51
|
Abstract
Extracellular ATP and adenosine have immunoregulatory roles during inflammation. Elevated extracellular ATP is known to exacerbate GVHD, and the pharmacologic activation of the adenosine A2A receptor is protective. However, the role of endogenous adenosine is unknown. We used gene-targeted mice and a pharmacologic inhibitor to test the role of adenosine generated by CD73/ecto-5'-nucleotidase in GVHD. In allogeneic transplants, both donor and recipient CD73 were protective, with recipient CD73 playing the dominant role. CD73 deficiency led to enhanced T-cell expansion and IFN-γ and IL-6 production, and the migratory capacity of Cd73-/- T cells in vitro was increased. However, the number of regulatory T cells and expression of costimulatory molecules on antigen-presenting cells were unchanged. A2A receptor deficiency led to increased numbers of allogeneic T cells, suggesting that signaling through the A2A receptor via CD73-generated adenosine is a significant part of the mechanism by which CD73 limits the severity of GVHD. Pharmacologic blockade of CD73 also enhanced graft-versus-tumor activity. These data have clinical implications, as both the severity of GVHD and the strength of an alloimmune antitumor response could be manipulated by enhancing or blocking CD73 activity or adenosine receptor signaling depending on the clinical indication.
Collapse
|
52
|
Spleen tyrosine kinase (Syk) is a potent target for GvHD prevention at different cellular levels. Leukemia 2012; 26:1617-29. [DOI: 10.1038/leu.2012.10] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
53
|
Kish DD, Gorbachev AV, Fairchild RL. IL-1 receptor signaling is required at multiple stages of sensitization and elicitation of the contact hypersensitivity response. THE JOURNAL OF IMMUNOLOGY 2012; 188:1761-71. [PMID: 22238457 DOI: 10.4049/jimmunol.1100928] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Contact hypersensitivity (CHS) is a CD8 T cell-mediated response to hapten skin sensitization and challenge. The points at which IL-1R signaling is required during this complex, multistep immune response have not been clearly delineated. The role of IL-1R signaling during 2, 4 dinitro-1-fluorobenezene (DNFB) sensitization to induce hapten-specific CD8 effector T cells and in the trafficking of the effector T cells to the DNFB challenge site to elicit the response were investigated using IL-1R deficient mice. DNFB-sensitized IL-1R(-/-) mice had low CHS responses to hapten challenge that were caused in part by marked decreases in hapten-specific CD8 T cell development to IL-17- and IFN-γ-producing cells during sensitization. Hapten-primed wild type CD8 T cell transfer to naive IL-1R(-/-) mice did not result in T cell activation in response to hapten challenge, indicating a need for IL-1R signaling for the localization or activation, or both, of the CD8 T cells at the challenge site. Decreased CD8 T cell priming in sensitized IL-1R(-/-) mice was associated with marked decreases in hapten-presenting dendritic cell migration from the sensitized skin to draining lymph nodes. Transfer of hapten-presenting dendritic cells from wild type donors to naive IL-1R(-/-) mice resulted in decreased numbers of the dendritic cells in the draining lymph nodes and decreased priming of hapten-specific CD8 T cells compared with dendritic cell transfer to naive wild type recipients. These results indicate that IL-1R signaling is required at multiple steps during the course of sensitization and challenge to elicit CHS.
Collapse
Affiliation(s)
- Danielle D Kish
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195-0001, USA
| | | | | |
Collapse
|
54
|
Peiser M, Tralau T, Heidler J, Api AM, Arts JHE, Basketter DA, English J, Diepgen TL, Fuhlbrigge RC, Gaspari AA, Johansen JD, Karlberg AT, Kimber I, Lepoittevin JP, Liebsch M, Maibach HI, Martin SF, Merk HF, Platzek T, Rustemeyer T, Schnuch A, Vandebriel RJ, White IR, Luch A. Allergic contact dermatitis: epidemiology, molecular mechanisms, in vitro methods and regulatory aspects. Current knowledge assembled at an international workshop at BfR, Germany. Cell Mol Life Sci 2011; 69:763-81. [PMID: 21997384 PMCID: PMC3276771 DOI: 10.1007/s00018-011-0846-8] [Citation(s) in RCA: 232] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 08/29/2011] [Accepted: 09/20/2011] [Indexed: 12/13/2022]
Abstract
Contact allergies are complex diseases, and one of the important challenges for public health and immunology. The German ‘Federal Institute for Risk Assessment’ hosted an ‘International Workshop on Contact Dermatitis’. The scope of the workshop was to discuss new discoveries and developments in the field of contact dermatitis. This included the epidemiology and molecular biology of contact allergy, as well as the development of new in vitro methods. Furthermore, it considered regulatory aspects aiming to reduce exposure to contact sensitisers. An estimated 15–20% of the general population suffers from contact allergy. Workplace exposure, age, sex, use of consumer products and genetic predispositions were identified as the most important risk factors. Research highlights included: advances in understanding of immune responses to contact sensitisers, the importance of autoxidation or enzyme-mediated oxidation for the activation of chemicals, the mechanisms through which hapten-protein conjugates are formed and the development of novel in vitro strategies for the identification of skin-sensitising chemicals. Dendritic cell cultures and structure-activity relationships are being developed to identify potential contact allergens. However, the local lymph node assay (LLNA) presently remains the validated method of choice for hazard identification and characterisation. At the workshop the use of the LLNA for regulatory purposes and for quantitative risk assessment was also discussed.
Collapse
Affiliation(s)
- M. Peiser
- Department of Product Safety, German Federal Institute for Risk Assessment (BfR), Thielallee 88-92, 14195 Berlin, Germany
| | - T. Tralau
- Department of Product Safety, German Federal Institute for Risk Assessment (BfR), Thielallee 88-92, 14195 Berlin, Germany
| | - J. Heidler
- Department of Product Safety, German Federal Institute for Risk Assessment (BfR), Thielallee 88-92, 14195 Berlin, Germany
| | - A. M. Api
- Research Institute for Fragrance Materials, Hackensack, NJ USA
| | | | | | - J. English
- Nottingham University Hospitals, Nottingham, UK
| | - T. L. Diepgen
- Department of Social Medicine, Occupational and Environmental Dermatology, University of Heidelberg, Heidelberg, Germany
| | | | - A. A. Gaspari
- School of Medicine, University of Maryland, Baltimore, MD USA
| | - J. D. Johansen
- Department of Derma-allergology, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - A. T. Karlberg
- Department of Chemistry, Dermatochemistry and Skin Allergy, University of Gothenburg, Gothenburg, Sweden
| | - I. Kimber
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | - M. Liebsch
- Department of Experimental Toxicology and ZEBET, Center for Alternatives to Animal Testing, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - H. I. Maibach
- Department of Dermatology, University of California San Francisco, San Francisco, CA USA
| | - S. F. Martin
- Allergy Research Group, Department of Dermatology, University Medical Center Freiburg, Freiburg, Germany
| | - H. F. Merk
- Department of Dermatology and Allergology, University Hospitals Aachen, Aachen, Germany
| | - T. Platzek
- Department of Product Safety, German Federal Institute for Risk Assessment (BfR), Thielallee 88-92, 14195 Berlin, Germany
| | - T. Rustemeyer
- VU University Medical Center, Amsterdam, The Netherlands
| | - A. Schnuch
- Department of Dermatology, University of Göttingen, Göttingen, Germany
| | - R. J. Vandebriel
- National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - I. R. White
- St. John’s Institute of Dermatology, St. Thomas’ Hospital, London, UK
| | - A. Luch
- Department of Product Safety, German Federal Institute for Risk Assessment (BfR), Thielallee 88-92, 14195 Berlin, Germany
- Department of Experimental Toxicology and ZEBET, Center for Alternatives to Animal Testing, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| |
Collapse
|
55
|
Martin SF, Esser PR, Weber FC, Jakob T, Freudenberg MA, Schmidt M, Goebeler M. Mechanisms of chemical-induced innate immunity in allergic contact dermatitis. Allergy 2011; 66:1152-63. [PMID: 21599706 DOI: 10.1111/j.1398-9995.2011.02652.x] [Citation(s) in RCA: 201] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Allergic contact dermatitis (ACD) is one of the most prevalent occupational skin diseases and causes severe and long-lasting health problems in the case of chronification. It is initiated by an innate inflammatory immune response to skin contact with low molecular weight chemicals that results in the priming of chemical-specific, skin-homing CD8(+) Tc1/Tc17 and CD4(+) Th1/Th17 cells. Following this sensitization step, T lymphocytes infiltrate the inflamed skin upon challenge with the same chemical. The T cells then exert cytotoxic function and secrete inflammatory mediators to produce an eczematous skin reaction. The recent characterization of the mechanisms underlying the innate inflammatory response has revealed that contact allergens activate innate effector mechanisms and signalling pathways that are also involved in anti-infectious immunity. This emerging analogy implies infection as a potential trigger or amplifier of the sensitization to contact allergens. Moreover, new mechanistic insights into the induction of ACD identify potential targets for preventive and therapeutic intervention. We summarize here the latest findings in this area of research.
