1
|
Wang S, Chen Y, Ling Z, Li J, Hu J, He F, Chen Q. The role of dendritic cells in the immunomodulation to implanted biomaterials. Int J Oral Sci 2022; 14:52. [PMCID: PMC9636170 DOI: 10.1038/s41368-022-00203-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Considering the substantial role played by dendritic cells (DCs) in the immune system to bridge innate and adaptive immunity, studies on DC-mediated immunity toward biomaterials principally center on their adjuvant effects in facilitating the adaptive immunity of codelivered antigens. However, the effect of the intrinsic properties of biomaterials on dendritic cells has not been clarified. Recently, researchers have begun to investigate and found that biomaterials that are nonadjuvant could also regulate the immune function of DCs and thus affect subsequent tissue regeneration. In the case of proteins adsorbed onto biomaterial surfaces, their intrinsic properties can direct their orientation and conformation, forming “biomaterial-associated molecular patterns (BAMPs)”. Thus, in this review, we focused on the intrinsic physiochemical properties of biomaterials in the absence of antigens that affect DC immune function and summarized the underlying signaling pathways. Moreover, we preliminarily clarified the specific composition of BAMPs and the interplay between some key molecules and DCs, such as heat shock proteins (HSPs) and high mobility group box 1 (HMGB1). This review provides a new direction for future biomaterial design, through which modulation of host immune responses is applicable to tissue engineering and immunotherapy.
Collapse
Affiliation(s)
- Siyuan Wang
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Yanqi Chen
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Zhaoting Ling
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Jia Li
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Jun Hu
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Fuming He
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Qianming Chen
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| |
Collapse
|
2
|
Keratinocytes Share Gene Expression Fingerprint with Epidermal Langerhans Cells via mRNA Transfer. J Invest Dermatol 2019; 139:2313-2323.e8. [DOI: 10.1016/j.jid.2019.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 01/13/2023]
|
3
|
Thomson AW, Metes DM, Ezzelarab MB, Raïch-Regué D. Regulatory dendritic cells for human organ transplantation. Transplant Rev (Orlando) 2019; 33:130-136. [PMID: 31130302 DOI: 10.1016/j.trre.2019.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/09/2019] [Indexed: 12/30/2022]
Abstract
Current immunosuppressive (IS) regimens used to prevent organ allograft rejection have well-recognized side effects, that include enhanced risk of infection and certain types of cancer, metabolic disorders, cardiovascular disease, renal complications and failure to control chronic allograft rejection. The life-long dependency of patients on these IS agents reflects their inability to induce donor-specific tolerance. Extensive studies in rodent and non-human primate models have demonstrated the ability of adoptively-transferred regulatory immune cells (either regulatory myeloid cells or regulatory T cells) to promote transplant tolerance. Consequently, there is considerable interest in the potential of regulatory immune cell therapy to allow safe minimization/complete withdrawal of immunosuppression and the promotion of organ transplant tolerance in the clinic. Here, we review the properties of regulatory dendritic cells (DCreg) with a focus on the approaches being taken to generate human DCreg for clinical testing. We also document the early phase clinical trials that are underway to assess DCreg therapy in clinical organ transplantation as well as in autoimmune disorders.
Collapse
Affiliation(s)
- Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Diana M Metes
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mohamed B Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dalia Raïch-Regué
- Nephropathies Research Group, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
4
|
Leifer CA. Dendritic cells in host response to biologic scaffolds. Semin Immunol 2017; 29:41-48. [PMID: 28214177 DOI: 10.1016/j.smim.2017.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 01/23/2017] [Accepted: 01/31/2017] [Indexed: 02/03/2023]
Abstract
Tissue regeneration and repair require a highly complex and orchestrated series of events that require inflammation, but can be compromised when inflammation is excessive or becomes chronic. Macrophages are one of the first cells to contact and respond to implanted materials, and mediate the inflammatory response. The series of events following macrophage association with biomaterials has been well-studied. Dendritic cells (DCs) also directly interact with biomaterials, are critical for specific immune responses, and can be activated in response to interactions with biomaterials. Yet, much less is known about the responses by DCs. This review discusses what we know about DC response to biomaterials, the underlying mechanisms involved, and how DCs can be influenced by the macrophage response to biomaterials. Lastly, I will discuss how biomaterials can be manipulated to enhance or suppress DC function to promote a specific desirable immune response - a major goal for implantable biologically active therapeutics.
