclerosis, Devic disease (also called neuromyelitis optica). Like in Devic disease, the inflammatory, demyelinating lesions were located in the optic nerve and spinal cord, sparing brain and cerebellum, and the murine lesions showed histological similarity with their human correlates. OSE mice have recombination-competent immune cells expressing a TCR-alphabeta specific for myelin oligodendrocyte glycoprotein (MOG) aa 35-55 peptide in the context of I-Ab along with an Ig J region replaced by the recombined heavy chain of a monoclonal antibody binding to a conformational epitope on MOG. OSE mouse B cells bound even high dilutions of recombinant MOG, but not MOG peptide, and processed and presented it to autologous T cells. In addition, in OSE mice, but not in single-transgenic parental mice, anti-MOG antibodies were switched from IgM to IgG1.
Collapse
MESH Headings
- Animals
- Autoimmunity
- B-Lymphocytes/immunology
- Cell Division
- Crosses, Genetic
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Immunoglobulin Heavy Chains/genetics
- Lymphocyte Subsets/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Myelin Proteins
- Myelin-Associated Glycoprotein/genetics
- Myelin-Oligodendrocyte Glycoprotein
- Optic Nerve/pathology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Spinal Cord/pathology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Gurumoorthy Krishnamoorthy
- Department of Neuroimmunology, Max Planck Institute for Neurobiology, Martinsried, Germany.
Division of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Max Planck Institute for Neurobiology, Martinsried, Germany.
Division of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Hartmut Wekerle
- Department of Neuroimmunology, Max Planck Institute for Neurobiology, Martinsried, Germany.
Division of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Andreas Holz
- Department of Neuroimmunology, Max Planck Institute for Neurobiology, Martinsried, Germany.
Division of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
1062
|
Eun SY, O'Connor BP, Wong AW, van Deventer HW, Taxman DJ, Reed W, Li P, Blum JS, McKinnon KP, Ting JPY. Cutting edge: rho activation and actin polarization are dependent on plexin-A1 in dendritic cells. THE JOURNAL OF IMMUNOLOGY 2006; 177:4271-5. [PMID: 16982860 DOI: 10.4049/jimmunol.177.7.4271] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We recently identified expression of the semaphorin receptor, plexin-A1, in dendritic cells (DCs); however, its function in these cells remains to be elucidated. To investigate function and maximize physiological relevance, we devised a retroviral approach to ablate plexin-A1 gene expression using small hairpin RNA (shRNA) in primary bone marrow-derived DCs. We show that plexin-A1 localizes within the cytoplasm of immature DCs, becomes membrane-associated, and is enriched at the immune synapse in mature DCs. Reducing plexin-A1 expression with shRNA greatly reduced actin polarization as well as Rho activation without affecting Rac or Cdc42 activation. A Rho inhibitor, C3, also reduced actin polarization. These changes were accompanied by the near-ablation of T cell activation. We propose a mechanism of adaptive immune regulation in which plexin-A1 controls Rho activation and actin cytoskeletal rearrangements in DCs that is associated with enhanced DC-T cell interactions.
Collapse
Affiliation(s)
- So-Young Eun
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1063
|
Wells JW, Cowled CJ, Giorgini A, Kemeny DM, Noble A. Regulation of allergic airway inflammation by class I-restricted allergen presentation and CD8 T-cell infiltration. J Allergy Clin Immunol 2006; 119:226-34. [PMID: 17208606 DOI: 10.1016/j.jaci.2006.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 08/31/2006] [Accepted: 09/06/2006] [Indexed: 11/23/2022]
Abstract
BACKGROUND CD8 T cells are known to respond to exogenous antigens through cross-presentation. The importance of the CD8 cell response in the lung after inhalation of allergen and its effects on asthmatic inflammation are less clear. OBJECTIVE We sought to determine the dynamics, nature, and immunoregulatory activities of the class I CD8 T-cell response to inhaled allergen. METHODS We studied a murine model of respiratory allergen sensitization, adoptive transfer of transgenic T cells, and flow cytometric analysis of lung infiltrates. RESULTS Class I-restricted CD8 T cells responded rapidly to inhaled allergen and dominated the acute infiltration of T cells into the lung after secondary exposure. CD8 cells in the lung expressed a type 1 phenotype and suppressed the systemic IgE response to subsequent immunization. Dendritic cells purified from conducting airways or lung tissue were highly efficient at cross-presentation of antigen into the class I pathway after intranasal challenge. Adoptive transfer of transgenic antigen-specific CD8, but not CD4, cells resulted in increased IL-12 levels and reduced IL-13 and IL-5 levels in bronchoalveolar lavage fluid, coupled with substantially reduced airway eosinophilia after repeated allergen inhalation, a process mimicked by intranasal administration of IL-12 and inhibited by anti-IL-12 antibody. CONCLUSION The data suggest that CD8 cells specific for inhaled allergens are generated in draining lymph nodes but suppress allergic airway inflammation through induction of IL-12 in the lung during interaction with respiratory dendritic cells. CLINICAL IMPLICATIONS Novel peptide immunotherapeutics targeting the class I-restricted CD8 T-cell response to allergen represent a promising strategy for extrinsic asthma.
Collapse
|
1064
|
Buckler JL, Walsh PT, Porrett PM, Choi Y, Turka LA. Cutting edge: T cell requirement for CD28 costimulation is due to negative regulation of TCR signals by PTEN. THE JOURNAL OF IMMUNOLOGY 2006; 177:4262-6. [PMID: 16982858 DOI: 10.4049/jimmunol.177.7.4262] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Recent studies suggest that the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) plays a critical role in the maintenance of self-tolerance. Using T cell-specific PTEN knockout mice (PTENDeltaT), we have identified a novel mechanism by which PTEN regulates T cell tolerance. We found that TCR stimulation alone, without CD28 costimulation, is sufficient to induce hyperactivation of the PI3K pathway, which leads to enhanced IL-2 production by naive PTENDeltaT T cells. Importantly, as a result of this increased response to TCR stimulation, PTENDeltaT CD4(+) T cells no longer require CD28 costimulation for in vitro or in vivo expansion. In fact, unlike wild-type T cells, PTENDeltaT CD4(+) T cells are not anergized by delivery of TCR stimulation alone. These data suggest that by negatively regulating TCR signals, PTEN imposes a requirement for CD28 costimulation, thus defining a novel mechanism for its role in self-tolerance.
Collapse
Affiliation(s)
- Jodi L Buckler
- Department of Medicine, University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA 19104-6144, USA
| | | | | | | | | |
Collapse
|
1065
|
Li MO, Sanjabi S, Flavell RA. Transforming growth factor-beta controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity 2006; 25:455-71. [PMID: 16973386 DOI: 10.1016/j.immuni.2006.07.011] [Citation(s) in RCA: 638] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2006] [Revised: 06/22/2006] [Accepted: 07/12/2006] [Indexed: 10/24/2022]
Abstract
The role of transforming growth factor-beta (TGF-beta) in inhibiting T cell functions has been studied with dominant-negative TGF-beta receptor transgenic models; however, the full impact of TGF-beta signaling on T cells and the mechanisms by which TGF-beta signals remain poorly understood. Here we show that mice with T cell-specific deletion of TGF-beta receptor II developed lethal inflammation associated with T cell activation and differentiation. In addition, TGF-beta signaling positively regulated T cell development and homeostasis. Development of CD8+ T cells and NKT cells, maintenance of peripheral Foxp3-expressing regulatory T cells, and survival of CD4+ T cells all depended on TGF-beta signaling. Both T helper 1 (Th1) differentiation and survival of activated CD4+ T cells required T-bet, the TGF-beta-regulated transcription factor, which controlled CD122 expression and IL-15 signaling in Th1 cells. This study reveals pleiotropic functions of TGF-beta signaling in T cells that may ensure a diverse and self-tolerant T cell repertoire in vivo.