Collapse
Affiliation(s)
- S F Martin
- Allergy Research Group, Department of Dermatology, University Medical Center Freiburg, Hauptstrasse 7, Freiburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
56
|
Abstract
Irrespective of the immune status, the vast majority of all lymphocytes reside in peripheral tissues whereas those present in blood only amount to a small fraction of the total. It has been estimated that T cells in healthy human skin outnumber those present in blood by at least a factor of two. How lymphocytes within these two compartments relate to each other is not well understood. However, mounting evidence suggest that the study of T cell subsets present in peripheral blood does not reflect the function of their counterparts at peripheral sites. This is especially true under steady-state conditions whereby long-lived memory T cells in healthy tissues, notably those in epithelial tissues at body surfaces, are thought to fulfill a critical immune surveillance function by contributing to the first line of defense against a series of local threats, including microbes, tumors, and toxins, and by participating in wound healing. The relative scarcity of information regarding peripheral T cells and the factors regulating their localization is primarily due to inherent difficulties in obtaining healthy tissue for the extraction and study of immune cells on a routine basis. This is most certainly true for humans. Here, we review our current understanding of T cell homing to human skin and compare it when possible with gut-selective homing. We also discuss candidate chemokines that may account for the tissue selectivity in this process and present a model whereby CCR8, and its ligand CCL1, selectively regulate the homeostatic migration of memory lymphocytes to skin tissue.
Collapse
Affiliation(s)
- Michelle L McCully
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University Cardiff, UK
| | | |
Collapse
|
57
|
Sigmundsdottir H. From the bench to the clinic: New aspects on immunoregulation by vitamin D analogs. DERMATO-ENDOCRINOLOGY 2011; 3:187-92. [PMID: 22110778 DOI: 10.4161/derm.3.3.15115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 02/08/2011] [Accepted: 02/09/2011] [Indexed: 01/08/2023]
Abstract
The skin is a vital organ that plays a crucial role in defending us from pathogens. Multiple players from the innate and adaptive immune system are involved, such as neutrophils, dendritic cells, lymphocytes and antimicrobial peptides. Chronic inflammatory skin diseases can be mediated by inflammatory T cells and their interactions with other cells in the skin. Vitamin D is generated in the skin upon sun exposure and has a variety of effects. Vitamin D and its analogs have been used with success in treating mild to moderate T cell-mediated skin diseases, but how they mediate the beneficial effects is not well understood. In the recent years, emerging evidence is rising that vitamin D analogs and its modulation on the immune system plays a major role. It has been shown that vitamin D analogs can induce the generation of regulatory T cells, which are able to suppress proliferation and alter the function of inflammatory T cells. This may help explain the therapeutic effects that are observed and at the same time give hope that in combination with other therapy or used alone, vitamin D analogs may be helpful when treating more severe forms of the diseases.
Collapse
Affiliation(s)
- Hekla Sigmundsdottir
- Department of Hematology Laboratory; Landspitali-The National University Hospital of Iceland; Hringbraut; and Faculty of Medicine; School of Health Sciences; University of Iceland; Reykjavik, Iceland
| |
Collapse
|
58
|
Wang S, Villablanca EJ, De Calisto J, Gomes DCO, Nguyen DD, Mizoguchi E, Kagan JC, Reinecker HC, Hacohen N, Nagler C, Xavier RJ, Rossi-Bergmann B, Chen YB, Blomhoff R, Snapper SB, Mora JR. MyD88-dependent TLR1/2 signals educate dendritic cells with gut-specific imprinting properties. THE JOURNAL OF IMMUNOLOGY 2011; 187:141-50. [PMID: 21646294 DOI: 10.4049/jimmunol.1003740] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gut-associated dendritic cells (DC) synthesize all-trans retinoic acid, which is required for inducing gut-tropic lymphocytes. Gut-associated DC from MyD88(-/-) mice, which lack most TLR signals, expressed low levels of retinal dehydrogenases (critical enzymes for all-trans retinoic acid biosynthesis) and were significantly impaired in their ability to induce gut-homing T cells. Pretreatment of extraintestinal DC with a TLR1/2 agonist was sufficient to induce retinal dehydrogenases and to confer these DC with the capacity to induce gut-homing lymphocytes via a mechanism dependent on MyD88 and JNK/MAPK. Moreover, gut-associated DC from TLR2(-/-) mice, or from mice in which JNK was pharmacologically blocked, were impaired in their education to imprint gut-homing T cells, which correlated with a decreased induction of gut-tropic T cells in TLR2(-/-) mice upon immunization. Thus, MyD88-dependent TLR2 signals are necessary and sufficient to educate DC with gut-specific imprinting properties and contribute in vivo to the generation of gut-tropic T cells.
Collapse
Affiliation(s)
- Sen Wang
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Oral exposure to Trypanosoma cruzi elicits a systemic CD8⁺ T cell response and protection against heterotopic challenge. Infect Immun 2011; 79:3397-406. [PMID: 21628516 DOI: 10.1128/iai.01080-10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Trypanosoma cruzi infects millions of people in Latin America and often leads to the development of Chagas disease. T. cruzi infection can be acquired at or near the bite site of the triatomine vector, but per os infection is also a well-documented mode of transmission, as evidenced by recent microepidemics of acute Chagas disease attributed to the consumption of parasite-contaminated foods and liquids. It would also be convenient to deliver vaccines for T. cruzi by the oral route, particularly live parasite vaccines intended for the immunization of reservoir hosts. For these reasons, we were interested in better understanding immunity to T. cruzi following oral infection or oral vaccination, knowing that the route of infection and site of antigen encounter can have substantial effects on the ensuing immune response. Here, we show that the route of infection does not alter the ability of T. cruzi to establish infection in muscle tissue nor does it impair the generation of a robust CD8(+) T cell response. Importantly, oral vaccination with attenuated parasites provides protection against wild-type (WT) T. cruzi challenge. These results strongly support the development of whole-organism-based vaccines targeting reservoir species as a means to alleviate the burden of Chagas disease in affected regions.
Collapse
|
60
|
Oyoshi MK, Elkhal A, Scott JE, Wurbel MA, Hornick JL, Campbell JJ, Geha RS. Epicutaneous challenge of orally immunized mice redirects antigen-specific gut-homing T cells to the skin. J Clin Invest 2011; 121:2210-20. [PMID: 21537081 DOI: 10.1172/jci43586] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 03/02/2011] [Indexed: 12/31/2022] Open
Abstract
Patients with atopic dermatitis (AD) often suffer from food allergy and develop flares upon skin contact with food allergens. However, it is unclear whether T cells sensitized to allergens in the gut promote this skin inflammation. To address this question, we orally immunized WT mice and mice lacking the skin-homing chemokine receptor Ccr4 (Ccr4-/- mice) with OVA and then challenged them epicutaneously with antigen. Allergic skin inflammation developed in the WT mice but not in the mutants and was characterized by epidermal thickening, dermal infiltration by eosinophils and CD4+ T cells, and upregulation of Th2 cytokines. T cells purified from mesenteric lymph nodes (MLNs) of orally immunized WT mice transferred allergic skin inflammation to naive recipients cutaneously challenged with antigen, but this effect was lost in T cells purified from Ccr4-/- mice. In addition, the ability of adoptively transferred OVA-activated T cells to home to the skin following cutaneous OVA challenge was ablated in mice that lacked lymph nodes. These results indicate that cutaneous exposure to food antigens can reprogram gut-homing effector T cells in LNs to express skin-homing receptors, eliciting skin lesions upon food allergen contact in orally sensitized AD patients.
Collapse
Affiliation(s)
- Michiko K Oyoshi
- Division of Immunology, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
61
|
Lass C, Merfort I, Martin SF. In vitro and in vivo analysis of pro- and anti-inflammatory effects of weak and strong contact allergens. Exp Dermatol 2011; 19:1007-13. [PMID: 20701630 DOI: 10.1111/j.1600-0625.2010.01136.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Inflammation is a crucial step in the development of allergic contact dermatitis. The primary contact with chemical allergens, called sensitization, and the secondary contact, called elicitation, result in an inflammatory response in the skin. The ability of contact allergens to induce allergic contact dermatitis correlates to a great extent with their inflammatory potential. Therefore, the analysis of the sensitizing potential of a putative contact allergen should include the examination of its ability and potency to cause an inflammation. In this study, we examined the inflammatory potential of different weak contact allergens and of the strong sensitizer 2,4,6-trinitrochlorobenzene (TNCB) in vitro and in vivo using the contact hypersensitivity model, the mouse model for allergic contact dermatitis. Cytokine induction was analysed by PCR and ELISA to determine mRNA and protein levels, respectively. Inflammation-dependent recruitment of skin-homing effector T cells was measured in correlation with the other methods. We show that the sensitizing potential of a contact allergen correlates with the strength of the inflammatory response. The different methods used gave similar results. Quantitative cytokine profiling may be used to determine the sensitizing potential of chemicals for hazard identification and risk assessment.
Collapse
Affiliation(s)
- Christian Lass
- Allergy Research Group, Department of Dermatology, University Medical Center Freiburg, Hauptstrasse, Freiburg, Germany
| | | | | |
Collapse
|
62
|
Koscielny A, Engel D, Maurer J, Hirner A, Kurts C, Kalff JC. Impact of CCR7 on the gastrointestinal field effect. Am J Physiol Gastrointest Liver Physiol 2011; 300:G665-75. [PMID: 21292999 DOI: 10.1152/ajpgi.00224.2010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Standardized intestinal manipulation (IM) leads to local bowel wall inflammation subsequently spreading over the entire gastrointestinal tract. Previously, we demonstrated that this so-called gastrointestinal field effect (FE) is immune mediated. This study aimed to investigate the role of CCR7 in IM-induced FE. Since CCR7 is expressed on activated dendritic cells and T cells and is well known to control their migration, we hypothesized that lack of CCR7 reduces or abolishes FE. Small bowel muscularis and colonic muscularis from CCR7(-/-) and wild-type (WT) mice were obtained after IM of the jejunum or sham operation. FE was analyzed by measuring gastrointestinal transit time of orally given fluorescent dextran (geometric center), colonic transit time, infiltration of MPO-positive cells, and circular smooth muscle contractility. Furthermore, mRNA levels of the inflammatory cytokine IL-6 were determined by RT-PCR. The number of dendritic cells and CD3+CD25+ T cells separately isolated from jejunum and colon was determined in mice after IM and sham operation. There was no significant difference in IL-6 mRNA upregulation in colonic muscularis between sham-operated WT and CCR7(-/-) mice after IM. Contractility of circular muscularis strips of the colon was significantly improved in CCR7(-/-) animals following IM and did not vary significantly from sham-operated animals. Additionally, inflammation of the colon determined by the number of MPO-positive cells and colonic transit time was significantly reduced in CCR7(-/-) mice. In contrast, jejunal contractility and jejunal inflammation of transgenic mice did not differ significantly from WT mice after IM. These data are supported by a significant increase of CD3+CD25+ T cells in the colonic muscularis of WT mice after IM, which could not be observed in CCR7(-/-) mice. These data demonstrate that CCR7 is required for FE and postoperative ileus. CCR7 indirectly affects FE by inhibiting migration of activated dendritic cells and of T cells from the jejunum to the colon. These findings support the critical role of the adaptive immune system in FE.
