51
|
Reinhold A, Reimann S, Reinhold D, Schraven B, Togni M. Expression of SKAP-HOM in DCs is required for an optimal immune response in vivo. J Leukoc Biol 2009; 86:61-71. [PMID: 19369640 DOI: 10.1189/jlb.0608344] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The cytosolic adaptor molecule SKAP-HOM, similar to the T cell-specific homologue SKAP55, interacts directly with ADAP, and both molecules are involved in inside-out signaling. Previous studies have shown that in the absence of SKAP-HOM, antigen receptor-triggered integrin-mediated adhesion is impaired severely in B cells but not in T cells. In addition, loss of SKAP-HOM results in a less severe clinical course of EAE. DCs are the most potent APCs and express SKAP-HOM. However, the role of SKAP-HOM in DCs remains unknown. Here, we assessed whether the reduced severity of EAE observed in SKAP-HOM-deficient mice is at least partially a result of an impaired cooperation between APCs and T cells. We demonstrate that migration of LC in vivo and the spontaneous motility of BMDCs in vitro are increased in the absence of SKAP-HOM. In contrast, triggering of the integrin results in a drastic decrease of DC motility and in enhanced actin polymerization in SKAP-HOM-deficient DCs. Furthermore, the antigen-dependent conjugate formed between wild-type T cells and SKAP-HOM(-/-) DCs is delayed in comparison with wild-type DCs. Strikingly, fewer antigen-specific T cells are induced by immunization with SKAP-HOM(-/-) BMDCs as compared with wild-type BMDCs in vivo. Thus, these findings suggest that SKAP-HOM expression in DCs is required for the induction of an optimal immune response.
Collapse
Affiliation(s)
- Annegret Reinhold
- Institute of Molecular and Clinical Immunology, Otto von Guericke University, Magdeburg, Germany
| | | | | | | | | |
Collapse
|
52
|
Boggiatto PM, Jie F, Ghosh M, Gibson-Corley KN, Ramer-Tait AE, Jones DE, Petersen CA. Altered dendritic cell phenotype in response to Leishmania amazonensis amastigote infection is mediated by MAP kinase, ERK. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1818-26. [PMID: 19349356 DOI: 10.2353/ajpath.2009.080905] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Initiation of productive immune responses against Leishmania depends on the successful transition of dendritic cells (DC) from an immature to a mature phenotype. This process is characterized by high CD40 surface expression as well as interleukin-12 production, which are frequently seen in response to L. major infection. In vivo footpad infection of C3HeB/FeJ mice for 7 days with L. amazonensis promoted an immature CD11c(+) DC phenotype characterized by both significantly low CD40 surface expression and significantly decreased interleukin-12p40 production compared with L. major infection of these same mice. In vitro infection of bone marrow-derived dendritic cells with L. amazonensis amastigotes resulted in rapid and significant phosphorylation of the mitogen activated protein kinase, extracellular signal-regulated kinase 1/2, observed within minutes of exposure to the parasite. Infection with L. amazonensis promastigotes led to increased 1/2 phosphorylation after 4 hours of infection compared with L. major infection, which correlated with promastigote transformation into amastigotes. Treatment of bone marrow-derived dendritic cells with a mitogen activated protein kinase kinase-specific inhibitor, PD98059, led to regained surface CD40 expression and interleukin-12p40 production following L. amazonensis amastigote infection compared with non-treated, infected DC. Treatment of L. amazonensis-infected mice with the highly-specific mitogen activated protein kinase kinase inhibitor, CI-1040, enhanced surface CD40 expression on CD11c(+) DC obtained from the draining lymph node. L. amazonensis amastigotes, through activation of extracellular signal-regulated kinase 1/2, inhibit the ability of DC to undergo proper maturation both in vitro and in vivo.
Collapse
|
53
|
Segura E, Villadangos JA. Antigen presentation by dendritic cells in vivo. Curr Opin Immunol 2009; 21:105-10. [PMID: 19342210 DOI: 10.1016/j.coi.2009.03.011] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 02/12/2009] [Accepted: 03/09/2009] [Indexed: 10/21/2022]
Abstract
Dendritic cells (DC) are heterogenous, comprising several subpopulations of migratory and lymphoid-organ-resident types. Recent studies addressing the role of each subset in antigen presentation in vivo have revealed a complex division of labor within the DC network. In addition to CD8(+) DC, migratory lung or dermal DC can cross-present antigen in vivo. Migratory DC also transport to the lymph nodes antigens that can be transferred to resident DC for presentation. In inflammatory conditions, the antigen-presentation abilities of DC can be severely impaired, but an additional population of monocyte-derived DC then comes into play. Understanding the contribution of each DC subset to a physiological immune response is particularly relevant for rational vaccine design.
Collapse
|
54
|
Poly(I:C)-Treated human langerhans cells promote the differentiation of CD4+ T cells producing IFN-gamma and IL-10. J Invest Dermatol 2009; 129:1963-71. [PMID: 19242516 DOI: 10.1038/jid.2009.21] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Epidermal Langerhans cells (LCs) are the first dendritic cells to encounter skin pathogens. However, their function has recently been challenged, especially in the initiation of T-cell responses to viral antigens. We have previously reported that fresh immature human LCs express mRNA encoding TLR3. Here we analyze the response of highly purified human LCs to poly(I:C), a synthetic mimetic of viral dsRNA recognized by TLR3. We show that LCs exposed for 2 days to poly(I:C) under serum-free conditions up-regulated co-stimulatory molecules, a process associated with increased allostimulatory capacity. Furthermore, poly(I:C) significantly enhanced LC survival and induced them to produce CXCL10, IL-6, and IL-12 p40. Bioactive IL-12 p70, IL-1beta, IL-15, IL-18, and IL-23 were never detected, even after CD40 ligation. LC incubation in the presence of bafilomycin completely reversed the effect of poly(I:C) on LC phenotypic activation and survival, indicating that endosomal TLR3 is involved in this process. Most interestingly, we report here that poly(I:C)-treated LCs favored alloreactive CD4(+) T-cell differentiation toward a Th1 profile and concomitant differentiation of IL-10-producing CD4(+) T cells that might limit, at another time, the inflammatory response and subsequent tissue damage.