Collapse
Affiliation(s)
- Ming O Li
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | |
Collapse
|
1066
|
Okkenhaug K, Patton DT, Bilancio A, Garçon F, Rowan WC, Vanhaesebroeck B. The p110delta isoform of phosphoinositide 3-kinase controls clonal expansion and differentiation of Th cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:5122-8. [PMID: 17015696 DOI: 10.4049/jimmunol.177.8.5122] [Citation(s) in RCA: 175] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of PI3K in T cell activation and costimulation has been controversial. We previously reported that a kinase-inactivating mutation (D910A) in the p110delta isoform of PI3K results in normal T cell development, but impaired TCR-stimulated cell proliferation in vitro. This proliferative defect can be overcome by providing CD28 costimulation, which raises the question as to whether p110delta activity plays a role in T cell activation in vivo, which occurs primarily in the context of costimulation. In this study, we show that the PI3K signaling pathway in CD28-costimulated p110delta D910A/D910A T cells is impaired, but that ERK phosphorylation and NF-kappaB nuclear translocation are unaffected. Under in vitro conditions of physiological Ag presentation and costimulation, p110delta D910A/D910A T cells showed normal survival, but underwent fewer divisions. Differentiation along the Th1 and Th2 lineages was impaired in p110delta D910A/D910A T cells and could not be rescued by exogenous cytokines in vitro. Adoptive transfer and immunization experiments in mice revealed that clonal expansion and differentiation in response to Ag and physiological costimulation were also compromised. Thus, p110delta contributes significantly to Th cell expansion and differentiation in vitro and in vivo, also in the context of CD28 costimulation.
Collapse
Affiliation(s)
- Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom.
| | | | | | | | | | | |
Collapse
|
1067
|
Taraban VY, Rowley TF, Tough DF, Al-Shamkhani A. Requirement for CD70 in CD4+ Th cell-dependent and innate receptor-mediated CD8+ T cell priming. THE JOURNAL OF IMMUNOLOGY 2006; 177:2969-75. [PMID: 16920932 DOI: 10.4049/jimmunol.177.5.2969] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cell (DC) conditioning by CD4+ T cells, or via engagement of innate receptors, is thought to be essential for CD8+ T cell priming. However, the molecular features that distinguish a conditioned DC from an unconditioned DC are poorly defined. In this study, we investigate the role of CD70, a member of the TNF superfamily that is expressed on activated DC, in CD4+ Th-dependent and -independent CD8+ T cell responses. We demonstrate that CD70 is required for CD4+ T cell-dependent priming of CD8+ T cells as well as priming mediated by the viral signature, dsRNA. Accordingly, mice that were subjected to CD70 blockade during the primary response fail to generate a memory CD8+ T cell response. Furthermore, we find that CD70 is dispensable for CD4+ T cell expansion and help for B cells, thus suggesting a direct role for CD70 in CD8+ T cell priming. Our results show that the innate and adaptive (CD4+ T cells) arms of the immune system use a common signaling pathway in driving CD8+ T cell responses and suggest that expression of CD70 on DC represents the hallmark of conditioned DC.
Collapse
Affiliation(s)
- Vadim Y Taraban
- Tenovus Research Laboratory, Cancer Sciences Division, University of Southampton School of Medicine, Southampton General Hospital, Southampton, United Kingdom
| | | | | | | |
Collapse
|
1068
|
Yoshida M, Kobayashi K, Kuo TT, Bry L, Glickman JN, Claypool SM, Kaser A, Nagaishi T, Higgins DE, Mizoguchi E, Wakatsuki Y, Roopenian DC, Mizoguchi A, Lencer WI, Blumberg RS. Neonatal Fc receptor for IgG regulates mucosal immune responses to luminal bacteria. J Clin Invest 2006; 116:2142-2151. [PMID: 16841095 PMCID: PMC1501111 DOI: 10.1172/jci27821] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Accepted: 05/16/2006] [Indexed: 01/22/2023] Open
Abstract
The neonatal Fc receptor for IgG (FcRn) plays a major role in regulating host IgG levels and transporting IgG and associated antigens across polarized epithelial barriers. Selective expression of FcRn in the epithelium is shown here to be associated with secretion of IgG into the lumen that allows for defense against an epithelium-associated pathogen (Citrobacter rodentium). This pathway of host resistance to a bacterial pathogen as mediated by FcRn involves retrieval of bacterial antigens from the lumen and initiation of adaptive immune responses in regional lymphoid structures. Epithelial-associated FcRn, through its ability to secrete and absorb IgG, may thus integrate luminal antigen encounters with systemic immune compartments and as such provide essential host defense and immunoregulatory functions at the mucosal surfaces.
Collapse
Affiliation(s)
- Masaru Yoshida
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Kanna Kobayashi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Timothy T. Kuo
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Lynn Bry
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Jonathan N. Glickman
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Steven M. Claypool
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Arthur Kaser
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Takashi Nagaishi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Darren E. Higgins
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Emiko Mizoguchi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Yoshio Wakatsuki
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Derry C. Roopenian
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Atsushi Mizoguchi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Wayne I. Lencer
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Richard S. Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| |
Collapse
|
1069
|
Li H, Oliver T, Jia W, He YW. Efficient dendritic cell priming of T lymphocytes depends on the extracellular matrix protein mindin. EMBO J 2006; 25:4097-107. [PMID: 16917498 PMCID: PMC1560362 DOI: 10.1038/sj.emboj.7601289] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2006] [Accepted: 07/25/2006] [Indexed: 01/01/2023] Open
Abstract
Rho guanosine triphosphatases (GTPases) regulate multiple aspects of dendritic cell (DC) function, but what regulates the expression of Rho GTPases in DCs is unknown. Here, we show that the extracellular matrix protein mindin regulates the expression of Rho GTPases in DCs. Mindin(-/-) mice displayed defective CD4+ T-cell priming and impaired humoral immune responses to T-dependent antigens. Mindin(-/-) DCs had reduced expression of Rac1/2 and impaired priming capacity owing to inefficient engagement with T lymphocytes. Ectopic Rac1 expression restored the priming capability of Mindin(-/-) DCs. Furthermore, we show that DC adhesion to mindin matrix was blocked by antibodies to alpha4, alpha5 and beta1 integrins. DCs lacking beta1 integrin had reduced adhesion to mindin matrix, decreased expression of Rac1/2 and impaired priming capacity. These results suggest that mindin-integrin interactions play a key role in regulating Rho GTPase expression in DCs and DC priming of T lymphocytes.
Collapse
Affiliation(s)
- Hong Li
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Tim Oliver
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Wei Jia
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - You-Wen He
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
1070
|
Reinhardt RL, Hong S, Kang SJ, Wang ZE, Locksley RM. Visualization of IL-12/23p40 in vivo reveals immunostimulatory dendritic cell migrants that promote Th1 differentiation. THE JOURNAL OF IMMUNOLOGY 2006; 177:1618-27. [PMID: 16849470 DOI: 10.4049/jimmunol.177.3.1618] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IL-12p40 is induced in macrophages and dendritic cells (DC) after activation by microbial TLR ligands and cytokines and constitutes a component of IL-12 and IL-23. In an effort to understand the location and kinetics of these cytokines during the course of an immune response, we generated knockin (gene-targeted) mice that express the p40 gene linked via a viral internal ribosome entry site element with fluorescent reporters, eYFP or eGFP. Macrophages and DC from these mice faithfully reported biallelic p40 induction using the fluorescent marker. s.c. inoculation with Listeria monocytogenes or LPS led to a rapid, but transient, accumulation of p40-expressing DC in draining lymph nodes, which could be blocked by the addition of pertussis toxin. In situ analysis also revealed the accumulation of IL-12p40 protein around high endothelial venules located in close proximity to p40-expressing DC. Consistent with the in vivo findings, in vitro-activated DC that expressed p40 migrated to draining lymph nodes and promoted Th1 differentiation more efficiently than DC that did not express p40. Accordingly, these mice provide a valuable tool for tracking critical functions of DC in vivo and should bestow a useful reagent for exploring the effector biology of these cells in models of infectious disease, cancer immunity, and vaccine development.