Collapse
Affiliation(s)
- A Koscielny
- Department of Surgery, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany.
| | | | | | | | | | | |
Collapse
|
63
|
Epigenetic modification of the human CCR6 gene is associated with stable CCR6 expression in T cells. Blood 2011; 117:2839-46. [PMID: 21228329 DOI: 10.1182/blood-2010-06-293027] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
CCR6 is a chemokine receptor expressed on Th17 cells and regulatory T cells that is induced by T-cell priming with certain cytokines, but how its expression and stability are regulated at the molecular level is largely unknown. Here, we identified and characterized a noncoding region of the human CCR6 locus that displayed unmethylated CpG motifs (differentially methylated region [DMR]) selectively in CCR6(+) lymphocytes. CCR6 expression on circulating CD4(+) T cells was stable on cytokine-induced proliferation but partially down-regulated on T-cell receptor stimulation. However, CCR6 down-regulation was mostly transient, and the DMR within the CCR6 locus remained demethylated. Notably, in vitro induction of CCR6 expression with cytokines in T-cell receptor-activated naive CD4(+) T cells was not associated with a demethylated DMR and resulted in unstable CCR6 expression. Conversely, treatment with the DNA methylation inhibitor 5'-azacytidine induced demethylation of the DMR and led to increased and stable CCR6 expression. Finally, when cloned into a reporter gene plasmid, the DMR displayed transcriptional activity in memory T cells that was suppressed by DNA methylation. In summary, we have identified a noncoding region of the human CCR6 gene with methylation-sensitive transcriptional activity in CCR6(+) T cells that controls stable CCR6 expression via epigenetic mechanisms.
Collapse
|
64
|
Iwata M, Yokota A. Retinoic acid production by intestinal dendritic cells. VITAMINS AND HORMONES 2011; 86:127-52. [PMID: 21419270 DOI: 10.1016/b978-0-12-386960-9.00006-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Subpopulations of dendritic cells (DCs) in the small intestine and its related lymphoid organs can produce retinoic acid (RA) from vitamin A (retinol). Through the RA production, these DCs play a pivotal role in imprinting lymphocytes with gut-homing specificity, and contribute to the development of immune tolerance by enhancing the differentiation of Foxp3(+) regulatory T cells and inhibiting that of inflammatory Th17 cells. The RA-producing capacity in these DCs mostly depends on the expression of retinal dehydrogenase 2 (RALDH2, ALDH1A2). It is likely that the RALDH2 expression is induced in DCs by the microenvironmental factors in the small intestine and its related lymphoid organs. The major factor responsible for the RALDH2 expression appears to be GM-CSF. RA itself is essential for the GM-CSF-induced RALDH2 expression. IL-4 and IL-13 also enhance RALDH2 expression, but are dispensable. Toll-like receptor-mediated signals can also enhance the GM-CSF-induced RALDH2 expression in immature DCs.
Collapse
Affiliation(s)
- Makoto Iwata
- Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University, Sanuki-shi, Kagawa, Japan
| | | |
Collapse
|
65
|
Adjuvants and delivery systems in veterinary vaccinology: current state and future developments. Arch Virol 2010; 156:183-202. [PMID: 21170730 DOI: 10.1007/s00705-010-0863-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Accepted: 11/13/2010] [Indexed: 12/20/2022]
Abstract
Modern adjuvants should induce strong and balanced immune responses, and it is often desirable to induce specific types of immunity. As an example, efficient Th1-immunity-inducing adjuvants are highly in demand. Such adjuvants promote good cell-mediated immunity against subunit vaccines that have low immunogenicity themselves. The development of such adjuvants may take advantage of the increased knowledge of the molecular mechanisms and factors controlling these responses. However, knowledge of such molecular details of immune mechanisms is relatively scarce for species other than humans and laboratory rodents, and in addition, there are special considerations pertaining to the use of adjuvants in veterinary animals, such as production and companion animals. With a focus on veterinary animals, this review highlights a number of approaches being pursued, including cytokines, CpG oligonucleotides, microparticles and liposomes.
Collapse
|
66
|
CXCL12 Mediates Immunosuppression in the Lymphoma Microenvironment after Allogeneic Transplantation of Hematopoietic Cells. Cancer Res 2010; 70:10170-81. [DOI: 10.1158/0008-5472.can-10-1943] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
67
|
Ohoka Y, Yokota A, Takeuchi H, Maeda N, Iwata M. Retinoic acid-induced CCR9 expression requires transient TCR stimulation and cooperativity between NFATc2 and the retinoic acid receptor/retinoid X receptor complex. THE JOURNAL OF IMMUNOLOGY 2010; 186:733-44. [PMID: 21148038 DOI: 10.4049/jimmunol.1000913] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Retinoic acid (RA) imprints gut-homing specificity on T cells upon activation by inducing the expression of chemokine receptor CCR9 and integrin α4β7. CCR9 expression seemed to be more highly dependent on RA than was the α4β7 expression, but its molecular mechanism remained unclear. In this article, we show that NFAT isoforms NFATc1 and NFATc2 directly interact with RA receptor (RAR) and retinoid X receptor (RXR) but play differential roles in RA-induced CCR9 expression on murine naive CD4(+) T cells. TCR stimulation for 6-24 h was required for the acquisition of responsiveness to RA and induced activation of NFATc1 and NFATc2. However, RA failed to induce CCR9 expression as long as TCR stimulation continued. After terminating TCR stimulation or adding cyclosporin A to the culture, Ccr9 gene transcription was induced, accompanied by inactivation of NFATc1 and sustained activation of NFATc2. Reporter and DNA-affinity precipitation assays demonstrated that the binding of NFATc2 to two NFAT-binding sites and that of the RAR/RXR complex to an RA response element half-site in the 5'-flanking region of the mouse Ccr9 gene were critical for RA-induced promoter activity. NFATc2 directly bound to RARα and RXRα, and it enhanced the binding of RARα to the RA response element half-site. NFATc1 also bound to the NFAT-binding sites and directly to RARα and RXRα, but it inhibited the NFATc2-dependent promoter activity. These results suggest that the cooperativity between NFATc2 and the RAR/RXR complex is essential for CCR9 expression on T cells and that NFATc1 interferes with the action of NFATc2.
Collapse
Affiliation(s)
- Yoshiharu Ohoka
- Laboratory of Biodefense Research, Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University, Kagawa 769-2193, Japan.
| | | | | | | | | |
Collapse
|
68
|
Hart AL, Ng SC, Mann E, Al-Hassi HO, Bernardo D, Knight SC. Homing of immune cells: role in homeostasis and intestinal inflammation. Inflamm Bowel Dis 2010; 16:1969-77. [PMID: 20848507 DOI: 10.1002/ibd.21304] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Rather like a satellite navigation system directing a vehicle to a particular destination defined by post-code, immune cells have homing molecules or "immune post-codes" enabling them to be recruited to specific organs, such as the intestine or skin. An efficient system would be designed such that the site of entry of an antigen influences the homing of effector T cells back to the appropriate organ. For example, to mount an immune response against an intestinal pathogen, T cells with a propensity to home to the gut to clear the infection would be induced. In health, there is such a sophisticated and finely tuned system in operation, enabling an appropriate balance of immune activity in different anatomical compartments. In disease states such as inflammatory bowel disease (IBD), which is characterized by intestinal inflammation and often an inflammatory process involving other organs such as skin, joints, liver, and eye, there is accumulating evidence that there is malfunction of this immune cell trafficking system. The clinical importance of dysregulated immune cell trafficking in IBD is reflected in recently proven efficacious therapies that target trafficking pathways such as natalizumab, an α4 integrin antibody, and Traficet-EN, a chemokine receptor-9 (CCR9) antagonist. Here we review the mechanisms involved in the homing of immune cells to different tissues, in particular the intestine, and focus on alterations in immune cell homing pathways in IBD. Unraveling the mechanisms underlying the immune post-code system would assist in achieving the goal of tissue-specific immunotherapy.