Collapse
|
55
|
Sojka DK, Lazarski CA, Huang YH, Bromberg I, Hughson A, Fowell DJ. Regulation of immunity at tissue sites of inflammation. Immunol Res 2009; 45:239-50. [DOI: 10.1007/s12026-009-8105-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
56
|
Carvalho LP, Pearce EJ, Scott P. Functional dichotomy of dendritic cells following interaction with Leishmania braziliensis: infected cells produce high levels of TNF-alpha, whereas bystander dendritic cells are activated to promote T cell responses. THE JOURNAL OF IMMUNOLOGY 2009; 181:6473-80. [PMID: 18941238 DOI: 10.4049/jimmunol.181.9.6473] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Leishmania braziliensis infections are often associated with exaggerated immune responses that can sometimes lead to severe disease associated with high levels of IFN-gamma and TNF-alpha. To explore the role played by dendritic cells (DCs) in these responses, we characterized DCs that were exposed to L. braziliensis. We found that DCs cultured with L. braziliensis parasites up-regulated DC activation markers and produced IL-12 and TNF-alpha. However, not all DCs in the culture became infected, and an analysis of infected and uninfected DCs demonstrated that the up-regulation of activation markers and IL-12 production was primarily confined to the uninfected (bystander) DCs. Further studies with Transwell chambers and parasite fractions indicated that the activation of bystander DCs was mediated by a soluble parasite product, in a type 1 IFN- and MyD88-independent, but TNF-alpha-dependent fashion, and that the activated DCs were more efficient at presenting Ag than control DCs. In contrast, L. braziliensis-infected DCs failed to up-regulate activation markers, but exhibited a dramatic enhancement in their ability to produce TNF-alpha in response to LPS as compared with uninfected DCs. These findings uncover a dual role for DCs in L. braziliensis infection: T cell activation by bystander DCs due to enhanced Ag-presenting capacity following exposure to soluble parasite products, and increased production of TNF-alpha by infected cells that may contribute to the local control of the parasites, but concomitantly induce immunopathology.
Collapse
Affiliation(s)
- Lucas P Carvalho
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19103, USA
| | | | | |
Collapse
|
57
|
Ng LG, Hsu A, Mandell MA, Roediger B, Hoeller C, Mrass P, Iparraguirre A, Cavanagh LL, Triccas JA, Beverley SM, Scott P, Weninger W. Migratory dermal dendritic cells act as rapid sensors of protozoan parasites. PLoS Pathog 2008; 4:e1000222. [PMID: 19043558 PMCID: PMC2583051 DOI: 10.1371/journal.ppat.1000222] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Accepted: 10/29/2008] [Indexed: 11/19/2022] Open
Abstract
Dendritic cells (DC), including those of the skin, act as sentinels for intruding microorganisms. In the epidermis, DC (termed Langerhans cells, LC) are sessile and screen their microenvironment through occasional movements of their dendrites. The spatio-temporal orchestration of antigen encounter by dermal DC (DDC) is not known. Since these cells are thought to be instrumental in the initiation of immune responses during infection, we investigated their behavior directly within their natural microenvironment using intravital two-photon microscopy. Surprisingly, we found that, under homeostatic conditions, DDC were highly motile, continuously crawling through the interstitial space in a Galpha(i) protein-coupled receptor-dependent manner. However, within minutes after intradermal delivery of the protozoan parasite Leishmania major, DDC became immobile and incorporated multiple parasites into cytosolic vacuoles. Parasite uptake occurred through the extension of long, highly dynamic pseudopods capable of tracking and engulfing parasites. This was then followed by rapid dendrite retraction towards the cell body. DDC were proficient at discriminating between parasites and inert particles, and parasite uptake was independent of the presence of neutrophils. Together, our study has visualized the dynamics and microenvironmental context of parasite encounter by an innate immune cell subset during the initiation of the immune response. Our results uncover a unique migratory tissue surveillance program of DDC that ensures the rapid detection of pathogens.
Collapse
Affiliation(s)
- Lai Guan Ng
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - Alice Hsu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael A. Mandell
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ben Roediger
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - Christoph Hoeller
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Paulus Mrass
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Amaya Iparraguirre
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Lois L. Cavanagh
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - James A. Triccas
- Microbial Pathogenesis and Immunity Group, Discipline of Infectious Diseases and Immunology, University of Sydney, Camperdown, New South Wales, Australia
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Wolfgang Weninger
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
- Discipline of Dermatology, University of Sydney, Camperdown, New South Wales, Australia
- * E-mail:
| |
Collapse
|
58
|
Nicolas JF, Guy B. Intradermal, epidermal and transcutaneous vaccination: from immunology to clinical practice. Expert Rev Vaccines 2008; 7:1201-14. [PMID: 18844594 DOI: 10.1586/14760584.7.8.1201] [Citation(s) in RCA: 183] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The dermis and epidermis are alternative sites for prophylactic vaccination that have received renewed interest in recent years, not only because of the ease of access to the skin, but also its unique immunological properties. This review discusses the characteristics of the skin, current knowledge on skin immunity and clinical experience with cutaneous immunization against infectious diseases, with a special focus on intradermal immunization. The most widely accepted paradigm explaining the efficacy of cutaneous immunization is reviewed and recent research suggesting where this paradigm may need some refinement is highlighted. Clinical investigations that have concentrated on the intradermal route to vaccinate against influenza, rabies or hepatitis B support the current knowledge on skin immunity and, when combined with recent progress made in the development of user-friendly injection systems, have stimulated the ongoing clinical development of novel vaccines.