Collapse
Affiliation(s)
- R Lee Reinhardt
- Howard Hughes Medical Institute, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
1071
|
McKenna KC, Kapp JA. Accumulation of immunosuppressive CD11b+ myeloid cells correlates with the failure to prevent tumor growth in the anterior chamber of the eye. THE JOURNAL OF IMMUNOLOGY 2006; 177:1599-608. [PMID: 16849468 DOI: 10.4049/jimmunol.177.3.1599] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The purpose of these studies is to determine why an immunogenic tumor grows unchecked in the anterior chamber (a.c.) of the eye. The OVA-expressing EL4 tumor, E.G7-OVA, was injected into the a.c. or skin of immunocompetent and immunodeficient mice. Tumor growth and tumor-specific immune responses were monitored. Ocular tumor-infiltrating leukocytes were characterized phenotypically and functionally. Growth of E.G7-OVA was inhibited when limiting numbers of cells were injected in the skin but not in the a.c. of C57BL/6 mice, although both routes primed OVA-specific immune responses, which prevented the growth of a subsequent injection with E.G7-OVA in the skin or opposite eye. Tumor regression was OVA-specific because growth of the parental EL-4 tumor was not inhibited in primed mice. E.G7-OVA growth in the skin was not inhibited in immunodeficient Rag(-/-) or CD8 T cell-deficient mice, suggesting that CD8(+) CTLs mediate tumor elimination. CD8(+) T cell numbers were significantly increased in eyes of mice primed with E.G7-OVA, but few were detected in primary ocular tumors. Nevertheless, growth of E.G7-OVA was retarded in the a.c. of TCR-transgenic OT-I mice, and CD8(+) T cell numbers were increased within eyes, suggesting that tumor-specific CD8(+) CTLs migrated into and controlled primary ocular tumor growth. E.G7-OVA did not lose antigenicity or become immunosuppressive after 13 days of growth in the eye. However, CD11b(+) cells accumulated in primary ocular tumors and contained potent immunosuppressive activity when assayed in vitro. Thus, CD11b(+) cells that accumulate within the eye as tumors develop in the a.c. may contribute to immune evasion by primary ocular tumors by inhibiting CTLs within the eye.
Collapse
Affiliation(s)
- Kyle C McKenna
- Department of Ophthalmology, Emory University, Atlanta, GA 30322, USA.
| | | |
Collapse
|
1072
|
Cruz A, Khader SA, Torrado E, Fraga A, Pearl JE, Pedrosa J, Cooper AM, Castro AG. Cutting edge: IFN-gamma regulates the induction and expansion of IL-17-producing CD4 T cells during mycobacterial infection. THE JOURNAL OF IMMUNOLOGY 2006; 177:1416-20. [PMID: 16849446 DOI: 10.4049/jimmunol.177.3.1416] [Citation(s) in RCA: 223] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
T cell responses are important to the control of infection but are deleterious if not regulated. IFN-gamma-deficient mice infected with mycobacteria exhibit enhanced accumulation of activated effector T cells and neutrophils within granulomatous lesions. These cells do not control bacterial growth and compromise the integrity of the infected tissue. We show that IFN-gamma-deficient mice have increased numbers of IL-17-producing T cells following infection with Mycobacterium bovis bacille Calmette Guérin. Furthermore, exogenous IFN-gamma increases IL-12 and decreases IL-23 production by bacille Calmette Guérin-infected bone marrow-derived dendritic cells and reduces the frequency of IL-17-producing T cells induced by these bone marrow-derived dendritic cells. These data support the hypothesis that, during mycobacterial infection, both IFN-gamma- and IL-17-producing T cells are induced, but that IFN-gamma serves to limit the IL-17-producing T cell population. This counterregulation pathway may be an important factor in limiting mycobacterially associated immune-mediated pathology.
Collapse
Affiliation(s)
- Andrea Cruz
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
1073
|
Bonasio R, Scimone ML, Schaerli P, Grabie N, Lichtman AH, von Andrian UH. Clonal deletion of thymocytes by circulating dendritic cells homing to the thymus. Nat Immunol 2006; 7:1092-100. [PMID: 16951687 DOI: 10.1038/ni1385] [Citation(s) in RCA: 315] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Accepted: 08/01/2006] [Indexed: 11/08/2022]
Abstract
Dendritic cell (DC) presentation of self antigen to thymocytes is essential to the establishment of central tolerance. We show here that circulating DCs were recruited to the thymic medulla through a three-step adhesion cascade involving P-selectin, interactions of the integrin VLA-4 with its ligand VCAM-1, and pertussis toxin-sensitive chemoattractant signaling. Ovalbumin-specific OT-II thymocytes were selectively deleted after intravenous injection of antigen-loaded exogenous DCs. We documented migration of endogenous DCs to the thymus in parabiotic mice and after painting mouse skin with fluorescein isothiocyanate. Antibody to VLA-4 blocked the accumulation of peripheral tissue-derived DCs in the thymus and also inhibited the deletion of OT-II thymocytes in mice expressing membrane-bound ovalbumin in cardiac myocytes. These findings identify a migratory route by which peripheral DCs may contribute to central tolerance.
Collapse
Affiliation(s)
- Roberto Bonasio
- The CBR Institute for Biomedical Research, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, USA
| | | | | | | | | | | |
Collapse
|
1074
|
Dullaers M, Van Meirvenne S, Heirman C, Straetman L, Bonehill A, Aerts JL, Thielemans K, Breckpot K. Induction of effective therapeutic antitumor immunity by direct in vivo administration of lentiviral vectors. Gene Ther 2006; 13:630-40. [PMID: 16355115 DOI: 10.1038/sj.gt.3302697] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Ex vivo lentivirally transduced dendritic cells (DC) have been described to induce CD8+ and CD4+ T-cell responses against various tumor-associated antigens (TAAs) in vitro and in vivo. We report here that direct administration of ovalbumin (OVA) encoding lentiviral vectors caused in vivo transduction of cells that were found in draining lymph nodes (LNs) and induced potent anti-OVA cytotoxic T cells similar to those elicited by ex vivo transduced DC. The cytotoxic T-lymphocyte (CTL) response following direct injection of lentiviral vectors was highly effective in eliminating target cells in vivo up to 30 days after immunization and was efficiently recalled after a boost immunization. Injection of lentiviral vectors furthermore activated OVA-specific CD4+ T cells and this CD4 help was shown to be necessary for an adequate primary and memory CTL response. When tested in therapeutic tumor experiments with OVA+ melanoma cells, direct administration of lentiviral vectors slowed down tumor growth to a comparable extent with the highest dose of ex vivo transduced DC. Taken together, these data indicate that direct in vivo administration of lentiviral vectors encoding TAAs has strong potential for anticancer vaccination.
Collapse
Affiliation(s)
- M Dullaers
- Laboratory of Molecular and Cellular Therapy, Department of Physiology-Immunology, Medical School of the Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
1075
|
Rudd ML, Tua-Smith A, Straus DB. Lck SH3 domain function is required for T-cell receptor signals regulating thymocyte development. Mol Cell Biol 2006; 26:7892-900. [PMID: 16923964 PMCID: PMC1636743 DOI: 10.1128/mcb.00968-06] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Thymocyte development is shaped by signals from the T-cell antigen receptor. The strength of receptor signaling determines developmental progression as well as deletion of self-reactive T cells. Receptor stimulation of the extracellular signal-regulated kinase (ERK) pathway plays an important regulatory role during thymocyte development. However, it is unclear how differences in receptor signaling are translated into distinctive activation of the ERK pathway. We have investigated the potential role of the Lck tyrosine kinase in regulating intracellular signaling during thymocyte development. While Lck is known to be critical for initial T-cell receptor signaling events, it may have an independent role in regulating intracellular signaling through the function of its SH3 domain. To determine whether such a regulatory mechanism functions during thymocyte development, we generated mice in which the normal lck allele is replaced with an lck SH3 domain mutant. Analysis of these mice revealed that both early thymocyte development and maturation of CD4(+) and CD8(+) lineages is impaired. Investigation of thymocyte responses to antigen receptor stimulation showed a significant reduction in proliferation and ERK pathway activation, although initial signaling events were intact. These findings indicate that Lck SH3 domain function may provide a means to independently couple receptor signaling to regulation of the ERK pathway during thymocyte development.