Collapse
Affiliation(s)
- Cynthia A Leifer
- Department of Microbiology and Immunology College of Veterinary Medicine, C5-153 Cornell University, Ithaca, NY, USA.
| |
Collapse
|
5
|
Prolongation of kidney allograft survival regulated by indoleamine 2, 3-dioxygenase in immature dendritic cells generated from recipient type bone marrow progenitors. Mol Immunol 2016; 79:22-31. [PMID: 27689750 DOI: 10.1016/j.molimm.2016.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 12/13/2022]
Abstract
Immature dendritic cells (iDCs) are bone marrow-derived professional antigen-presenting cells, exhibit very low levels of the co-stimulatory molecules CD80 (B7-1), CD86 (B7-2), and CD40 and major histocompatibility complex (MHC) class II and play a critical role in triggering antigen-specific immunotolerance. The enzyme indoleamine 2, 3-dioxygenase (IDO) is a cytosolic tryptophan catabolism rate-limiting step enzyme. IDO secreted by DCs shows an association with the suppression of T-cell responses and promotion of tolerance. In this study, BN rat recipients were pre-injected with donor renal alloantigen-treated recipient iDCs before kidney transplantation. The renal allograft exhibited a lighter renal rejection response, prolonged graft survival time, and an increasing content of CD4+CD25+Foxp3+ regulatory T cells (Tregs). Additionally, up-regulated secretion of Th2 cytokines were found in recipient sera post-transplantation. Transfection of si-IDO1 RNA into renal-antigen-treated recipient iDCs reversed these changes, which suggested that IDO channel signaling may be involved in iDC-induced allograft immunotolerance. These results suggested that iDC-induced and IDO-mediated allograft immunotolerance might be a potentially feasible tactic to prolong allograft survival, in addition to immunosuppressive drugs.
Collapse
|
6
|
Nizza ST, Campbell JJ. CD11b+ migratory dendritic cells mediate CD8 T cell cross-priming and cutaneous imprinting after topical immunization. PLoS One 2014; 9:e91054. [PMID: 24618819 PMCID: PMC3949685 DOI: 10.1371/journal.pone.0091054] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/07/2014] [Indexed: 11/23/2022] Open
Abstract
Topical antigen application is a focus of current vaccine research. This immunization route mimics natural antigen exposure across a barrier tissue and generates T cells imprinted for skin-selective homing. Soluble antigens introduced through this route require cross-presentation by DC to generate CD8 T cell responses. Here we have explored the relative contribution of various skin-derived DC subsets to cross-priming and skin-selective imprinting. In our model, DC acquire soluble Ag in vivo from immunized murine skin for cross-presentation to naïve CD8 T cells ex vivo. We find CD11b+ migratory DC to be the relevant cross-priming DC in this model. Both Langerin+ and Langerin- CD11b+ migratory DC can cross-present antigen in our system, but only the Langerin+ subset can induce expression of the skin-selective addressin E-selectin ligand. Thus, the CD11b+ Langerin+ migratory DC population, comprised primarily of Langerhans cells, both cross-primes naïve CD8 T cells and imprints them with skin-homing capabilities.
Collapse
Affiliation(s)
- Suzanne T. Nizza
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, United States of America
| | - James J. Campbell
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Department of Dermatology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
7
|
Seneschal J, Clark RA, Gehad A, Baecher-Allan CM, Kupper TS. Human epidermal Langerhans cells maintain immune homeostasis in skin by activating skin resident regulatory T cells. Immunity 2012; 36:873-84. [PMID: 22560445 PMCID: PMC3716276 DOI: 10.1016/j.immuni.2012.03.018] [Citation(s) in RCA: 328] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 09/20/2011] [Accepted: 03/09/2012] [Indexed: 12/18/2022]
Abstract
Recent studies have demonstrated that the skin of a normal adult human contains 10-20 billion resident memory T cells, including various helper, cytotoxic, and regulatory T cell subsets, that are poised to respond to environmental antigens. Using only autologous human tissues, we report that both in vitro and in vivo, resting epidermal Langerhan cells (LCs) selectively and specifically induced the activation and proliferation of skin resident regulatory T (Treg) cells, a minor subset of skin resident memory T cells. In the presence of foreign pathogen, however, the same LCs activated and induced proliferation of effector memory T (Tem) cells and limited Treg cells' activation. These underappreciated properties of LCs, namely maintenance of tolerance in normal skin, and activation of protective skin resident memory T cells upon infectious challenge, help clarify the role of LCs in skin.