Collapse
Affiliation(s)
- Jean-François Nicolas
- University Lyon 1, UFR Lyon-Sud, IFR 128 BioSciences Lyon-Gerland, Institut National de la Santé et de la Recherche Médicale U503, 21 Avenue Tony Garnier, Lyon Cedex 07, Lyon 69365, France.
| | | |
Collapse
|
59
|
Abstract
Professional antigen-presenting cells (APCs) are sentinel cells of the immune system that present antigen to T lymphocytes and mediate an appropriate immune response. It is therefore surprising that knowledge of the professional APCs in human lymph nodes is limited. Using 3-color immunohistochemistry, we have identified APCs in human lymph nodes, excluding plasmacytoid APCs, that fall into 2 nonoverlapping classes: (1) CD209+ APCs, coexpressing combinations of CD206, CD14, and CD68, that occupied the medullary cords, lined the capsule and trabeculae and were also scattered throughout the diffuse T-lymphocyte areas of the paracortex; and (2) APCs expressing combinations of CD1a, CD207, and CD208, that were always restricted to the paracortex. Surprisingly, this second class of APCs was almost entirely absent from many lymph nodes. Our data suggest that most CD208+ cells, often referred to as "interdigitating cells," derive from migratory APCs, and that the major APC subset consistently resident in the paracortex of human lymph nodes is the CD209+ subset. All APC subsets were demonstrated to be in close contact with the fibroreticular network. The identification of 2 distinct APC populations in the paracortex of human lymph nodes has important implications for understanding T-lymphocyte responses and optimizing vaccine design.
Collapse
|
60
|
The absence of cutaneous lymph nodes results in a Th2 response and increased susceptibility to Leishmania major infection in mice. Infect Immun 2008; 76:4241-50. [PMID: 18625738 DOI: 10.1128/iai.01714-07] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lymph nodes (LNs) are important sentinel organs where antigen-presenting cells interact with T cells to induce adaptive immune responses. In cutaneous infection of mice with Leishmania major, resistance depends on the induction of a T-helper-cell-1 (Th1)-mediated cellular immune response in draining, peripheral LNs. We investigated whether draining, peripheral LNs are absolutely required for resistance against L. major infection. We investigated the course of experimental leishmaniasis in wild-type (wt) mice lacking peripheral LNs (pLNs), which we generated by in utero blockade of membrane-bound lymphotoxin, and in mice lacking pLNs or all LNs due to genetic deletion of lymphotoxin ligands or receptors. wt mice of the resistant C57BL/6 strain without local skin-draining LNs were still able to generate specific T-cell responses, but this yielded Th2 cells. This switch to a Th2 response resulted in severe systemic infection. We also confirmed these results with mice lacking pLNs due to genetic depletion of lymphotoxin-beta. The complete absence of LNs due to a genetic depletion of the lymphotoxin-beta receptor also resulted in a marked deterioration of disease and a Th2 response. Thus, in the absence of pLNs, an L. major-specific Th2 response is induced in the remaining secondary lymphoid organs, such as the spleen and non-skin-draining LNs. This indicates a critical requirement for pLNs to induce protective Th1 immunity and suggests that whether Th1 or Th2 priming to the same antigen occurs depends on the site of the primary antigen recognition.
Collapse
|
61
|
Skallová A, Iezzi G, Ampenberger F, Kopf M, Kopecky J. Tick saliva inhibits dendritic cell migration, maturation, and function while promoting development of Th2 responses. THE JOURNAL OF IMMUNOLOGY 2008; 180:6186-92. [PMID: 18424740 DOI: 10.4049/jimmunol.180.9.6186] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Similarly to other blood-feeding arthropods, ticks have evolved immunosuppressive mechanisms enabling them to overcome the host immune system. Although the immunomodulatory effect of tick saliva on several cell populations of the immune system has been extensively studied, little is known about its impact on dendritic cells (DCs). We have examined the effect of Ixodes ricinus tick saliva on DC function in vitro and in vivo. Exposure of DCs to tick saliva in vitro resulted in impaired maturation, upon CD40 or TLR9, TLR3 and TLR7 ligation, as well as reduced Ag presentation capacity. Administration of tick saliva in vivo significantly inhibited maturation and early migration of DCs from inflamed skin to draining lymph nodes, and decreased the capacity of lymph node DCs to present soluble Ag to specific T cells. Moreover, saliva-exposed DCs failed to induce efficient Th1 and Th17 polarization and promoted development of Th2 responses. Our data reveal a complex inhibitory effect exerted by tick saliva on DC function. Given the role of DCs as the key instigators of adaptive immune responses, alteration of their function might represent a major mechanism of tick-mediated immune evasion.
Collapse
Affiliation(s)
- Anna Skallová
- Institute of Parasitology, Biology Centre AS CR, Branisovska 31, Ceské Budejovice, Czech Republic
| | | | | | | | | |
Collapse
|
62
|
dos Santos MST, Vaz Cardoso LP, Nascimento GR, Lino RDS, Dorta ML, de Oliveira MAP, Ribeiro-Dias F. Leishmania major: recruitment of Gr-1+ cells into draining lymph nodes during infection is important for early IL-12 and IFN gamma production. Exp Parasitol 2008; 119:403-10. [PMID: 18501355 DOI: 10.1016/j.exppara.2008.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Revised: 03/30/2008] [Accepted: 04/04/2008] [Indexed: 10/22/2022]
Abstract
The production of interleukin-12 and interferon-gamma is a key event for controlling leishmaniasis. Here, we tested the hypothesis that after murine infection with Leishmania major, cell migration into draining lymph nodes is crucial for early production of those cytokines. We showed that inflammatory cells carrying the marker of recently migrated cells, the Gr-1 antigen, including polymorphonuclear and mononuclear cells, migrate rapidly into the site of promastigote infection and, subsequently, into draining lymph nodes. Treatment with RB6-8C5 monoclonal antibody reduced local inflammation and migration of Gr-1+ cells into the draining lymph nodes. This reduction was associated with a decrease of interleukin-12 production by draining lymph node cells from BALB/c mice but not C57BL/6 mice. Additionally, interferon-gamma was also reduced in both mouse strains after depletion of Gr-1+ cells, suggesting that these cells are important for early interleukin-12 and interferon-gamma production. Our findings suggest that recently migrated myeloid cells, more than resident cells, are the major source of the early IL-12 production after L. major infection.