Collapse
Affiliation(s)
- Meghan L Rudd
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | |
Collapse
|
1076
|
Kessels HWHG, Schepers K, van den Boom MD, Topham DJ, Schumacher TNM. Generation of T cell help through a MHC class I-restricted TCR. THE JOURNAL OF IMMUNOLOGY 2006; 177:976-82. [PMID: 16818753 DOI: 10.4049/jimmunol.177.2.976] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD4+ T cells that are activated by a MHC class II/peptide encounter can induce maturation of APCs and promote cytotoxic CD8+ T cell responses. Unfortunately, the number of well-defined tumor-specific CD4+ T cell epitopes that can be exploited for adoptive immunotherapy is limited. To determine whether Th cell responses can be generated by redirecting CD4+ T cells to MHC class I ligands, we have introduced MHC class I-restricted TCRs into postthymic murine CD4+ T cells and examined CD4+ T cell activation and helper function in vitro and in vivo. These experiments indicate that Ag-specific CD4+ T cell help can be induced by the engagement of MHC class I-restricted TCRs in peripheral CD4+ T cells but that it is highly dependent on the coreceptor function of the CD8beta-chain. The ability to generate Th cell immunity by infusion of MHC class I-restricted Th cells may prove useful for the induction of tumor-specific T cell immunity in cases where MHC class II-associated epitopes are lacking.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- CD8 Antigens/physiology
- Cell Differentiation/immunology
- Cell Line
- Cell Proliferation
- Dimerization
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/metabolism
- Intracellular Fluid/immunology
- Intracellular Fluid/metabolism
- Ligands
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/physiology
- Signal Transduction/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
- Helmut W H G Kessels
- Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, CX 1066 Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
1077
|
Bertrand JY, Desanti GE, Lo-Man R, Leclerc C, Cumano A, Golub R. Fetal spleen stroma drives macrophage commitment. Development 2006; 133:3619-28. [PMID: 16914502 DOI: 10.1242/dev.02510] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The role of the fetal spleen in hematopoeisis remains largely unknown. In this particular environment, we show that hematopoietic stem cells do not proliferate, but that they lose multipotency and differentiate exclusively into mature macrophages. B lymphocytes in the spleen derive from committed B cell precursors that are likely to have immigrated from the fetal liver. We developed fetal spleen stromal cell lines that are unique in their capacity to expand myeloid precursors, resulting in large numbers of mature macrophages. These lines secrete high levels of anti-inflammatory molecules. By phenotype, fetal splenic macrophages are reminiscent of their adult counterparts found in the red pulp. We postulate that F4/80(+) splenic macrophages participate in fetal erythropoiesis, as well as in the formation of the splenic architecture.
Collapse
Affiliation(s)
- Julien Y Bertrand
- Unité du Développement des Lymphocytes, INSERM U668, Institut Pasteur, 25, Rue du Dr Roux, 75724 Paris cedex 15, France
| | | | | | | | | | | |
Collapse
|
1078
|
Stachowiak AN, Wang Y, Huang YC, Irvine DJ. Homeostatic Lymphoid Chemokines Synergize with Adhesion Ligands to Trigger T and B Lymphocyte Chemokinesis. THE JOURNAL OF IMMUNOLOGY 2006; 177:2340-8. [PMID: 16887995 DOI: 10.4049/jimmunol.177.4.2340] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Homeostatic chemokines such as CCL19, CCL21, and CXCL13 are known to elicit chemotaxis from naive T and B cells and play a critical role in lymphocyte homing to appropriate zones within secondary lymphoid organs (SLO). Here we tested whether CCL21 and CXCL13 modulate murine lymphocyte motility in the absence of concentration gradients, using videomicroscopy to directly observe the migration of single cells. CCL21 treatment of T cells induced rapid polarization and sustained random migration with average speeds of 5.16 +/- 2.08 microm/min; B cell migration (average velocity 4.10 +/- 1.58 microm/min) was similarly induced by CXCL13. Migration required the presence of both chemokine and adhesion ligands and was sustained for >24 h. Furthermore, in in vitro assays modeling the relative infrequency of Ag-specific T cell-dendritic cell (DC) encounters during primary immune responses, we found that CCL21 addition to T-DC cocultures accelerated the kinetics of CD69 up-regulation and enhanced by 2-fold the proliferation of Ag-specific T cells in a manner dependent on G-protein-coupled receptor signaling in T cells. These results suggest that homeostatic chemokines could substantially impact the dynamics and priming of lymphocytes within SLO even in the absence of significant concentration gradients.
Collapse
Affiliation(s)
- Agnieszka N Stachowiak
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Boston, MA 02139, USA
| | | | | | | |
Collapse
|
1079
|
Iezzi G, Boni A, Degl'Innocenti E, Grioni M, Bertilaccio MTS, Bellone M. Type 2 Cytotoxic T Lymphocytes Modulate the Activity of Dendritic Cells Toward Type 2 Immune Responses. THE JOURNAL OF IMMUNOLOGY 2006; 177:2131-7. [PMID: 16887972 DOI: 10.4049/jimmunol.177.4.2131] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Activated CD8+ T cells can differentiate into type 1 (Tc1) cells, producing mainly IFN-gamma, and type 2 (Tc2) cells, producing mostly IL-4, IL-5, and IL-10. Tc1 cells are potent CTL involved in the defense against intracellular pathogens and cancer cells. The role of Tc2 cells in the immune response is largely unknown, although their presence in chronic infections, cancer, and autoimmune diseases is associated with disease severity and progression. Here, we show that mouse Tc2 cells modify, through a cell-to-cell contact mechanism, the function of bone marrow-derived dendritic cells (DC). Indeed, Tc2-conditioned DC displayed a reduced expression of MHC class II and costimulatory molecules, produced IL-10 instead of IL-12, and favored the differentiation of both naive CD4+ and CD8+ T cells toward type 2 cells in the absence of added polarizing cytokines. The novel function for Tc2 cells suggests a type 2 loop in which Tc2 cells modify DC function and favor differentiation of naive T cells to type 2 cells. The type 2 loop may at least in part explain the unexpected high frequency of type 2 cells during a chronic exposure to the Ag.
Collapse
Affiliation(s)
- Giandomenica Iezzi
- Cancer Immunotherapy and Gene Therapy Program, Istituto Scientifico H San Raffaele, Via Olgettina 58, 20132 Milan, Italy
| | | | | | | | | | | |
Collapse
|
1080
|
Lüthje K, Cramer SO, Ehrlich S, Veit A, Steeg C, Fleischer B, Bonin AV, Breloer M. Transgenic expression of a CD83-immunoglobulin fusion protein impairs the development of immune-competent CD4-positive T cells. Eur J Immunol 2006; 36:2035-45. [PMID: 16841299 DOI: 10.1002/eji.200636068] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The murine transmembrane glycoprotein CD83 is an important regulator for both thymic T cell maturation and peripheral T cell response. CD83 deficiency leads to a block in the thymic maturation of CD4-positive T cells, and interference with peripheral CD83/CD83 ligand interaction by addition of soluble CD83 suppresses immune responses in vivo and in vitro. Here we report the generation of a mouse transgenic for a fusion protein consisting of the extracellular domain of murine CD83 fused to the constant part of human IgG1 heavy chain. Thymic selection of CD4-positive T cells was unchanged in CD83Ig transgenic and in CD83Ig/OT-2 double-transgenic mice. However, thymic and peripheral CD4-positive T cells derived from CD83Ig/OT-2 transgenic mice displayed a reduced cytokine response to antigenic stimulation in vitro, whereas CD83Ig/OT-1-derived CD8-positive T cells showed normal cytokine secretion. The T cell defect was relevant in vivo, since a sub-lethal infection with Trypanosoma cruzi led to an increased parasitemia and reduced survival rate of CD83Ig transgenic mice compared to wild-type C57BL/6 mice. In contrast, in vivo application of recombinant CD83Ig did not result in an increase in parasitemia. Taken together our data suggest that thymic selection in the presence of CD83Ig leads to an intrinsic T cell defect of CD4-positive T cells resembling the phenotype described for CD4-positive T cells derived from CD83-deficient mouse strains.
Collapse
Affiliation(s)
- Katja Lüthje
- Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
1081
|
Abstract
Although thymic ectopy has long been recognized in humans, the functional activity or potential immunological significance of this thymic tissue is unknown. In this study, we describe murine thymic ectopy, cervical thymic tissue that possesses the same general organization as the thoracic thymus, that is able to support T cell differentiation, and that can export T cells to the periphery. Unexpectedly, the pattern of autoantigen expression by ectopic thymic tissue differs from that of the thoracic thymus, raising the possibility that these two thymic environments may project different versions of self.