Collapse
Affiliation(s)
- Julien Seneschal
- Department of Dermatology/Harvard Skin Disease Research Center,
Brigham and Women’s Hospital, Boston, MA 02115
- Department of Dermatology and Pediatric Dermatology/ University of
Bordeaux, INSERM U1035 and CIRID UMR 5164, Bordeaux, France
| | - Rachael A. Clark
- Department of Dermatology/Harvard Skin Disease Research Center,
Brigham and Women’s Hospital, Boston, MA 02115
| | - Ahmed Gehad
- Department of Dermatology/Harvard Skin Disease Research Center,
Brigham and Women’s Hospital, Boston, MA 02115
| | - Clare M. Baecher-Allan
- Department of Dermatology/Harvard Skin Disease Research Center,
Brigham and Women’s Hospital, Boston, MA 02115
| | - Thomas S. Kupper
- Department of Dermatology/Harvard Skin Disease Research Center,
Brigham and Women’s Hospital, Boston, MA 02115
| |
Collapse
|
8
|
Gleisner MA, Rosemblatt M, Fierro JA, Bono MR. Delivery of alloantigens via apoptotic cells generates dendritic cells with an immature tolerogenic phenotype. Transplant Proc 2012; 43:2325-33. [PMID: 21839264 DOI: 10.1016/j.transproceed.2011.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND Dendritic cells (DCs) are professional antigen-presenting cells able to induce immunity or tolerance. The interactions of immature DCs with naive T lymphocytes induce peripheral tolerance through mechanisms that include anergy or deletion of lymphocytes or the generation of regulatory T cells. Because of the central role of DCs in the immune response, they are potential targets for the induction of experimental tolerance. Thus, the generation of immature (tolerogenic) DCs able to capture and present alloantigens to T cells represents an important aim in our efforts to achieve better transplant acceptance. METHODS In this work, we generated immature DCs by using vitamin D(3) (VD3) during the process of DC differentiation. RESULTS The VD3-DCs showed an immature phenotype characterized by a low expression of major histocompatibility complex antigens of class II, CD86, and CD80 molecules and the secretion of a tolerogenic cytokine pattern. Furthermore, we showed that VD3-DCs phagocytose apoptotic allogeneic cells efficiently without inducing DC maturation or activation. Most important, our experiments demonstrated that mice treated with VD3 produce immature DCs in vivo, and that DCs from VD3-treated mice immunized with allogeneic apoptotic cells maintained their tolerogenic phenotype. CONCLUSION Our results show that allogeneic apoptotic cells in combination with VD3 generate DCs with tolerogenic characteristics that could be used to induce tolerance towards alloantigens.
Collapse
Affiliation(s)
- M A Gleisner
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | | | | | | |
Collapse
|
9
|
del Rio ML, Cote-Sierra J, Rodriguez-Barbosa JI. Flt3L-mobilized dendritic cells bearing H2-Kbm1 apoptotic cells do not induce cross-tolerance to CD8+ T cells across a class I MHC mismatched barrier. Transpl Int 2011; 24:501-13. [PMID: 21276089 DOI: 10.1111/j.1432-2277.2011.01220.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tolerization of allogeneic CD8(+) T cells is still a pending issue in the field of transplantation research to achieve long-term survival. To test whether dendritic cells (DC) bearing allogeneic major histocompatibility complex (MHC) class I mismatched apoptotic cells could induce cross-tolerance to alloreactive CD8(+) T cells, the following experimental strategy was devised. Rag2/γ(c) KO B6 mice were treated with Fms-like tyrosine kinase 3 ligand (Flt3L)-transduced B16 melanoma cells to drive a rapid expansion and mobilization of DC in vivo. Of all DC populations expanded, splenic CD11c(+) CD103(+) CD8α(+) DC were selectively involved in the process of antigen clearance of X-ray irradiated apoptotic thymocytes in vivo. Considering that CD11c(+) CD103(+) CD8α(+) DC selectively take up apoptotic cells and that they are highly specialized in cross-presenting antigen to CD8(+) T cells, we investigated whether B6 mice adoptively transferred with Flt3L-derived DC loaded with donor-derived apoptotic thymocytes could induce tolerance to bm1 skin allografts. Our findings on host anti-donor alloresponse, as revealed by skin allograft survival and cytotoxic T lymphocyte assays, indicated that the administration of syngeneic DC presenting K(bm1) donor-derived allopeptides through the indirect pathway of antigen presentation was not sufficient to induce cross-tolerance to alloreactive CD8(+) T cells responding to bm1 alloantigens in a murine model of skin allograft transplantation across an MHC class I mismatched barrier.