Collapse
|
63
|
Manolova V, Flace A, Bauer M, Schwarz K, Saudan P, Bachmann M. Nanoparticles target distinct dendritic cell populations according to their size. Eur J Immunol 2008; 38:1404-13. [DOI: 10.1002/eji.200737984] [Citation(s) in RCA: 890] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
64
|
Abstract
The priming of a T cell results from its physical interaction with a dendritic cell (DC) that presents the cognate antigenic peptide. The success rate of such interactions is extremely low, because the precursor frequency of a naive T cell recognizing a specific antigen is in the range of 1:10(5)-10(6). To make this principle practicable, encounter frequencies between DCs and T cells are maximized within lymph nodes (LNs) that are compact immunological projections of the peripheral tissue they drain. But LNs are more than passive meeting places for DCs that immigrated from the tissue and lymphocytes that recirculated via the blood. The microanatomy of the LN stroma actively organizes the cellular encounters by providing preformed migration tracks that create dynamic but highly ordered movement patterns. LN architecture further acts as a sophisticated filtration system that sieves the incoming interstitial fluid at different levels and guarantees that immunologically relevant antigens are loaded on DCs or B cells while inert substances are channeled back into the blood circulation. This review focuses on the non-hematopoietic infrastructure of the lymph node. We describe the association between fibroblastic reticular cell, conduit, DC, and T cell as the essential functional unit of the T-cell cortex.
Collapse
Affiliation(s)
- Tim Lämmermann
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | |
Collapse
|
65
|
Abstract
Dendritic cells (DCs) play a central role in T-cell activation and the control of the inherent autoreactivity of the T-cell compartment. Pleiotropic DC functions are likely associated with discrete DC subsets. However, the latter remain largely defined by phenotype and unique anatomic location, rather than function. The investigation of DC involvement in complex phenomena that rely on multicellular interactions, such as immuno-stimulation and tolerization calls for an assessment of DC functions within physiological context. Given the highly dynamic DC compartment, the method of choice to study in vivo DC functions is their conditional ablation in the intact organism. Here, we summarize the recent progress in this field highlighting pitfalls and prospects of the approach.
Collapse
|
66
|
Wang L, Bursch LS, Kissenpfennig A, Malissen B, Jameson SC, Hogquist KA. Langerin Expressing Cells Promote Skin Immune Responses under Defined Conditions. THE JOURNAL OF IMMUNOLOGY 2008; 180:4722-7. [DOI: 10.4049/jimmunol.180.7.4722] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
67
|
Abstract
Toll-like receptors exist as highly conserved pathogen sensors throughout the animal kingdom and they represent a key family of molecules bridging the ancient innate and adaptive immune systems. The first molecules of adaptive immunity appeared in the cartilaginous fishes and, with these, major histocompatibility proteins and cells expressing these molecules, and thus, by definition, the advent of antigen-presenting cells and the "professional" antigen-presenting cells, the dendritic cells. Dendritic cells themselves are highly specialized subsets of cells with the major roles of antigen presentation and stimulation of lymphocytes. The dendritic cell functions of inducing immunity are regulated by their own activation status, which is governed by their encounter with pathogen-associated molecular patterns that signal through pattern recognition receptors, including Toll-like receptors, expressed at the surface and within the cytoplasm and endosomal membranes of dendritic cells. Thus although dendritic cells play a crucial role in the induction of adaptive immunity, the adaptive response is itself initiated at the level of ancient receptors of the innate immune system. A further degree in the complexity of dendritic cell activation is established by the fact that not all dendritic cells are equal. Dendritic cells exist as multiple subsets that vary in location, function, and phenotype. Distinct dendritic cell subsets display great variation in the type of Toll-like receptors expressed and consequently variation in the type of pathogens sensed and the subsequent type of immune responses initiated.
Collapse
|
68
|
Bursch LS, Wang L, Igyarto B, Kissenpfennig A, Malissen B, Kaplan DH, Hogquist KA. Identification of a novel population of Langerin+ dendritic cells. ACTA ACUST UNITED AC 2007; 204:3147-56. [PMID: 18086865 PMCID: PMC2150989 DOI: 10.1084/jem.20071966] [Citation(s) in RCA: 388] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Langerhans cells (LCs) are antigen-presenting cells that reside in the epidermis of the skin and traffic to lymph nodes (LNs). The general role of these cells in skin immune responses is not clear because distinct models of LC depletion resulted in opposite conclusions about their role in contact hypersensitivity (CHS) responses. While comparing these models, we discovered a novel population of LCs that resides in the dermis and does not represent migrating epidermal LCs, as previously thought. Unlike epidermal LCs, dermal Langerin(+) dendritic cells (DCs) were radiosensitive and displayed a distinct cell surface phenotype. Dermal Langerin(+) DCs migrate from the skin to the LNs after inflammation and in the steady state, and represent the majority of Langerin(+) DCs in skin draining LNs. Both epidermal and dermal Langerin(+) DCs were depleted by treatment with diphtheria toxin in Lang-DTREGFP knock-in mice. In contrast, transgenic hLang-DTA mice lack epidermal LCs, but have normal numbers of dermal Langerin(+) DCs. CHS responses were abrogated upon depletion of both epidermal and dermal LCs, but were unaffected in the absence of only epidermal LCs. This suggests that dermal LCs can mediate CHS and provides an explanation for previous differences observed in the two-model systems.