Collapse
Affiliation(s)
- Ailsa L Hart
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St Mark's Campus, Harrow, Middlesex, UK.
| | | | | | | | | | | |
Collapse
|
69
|
Kapp K, Maul J, Hostmann A, Mundt P, Preiss JC, Wenzel A, Thiel A, Zeitz M, Ullrich R, Duchmann R. Modulation of systemic antigen-specific immune responses by oral antigen in humans. Eur J Immunol 2010; 40:3128-37. [PMID: 20957752 DOI: 10.1002/eji.201040701] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Revised: 07/21/2010] [Accepted: 08/16/2010] [Indexed: 11/06/2022]
Abstract
Oral antigen uptake can induce systemic immune responses ranging from tolerance to immunity. However, the underlying mechanisms are poorly understood, especially in humans. Here, keyhole limpet hemocyanin (KLH), a neoantigen which has been used in earlier studies of oral tolerance, was fed in a repeated low-dose and a single high-dose protocol to healthy volunteers. KLH-specific CD4(+) T-cell proliferation and cytokine production, as well as KLH-specific serum Ab and the effects of oral KLH on a subsequent parenterally induced systemic immune response, were analyzed. Repeated low-dose oral KLH alone induced antigen-specific CD4(+) T cells positive predominantly for the gut-homing receptor integrin β7 and the cytokines IL-2 and TNF-α; some CD4(+) T cells also produced IL-4. Oral feeding of KLH accelerated a subsequent parenterally induced systemic CD4(+) T-cell response. The cytokine pattern of KLH-specific CD4(+) T cells shifted toward more IL-4- and IL-10- and less IFN-γ-, IL-2- and TNF-α-producing cells. The parenterally induced systemic KLH-specific B-cell response was accelerated and amplified by oral KLH. The impact of single high-dose oral KLH on antigen-specific immune responses was less pronounced compared with repeated low-dose oral KLH. These findings suggest that oral antigen can effectively modulate subsequently induced systemic antigen-specific immune responses. Immunomodulation by oral antigen may offer new therapeutic strategies for Th type1-mediated inflammatory diseases and for the development of vaccination strategies.
Collapse
Affiliation(s)
- Kerstin Kapp
- Charité-Campus Benjamin Franklin, Medizinische Klinik I (Gastroenterologie, Infektiologie, Rheumatologie), Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Ng SC, Kamm MA, Stagg AJ, Knight SC. Intestinal dendritic cells: their role in bacterial recognition, lymphocyte homing, and intestinal inflammation. Inflamm Bowel Dis 2010; 16:1787-807. [PMID: 20222140 DOI: 10.1002/ibd.21247] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) play a key role in discriminating between commensal microorganisms and potentially harmful pathogens and in maintaining the balance between tolerance and active immunity. The regulatory role of DC is of particular importance in the gut where the immune system lies in intimate contact with the highly antigenic external environment. Intestinal DC constantly survey the luminal microenvironment. They act as sentinels, acquiring antigens in peripheral tissues before migrating to secondary lymphoid organs to activate naive T cells. They are also sensors, responding to a spectrum of environmental cues by extensive differentiation or maturation. Recent studies have begun to elucidate mechanisms for functional specializations of DC in the intestine that may include the involvement of retinoic acid and transforming growth factor-β. Specialized CD103(+) intestinal DC can promote the differentiation of Foxp3(+) regulatory T cells via a retinoic acid-dependent process. Different DC outcomes are, in part, influenced by their exposure to microbial stimuli. Evidence is also emerging of the close interaction between bacteria, epithelial cells, and DC in the maintenance of intestinal immune homeostasis. Here we review recent advances of functionally specialized intestinal DC and their mechanisms of antigen uptake and recognition. We also discuss the interaction of DC with intestinal microbiota and their ability to orchestrate protective immunity and immune tolerance in the host. Lastly, we describe how DC functions are altered in intestinal inflammation and their emerging potential as a therapeutic target in inflammatory bowel disease.
Collapse
Affiliation(s)
- S C Ng
- Antigen Presentation Research Group, Faculty of Medicine, Imperial College London, Northwick Park and St Mark's Campus, Harrow, UK
| | | | | | | |
Collapse
|
71
|
Takeuchi H, Yokota A, Ohoka Y, Kagechika H, Kato C, Song SY, Iwata M. Efficient induction of CCR9 on T cells requires coactivation of retinoic acid receptors and retinoid X receptors (RXRs): exaggerated T Cell homing to the intestine by RXR activation with organotins. THE JOURNAL OF IMMUNOLOGY 2010; 185:5289-99. [PMID: 20881191 DOI: 10.4049/jimmunol.1000101] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The active vitamin A metabolite retinoic acid (RA) imprints gut-homing specificity on lymphocytes upon activation by inducing the expression of α4β7 integrin and CCR9. RA receptor (RAR) activation is essential for their expression, whereas retinoid X receptor (RXR) activation is not essential for α4β7 expression. However, it remains unclear whether RXR activation affects the RA-dependent CCR9 expression on T cells and their gut homing. The major physiological RA, all-trans-RA, binds to RAR but not to RXR at physiological concentrations. Cell-surface CCR9 expression was often induced on a limited population of murine naive CD4(+) T cells by all-trans-RA or the RAR agonist Am80 alone upon CD3/CD28-mediated activation in vitro, but it was markedly enhanced by adding the RXR agonist PA024 or the RXR-binding environmental chemicals tributyltin and triphenyltin. Accordingly, CD4(+) T cells treated with the combination of all-trans-RA and tributyltin migrated into the small intestine upon adoptive transfer much more efficiently than did those treated with all-trans-RA alone. Furthermore, naive TCR transgenic CD4(+) T cells transferred into wild-type recipients migrated into the small intestinal lamina propria following i.p. injection of Ag, and the migration was enhanced by i.p. injection of PA024. We also show that PA024 markedly enhanced the all-trans-RA-induced CCR9 expression on naturally occurring naive-like regulatory T cells upon activation, resulting in the expression of high levels of α4β7, CCR9, and Foxp3. These results suggest that RXR activation enhances the RAR-dependent expression of CCR9 on T cells and their homing capacity to the small intestine.
Collapse
Affiliation(s)
- Hajime Takeuchi
- Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University, Kagawa, Japan
| | | | | | | | | | | | | |
Collapse
|
72
|
Abstract
Chemokines constitute a large family of low-molecular-weight proteins ( approximately 10 kDa in size), recognized primarily for their role in directing leukocyte migration under both homeostatic and inflammatory settings. The chemokine CCL25 displays a unique and highly restricted expression pattern compared with other chemokine family members. In the steady state, CCL25 is expressed at high levels primarily in the thymus and small intestine, while its sole functional receptor, CCR9, is expressed on subsets of developing thymocytes and intestinal lymphocytes. Mice that are deficient in CCR9 show relatively normal thymocyte development; however, in competitive transfer experiments, CCR9(-/-) bone-marrow cells are severely disadvantaged in their ability to generate mature T cells compared with wildtype cells. Indeed, expression data and analysis of genetically modified mice suggest that CCL25/CCR9 may be involved in multiple stages of thymocyte development. Recent in vivo studies have demonstrated a role for CCL25/CCR9 in mediating lymphocyte recruitment to the small intestine and in the development of the small intestinal T-cell receptor-gammadelta T-cell compartment. Finally, CCL25 is expressed in the small intestine of Crohn's disease patients and, in certain inflammatory conditions, outside the small intestine. Together, these results suggest an important role for CCL25/CCR9 in T-cell development and small intestinal immunity and suggest that targeting the CCL25/CCR9 pathway may provide a means to modulate small intestinal immune responses.
Collapse
|
73
|
Sigmundsdottir H. Improving topical treatments for skin diseases. Trends Pharmacol Sci 2010; 31:239-45. [DOI: 10.1016/j.tips.2010.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 03/24/2010] [Accepted: 03/30/2010] [Indexed: 11/15/2022]
|
74
|
Abstract
The development of immune memory mediated by T lymphocytes is central to durable, long-lasting protective immunity. A key issue in the field is how to direct the generation and persistence of memory T cells to elicit the appropriate secondary response to provide protection to a specific pathogen. Two prevailing views have emerged; that cellular and molecular regulators control the lineage fate and functional capacities of memory T cells early after priming, or alternatively, that populations of memory T cells are inherently plastic and subject to alterations in function and/or survival at many stages during their long-term maintenance. Here, we will review current findings in CD4 T-cell memory that suggest inherent plasticity in populations of memory CD4 T cells at all stages of their development--originating with their generation from multiple types of primed CD4 T cells, during their persistence and homeostatic turnover in response to T-cell receptor signals, and also following secondary challenge. These multiple aspects of memory CD4 T-cell flexibility contrast the more defined lineages and functions ascribed to memory CD8 T cells, suggesting a dynamic nature to memory CD4 T-cell populations and responses. The flexible attributes of CD4 T-cell memory suggest opportunities and mechanisms for therapeutic manipulation at all phases of immune memory development, maintenance and recall.
Collapse
Affiliation(s)
- Jason R Lees
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | |
Collapse
|
75
|
Ferguson AR, Engelhard VH. CD8 T cells activated in distinct lymphoid organs differentially express adhesion proteins and coexpress multiple chemokine receptors. THE JOURNAL OF IMMUNOLOGY 2010; 184:4079-86. [PMID: 20212096 DOI: 10.4049/jimmunol.0901903] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Previous work from this laboratory showed that generation of memory CD8 T cells by different immunization routes correlates with control of tumors growing in distinct sites. We hypothesized that effector CD8 T cell expression of adhesion proteins and chemokine receptors would be influenced by activation in different secondary lymphoid organs. In this report, CD8 T cells were activated by immunization with bone marrow-derived dendritic cells via i.p., i.v., or s.c. routes. Three distinct populations of activated CD8 T cells arise in mesenteric, axillary/brachial, and mediastinal lymph nodes and spleen based on differential expression of alpha4beta7 integrin, E-selectin ligand, and alpha4beta1 integrin, respectively. In contrast, three subsets of CD8 T cells defined by differential expression of P-selectin ligand and chemokine receptors were induced irrespective of activation site. The majority of activated CD8 T cells expressed CXCR3, with one subset additionally expressing P-selectin ligand, and another subset additionally expressing CCR3, CCR4, CCR5, CCR6, and CCR9. In the mesenteric lymph node, a fourth subset expressed CCR9 and CXCR3 in the absence of CCR5. Similar homing receptor profiles were induced in the same sites after localized vaccinia immunization. Homing receptor expression on CD8 T cells activated in vitro was distinct, revealing influences of both dendritic cells and the lymphoid microenvironment. Collectively, these results identify previously undescribed populations of activated CD8 T cells based on adhesion protein expression and coexpression of chemokine receptors that arise after activation in distinct secondary lymphoid organs.