Collapse
Affiliation(s)
- Maria-Luisa del Rio
- Immunobiology Section, Institute of Biomedicine, University of León, León, Spain
| | | | | |
Collapse
|
10
|
Dendritic cells expressing soluble CTLA4Ig prolong antigen-specific skin graft survival. Immunol Cell Biol 2010; 88:846-50. [PMID: 20404834 DOI: 10.1038/icb.2010.58] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) and CTLA4Ig are important in regulating T-cell responses and therefore represent potential therapeutic tools in transplantation. In this study, CTLA4Ig was expressed in a C57BL/6 murine DC line (JAWS II) by lentiviral transduction and these cells were used to examine T-cell immunomodulatory effects in vitro and in vivo. A lower stimulation index to C57BL/6 was observed with splenocytes from BALB/c mice primed with JAWS II-CTLA4Ig compared with control JAWS II-green fluorescent protein (JAWS II-GFP). Mice primed with JAWS II-CTLA4Ig cells had significantly prolonged antigen-specific C57BL/6 skin graft survival compared with either JAWS II-GFP-primed or naïve mice (median 13, 11 and 11 days, respectively, P=0.0001). Furthermore, JAWS II-CTLA4Ig-primed mice that had been previously transplanted with skin grafts were re-transplanted with skin grafts 6 months later without immune manipulation. These mice demonstrated specific prolongation of second-set rejection responses, indicating systemic immune modulation induced by genetically modified DC. The mechanism was not due to expression of indoleamine 2,3-dioxygenase or induction of circulating regulatory T cells as assessed by flow cytometry of the peripheral blood lymphocytes. This potent effect demonstrated with skin grafts and second-set responses highlights the potential use of this strategy for transplantation more generally.
Collapse
|
11
|
Anti-CD45RB monoclonal antibody induces immunologic toleration by suppressing dendritic cells. Transpl Immunol 2009; 21:136-9. [DOI: 10.1016/j.trim.2009.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Revised: 04/07/2009] [Accepted: 04/10/2009] [Indexed: 11/22/2022]
|
12
|
Mutyambizi K, Berger CL, Edelson RL. The balance between immunity and tolerance: the role of Langerhans cells. Cell Mol Life Sci 2009; 66:831-40. [PMID: 19002380 PMCID: PMC4075167 DOI: 10.1007/s00018-008-8470-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Langerhans cells are immature skin-homing dendritic cells that furnish the epidermis with an immune surveillance system, and translate information between the internal and external milieu. Dendritic cells, in particular Langerhans cells, are gaining prominence as one of the potential principal players orchestrating the decision between immunity and tolerance. Langerhans cells capture aberrant self-antigen and pathogen-derived antigen for display to the efferent immune response. Recent evidence suggests redundancy in the antigen-presenting function of Langerhans cells, with dermal dendritic subsets capable of fulfilling an analogous role. There is mounting evidence that Langerhans cells can cross-prime T cells to recognize antigens. Langerhans cells are proposed to stimulate T regulatory cells, and are implicated in the pathogenesis of cutaneous T cell lymphoma.The phenotype of Langerhans cells, which may be tolerogenic or immunogenic, appears to depend on their state of maturity, inciting immunogen and cytokine environment, offering the potential for manipulation in immunotherapy.