Collapse
Affiliation(s)
- James Dooley
- Department of Biological Structure, University of Washinton School of Medicine, Seattle, WA 98195, USA
| | | | | | | |
Collapse
|
1082
|
Leithäuser F, Meinhardt-Krajina T, Fink K, Wotschke B, Möller P, Reimann J. Foxp3-expressing CD103+ regulatory T cells accumulate in dendritic cell aggregates of the colonic mucosa in murine transfer colitis. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1898-909. [PMID: 16723705 PMCID: PMC1606612 DOI: 10.2353/ajpath.2006.050228] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Little is known of the anatomical compartmentalization of colitogenic or regulatory T-cell responses in the murine transfer colitis model. Therefore, we analyzed the putative function of large intestinal dendritic cell (DC) aggregates, to which donor CD4+ T cells selectively home before colitis becomes manifest. The co-stimulatory molecules MHC-II, CD40, CD80, and CD86 were expressed in DC aggregates. IL-23 was primarily absent from DC aggregates at all stages of disease but was expressed at high levels in the severely inflamed lamina propria. Interferon-gamma was up-regulated in the lamina propria during early and advanced disease, whereas in DC aggregates it was detectable to a significant degree only in fully developed colitis. In contrast, Foxp3, a marker of regulatory T cells, was expressed in DC aggregates on T-cell transfer, coinciding with the appearance of CD103+ CD25- T cells in these clusters. Foxp3 was enriched in the CD103+ T-cell fraction isolated from the lamina propria of diseased mice. T-cell grafts depleted of CD103+ T cells generated similar numbers of colonic CD103+ T cells as unfractionated T cells. We conclude that DC aggregates are structures involved in the expansion and/or differentiation of CD103+ CD25- CD4+ Foxp3-expressing regulatory T cells.
Collapse
Affiliation(s)
- Frank Leithäuser
- Department of Pathology, University of Ulm, Albert Einstein Allee 11, 89081 Ulm, Germany.
| | | | | | | | | | | |
Collapse
|
1083
|
Garrod KR, Chang CK, Liu FC, Brennan TV, Foster RD, Kang SM. Targeted Lymphoid Homing of Dendritic Cells Is Required for Prolongation of Allograft Survival. THE JOURNAL OF IMMUNOLOGY 2006; 177:863-8. [DOI: 10.4049/jimmunol.177.2.863] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
1084
|
Khader SA, Partida-Sanchez S, Bell G, Jelley-Gibbs DM, Swain S, Pearl JE, Ghilardi N, Desauvage FJ, Lund FE, Cooper AM. Interleukin 12p40 is required for dendritic cell migration and T cell priming after Mycobacterium tuberculosis infection. ACTA ACUST UNITED AC 2006; 203:1805-15. [PMID: 16818672 PMCID: PMC2118335 DOI: 10.1084/jem.20052545] [Citation(s) in RCA: 248] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Migration of dendritic cells (DCs) to the draining lymph node (DLN) is required for the activation of naive T cells. We show here that migration of DCs from the lung to the DLN after Mycobacterium tuberculosis (Mtb) exposure is defective in mice lacking interleukin (IL)-12p40. This defect compromises the ability of IL-12p40–deficient DCs to activate naive T cells in vivo; however, DCs that express IL-12p40 alone can activate naive T cells. Treatment of IL-12p40–deficient DCs with IL-12p40 homodimer (IL-12(p40)2) restores Mtb-induced DC migration and the ability of IL-12p40–deficient DCs to activate naive T cells. These data define a novel and fundamental role for IL-12p40 in the pathogen-induced activation of pulmonary DCs.
Collapse
|
1085
|
Datta SK, Okamoto S, Hayashi T, Shin SS, Mihajlov I, Fermin A, Guiney DG, Fierer J, Raz E. Vaccination with Irradiated Listeria Induces Protective T Cell Immunity. Immunity 2006; 25:143-52. [PMID: 16860763 DOI: 10.1016/j.immuni.2006.05.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 04/03/2006] [Accepted: 05/17/2006] [Indexed: 12/14/2022]
Abstract
We evaluated gamma-irradiated Listeria monocytogenes as a killed bacterial vaccine, testing the hypothesis that irradiation preserves antigenic and adjuvant structures destroyed by traditional heat or chemical inactivation. Irradiated Listeria monocytogenes (LM), unlike heat-killed LM, efficiently activated dendritic cells via Toll-like receptors and induced protective T cell responses in mice. Like live LM, irradiated LM induced Toll-like-receptor-independent T cell priming. Cross-presentation of irradiated listerial antigens to CD8(+) T cells involved TAP- and proteasome-dependent cytosolic antigen processing. These results establish that killed LM can induce protective T cell responses, previously thought to require live infection. gamma-irradiation may be potentially applied to numerous bacterial vaccine candidates, and irradiated bacteria could serve as a vaccine platform for recombinant antigens derived from other pathogens, allergens, or tumors.
Collapse
Affiliation(s)
- Sandip K Datta
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
1086
|
Schnorrer P, Behrens GMN, Wilson NS, Pooley JL, Smith CM, El-Sukkari D, Davey G, Kupresanin F, Li M, Maraskovsky E, Belz GT, Carbone FR, Shortman K, Heath WR, Villadangos JA. The dominant role of CD8+ dendritic cells in cross-presentation is not dictated by antigen capture. Proc Natl Acad Sci U S A 2006; 103:10729-34. [PMID: 16807294 PMCID: PMC1502299 DOI: 10.1073/pnas.0601956103] [Citation(s) in RCA: 309] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Mouse spleens contain three populations of conventional (CD11c(high)) dendritic cells (DCs) that play distinct functions. The CD8(+) DC are unique in that they can present exogenous antigens on their MHC class I molecules, a process known as cross-presentation. It is unclear whether this special ability is because only the CD8(+) DC can capture the antigens used in cross-presentation assays, or because this is the only DC population that possesses specialized machinery for cross-presentation. To solve this important question we examined the splenic DC subsets for their ability to both present via MHC class II molecules and cross-present via MHC class I using four different forms of the model antigen ovalbumin (OVA). These forms include a cell-associated form, a soluble form, OVA expressed in bacteria, or OVA bound to latex beads. With the exception of bacterial antigen, which was poorly cross-presented by all DC, all antigenic forms were cross-presented much more efficiently by the CD8(+) DC. This pattern could not be attributed simply to a difference in antigen capture because all DC subsets presented the antigen via MHC class II. Indeed, direct assessments of endocytosis showed that CD8(+) and CD8(-) DC captured comparable amounts of soluble and bead-associated antigen, yet only the CD8(+) DC cross-presented these antigenic forms. Our results indicate that cross-presentation requires specialized machinery that is expressed by CD8(+) DC but largely absent from CD8(-) DC. This conclusion has important implications for the design of vaccination strategies based on antigen targeting to DC.
Collapse
Affiliation(s)
| | | | - Nicholas S. Wilson
- *Immunology Division and
- Cooperative Research Centre for Vaccine Technology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia; Departments of
- Medical Biology and
| | | | - Christopher M. Smith
- *Immunology Division and
- Cooperative Research Centre for Vaccine Technology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia; Departments of
- Medical Biology and
| | | | | | | | - Ming Li
- *Immunology Division and
- Medical Biology and
| | | | - Gabrielle T. Belz
- *Immunology Division and
- Cooperative Research Centre for Vaccine Technology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia; Departments of
| | - Francis R. Carbone
- Microbiology and Immunology, University of Melbourne, Parkville, Victoria 3010, Australia; and
| | | | - William R. Heath
- *Immunology Division and
- Cooperative Research Centre for Vaccine Technology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia; Departments of
- To whom correspondence may be addressed at:
Immunology Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia. E-mail:
or
| | - Jose A. Villadangos
- *Immunology Division and
- Cooperative Research Centre for Vaccine Technology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia; Departments of
- To whom correspondence may be addressed at:
Immunology Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia. E-mail:
or
| |
Collapse
|
1087
|
Soroosh P, Ine S, Sugamura K, Ishii N. OX40-OX40 ligand interaction through T cell-T cell contact contributes to CD4 T cell longevity. THE JOURNAL OF IMMUNOLOGY 2006; 176:5975-87. [PMID: 16670306 DOI: 10.4049/jimmunol.176.10.5975] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Signals through the OX40 costimulatory receptor on naive CD4 T cells are essential for full-fledged CD4 T cell activation and the generation of CD4 memory T cells. Because the ligand for OX40 is mainly expressed by APCs, including activated B cells, dendritic cells, and Langerhans cells, the OX40-OX40 ligand (OX40L) interaction has been thought to participate in T cell-APC interactions. Although several reports have revealed the expression of OX40L on T cells, the functional significance of its expression on them is still unclear. In this study, we demonstrate that Ag stimulation induced an increase in the surface expression and transcript levels of OX40L in CD4 T cells. Upon contact with OX40-expressing T cells, the cell surface expression of OX40L on CD4 T cells was markedly down-regulated, suggesting that OX40-OX40L binding occurs through a novel T cell-T cell interaction. To investigate the function of this phenomenon, we examined the proliferative response and survival of OX40L-deficient CD4 T cells when challenged with Ag. In vitro studies demonstrated markedly less CD3-induced proliferation of OX40L-deficient CD4 T cells compared with wild-type CD4 T cells. When using TCR transgenic CD4 T cells upon Ag stimulation, survival of OX40L-deficient T cells was impaired. Furthermore, we show that upon antigenic stimulation, fewer OX40L-deficient CD4 T cells than wild-type cells survived following transfer into wild-type and sublethally irradiated recipient mice. Taken together, our findings indicate that OX40L-expressing T cells have an autonomous machinery that provides OX40 signals through a T cell-T cell circuit, creating an additional mechanism for sustaining CD4 T cell longevity.