Collapse
Affiliation(s)
- Andrew R Ferguson
- Department of Microbiology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | |
Collapse
|
76
|
Homing in on acute graft vs. host disease: tissue-specific T regulatory and Th17 cells. Curr Top Microbiol Immunol 2010; 341:121-46. [PMID: 20563712 DOI: 10.1007/82_2010_24] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acute graft vs. host disease (aGVHD) is a major limitation of hematopoietic stem cell transplantation (HSCT), and it causes significant morbidity and mortality for this patient population. This immune-mediated injury occurs unpredictably and is caused by donor-derived T cells reacting to recipient alloantigens. Although donor Th1 cells play a critical role in aGVHD generation, numerous arms of both the innate and the adaptive immune systems along with determinants of lymphocyte trafficking are likely involved in the multifaceted cascade of immunological events that culminates in clinical aGVHD. T regulatory and Th17 cells are T cell subsets distinct from Th1 cells that are likely involved with aGVHD. Regulatory T cells (Tregs) have been implicated in the prevention of aGVHD in both mouse and man, while Th17 cells may modulate early inflammatory responses associated with aGVHD, especially those involving the skin and the lungs. Interestingly, these two lymphocyte subsets appear to be reciprocally regulated in part through retinoic acid, through cytokines such as IL-6, and via interactions with dendritic cells. Another area under tight regulation appears to be the homing of lymphocytes to lymph nodes, skin, and gut. Adhesion molecules including chemokine receptors, selectins, and integrins may identify specific T cell subsets with unique migratory functional properties during HSCT. Controlling the migration patterns of Th17 cells and Tregs represents a potential therapeutic target. A major goal of HSCT research will be to develop approaches to pharmacologically manipulate T cell subsets in vivo or to select, expand, and infuse T cell subsets that will maximize the targeted graft vs. tumor effect while minimizing the potentially fatal side effects of aGVHD. A better understanding of Tregs and their tissue specificity should lead to improvement in the success of HSCT.
Collapse
|
77
|
Molenaar R, Greuter M, van der Marel APJ, Roozendaal R, Martin SF, Edele F, Huehn J, Förster R, O'Toole T, Jansen W, Eestermans IL, Kraal G, Mebius RE. Lymph node stromal cells support dendritic cell-induced gut-homing of T cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:6395-402. [PMID: 19841174 DOI: 10.4049/jimmunol.0900311] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
T cells are imprinted to express tissue-specific homing receptors upon activation in tissue-draining lymph nodes, resulting in their migration to the site of Ag entry. Expression of gut-homing molecules alpha(4)beta(7) and CCR9 is induced by retinoic acid, a vitamin A metabolite produced by retinal dehydrogenases, which are specifically expressed in dendritic cells as well as stromal cells in mucosa-draining lymph nodes. In this study, we demonstrate that mesenteric lymph node stromal cell-derived retinoic acid can directly induce the expression of gut-homing molecules on proliferating T cells, a process strongly enhanced by bone marrow-derived dendritic cells in vitro. Therefore, cooperation of sessile lymph node stromal cells with mobile dendritic cells warrants the imprinting of tissue specific homing receptors on activated T cells.
Collapse
Affiliation(s)
- Rosalie Molenaar
- Department of Molecular Cell Biology and Immunology, VU (Vrije Universiteit) University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Averbeck M, Gebhardt C, Simon JC, Treudler R. Resolution of both persistent eczema and implant failure following removal of nickel containing implant. J Eur Acad Dermatol Venereol 2009; 23:1215-6. [DOI: 10.1111/j.1468-3083.2009.03120.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
79
|
Carlow DA, Gossens K, Naus S, Veerman KM, Seo W, Ziltener HJ. PSGL-1 function in immunity and steady state homeostasis. Immunol Rev 2009; 230:75-96. [PMID: 19594630 DOI: 10.1111/j.1600-065x.2009.00797.x] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The substantial importance of P-selectin glycoprotein ligand 1 (PSGL-1) in leukocyte trafficking has continued to emerge beyond its initial identification as a selectin ligand. PSGL-1 seemed to be a relatively simple molecule with an extracellular mucin domain extended as a flexible rod, teleologically consistent with its primary role in tethering leukocytes to endothelial selectins. The rolling interaction between leukocyte and endothelium mediated by this selectin-PSGL-1 interaction requires branched O-glycan extensions on specific PSGL-1 amino acid residues. In some cells, such as neutrophils, the glycosyltransferases involved in formation of the O-glycans are constitutively expressed, while in other cells, such as T cells, they are expressed only after appropriate activation. Thus, PSGL-1 supports leukocyte recruitment in both innate and adaptive arms of the immune response. A complex array of amino acids within the selectins engage multiple sugar residues of the branched O-glycans on PSGL-1 and provide the molecular interactions responsible for the velcro-like catch bonds that support leukocyte rolling. Such binding of PSGL-1 can also induce signaling events that influence cell phenotype and function. Scrutiny of PSGL-1 has revealed a better understanding of how it performs as a selectin ligand and yielded unexpected insights that extend its scope from supporting leukocyte rolling in inflammatory settings to homeostasis including stem cell homing to the thymus and mature T-cell homing to secondary lymphoid organs. PSGL-1 has been found to bind homeostatic chemokines CCL19 and CCL21 and to support the chemotactic response to these chemokines. Surprisingly, the O-glycan modifications of PSGL-1 that support rolling mediated by selectins in inflammatory conditions interfere with PSGL-1 binding to homeostatic chemokines and thereby limit responsiveness to the chemotactic cues used in steady state T-cell traffic. The multi-level influence of PSGL-1 on cell traffic in both inflammatory and steady state settings is therefore substantially determined by the orchestrated addition of O-glycans. However, central as specific O-glycosylation is to PSGL-1 function, in vivo regulation of PSGL-1 glycosylation in T cells remains poorly understood. It is our purpose herein to review what is known, and not known, of PSGL-1 glycosylation and to update understanding of PSGL-1 functional scope.
Collapse
Affiliation(s)
- Douglas A Carlow
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
80
|
Pabst O, Wahl B, Bernhardt G, Hammerschmidt SI. Mesenteric lymph node stroma cells in the generation of intestinal immune responses. J Mol Med (Berl) 2009; 87:945-51. [PMID: 19649572 DOI: 10.1007/s00109-009-0502-z] [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] [Received: 04/30/2009] [Revised: 06/25/2009] [Accepted: 07/06/2009] [Indexed: 12/23/2022]
Abstract
Lymph nodes at different anatomical locations share similar architecture and operate on the basis of identical principles. Still, the quality of immune responses is modified substantially by the local peculiarities at the site of its induction. Here, we discuss how lymph node stroma cells contribute to functional differences between various lymph nodes, thus helping to explain why and how an immune response induced in skin draining peripheral lymph nodes differs from that elicited in the gut draining mesenteric lymph nodes. Stroma cells constitute a major part of the lymph node scaffold and control the flow of immune cells as well as soluble substances within the organ. Moreover, stroma cells express cytokines, chemokines as well as adhesion factors and thereby actively influence immune status. Lymph node transplantations and adoptive transfers of dendritic cells demonstrated that regional lymph node stroma cells differ in their ability to support mucosal tolerance, the induction of tissue tropism, and humoral immunity. This suggests that stroma cells shape tissue-specific immune responses and equip lymph nodes with unique functional properties that might originate during lymph node organogenesis.
Collapse
Affiliation(s)
- Oliver Pabst
- Institute of Immunology, Hannover Medical School, Hannover, Germany.
| | | | | | | |
Collapse
|
81
|
Agace WW. T-cell recruitment to the intestinal mucosa. Trends Immunol 2009; 29:514-22. [PMID: 18838302 DOI: 10.1016/j.it.2008.08.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 08/27/2008] [Accepted: 08/28/2008] [Indexed: 01/02/2023]
Abstract
The intestinal epithelium and underlying lamina propria contains large numbers of T cells that play an important role in maintaining intestinal homeostasis and defense against intestinal pathogens. Recent years have seen several significant advances in our understanding of the mechanisms regulating T-cell localization to the intestinal mucosa. For instance, we now know that the small intestine 'imprints' gut homing properties on T cells by inducing the expression of specific integrins and chemokine receptors. Further studies have identified distinct subsets of intestinal dendritic cells that use retinoic acid to generate both gut-tropic and regulatory T cells. As our understanding of the mechanisms regulating the generation of gut tropic T-cell populations evolves, the possibility of targeting these processes for mucosal vaccine development and treatment of intestinal immune pathology become more apparent.