Collapse
Affiliation(s)
- K. Mutyambizi
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar Street, New Haven, Connecticut 06520 USA
| | - C. L. Berger
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar Street, New Haven, Connecticut 06520 USA
| | - R. L. Edelson
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar Street, New Haven, Connecticut 06520 USA
- Yale Cancer Center (YCC), School of Medicine, Yale University, New Haven, USA
| |
Collapse
|
13
|
Fischer R, Turnquist HR, Taner T, Thomson AW. Use of rapamycin in the induction of tolerogenic dendritic cells. Handb Exp Pharmacol 2009:215-32. [PMID: 19031028 DOI: 10.1007/978-3-540-71029-5_10] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rapamycin (RAPA), a macrocyclic triene antibiotic pro-drug, is a clinically-utilized 'tolerance-sparing' immunosuppressant that inhibits the activity of T, B, and NK cells. Furthermore, maturation-resistance and tolerogenic properties of dendritic cells (DC) can be supported and preserved by conditioning with RAPA. Propagation of murine bone marrow (BM)-derived myeloid DC (mDC) in clinically relevant concentrations of RAPA (RAPA-DC) generates phenotypically immature DC with low levels of MHC and significantly reduced co-stimulatory molecules (especially CD86), even when exposed to inflammatory stimuli. RAPA-DC are weak stimulators of T cells and induce hyporesponsiveness and apoptosis in allo-reactive T cells. An interesting observation has been that RAPA-DC retain the ability to stimulate and enrich the regulatory T cells (Treg). Presumably as a result of these properties, alloantigen (alloAg)-pulsed recipient-derived DC are effective in subverting anti-allograft immune responses in rodent transplant models, making them an attractive subject for further investigation of their tolerance-promoting potential.
Collapse
Affiliation(s)
- Ryan Fischer
- Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
14
|
Thomson AW, Robbins PD. Tolerogenic dendritic cells for autoimmune disease and transplantation. Ann Rheum Dis 2008; 67 Suppl 3:iii90-6. [PMID: 19022823 DOI: 10.1136/ard.2008.099176] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dendritic leucocytes are professional antigen-presenting cells with inherent tolerogenic properties and are regarded as critical regulators of innate and adaptive immunity. Modification of dendritic cells (DCs) in the laboratory can enhance and stabilise their tolerogenic properties. Numerous reports suggest that such immature, maturation-resistant or "alternatively activated" DCs can regulate autoreactive or alloreactive T-cell responses and promote or restore antigen-specific tolerance in experimental animal models. The first clinical testing of tolerogenic DCs in human autoimmune disease, including rheumatoid arthritis, is imminent. Herein the properties of tolerogenic DCs and prospects for their testing in chronic inflammatory disease and transplantation are reviewed.
Collapse
Affiliation(s)
- A W Thomson
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.
| | | |
Collapse
|
15
|
Fiorina P, Jurewicz M, Vergani A, Augello A, Paez J, Ricchiuti V, Tchipachvili V, Sayegh MH, Abdi R. Phenotypic and functional differences between wild-type and CCR2-/- dendritic cells: implications for islet transplantation. Transplantation 2008; 85:1030-8. [PMID: 18408585 DOI: 10.1097/tp.0b013e31816843a0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Trafficking of dendritic cells (DC), the primary regulators of alloimmune responses, is controlled by chemokines. Here, we provide evidence that lack of CCR2 could lead to the generation of functionally and phenotypically different DC, which in part could explain the benefits observed in transplanting islets in CCR2 recipients. METHODS AND RESULTS We show that, in contrast to the in vitro DC maturation model, in vivo DC maturation is accompanied by an increase in the expression of CCR2. Compared with wild-type (WT), DC generated in vitro from CCR2 mice, and DC extracted from CCR2 naïve mice or from CCR2 recipients of islet allografts, display lesser allostimulatory capacity. Compared with WT DC, CCR2 DC produce more IL-4 and induce more IL-4-producing T cells. CCR2 DC also promote the generation of regulatory T cells that more efficiently suppress T cell proliferative responses by mixed leukocyte reaction. Similarly, the percentage of CD4CD25FoxP3 cells were found to be higher in CCR2 recipients of islet allografts than in WT recipients. CONCLUSIONS In summary, lack of CCR2 interferes with the allostimulatory capacity of DC and promotes the generation of regulatory T cells. This is the first demonstration of a mechanistic link between targeting a specific chemokine pathway and the DC-regulatory T cell axis in alloimmunity.