Collapse
Affiliation(s)
- Pejman Soroosh
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | |
Collapse
|
1088
|
Ohmura-Hoshino M, Matsuki Y, Aoki M, Goto E, Mito M, Uematsu M, Kakiuchi T, Hotta H, Ishido S. Inhibition of MHC Class II Expression and Immune Responses by c-MIR. THE JOURNAL OF IMMUNOLOGY 2006; 177:341-54. [PMID: 16785530 DOI: 10.4049/jimmunol.177.1.341] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We previously reported a novel E3 ubiquitin ligase (E3), designated as c-MIR, which targets B7-2 to lysosomal degradation and down-regulates the B7-2 surface expression through ubiquitination of its cytoplasmic tail. B7-2 is well known as a costimulatory molecule for Ag presentation, suggesting that the manipulation of c-MIR expression modulates immune responses in vivo. To examine this hypothesis, we generated genetically modified mice in which c-MIR was expressed under an invariant chain (Ii) promoter. Dendritic cells derived from genetically engineered mice showed low ability to present Ags. In addition, these mice showed resistance to the onset of experimental autoimmune encephalomyelitis and an impaired development of CD4 T cells in the thymus and the periphery. These findings led us to conclude that MHC class II (MHC II) is an additional target for c-MIR. Indeed, forced expression of c-MIR in several B cell lines down-regulated the surface expression of MHC II, and down-regulation was found to depend on the presence of a single lysine residue in the cytoplasmic tail of the I-A beta-chain. In a reconstitution system using 293T cells, we found that the lysine residue at position 225 in the I-A beta-chain was ubiquitinated by c-MIR. To our knowledge, c-MIR is the first example of an E3 that is capable of inhibiting MHC II expression. Our findings suggest that c-MIR might potently regulate immune responses in vivo.
Collapse
Affiliation(s)
- Mari Ohmura-Hoshino
- Laboratory for Infectious Immunity, The Institute of Physical and Chemical Research (RIKEN), Research Center for Allergy and Immunology, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
1089
|
Mueller SN, Jones CM, Stock AT, Suter M, Heath WR, Carbone FR. CD4+ T Cells Can Protect APC from CTL-Mediated Elimination. THE JOURNAL OF IMMUNOLOGY 2006; 176:7379-84. [PMID: 16751382 DOI: 10.4049/jimmunol.176.12.7379] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Professional APC play a central role in generating antiviral CD8(+) CTL immunity. However, the fate of such APC following interaction with these same CTL remains poorly understood. We have shown previously that prolonged Ag presentation persists in the presence of a strong CTL response following HSV infection. In this study, we examined the mechanism of survival of APC in vivo when presenting an immunodominant determinant from HSV. We show that transferred peptide-labeled dendritic cells were eliminated from draining lymph nodes in the presence of HSV-specific CTL. Maturation of dendritic cells with LPS or anti-CD40 before injection protected against CTL lysis in vivo. Furthermore, endogenous APC could be eliminated from draining lymph nodes early after HSV infection by adoptive transfer of HSV-specific CTL, yet the cotransfer of significant virus-specific CD4(+) T cell help promoted prolonged Ag presentation. This suggests that Th cells may assist in prolonging class I-restricted Ag presentation, potentially enhancing CTL recruitment and allowing more efficient T cell priming.
Collapse
Affiliation(s)
- Scott N Mueller
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Australia
| | | | | | | | | | | |
Collapse
|
1090
|
Huddleston SJ, Hays WS, Filatenkov A, Ingulli E, Jenkins MK. CD154+ graft antigen-specific CD4+ T cells are sufficient for chronic rejection of minor antigen incompatible heart grafts. Am J Transplant 2006; 6:1312-9. [PMID: 16686755 DOI: 10.1111/j.1600-6143.2006.01309.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We used a defined model system to address the role of minor histocompatibility antigen-specific CD4+ T cells in chronic rejection. The coronary arteries of vascularized heart grafts expressing the model antigen ovalbumin developed intimal hyperplasia in normal recipients and those lacking CD8+ T cells but not in those lacking CD4+ T cells. Furthermore, purified ovalbumin-specific CD4+ T cells from T-cell antigen receptor transgenic mice caused intimal hyperplasia in ovalbumin-expressing heart grafts in lymphocyte-deficient mice. The graft antigen-specific CD4+ T cells only caused intimal hyperplasia when expressing CD154 and were found in the intima of the affected coronary arteries along with CD40+ cells, CD11c+ dendritic cells and CD11b+, Gr-1+ monocytes. These results show that minor histocompatibility antigen-specific CD4+ T cells are required to cause the classical vascular changes of chronic rejection. They are capable of doing so without contributions from other lymphocytes, and may cause intimal hyperplasia by using CD154 to stimulate other non-lymphoid cells in the intima.
Collapse
Affiliation(s)
- S J Huddleston
- Department of Surgery, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| | | | | | | | | |
Collapse
|
1091
|
Takegahara N, Takamatsu H, Toyofuku T, Tsujimura T, Okuno T, Yukawa K, Mizui M, Yamamoto M, Prasad DVR, Suzuki K, Ishii M, Terai K, Moriya M, Nakatsuji Y, Sakoda S, Sato S, Akira S, Takeda K, Inui M, Takai T, Ikawa M, Okabe M, Kumanogoh A, Kikutani H. Plexin-A1 and its interaction with DAP12 in immune responses and bone homeostasis. Nat Cell Biol 2006; 8:615-22. [PMID: 16715077 DOI: 10.1038/ncb1416] [Citation(s) in RCA: 201] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Accepted: 04/28/2006] [Indexed: 01/08/2023]
Abstract
Semaphorins and their receptors have diverse functions in axon guidance, organogenesis, vascularization and/or angiogenesis, oncogenesis and regulation of immune responses. The primary receptors for semaphorins are members of the plexin family. In particular, plexin-A1, together with ligand-binding neuropilins, transduces repulsive axon guidance signals for soluble class III semaphorins, whereas plexin-A1 has multiple functions in chick cardiogenesis as a receptor for the transmembrane semaphorin, Sema6D, independent of neuropilins. Additionally, plexin-A1 has been implicated in dendritic cell function in the immune system. However, the role of plexin-A1 in vivo, and the mechanisms underlying its pleiotropic functions, remain unclear. Here, we generated plexin-A1-deficient (plexin-A1(-/-)) mice and identified its important roles, not only in immune responses, but also in bone homeostasis. Furthermore, we show that plexin-A1 associates with the triggering receptor expressed on myeloid cells-2 (Trem-2), linking semaphorin-signalling to the immuno-receptor tyrosine-based activation motif (ITAM)-bearing adaptor protein, DAP12. These findings reveal an unexpected role for plexin-A1 and present a novel signalling mechanism for exerting the pleiotropic functions of semaphorins.