Collapse
|
82
|
Knieke K, Hoff H, Maszyna F, Kolar P, Schrage A, Hamann A, Debes GF, Brunner-Weinzierl MC. CD152 (CTLA-4) determines CD4 T cell migration in vitro and in vivo. PLoS One 2009; 4:e5702. [PMID: 19479036 PMCID: PMC2682661 DOI: 10.1371/journal.pone.0005702] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Accepted: 04/29/2009] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Migration of antigen-experienced T cells to secondary lymphoid organs and the site of antigenic-challenge is a mandatory prerequisite for the precise functioning of adaptive immune responses. The surface molecule CD152 (CTLA-4) is mostly considered as a negative regulator of T cell activation during immune responses. It is currently unknown whether CD152 can also influence chemokine-driven T cell migration. METHODOLOGY/PRINCIPAL FINDINGS We analyzed the consequences of CD152 signaling on Th cell migration using chemotaxis assays in vitro and radioactive cell tracking in vivo. We show here that the genetic and serological inactivation of CD152 in Th1 cells reduced migration towards CCL4, CXCL12 and CCL19, but not CXCL9, in a G-protein dependent manner. In addition, retroviral transduction of CD152 cDNA into CD152 negative cells restored Th1 cell migration. Crosslinking of CD152 together with CD3 and CD28 stimulation on activated Th1 cells increased expression of the chemokine receptors CCR5 and CCR7, which in turn enhanced cell migration. Using sensitive liposome technology, we show that mature dendritic cells but not activated B cells were potent at inducing surface CD152 expression and the CD152-mediated migration-enhancing signals. Importantly, migration of CD152 positive Th1 lymphocytes in in vivo experiments increased more than 200% as compared to CD152 negative counterparts showing that indeed CD152 orchestrates specific migration of selected Th1 cells to sites of inflammation and antigenic challenge in vivo. CONCLUSIONS/SIGNIFICANCE We show here, that CD152 signaling does not just silence cells, but selects individual ones for migration. This novel activity of CD152 adds to the already significant role of CD152 in controlling peripheral immune responses by allowing T cells to localize correctly during infection. It also suggests that interference with CD152 signaling provides a tool for altering the cellular composition at sites of inflammation and antigenic challenge.
Collapse
Affiliation(s)
- Karin Knieke
- Experimentelle Pädiatrie, Universitätskinderklinik – Otto-von-Guericke Universität, Magdeburg, Germany
- Deutsches Rheuma-Forschungszentrum Berlin and Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinischer Immunologie, CCM, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Holger Hoff
- Deutsches Rheuma-Forschungszentrum Berlin and Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinischer Immunologie, CCM, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Maszyna
- Deutsches Rheuma-Forschungszentrum Berlin and Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinischer Immunologie, CCM, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Paula Kolar
- Deutsches Rheuma-Forschungszentrum Berlin and Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinischer Immunologie, CCM, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Arnhild Schrage
- Deutsches Rheuma-Forschungszentrum Berlin and Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinischer Immunologie, CCM, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Alf Hamann
- Deutsches Rheuma-Forschungszentrum Berlin and Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinischer Immunologie, CCM, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Gudrun F. Debes
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Monika C. Brunner-Weinzierl
- Experimentelle Pädiatrie, Universitätskinderklinik – Otto-von-Guericke Universität, Magdeburg, Germany
- Deutsches Rheuma-Forschungszentrum Berlin and Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinischer Immunologie, CCM, Charité -Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
83
|
Bianchi T, Pincus LB, Wurbel MA, Rich BE, Kupper TS, Fuhlbrigge RC, Boes M. Maintenance of peripheral tolerance through controlled tissue homing of antigen-specific T cells in K14-mOVA mice. THE JOURNAL OF IMMUNOLOGY 2009; 182:4665-74. [PMID: 19342642 DOI: 10.4049/jimmunol.0803628] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunological tolerance is crucial to avoid autoimmune and inflammatory diseases; however, the mechanisms involved are incompletely understood. To study peripheral tolerance to skin-associated Ags, we generated new transgenic mice expressing a membrane-bound form of OVA in skin under the human keratin 14 (K14) promoter (K14-mOVA mice). In contrast to other transgenic mice expressing similar self-Ags in skin, adoptive transfer of Ag-specific T cells does not induce inflammatory skin disease in our K14-mOVA mice. OVA-specific T cells transferred into K14-mOVA mice are activated in lymphoid tissues, undergo clonal expansion, and eventually acquire effector function. Importantly, these Ag-specific T cells selectively up-regulate expression of E-selectin ligand in cutaneous lymph nodes but not in mesenteric lymph nodes and spleen, demonstrating that expression of endogenous self-Ags in skin dictates imprinting of skin tissue homing in vivo. However, an additional inflammatory signal, here induced by tape stripping, is required in K14-mOVA mice to induce T cell migration to skin and development of inflammatory skin disease. Depletion of regulatory CD4(+)CD25(+) T cells did not provoke homing of transferred T cells to skin under steady-state conditions, indicating that these cells are not the key regulators for inhibiting T cell homing in K14-mOVA mice. Both skin-derived and lymph node-resident CD8alpha(+) dendritic cells are responsible for Ag presentation in vivo and induce tolerance to skin Ags, as we show by selective depletion of langerin(+) and CD11c(+) dendritic cells. Taken together, controlled skin homing of T cells is critical for the maintenance of peripheral immune tolerance to epidermal self-Ags.
Collapse
Affiliation(s)
- Teresa Bianchi
- Department of Dermatology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
84
|
Woodland DL, Kohlmeier JE. Migration, maintenance and recall of memory T cells in peripheral tissues. Nat Rev Immunol 2009; 9:153-61. [PMID: 19240755 DOI: 10.1038/nri2496] [Citation(s) in RCA: 303] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
After the resolution of an immune response, antigen-specific memory T cells persist at many sites in the body. The antigen-specific memory T-cell pool includes memory T cells that preferentially reside in peripheral tissues, such as the skin, gut and lungs, where they provide a first line of defence against secondary pathogen infection. Determining how peripheral memory T cells are regulated is essential for our understanding of host-pathogen interactions and for vaccine development. In this Review, we discuss recent insights into the generation, control and recall of peripheral T-cell memory responses.
Collapse
|
85
|
Abstract
Vitamins are essential constituents of our diet that have long been known to influence the immune system. Vitamins A and D have received particular attention in recent years as these vitamins have been shown to have an unexpected and crucial effect on the immune response. We present and discuss our current understanding of the essential roles of vitamins in modulating a broad range of immune processes, such as lymphocyte activation and proliferation, T-helper-cell differentiation, tissue-specific lymphocyte homing, the production of specific antibody isotypes and regulation of the immune response. Finally, we discuss the clinical potential of vitamin A and D metabolites for modulating tissue-specific immune responses and for preventing and/or treating inflammation and autoimmunity.
Collapse
|
86
|
Yokota A, Takeuchi H, Maeda N, Ohoka Y, Kato C, Song SY, Iwata M. GM-CSF and IL-4 synergistically trigger dendritic cells to acquire retinoic acid-producing capacity. Int Immunol 2009; 21:361-77. [PMID: 19190084 PMCID: PMC2660862 DOI: 10.1093/intimm/dxp003] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Retinoic acid (RA) produced by intestinal dendritic cells (DCs) imprints gut-homing specificity on lymphocytes and enhances Foxp3+ regulatory T-cell differentiation. The expression of aldehyde dehydrogenase (ALDH) 1A in these DCs is essential for the RA production. However, it remains unclear how the steady-state ALDH1A expression is induced under specific pathogen-free (SPF) conditions. Here, we found that bone marrow-derived dendritic cells (BM-DCs) generated with granulocyte-macrophage colony-stimulating factor (GM-CSF) expressed Aldh1a2, an isoform of Aldh1a, but that fms-related tyrosine kinase 3 ligand-generated BM-DCs did not. DCs from mesenteric lymph nodes (MLN) and Peyer's patches (PP) of normal SPF mice expressed ALDH1A2, but not the other known RA-producing enzymes. Employing a flow cytometric method, we detected ALDH activities in 10–30% of PP-DCs and MLN-DCs. They were CD11chighCD4−/lowCD8αintermediateCD11b−/low F4/80low/intermediateCD45RBlowCD86highMHC class IIhighB220−CD103+. Equivalent levels of aldehyde dehydrogenase activity (ALDHact) and ALDH1A2 expression were induced synergistically by GM-CSF and IL-4 in splenic DCs in vitro. In BM-DCs, however, additional signals via Toll-like receptors or RA receptors were required for inducing the equivalent levels. The generated ALDH1A2+ DCs triggered T cells to express gut-homing receptors or Foxp3. GM-CSF receptor-deficient or vitamin A-deficient mice exhibited marked reductions in the ALDHact in intestinal DCs and the T cell number in the intestinal lamina propria, whereas IL-4 receptor-mediated signals were dispensable. GM-CSF+CD11c−F4/80+ cells existed constitutively in the intestinal tissues. The results suggest that GM-CSF and RA itself are pivotal among multiple microenvironment factors that enable intestinal DCs to produce RA.
Collapse
Affiliation(s)
- Aya Yokota
- Laboratory of Biodefense Research, Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University, Sanuki-shi, Kagawa, Japan
| | | | | | | | | | | | | |
Collapse
|
87
|
Masopust D. Developing an HIV cytotoxic T-lymphocyte vaccine: issues of CD8 T-cell quantity, quality and location. J Intern Med 2009; 265:125-37. [PMID: 19093965 DOI: 10.1111/j.1365-2796.2008.02054.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Issues of quantity, quality and location impact the ability of CD8 T cells to mediate protection from infection. These issues are considered in light of human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) vaccination. Methods are reviewed that result in 100- to 1000-fold higher frequencies of vaccine-specific memory CD8 T cells than that achieved by current HIV/SIV vaccine approaches. Data demonstrating that location within mucosal tissues has a direct impact on memory CD8 T-cell function are discussed. Arguments are made that establishing memory CD8 T cells within mucosal sites of transmission, a priori to natural infection, may be essential for conferring optimal and rapid protection. Lastly, it is proposed that heterologous prime-boost vaccination with recombinant live replicating vectors, which has the potential to induce tremendous numbers of cytolytic memory CD8 T cells within mucosal tissues, would provide a far more stringent test of the hypothesis that memory CD8 T cells could, in principal, form the basis for a preventative HIV vaccine.