Collapse
Affiliation(s)
- Laura S Bursch
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | |
Collapse
|
69
|
Immunization with a lentivector that targets tumor antigen expression to dendritic cells induces potent CD8+ and CD4+ T-cell responses. J Virol 2007; 82:86-95. [PMID: 17959670 DOI: 10.1128/jvi.01289-07] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lentivectors stimulate potent immune responses to antigen transgenes and are being developed as novel genetic vaccines. To improve safety while retaining efficacy, we constructed a lentivector in which transgene expression was restricted to antigen-presenting cells using the mouse dectin-2 gene promoter. This lentivector expressed a green fluorescent protein (GFP) transgene in mouse bone marrow-derived dendritic cell cultures and in human skin-derived Langerhans and dermal dendritic cells. In mice GFP expression was detected in splenic dectin-2(+) cells after intravenous injection and in CD11c(+) dendritic cells in the draining lymph node after subcutaneous injection. A dectin-2 lentivector encoding the human melanoma antigen NY-ESO-1 primed an NY-ESO-1-specific CD8(+) T-cell response in HLA-A2 transgenic mice and stimulated a CD4(+) T-cell response to a newly identified NY-ESO-1 epitope presented by H2 I-A(b). As immunization with the optimal dose of the dectin-2 lentivector was similar to that stimulated by a lentivector containing a strong constitutive viral promoter, targeting antigen expression to dendritic cells can provide a safe and effective vaccine.
Collapse
|
70
|
Najar HM, Dutz JP. Topical TLR9 agonists induce more efficient cross-presentation of injected protein antigen than parenteral TLR9 agonists do. Eur J Immunol 2007; 37:2242-56. [PMID: 17634951 DOI: 10.1002/eji.200636212] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Topical application of adjuvant to the skin promotes the generation of immune responses to co-administered peptide or protein antigen. We demonstrate that topical administration of CpG adjuvant (a TLR9 agonist) induces the cross-presentation of, and antigen-specific CTL induction to, locally injected soluble protein antigen. C57BL/6 mice were immunized by subcutaneous or intramuscular injection with ovalbumin (OVA) protein as model antigen. Application of CpG to the local skin induced more efficient cross-presentation of the injected antigen than co-injected adjuvant. Robust antigen-specific CTL responses were generated, as determined by antigen-specific CTL enumeration using tetramers, IFN-gamma ELISPOT analysis and cytotoxicity assays. Long-term memory CTL responses were induced. Topical administration of adjuvant induced Langerhans cell migration, local type 1 IFN-dependent myxovirus-resistance protein A expression and bystander dendritic cell (DC) activation. Soluble antigen-bearing DC within the skin draining lymph nodes were mainly CD11chiCD11bhilangerinloDEC205lo. Topical administration did not result in the splenomegaly or systemic cytokine induction (including TNF-alpha, IL-12, IFN-gamma and MCP-1) noted with parenteral administration. Topical TLR9 family agonists may be used to modulate the immune response to soluble protein vaccines administered by standard percutaneous route. Topical adjuvant administration increases efficacy of CTL induction and reduces toxicity when compared to parenteral adjuvant administration.
Collapse
Affiliation(s)
- Hossain M Najar
- Department of Dermatology, and Child and Family Research Institute, University British Colombia, Vancouver, British Colombia, Canada
| | | |
Collapse
|
71
|
Wolf AJ, Linas B, Trevejo-Nuñez GJ, Kincaid E, Tamura T, Takatsu K, Ernst JD. Mycobacterium tuberculosis Infects Dendritic Cells with High Frequency and Impairs Their Function In Vivo. THE JOURNAL OF IMMUNOLOGY 2007; 179:2509-19. [PMID: 17675513 DOI: 10.4049/jimmunol.179.4.2509] [Citation(s) in RCA: 407] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is thought to reside in macrophages, although infected dendritic cells (DCs) have been observed. Thus, although cellular associations have been made, global characterization of the cells harboring Mtb is lacking. We have performed temporal and quantitative characterization of the cells harboring Mtb following aerosol infection of mice by using GFP-expressing bacteria and flow cytometry. We discovered that Mtb infects phagocytic cells of diverse phenotypes, that the predominant infected cell populations change with time, and that myeloid DCs are the major cell population infected with Mtb in the lungs and lymph nodes. We also found that the bacteria in the lung-draining lymph node are transported there from the lungs by a CCL19/21-dependent mechanism and that the transport of bacteria to the lymph node is a transient phenomenon despite chronic infection. In addition, we found that the lymph node cell subsets that are most efficacious in stimulating Mtb-specific, TCR-transgenic CD4(+) T lymphocytes are not infected with the bacteria and are scarce or absent from the lungs of infected mice. Finally, we found that the lung cell populations that are infected with Mtb at high frequency are relatively ineffective at stimulating Ag-specific CD4(+) T lymphocytes, and we have obtained evidence that live Mtb can inhibit MHC class II Ag presentation without a decrease in the surface expression of MHC class II. These results indicate that Mtb targets DC migration and Ag presentation in vivo to promote persistent infection.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- Biological Transport, Active/genetics
- Biological Transport, Active/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/microbiology
- CD4-Positive T-Lymphocytes/pathology
- Cell Movement/genetics
- Cell Movement/immunology
- Chemokine CCL19
- Chemokine CCL21
- Chemokines, CC/immunology
- Dendritic Cells/immunology
- Dendritic Cells/microbiology
- Dendritic Cells/pathology
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/immunology
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Lung/immunology
- Lung/microbiology
- Lung/pathology
- Lymph Nodes/immunology
- Lymph Nodes/microbiology
- Lymph Nodes/pathology
- Macrophages/immunology
- Macrophages/microbiology
- Macrophages/pathology
- Mice
- Mice, Transgenic
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/pathology
- Tuberculosis, Pulmonary/veterinary
Collapse
Affiliation(s)
- Andrea J Wolf
- Division of Infectious Diseases, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
72
|
de Heusch M, Blocklet D, Egrise D, Hauquier B, Vermeersch M, Goldman S, Moser M. Bidirectional MHC molecule exchange between migratory and resident dendritic cells. J Leukoc Biol 2007; 82:861-8. [PMID: 17634280 DOI: 10.1189/jlb.0307167] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Dendritic cells (DCs) loaded extracorporeally with antigen can be used as an adjuvant in vivo. In this work, we analyzed the migration of transferred DC and monitored the phenotype of new migrants in the draining lymph nodes. It is surprising that we found that a majority of resident DCs expressed donor MHC molecules and that a proportion of injected DCs acquired host MHC molecules. These observations suggest that a bidirectional MHC molecule exchange occurs between migratory and resident DCs, a mechanism that may amplify antigen presentation in vivo.