Collapse
Affiliation(s)
- Noriko Takegahara
- Department of Molecular Immunology and CREST program of JST, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1092
|
Dillon S, Agrawal S, Banerjee K, Letterio J, Denning TL, Oswald-Richter K, Kasprowicz DJ, Kellar K, Pare J, van Dyke T, Ziegler S, Unutmaz D, Pulendran B. Yeast zymosan, a stimulus for TLR2 and dectin-1, induces regulatory antigen-presenting cells and immunological tolerance. J Clin Invest 2006; 116:916-28. [PMID: 16543948 PMCID: PMC1401484 DOI: 10.1172/jci27203] [Citation(s) in RCA: 418] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 01/24/2006] [Indexed: 01/08/2023] Open
Abstract
Emerging evidence suggests critical roles for APCs in suppressing immune responses. Here, we show that zymosan, a stimulus for TLR2 and dectin-1, regulates cytokine secretion in DCs and macrophages to induce immunological tolerance. First, zymosan induces DCs to secrete abundant IL-10 but little IL-6 and IL-12(p70). Induction of IL-10 is dependent on TLR2- and dectin-1-mediated activation of ERK MAPK via a mechanism independent of the activation protein 1 (AP-1) transcription factor c-Fos. Such DCs stimulate antigen-specific CD4+ T cells poorly due to IL-10 and the lack of IL-6. Second, zymosan induces F4-80+ macrophages in the splenic red pulp to secrete TGF-beta. Consistent with these effects on APCs, injection of zymosan plus OVA into mice results in OVA-specific T cells that secrete little or no Th1 or Th2 cytokines, but secrete robust levels of IL-10, and are unresponsive to challenge with OVA plus adjuvant. Finally, coinjection of zymosan with OVA plus LPS suppresses the response to OVA via a mechanism dependent on IL-10, TGF-beta, and lack of IL-6. Together, our data demonstrate that zymosan stimulates IL-10+ IL-12(p70)- IL-6low regulatory DCs and TGF-beta+ macrophages to induce immunological tolerance. These data suggest several targets for pharmacological modulation of immune responses in various clinical settings.
Collapse
Affiliation(s)
- Stephanie Dillon
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sudhanshu Agrawal
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kaustuv Banerjee
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - John Letterio
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Timothy L. Denning
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kyra Oswald-Richter
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Deborah J. Kasprowicz
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kathryn Kellar
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jeff Pare
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Thomas van Dyke
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Derya Unutmaz
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Bali Pulendran
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
1093
|
Holst J, Vignali KM, Burton AR, Vignali DAA. Rapid analysis of T-cell selection in vivo using T cell-receptor retrogenic mice. Nat Methods 2006; 3:191-7. [PMID: 16489336 DOI: 10.1038/nmeth858] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2005] [Accepted: 01/20/2006] [Indexed: 01/09/2023]
Abstract
Although T-cell receptor (TCR) transgenic as well as knockout and knockin mice have had a large impact on our understanding of T-cell development, signal transduction and function, the need to cross these mice delays experiments considerably. Here we provide a methodology for the rapid expression of TCRs in mice using 2A peptide-linked multicistronic retroviral vectors to transduce stem cells of any background before adoptive transfer into RAG-1(-/-) mice. For simplicity, we refer to these as retrogenic mice. We demonstrate that these retrogenic mice are comparable to transgenic mice expressing three commonly used TCRs (OT-I, OT-II [corrected] and AND). We also show that retrogenic mice expressing male antigen-specific TCRs (HY, MataHari and Marilyn) facilitated the analysis of positive and negative selection in female and male mice, respectively. We examined various tolerance mechanisms in epitope-coupled TCR retrogenic mice. This powerful resource could expedite the identification of proteins involved in T-cell development and function.
Collapse
Affiliation(s)
- Jeff Holst
- Department of Immunology, St. Jude Children's Research Hospital, 332 N. Lauderdale, Memphis, Tennessee 38105, USA
| | | | | | | |
Collapse
|
1094
|
Stoitzner P, Tripp CH, Eberhart A, Price KM, Jung JY, Bursch L, Ronchese F, Romani N. Langerhans cells cross-present antigen derived from skin. Proc Natl Acad Sci U S A 2006; 103:7783-8. [PMID: 16672373 PMCID: PMC1472522 DOI: 10.1073/pnas.0509307103] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Dendritic cells (DC) efficiently cross-present exogenous antigen on MHC class I molecules to CD8+ T cells. However, little is known about cross-presentation by Langerhans cells (LC), the DCs of the epidermis. Therefore, we investigated this issue in detail. Isolated murine LCs were able to cross-present soluble ovalbumin protein on MHC-class I molecules to antigen-specific CD8+ T cells, albeit less potently than the CD8+ DC subsets from spleen. Furthermore, LCs cross-presented cell-associated ovalbumin peptide and protein expressed by neighboring keratinocytes. Use of transporter associated with antigen processing (TAP-1)-deficient mice suggested a TAP-dependent pathway. Similar observations were made with migratory LC. Antigen expressed in the epidermis was ingested by LCs during migration from the epidermis and presented to antigen-specific T cells in vitro. Cross-presentation of ovalbumin protein by LCs induced IFN-gamma production and cytotoxicity in antigen-specific CD8+ T cells. Additionally, epicutaneous application of ovalbumin protein induced in vivo proliferation of OT-I T cells in the draining lymph nodes; this was markedly enhanced when antigen was applied to inflamed, barrier-disrupted skin. Thus, LCs cross-present exogenous antigen to CD8+ T cells and induce effector functions, like cytokine production and cytotoxicity, and may thereby critically contribute in epicutaneous vaccination approaches.
Collapse
Affiliation(s)
- Patrizia Stoitzner
- Departments of *Dermatology and
- Malaghan Institute of Medical Research, Wellington 6005, New Zealand; and
| | | | - Andreas Eberhart
- Biochemical Pharmacology, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Kylie M. Price
- Malaghan Institute of Medical Research, Wellington 6005, New Zealand; and
| | - Jae Y. Jung
- Malaghan Institute of Medical Research, Wellington 6005, New Zealand; and
| | - Laura Bursch
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington 6005, New Zealand; and
| | - Nikolaus Romani
- Departments of *Dermatology and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
1095
|
Castellino F, Huang AY, Altan-Bonnet G, Stoll S, Scheinecker C, Germain RN. Chemokines enhance immunity by guiding naive CD8+ T cells to sites of CD4+ T cell-dendritic cell interaction. Nature 2006; 440:890-5. [PMID: 16612374 DOI: 10.1038/nature04651] [Citation(s) in RCA: 667] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Accepted: 02/15/2006] [Indexed: 01/19/2023]
Abstract
CD8+ T cells have a crucial role in resistance to pathogens and can kill malignant cells; however, some critical functions of these lymphocytes depend on helper activity provided by a distinct population of CD4+ T cells. Cooperation between these lymphocyte subsets involves recognition of antigens co-presented by the same dendritic cell, but the frequencies of such antigen-bearing cells early in an infection and of the relevant naive T cells are both low. This suggests that an active mechanism facilitates the necessary cell-cell associations. Here we demonstrate that after immunization but before antigen recognition, naive CD8+ T cells in immunogen-draining lymph nodes upregulate the chemokine receptor CCR5, permitting these cells to be attracted to sites of antigen-specific dendritic cell-CD4+ T cell interaction where the cognate chemokines CCL3 and CCL4 (also known as MIP-1alpha and MIP-1beta) are produced. Interference with this actively guided recruitment markedly reduces the ability of CD4+ T cells to promote memory CD8+ T-cell generation, indicating that an orchestrated series of differentiation events drives nonrandom cell-cell interactions within lymph nodes, optimizing CD8+ T-cell immune responses involving the few antigen-specific precursors present in the naive repertoire.