Collapse
Affiliation(s)
- D Masopust
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
88
|
Edele F, Molenaar R, Gütle D, Dudda JC, Jakob T, Homey B, Mebius R, Hornef M, Martin SF. Cutting edge: instructive role of peripheral tissue cells in the imprinting of T cell homing receptor patterns. THE JOURNAL OF IMMUNOLOGY 2008; 181:3745-9. [PMID: 18768825 DOI: 10.4049/jimmunol.181.6.3745] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tissue-specific homing of effector and memory T cells to skin and small intestine requires the imprinting of specific combinations of adhesion molecules and chemokine receptors by dendritic cells in the draining lymph nodes. In this study, we demonstrate that CD8(+) T cells activated by Ag-pulsed bone marrow-derived dendritic cells were induced to express the small intestine homing receptors alpha(4)beta(7) integrin and chemokine receptor CCR9 in coculture with small intestinal epithelial cells. In contrast, in coculture with dermal fibroblasts the skin-homing receptor E-selectin ligand was induced. Interestingly, the imprinting of gut homing receptors on anti-CD3/anti-CD28 stimulated T cells was induced by soluble factors produced by small intestinal epithelial cells. Retinoic acid was identified as a crucial factor. These findings show that peripheral tissue cells directly produce homing receptor imprinting factors and suggest that dendritic cells can acquire their imprinting potential already in the peripheral tissue of origin.
Collapse
Affiliation(s)
- Fanny Edele
- Allergy Research Group, Department of Dermatology, University Medical Center, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Sigmundsdottir H, Butcher EC. Environmental cues, dendritic cells and the programming of tissue-selective lymphocyte trafficking. Nat Immunol 2008; 9:981-7. [PMID: 18711435 DOI: 10.1038/ni.f.208] [Citation(s) in RCA: 249] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Lymphocytes are imprinted during activation with trafficking programs (combinations of adhesion and chemoattractant receptors) that target their migration to specific tissues and microenvironments. Cytokines contribute, but, for gut and skin, evolution has cleverly adapted external cues from food (vitamin A) and sunlight (ultraviolet-induced vitamin D3) to imprint lymphocyte homing to the small intestines and T cell migration into the epidermis. Dendritic cells are essential: they process the vitamins to their active metabolites (retinoic acid and 1,25(OH)(2)D3) for presentation with antigen to lymphocytes, and they help export environmental cues through lymphatics to draining lymph nodes, to program the trafficking and effector functions of naive T and B cells.
Collapse
Affiliation(s)
- Hekla Sigmundsdottir
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | |
Collapse
|
90
|
Hammerschmidt SI, Ahrendt M, Bode U, Wahl B, Kremmer E, Förster R, Pabst O. Stromal mesenteric lymph node cells are essential for the generation of gut-homing T cells in vivo. ACTA ACUST UNITED AC 2008; 205:2483-90. [PMID: 18852290 PMCID: PMC2571923 DOI: 10.1084/jem.20080039] [Citation(s) in RCA: 252] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
T cells primed in the gut-draining mesenteric lymph nodes (mLN) are imprinted to express alpha4beta7-integrin and chemokine receptor CCR9, thereby enabling lymphocytes to migrate to the small intestine. In vitro activation by intestinal dendritic cells (DC) or addition of retinoic acid (RA) is sufficient to instruct expression of these gut-homing molecules. We report that in vivo stroma cells, but not DC, allow the mLN to induce the generation of gut tropism. Peripheral LN (pLN) transplanted into the gut mesenteries fail to support the generation of gut-homing T cells, even though gut-derived DC enter the transplants and prime T cells. DC that fail to induce alpha4beta7-integrin and CCR9 in vitro readily induce these factors in vivo upon injection into mLN afferent lymphatics. Moreover, uniquely mesenteric but not pLN stroma cells express high levels of RA-producing enzymes and support induction of CCR9 on activated T cells in vitro. These results demonstrate a hitherto unrecognized contribution of stromal cell delivered signals, including RA, on the imprinting of tissue tropism in vivo.
Collapse
|
91
|
Iwata M. Retinoic acid production by intestinal dendritic cells and its role in T-cell trafficking. Semin Immunol 2008; 21:8-13. [PMID: 18849172 DOI: 10.1016/j.smim.2008.09.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 08/29/2008] [Accepted: 09/01/2008] [Indexed: 12/13/2022]
Abstract
Vitamin A deficiency causes a marked reduction in the number of T and B cells in the small intestinal tissues. The vitamin A metabolite retinoic acid imprints lymphocytes with gut-homing specificity upon antigenic stimulation. In the small intestinal lamina propria, Peyer's patches, and mesenteric lymph nodes, there are dendritic cells capable of producing retinoic acid. Their capacity depends on the expression of retinal dehydrogenases (RALDH). RALDH2, encoded by Aldh1a2, is a major isoform of RALDH in the intestinal dendritic cells under specific pathogen-free conditions, and can be induced by multiple factors constitutively present or induced in the small intestinal microenvironment.
Collapse
Affiliation(s)
- Makoto Iwata
- Laboratory of Biodefense Research, Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University, 1314-1 Shido, Sanuki-shi, Kagawa 769-2193, Japan.
| |
Collapse
|
92
|
Lass C, Vocanson M, Wagner S, Schempp CM, Nicolas JF, Merfort I, Martin SF. Anti-inflammatory and immune-regulatory mechanisms prevent contact hypersensitivity toArnica montanaL. Exp Dermatol 2008; 17:849-57. [DOI: 10.1111/j.1600-0625.2008.00717.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
93
|
Schepers K, Swart E, van Heijst JWJ, Gerlach C, Castrucci M, Sie D, Heimerikx M, Velds A, Kerkhoven RM, Arens R, Schumacher TNM. Dissecting T cell lineage relationships by cellular barcoding. ACTA ACUST UNITED AC 2008; 205:2309-18. [PMID: 18809713 PMCID: PMC2556794 DOI: 10.1084/jem.20072462] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
T cells, as well as other cell types, are composed of phenotypically and functionally distinct subsets. However, for many of these populations it is unclear whether they develop from common or separate progenitors. To address such issues, we developed a novel approach, termed cellular barcoding, that allows the dissection of lineage relationships. We demonstrate that the labeling of cells with unique identifiers coupled to a microarray-based detection system can be used to analyze family relationships between the progeny of such cells. To exemplify the potential of this technique, we studied migration patterns of families of antigen-specific CD8+ T cells in vivo. We demonstrate that progeny of individual T cells rapidly seed independent lymph nodes and that antigen-specific CD8+ T cells present at different effector sites are largely derived from a common pool of precursors. These data show how locally primed T cells disperse and provide a technology for kinship analysis with wider utility.
Collapse
Affiliation(s)
- Koen Schepers
- Division of Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Mora JR, von Andrian UH. Role of retinoic acid in the imprinting of gut-homing IgA-secreting cells. Semin Immunol 2008; 21:28-35. [PMID: 18804386 DOI: 10.1016/j.smim.2008.08.002] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 08/07/2008] [Accepted: 08/09/2008] [Indexed: 12/20/2022]
Abstract
Antibody-secreting cells (ASCs) lodging in the mucosa of the small intestine are derived from activated B cells that are thought to arise in gut-associated lymphoid tissues (GALT). Upon leaving the GALT, B cells return to the blood where they must express the gut-homing receptors alpha4beta7 and CCR9 in order to emigrate into the small bowel. Recent evidence indicates that gut-associated dendritic cells (DCs) in GALT induce gut-homing receptors on B cells via a mechanism that depends on the vitamin A metabolite retinoic acid (RA). In addition, although ASC associated with other mucosal tissues secrete IgA in an RA-independent fashion, the presence of high levels of RA in intestine and GALT can promote B cell class switching to IgA and thus, boost the production of IgA in the intestinal mucosa. Here, we discuss the role of RA in the imprinting of gut-homing ASC and the evidence linking RA with the generation of intestinal IgA-ASCs.
Collapse
Affiliation(s)
- J Rodrigo Mora
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRJ-815, Boston, MA 02114, USA.
| | | |
Collapse
|
95
|
Martin SF, Dudda JC, Bachtanian E, Lembo A, Liller S, Dürr C, Heimesaat MM, Bereswill S, Fejer G, Vassileva R, Jakob T, Freudenberg N, Termeer CC, Johner C, Galanos C, Freudenberg MA. Toll-like receptor and IL-12 signaling control susceptibility to contact hypersensitivity. ACTA ACUST UNITED AC 2008; 205:2151-62. [PMID: 18725520 PMCID: PMC2526208 DOI: 10.1084/jem.20070509] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Allergic contact hypersensitivity (CHS) is a T cell–mediated inflammatory skin disease. Interleukin (IL)-12 is considered to be important in the generation of the allergen-specific T cell response. Loss of IL-12 function in IL-12Rβ2–deficient mice, however, did not ameliorate the allergic immune response, suggesting alternate IL-12–independent pathways in the induction of CHS. Because exposure to contact allergens always takes place in the presence of microbial skin flora, we investigated the potential role of Toll-like receptors (TLRs) in the induction of CHS. Using mice deficient in TLR4, the receptor for bacterial lipopolysaccharide (LPS), IL-12 receptor (R) β2, or both, we show that the concomitant absence of TLR4 and IL-12Rβ2, but not the absence of TLR4 or IL-12Rβ2 alone, prevented DC-mediated sensitization, generation of effector T cells, and the subsequent CHS response to 2,4,6-trinitro-1-chlorobenzene (TNCB), oxazolone, and fluorescein isothiocyanate. Introduction of the TLR4 transgene into the TLR4/IL-12Rβ2 mutant restored the CHS inducibility, showing a requirement for TLR4 in IL-12–independent CHS induction. Furthermore, the concomitant absence of TLR2 and TLR4 prevented the induction of CHS to TNCB in IL-12–competent mice. Finally, CHS was inducible in germ-free wild-type and IL-12Rβ2–deficient mice, but not in germ-free TLR4/IL-12Rβ2 double deficient mice, suggesting that the necessary TLR activation may proceed via endogenous ligands.