Collapse
Affiliation(s)
- Magali de Heusch
- Institut de Biologie et Médecine Moléculaires, Université Libre de Bruxelles, Rue des Prof. Jeener et Brachet 12, 6041 Gosselies, Belgium
| | | | | | | | | | | | | |
Collapse
|
73
|
Abstract
Animal skin separates the inner world of the body from the largely hostile outside world and is actively involved in the defence against microbes. However, the skin is no perfect defence barrier and many microorganisms have managed to live on or within the skin as harmless passengers or as disease-causing pathogens. Microbes have evolved numerous strategies that allow them to gain access to the layers underneath the epidermis where they either multiply within the dermis or move to distant destinations within the body for replication. A number of viruses, bacteria and parasites use arthropod vectors, like ticks or mosquitoes, to deliver them into the dermis while taking their blood meal. Within the dermis, successful pathogens subvert the function of a variety of skin resident cells or cells of the innate immune system that rush to the site of infection. In this review several interactions with cells of the skin by medically relevant vector-borne pathogens are discussed to highlight the different ways in which these pathogens have come to survive within the skin and to usurp the defence mechanisms of the host for their own ends.
Collapse
Affiliation(s)
- Freddy Frischknecht
- Department of Parasitology, Hygiene Institute, Heidelberg University School of Medicine, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| |
Collapse
|
74
|
Radwanska M, Cutler AJ, Hoving JC, Magez S, Holscher C, Bohms A, Arendse B, Kirsch R, Hunig T, Alexander J, Kaye P, Brombacher F. Deletion of IL-4Ralpha on CD4 T cells renders BALB/c mice resistant to Leishmania major infection. PLoS Pathog 2007; 3:e68. [PMID: 17500591 PMCID: PMC1867380 DOI: 10.1371/journal.ppat.0030068] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Accepted: 03/27/2007] [Indexed: 11/18/2022] Open
Abstract
Effector responses induced by polarized CD4+ T helper 2 (Th2) cells drive nonhealing responses in BALB/c mice infected with Leishmania major. Th2 cytokines IL-4 and IL-13 are known susceptibility factors for L. major infection in BALB/c mice and induce their biological functions through a common receptor, the IL-4 receptor α chain (IL-4Rα). IL-4Rα–deficient BALB/c mice, however, remain susceptible to L. major infection, indicating that IL-4/IL-13 may induce protective responses. Therefore, the roles of polarized Th2 CD4+ T cells and IL-4/IL-13 responsiveness of non-CD4+ T cells in inducing nonhealer or healer responses have yet to be elucidated. CD4+ T cell–specific IL-4Rα (LckcreIL-4Rα−/lox) deficient BALB/c mice were generated and characterized to elucidate the importance of IL-4Rα signaling during cutaneous leishmaniasis in the absence of IL-4–responsive CD4+ T cells. Efficient deletion was confirmed by loss of IL-4Rα expression on CD4+ T cells and impaired IL-4–induced CD4+ T cell proliferation and Th2 differentiation. CD8+, γδ+, and NK–T cells expressed residual IL-4Rα, and representative non–T cell populations maintained IL-4/IL-13 responsiveness. In contrast to IL-4Rα−/lox BALB/c mice, which developed ulcerating lesions following infection with L. major, LckcreIL-4Rα−/lox mice were resistant and showed protection to rechallenge, similar to healer C57BL/6 mice. Resistance to L. major in LckcreIL-4Rα−/lox mice correlated with reduced numbers of IL-10–secreting cells and early IL-12p35 mRNA induction, leading to increased delayed type hypersensitivity responses, interferon-γ production, and elevated ratios of inducible nitric oxide synthase mRNA/parasite, similar to C57BL/6 mice. These data demonstrate that abrogation of IL-4 signaling in CD4+ T cells is required to transform nonhealer BALB/c mice to a healer phenotype. Furthermore, a beneficial role for IL-4Rα signaling in L. major infection is revealed in which IL-4/IL-13–responsive non-CD4+ T cells induce protective responses. Leishmaniasis is a disease induced by a protozoan parasite and transmitted by the sandfly. Several forms of infection are identified, and the different diseases have wide-ranging symptoms from localized cutaneous sores to visceral disease affecting many internal organs. Animal models of human cutaneous leishmaniasis have been established in which disease is induced by infecting mice subcutaneously with Leishmania major. Different strains of inbred mice have been found to be susceptible or resistant to L. major infection. “Healer” C57BL/6 mice control infection with transient lesion development. The protective response to infection in this strain is dominated by type 1 cytokines inducing parasite killing by nitric oxide. Conversely, “nonhealer” BALB/c mice are unable to control infection and develop nonhealing lesions associated with a dominant type 2 immune response driven by cytokines IL-4 and IL-13. However, mice deficient in IL-4/IL-13 signaling are not protected against development of cutaneous leishmaniasis. Here we describe a BALB/c mouse where the ability to polarize to a dominant type 2 response is removed by cell-specific deletion of the receptor for IL-4/IL-13 on CD4+ T cells. These mice are resistant to L. major infection similar to C57BL/6 mice, which highlights the role of T helper 2 cells in driving susceptibility and the protective role of IL-4/IL-13 signaling in non-CD4+ T cells in BALB/c mice.