Collapse
Affiliation(s)
- Flora Castellino
- Lymphocyte Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
1096
|
Foulds KE, Shen H. Clonal competition inhibits the proliferation and differentiation of adoptively transferred TCR transgenic CD4 T cells in response to infection. THE JOURNAL OF IMMUNOLOGY 2006; 176:3037-43. [PMID: 16493062 DOI: 10.4049/jimmunol.176.5.3037] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD4 and CD8 T cells have been shown to proliferate and differentiate to different extents following antigenic stimulation. CD4 T cells form a heterogenous pool of effector cells in various stages of division and differentiation, while nearly all responding CD8 T cells divide and differentiate to the same extent. We examined CD4 and CD8 T cell responses during bacterial infection by adoptive transfer of CFSE-labeled monoclonal and polyclonal T cells. Monoclonal and polyclonal CD8 T cells both divided extensively, whereas monoclonal CD4 T cells underwent limited division in comparison with polyclonal CD4 T cells. Titration studies revealed that the limited proliferation of transferred monoclonal CD4 T cells was due to inhibition by a high precursor frequency of clonal T cells. This unusually high precursor frequency of clonal CD4 T cells also inhibited the differentiation of these cells. These results suggest that the adoptive transfer of TCR transgenic CD4 T cells significantly underestimates the extent of proliferation and differentiation of CD4 T cells following infection.
Collapse
Affiliation(s)
- Kathryn E Foulds
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
1097
|
Crawford A, Macleod M, Schumacher T, Corlett L, Gray D. Primary T cell expansion and differentiation in vivo requires antigen presentation by B cells. THE JOURNAL OF IMMUNOLOGY 2006; 176:3498-506. [PMID: 16517718 DOI: 10.4049/jimmunol.176.6.3498] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
B cells are well documented as APC; however, their role in supporting and programming the T cell response in vivo is still unclear. Studies using B cell-deficient mice have given rise to contradictory results. We have used mixed BM chimeric mice to define the contribution that B cells make as APC. When the B cell compartment is deficient in MHC class II, while other APC are largely normal, T cell clonal expansion is significantly reduced and the differentiation of T cells into cytokine-secreting effector cells is impaired (in particular, Th2 cells). The development of the memory T cell populations is also decreased. Although MHC class II-mediated presentation by B cells was crucial for an optimal T cell response, neither a B cell-specific lack of CD40 (influencing costimulation) nor lymphotoxin alpha (influencing lymphoid tissue architecture) had any effect on the T cell response. We conclude that in vivo B cells provide extra and essential Ag presentation capacity over and above that provided by dendritic cells, optimizing expansion and allowing the generation of memory and effector T cells.
Collapse
Affiliation(s)
- Alison Crawford
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, King's Buildings, West Mains Road, Edinburgh EH9 3JT, United Kingdom
| | | | | | | | | |
Collapse
|
1098
|
Wingender G, Berg M, Jüngerkes F, Diehl L, Sullivan BA, Kronenberg M, Limmer A, Knolle PA. Immediate antigen-specific effector functions by TCR-transgenic CD8+ NKT cells. Eur J Immunol 2006; 36:570-82. [PMID: 16506291 DOI: 10.1002/eji.200535461] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Only recently have natural antigens for CD1d-dependent, invariant Valpha14+ natural killer T (iNKT) cells been identified. Similar data for CD1d-independent and CD8+ NKT cell populations are still missing. Here, we show that the MHC class I-restricted CD8+ TCR-transgenic mouse lines OT-I, P14 and H-Y contain a significant proportion of transgenic CD8+ NK1.1+ T cells. In liver, most of NK1.1+ T cells express CD8alphaalpha homodimers. Transgenic NKT cells did not bind invariant Valpha14-to-Jalpha18 TCR rearrangement (Valpha14i)-specific CD1d/alpha-galactosylceramide tetramers and the frequency of iNKT cells was severely reduced. The activated cell surface phenotype and the distribution of transgenic NKT cells in vivo were similar to that reported for iNKT cells. The OT-I and P14 CD8+ NKT cells recognized their cognate antigen in the context of H2-Kb and produced cytokines shortly after TCR stimulation. Importantly, transgenic NKT cells exerted immediate antigen-specific cytotoxicity in vitro and in vivo. Our results demonstrate the presence of transgenic CD8+ NKT cells in MHC class I-restricted TCR-transgenic animals, which are endowed with rapid antigen-specific effector functions. These data imply that experiments studying naive T cell function in TCR-transgenic animals should be interpreted with caution, and that such animals could be utilized for studying CD8+ NKT cell function in an antigen-specific manner.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- Antigens, CD1/immunology
- Antigens, CD1d
- Antigens, Ly
- Antigens, Surface/immunology
- CD8 Antigens/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Galactosylceramides/immunology
- Gene Rearrangement, T-Lymphocyte/immunology
- H-2 Antigens/immunology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Lectins, C-Type/immunology
- Liver/cytology
- Liver/immunology
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice
- Mice, Transgenic
- NK Cell Lectin-Like Receptor Subfamily B
- Proteins/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
Collapse
Affiliation(s)
- Gerhard Wingender
- Institute of Molecular Medicine and Experimental Immunology, Bonn, Germany
| | | | | | | | | | | | | | | |
Collapse
|
1099
|
Chaudhry UI, Katz SC, Kingham TP, Pillarisetty VG, Raab JR, Shah AB, DeMatteo RP. In vivo
overexpression of Flt3 ligand expands and activates murine spleen natural killer dendritic cells. FASEB J 2006; 20:982-4. [PMID: 16571772 DOI: 10.1096/fj.05-5411fje] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Natural killer dendritic cells (NKDC) are a unique class of murine immune cells that possess the characteristics of both natural killer (NK) cells and dendritic cells (DC). Because NKDC are able to secrete IFN-gamma, directly lyse tumor cells, and present antigen to naïve T cells, they have immunotherapeutic potential. The relative paucity of NKDC, however, impedes their detailed study. We have found that in vivo, overexpression of the hematopoietic cytokine Flt3 ligand (Flt3L) expands NKDC in various organs from 2-18 fold. Flt3L expanded splenic NKDC retain the ability to lyse tumor cells and become considerably more potent at activating naïve allogeneic and antigen-specific T cells. Compared to normal splenic NKDC, Flt3L-expanded splenic NKDC have a more mature phenotype, a slightly increased ability to capture and process antigen, and a similar cytokine profile. In vivo, we found that Flt3L-expanded splenic NKDC are more effective than normal splenic NKDC in stimulating antigen-specific CD8 T cells. Additionally, we show that NKDC are able to cross-present antigen in vivo. The ability to expand NKDC in vivo using Flt3L will facilitate further analysis of their unique biology. Moreover, Flt3L-expanded NKDC may have enhanced immunotherapeutic potential, given their increased ability to stimulate T cells.
Collapse
Affiliation(s)
- Umer I Chaudhry
- Hepatobiliary Service, Memorial Sloan-Kettering Cancer Center, Box 203, 1275 York Ave., New York, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
1100
|
Mattei F, Schiavoni G, Borghi P, Venditti M, Canini I, Sestili P, Pietraforte I, Morse HC, Ramoni C, Belardelli F, Gabriele L. ICSBP/IRF-8 differentially regulates antigen uptake during dendritic-cell development and affects antigen presentation to CD4+ T cells. Blood 2006; 108:609-17. [PMID: 16569763 DOI: 10.1182/blood-2005-11-4490] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Interferon consensus sequence-binding protein (ICSBP)/interferon regulatory factor 8 (IRF-8) is a transcription factor that plays critical roles in the differentiation of defined dendritic-cell (DC) populations and in the immune response to many pathogens. In this study, we show that splenic DCs (s-DCs) from ICSBP(-/-) mice are markedly defective in their ability to capture and to present exogenous antigens (Ags) to naive CD4(+) T lymphocytes. We found that CD8alpha(+) DCs and, to a lesser extent, CD8alpha(-) DCs from ICSBP(-/-) mice are impaired at internalizing Ags, either through a receptor-mediated pathway or by macropinocytosis, in spite of having a more immature phenotype than their wild-type (WT) counterparts. These features reflected a greatly impaired ability of ICSBP(-/-) s-DCs to present injected soluble ovalbumin (OVA) to OVA-specific CD4(+) T cells in vivo. Conversely, bone marrow (BM)-derived DCs from ICSBP(-/-) mice, in keeping with their immature phenotype, exhibited higher endocytic activity than WT cells. However, Ag-loaded ICSBP(-/-) BM-DCs were defective in priming Ag-specific CD4(+) T lymphocytes and failed to induce a contact hypersensitivity (CHS) response when injected into competent WT hosts. Together, these results indicate that, throughout the developmental program of DCs, ICSBP differentially controls Ag uptake and MHC class II (MHC-II) presentation affecting both functions only in differentiated peripheral DCs.
Collapse
Affiliation(s)
- Fabrizio Mattei
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|