Collapse
Affiliation(s)
- Stefan F Martin
- Allergy Research Group, Department of Dermatology, University Medical Center Freiburg, D-79104 Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Brinkman CC, Sheasley-O'Neill SL, Ferguson AR, Engelhard VH. Activated CD8 T cells redistribute to antigen-free lymph nodes and exhibit effector and memory characteristics. THE JOURNAL OF IMMUNOLOGY 2008; 181:1814-24. [PMID: 18641319 DOI: 10.4049/jimmunol.181.3.1814] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Exogenous dendritic cells display restricted trafficking when injected in vivo and stimulate CD8 T cell responses that are localized to a small number of lymphoid compartments. By examining these responses in the presence and absence of FTY720, a drug that causes sequestration of T cells in lymph nodes, we demonstrate that a significant fraction of divided CD8 T cells redistribute into Ag-free lymph nodes within 3 days of activation. Despite variation in the level of expression of CD62L, redistribution of these cells is CD62L-dependent. Redistributed CD8 T cells exhibit characteristics of differentiated effectors. However, when re-isolated from Ag-free lymph nodes 3 days after activation and transferred into naive mice, they persist for at least 3 wk and expand upon Ag challenge. Thus, CD8 T cells that redistribute to Ag-free lymph nodes 3 days after immunization contain memory precursors. We suggest that this redistribution process represents an important mechanism for establishment of lymph node resident central memory, and that redistribution to Ag-free nodes is an additional characteristic to be added to those that distinguish memory precursors from terminal effectors.
Collapse
Affiliation(s)
- C Colin Brinkman
- Department of Microbiology and Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
97
|
Jaensson E, Uronen-Hansson H, Pabst O, Eksteen B, Tian J, Coombes JL, Berg PL, Davidsson T, Powrie F, Johansson-Lindbom B, Agace WW. Small intestinal CD103+ dendritic cells display unique functional properties that are conserved between mice and humans. ACTA ACUST UNITED AC 2008; 205:2139-49. [PMID: 18710932 PMCID: PMC2526207 DOI: 10.1084/jem.20080414] [Citation(s) in RCA: 458] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A functionally distinct subset of CD103(+) dendritic cells (DCs) has recently been identified in murine mesenteric lymph nodes (MLN) that induces enhanced FoxP3(+) T cell differentiation, retinoic acid receptor signaling, and gut-homing receptor (CCR9 and alpha4beta7) expression in responding T cells. We show that this function is specific to small intestinal lamina propria (SI-LP) and MLN CD103(+) DCs. CD103(+) SI-LP DCs appeared to derive from circulating DC precursors that continually seed the SI-LP. BrdU pulse-chase experiments suggested that most CD103(+) DCs do not derive from a CD103(-) SI-LP DC intermediate. The majority of CD103(+) MLN DCs appear to represent a tissue-derived migratory population that plays a central role in presenting orally derived soluble antigen to CD8(+) and CD4(+) T cells. In contrast, most CD103(-) MLN DCs appear to derive from blood precursors, and these cells could proliferate within the MLN and present systemic soluble antigen. Critically, CD103(+) DCs with similar phenotype and functional properties were present in human MLN, and their selective ability to induce CCR9 was maintained by CD103(+) MLN DCs isolated from SB Crohn's patients. Thus, small intestinal CD103(+) DCs represent a potential novel target for regulating human intestinal inflammatory responses.
Collapse
Affiliation(s)
- Elin Jaensson
- Immunology Section, BMC D14, 221 84 Lund University, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Thompson JM, Nicholson MG, Whitmore AC, Zamora M, West A, Iwasaki A, Staats HF, Johnston RE. Nonmucosal alphavirus vaccination stimulates a mucosal inductive environment in the peripheral draining lymph node. THE JOURNAL OF IMMUNOLOGY 2008; 181:574-85. [PMID: 18566424 DOI: 10.4049/jimmunol.181.1.574] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The strongest mucosal immune responses are induced following mucosal Ag delivery and processing in the mucosal lymphoid tissues, and much is known regarding the immunological parameters which regulate immune induction via this pathway. Recently, experimental systems have been identified in which mucosal immune responses are induced following nonmucosal Ag delivery. One such system, footpad delivery of Venezuelan equine encephalitis virus replicon particles (VRP), led to the local production of IgA Abs directed against both expressed and codelivered Ags at multiple mucosal surfaces in mice. In contrast to the mucosal delivery pathway, little is known regarding the lymphoid structures and immunological components that are responsible for mucosal immune induction following nonmucosal delivery. In this study, we have used footpad delivery of VRP to probe the constituents of this alternative pathway for mucosal immune induction. Following nonmucosal VRP delivery, J chain-containing, polymeric IgA Abs were detected in the peripheral draining lymph node (DLN), at a time before IgA detection at mucosal surfaces. Further analysis of the VRP DLN revealed up-regulated alpha4beta7 integrin expression on DLN B cells, expression of mucosal addressin cell adhesion molecule 1 on the DLN high endothelia venules, and production of IL-6 and CC chemokines, all characteristics of mucosal lymphoid tissues. Taken together, these results implicate the peripheral DLN as an integral component of an alternative pathway for mucosal immune induction. A further understanding of the critical immunological and viral components of this pathway may significantly improve both our knowledge of viral-induced immunity and the efficacy of viral-based vaccines.
Collapse
Affiliation(s)
- Joseph M Thompson
- Department of Microbiology and Immunology, Carolina Vaccine Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Lembo A, Pelletier M, Iyer R, Timko M, Dudda JC, West TE, Wilson CB, Hajjar AM, Skerrett SJ. Administration of a synthetic TLR4 agonist protects mice from pneumonic tularemia. THE JOURNAL OF IMMUNOLOGY 2008; 180:7574-81. [PMID: 18490759 DOI: 10.4049/jimmunol.180.11.7574] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Francisella tularensis is a Gram-negative intracellular pathogen that causes the zoonosis tularemia. Because F. tularensis LPS causes weak TLR4 activation, we hypothesized that administration of a synthetic TLR4 agonist, aminoalkyl glucosaminide phosphate (AGP), would boost the innate immune system and compensate for reduced TLR4 stimulation. Intranasal administration of AGPs induced intrapulmonary production of proinflammatory cytokines and chemokines. Mice treated with AGPs before and after inhalation of Francisella novicida exhibited augmented cytokine and inflammatory responses to infection; reduced bacterial replication in lung, liver, and spleen; and increased survival, whereas all PBS-treated control mice died within 4 days of infection, all AGP-treated mice showed prolonged time-to-death, and 30-60% of AGP-treated mice survived. The protective effect of AGP was lost in mice lacking IFN-gamma. Long-term survivors developed specific Th1 splenocyte responses and specific Abs dominated by IgG2 isotypes. Survivors were fully protected from rechallenge with aerosolized F. novicida. Thus, preventive administration of AGP successfully modulated innate immune responses to aerosolized F. novicida, leading to protective immunity to pneumonic tularemia. This is the first report of the protective effect of a TLR ligand on resistance to F. novicida-induced pneumonic tularemia.
Collapse
Affiliation(s)
- Annalisa Lembo
- Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Dörrie J, Schaft N, Müller I, Wellner V, Schunder T, Hänig J, Oostingh GJ, Schön MP, Robert C, Kämpgen E, Schuler G. Introduction of functional chimeric E/L-selectin by RNA electroporation to target dendritic cells from blood to lymph nodes. Cancer Immunol Immunother 2008; 57:467-77. [PMID: 17768622 PMCID: PMC11041385 DOI: 10.1007/s00262-007-0385-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 07/30/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Inefficient migration of dendritic cells (DC) to regional lymph nodes (LN) upon intracutaneous injection is a major obstacle for effective DC vaccination. Intravenous vaccination is unfavorable, because DC cannot migrate directly from the blood into LN. METHODS To enable human monocyte-derived (mo)DC to enter LN directly from the blood, we manipulated them by RNA electroporation to express a human chimeric E/L-selectin (CD62E/CD62L) protein, which binds to peripheral node addressin expressed on high endothelial venules. RESULTS Transfection efficiency exceeded 95%, and high E/L-selectin surface expression was detected for >48 h. E/L-selectin RNA-transfected DC displayed an identical mature DC phenotype as mock-transfected DC. Furthermore, E/L-selectin-transfected DC maintained their normal CCR7-mediated migration capacity, and their ability to prime and expand functional cytotoxic T cells recognizing MelanA. Most importantly, E/L-selectin-RNA-transfected DC gained the capability to attach to and roll on sialyl-Lewis(X) in vitro. OUTLOOK The presented strategy can be readily translated into the clinic, as it involves no stable genetic manipulation or viral transformation, and allows targeting of a large number of LN.
Collapse
Affiliation(s)
- Jan Dörrie
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Ina Müller
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Verena Wellner
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Tanja Schunder
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Jens Hänig
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Gertie J. Oostingh
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine
and Department of Dermatology, University of Würzburg, Würzburg, Germany
| | - Michael P. Schön
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine
and Department of Dermatology, University of Würzburg, Würzburg, Germany
| | | | - Eckhart Kämpgen
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Gerold Schuler
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| |
Collapse
|