Collapse
Affiliation(s)
- Magdalena Radwanska
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Antony J Cutler
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - J. Claire Hoving
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Stefan Magez
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- VIB, Vrije Universiteit Brussel, Brussels, Belgium
| | - Christoph Holscher
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Andreas Bohms
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Berenice Arendse
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Richard Kirsch
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Thomas Hunig
- Institute for Virology and Immunobiology, University of Wurzburg, Wurzburg, Germany
| | - James Alexander
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Paul Kaye
- Immunology and Infection Unit, Department of Biology, University of York, York, United Kingdom
| | - Frank Brombacher
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
75
|
Zimmerli SC, Hauser C. Langerhans cells and lymph node dendritic cells express the tight junction component claudin-1. J Invest Dermatol 2007; 127:2381-90. [PMID: 17508021 DOI: 10.1038/sj.jid.5700882] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Claudin-1 is a critical structural component of tight junctions that have an important role in adhesive properties, barrier function, and paracellular transport of epithelia and other nonhematopoietic tissues. We found claudin-1 in murine CD207+ Langerhans cells (LC) residing in epidermis. Claudin-1 was not detected in other skin dendritic cells (DC). LC expressed claudin-1 in steady state and inflamed skin. Claudin-1 was demonstrated further in lymph node LC under steady state and inflammatory conditions, including after direct tracking with tetramethylrhodamine-isothiocyanate (TRITC). All subsets of skin draining lymph node DC defined by CD205, CD11b, CD11c, and CD8, including a presumably blood-borne lymph node resident CD8+CD207+ LC population, were claudin-1+. TRITC tracking demonstrated claudin-1 in CD207- skin migrant DC in the lymph node, suggesting upregulation of this molecule during migration or once arrived in the lymph node. Claudin-1 expression in CD207+ cells was confirmed at the protein and mRNA levels. Transforming growth factor-beta, a factor critical for the induction of LC in vitro and in vivo, stimulated the accumulation of claudin-1 mRNA and protein when added to bone marrow cells cultured with GM-CSF and IL-4. Claudin-1 may thus have an important function in adhesion and/or migration of LC.
Collapse
Affiliation(s)
- Simone C Zimmerli
- Allergy Unit, Division of Immunology and Allergy, Department of Internal Medicine, Geneva University Hospitals and Medical School, Geneva, Switzerland.
| | | |
Collapse
|
76
|
Henri S, Siret C, Machy P, Kissenpfennig A, Malissen B, Leserman L. Mature DC from skin and skin-draining LN retain the ability to acquire and efficiently present targeted antigen. Eur J Immunol 2007; 37:1184-93. [PMID: 17407099 DOI: 10.1002/eji.200636793] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Skin-draining LN contain several phenotypically distinguishable DC populations, which may be immature or mature. Mature DC are generally considered to have lost the capacity to acquire and present newly encountered Ag. Using antibody-opsonized liposomes as Ag carriers, we show that mature DC purified from skin explants are able to efficiently capture liposomes, process Ag encapsulated within them and activate Ag-specific CD4(+) T cells. Explant DC from mice with Langerhans cells (LC) expressing the primate diphtheria toxin receptor that were exposed to diphtheria toxin in vivo presented Ag as well as explant DC from wild-type mice, indicating that LC are not required and dermal DC are probably responsible for this presentation. We further show that all DC subtypes from LN that capture opsonized Ag are capable of cross-presenting it to CD8(+) T cells. Induction of additional maturation in vivo by LPS or treatment with double-stranded RNA did not alter the Ag presentation capacity of the skin or LN DC subtypes. These results suggest that mature DC present in skin-draining LN may play an important role in the induction of primary and/or secondary immune responses against Ag delivered to the LN that they take up by receptor-mediated endocytosis.
Collapse
Affiliation(s)
- Sandrine Henri
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, Faculté des Sciences de Luminy, Marseille, France.
| | | | | | | | | | | |
Collapse
|
77
|
León B, López-Bravo M, Ardavín C. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania. Immunity 2007; 26:519-31. [PMID: 17412618 DOI: 10.1016/j.immuni.2007.01.017] [Citation(s) in RCA: 500] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 01/22/2007] [Accepted: 01/29/2007] [Indexed: 01/24/2023]
Abstract
Infection-induced inflammatory reactions involve a strong increase in dendritic cells (DCs) at the infection site and draining lymph nodes (dLNs). Whether inflammatory DCs are recruited to these locations or differentiate locally, and what their functional relevance is, remain unclear. Here we showed that during Leishmania infection, monocytes were recruited to the dermis and differentiated into "dermal monocyte-derived DCs," which subsequently migrated into the dLNs. In addition, monocyte recruitment to the dLNs resulted in the differentiation into "LN monocyte-derived DCs." Analysis of the kinetics of monocyte differentiation into DCs, susceptibility to infection, IL-12 production, and L. major-specific T cell stimulation potential suggest that dermal monocyte-derived DCs controlled the induction of protective T helper 1 responses against Leishmania. Thus, the demonstration of monocyte differentiation potential into DCs during in vivo infection and of local DC differentiation in inflammatory foci suggests that de novo formed monocyte-derived DCs are essential in T cell immunity against pathogens.
Collapse
Affiliation(s)
- Beatriz León
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Universidad Autónoma, 28049 Madrid, Spain
| | | | | |
Collapse
|
78
|
Abstract
Studies in murine experimental Leishmania major infection have helped to understand the requirements for efficient development of T helper (Th)1/cytotoxic T (Tc)1-mediated protection against the parasite. As such they have revealed that Fc gamma receptor (Fc gamma R)I and Fc gamma RIII-mediated uptake of L. major amastigotes by dendritic cells (DC) is an important prerequisite for Th1 development. In addition, DC-derived cytokines contribute to adequate T-cell education. DC-based vaccines may thus provide an important tool for both the development of a prophylactic vaccine against leishmaniasis and - together with leishmanicidal drugs - for eliciting immune-deviating functions towards protective immunity in non-healing leishmaniasis. This review highlights recent advances in the understanding of the role of DC for the induction of Th1/Tc1-predominant immunity against L. major and how this knowledge may translate into clinical approaches.
Collapse
Affiliation(s)
- Esther von Stebut
- Department of Dermatology, Johannes Gutenberg-University Mainz, Mainz, Germany.
| |
Collapse
|
79
|
Leggatt GR, Frazer IH. HPV vaccines: the beginning of the end for cervical cancer. Curr Opin Immunol 2007; 19:232-8. [PMID: 17293100 DOI: 10.1016/j.coi.2007.01.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Accepted: 01/31/2007] [Indexed: 11/16/2022]
Abstract
Vaccines prophylactic against infection with human papillomavirus (HPV) are based on alum adjuvanted virus-like particles. Two such vaccines have recently been shown to prevent persistent HPV infection and associated cervical cancer precursor lesions. The genotype-specific neutralising antibody directed at conformational epitopes of the L1 major capsid protein is likely to mediate protection. Vaccines therapeutic for persisting HPV infection can eliminate transplantable tumors in animal models, but are of limited efficacy in mice grafted with skin that expresses HPV antigens or in humans. This paradox has been partially resolved by data clarifying the immunoregulatory role of skin cytokines (e.g. transforming growth factor-beta and interleukin-10) and the consequences of antigen presentation by subsets of skin-associated antigen-presenting cells.
Collapse
Affiliation(s)
- Graham R Leggatt
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | | |
Collapse
|
80
|
Lechner A, Ritter U, Varona R, Marquez G, Bogdan C, Körner H. Protective immunity and delayed type hypersensitivity reaction are uncoupled in experimental Leishmania major infection of CCR6-negative mice. Microbes Infect 2007; 9:291-9. [PMID: 17317260 DOI: 10.1016/j.micinf.2006.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Revised: 12/15/2006] [Accepted: 12/15/2006] [Indexed: 10/23/2022]
Abstract
The chemokine receptor CCR6 is expressed on naïve B cells, dendritic cell and T-cell subpopulations and is involved in cell navigation during organogenesis and recruitment in response to inflammatory stimuli. Gene-deficient C57BL/6 CCR6(-/-) mice infected with the protozoan parasite Leishmania (L.) major were able to mount a protective immune response and survived the infection. Whereas macrophage production of nitric oxide (NO), the key leishmanicidal effector molecule during the immune response to L. major, did not require CCR6, the migration of CD4(+) T cells to the site of infection was reduced in CCR6(-/-) mice. Furthermore, the induction of a T-cell-dependent delayed-type-hypersensitivity (DTH) reaction was defective in CCR6(-/-) mice, whereas resistance to re-infection was maintained in the absence of CCR6. We conclude that CCR6 contributes to the recruitment of T cells to the site of infection, but is largely dispensable for the control of L. major parasites during primary or secondary infection.
Collapse
Affiliation(s)
- Anja Lechner
- Nachwuchsgruppe 1 des Interdisziplinären Zentrums für Klinische Forschung am Nikolaus Fiebiger Zentrum der Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
81
|
Shamshiev AT, Ampenberger F, Ernst B, Rohrer L, Marsland BJ, Kopf M. Dyslipidemia inhibits Toll-like receptor-induced activation of CD8alpha-negative dendritic cells and protective Th1 type immunity. ACTA ACUST UNITED AC 2007; 204:441-52. [PMID: 17296788 PMCID: PMC2118729 DOI: 10.1084/jem.20061737] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Environmental factors, including diet, play a central role in influencing the balance of normal immune homeostasis; however, many of the cellular mechanisms maintaining this balance remain to be elucidated. Using mouse models of genetic and high-fat/cholesterol diet–induced dyslipidemia, we examined the influence of dyslipidemia on T cell and dendritic cell (DC) responses in vivo and in vitro. We show that dyslipidemia inhibited Toll-like receptor (TLR)–induced production of proinflammatory cytokines, including interleukin (IL)-12, IL-6, and tumor necrosis factor-α, as well as up-regulation of costimulatory molecules by CD8α− DCs, but not by CD8α+ DCs, in vivo. Decreased DC activation profoundly influenced T helper (Th) cell responses, leading to impaired Th1 and enhanced Th2 responses. As a consequence of this immune modulation, host resistance to Leishmania major was compromised. We found that oxidized low-density lipoprotein (oxLDL) was the key active component responsible for this effect, as it could directly uncouple TLR-mediated signaling on CD8α− myeloid DCs and inhibit NF-κB nuclear translocation. These results show that a dyslipidemic microenvironment can directly interfere with DC responses to pathogen-derived signals and skew the development of T cell–mediated immunity.
Collapse
Affiliation(s)
- Abdijapar T Shamshiev
- Molecular Biomedicine, Institute of Integrative Biology, Swiss Federal Institute of Technology Zürich, 8952 Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|