Collapse
Affiliation(s)
- Paolo Fiorina
- Transplantation Research Center, Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kuo YR, Huang CW, Goto S, Wang CT, Hsu LW, Lin YC, Yang KD, Chen CL, Lee WPA. Alloantigen-pulsed host dendritic cells induce T-cell regulation and prolong allograft survival in a rat model of hindlimb allotransplantation. J Surg Res 2008; 153:317-25. [PMID: 19101689 DOI: 10.1016/j.jss.2008.05.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 05/22/2008] [Accepted: 05/27/2008] [Indexed: 11/28/2022]
Abstract
BACKGROUND Composite tissue allotransplantation is restricted due to the risks presented by long-term therapeutic immunosuppression. This study is conducted to investigate whether treatment with recipient immature dendritic cells (DCs) pulsed with donor alloantigens can prolong allograft survival and induce T-cell regulation in a rodent model. MATERIALS AND METHODS Orthotopic hindlimb transplants from Brown-Norway (RT1(n)) to Lewis (RT1(1)) rats were performed (day 0). DCs were propagated from the recipient bone marrow and pulsed with the donor alloantigen lysate. Group 1 (control group) did not receive any treatment. Groups 2 and 3 received cyclosporine A (CsA) at a concentration of 10 and 16 mg.kg(-1).day(-1), respectively, on days 0-20 following composite tissue allotransplantation. Group 4 received antilymphocyte serum (i.p. administered 4 d before and 1 d after transplantation) therapy. Group 5 received combined treatment with CsA (10 mg.kg(-1).day(-1), days 0-20) and donor alloantigen-pulsed recipient DCs (i.v. administered on days 7, 14, and 21). Group 6 received combined treatment with CsA (10 mg.kg(-1).day(-1) on days 0-20), antilymphocyte serum (administered i.p. 4 d before and 1 d after transplantation), and DCs (administered i.v. on days 7, 14, and 21). Graft rejection was defined as epidermolysis/desquamation of the donor skin. The mixed lymphocyte reaction was performed to determine the donor T-cell reactivity. Tissue samples were biopsied to analyze the histological changes, and flow cytometry was performed to quantify the donor T-cells. RESULTS Allograft survival was significantly prolonged (>200 d) in Group 6 when compared with the other groups (P < 0.001). The mixed lymphocyte reaction performed for Group 6 revealed hyporesponsiveness of the T-cells to donor alloantigens. Flow cytometric analysis in Group 6 revealed a significant increase in the percentage of CD4(+)/CD25(+) and CD4(+)/foxP3(+) T-cells expression, and significant increase in the percentage of donor cells (RT1(n)) in the recipient peripheral blood. Immunohistochemical staining of allo-skin revealed a significant increase in the proportion of CD25(+) cells in the subcutaneous and dermis layers in Group 6, as compared to other groups. CONCLUSION Treatment with donor alloantigen-pulsed recipient immature DCs in combination with transient immunosuppression prolongs allograft survival and induced tolerance by inducing T-cell hyporesponsiveness to donor alloantigens and increasing the CD4(+)/CD25(+) T-cell population.
Collapse
Affiliation(s)
- Yur-Ren Kuo
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University, College of Medicine, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
In addition to the function as a physical barrier human skin has been shown to be an important immune organ displaying various defense mechanisms, which can be divided into three major functional compartments: (i) Epithelial defense, which is characterized by antimicrobial peptides and proteins (AP) and which can be induced in inflammatory lesions but also in the absence of inflammation. (ii) Innate-inflammatory immunity, which involves recognition of microbial compounds by particular receptors like Toll-like receptors (TLR) and subsequent activation of signalling pathways resulting in expression of pro-inflammatory cytokines and interferons, as well as genes of adaptive immunity. Interferon alpha (IFNalpha) produced by plasmacytoid dendritic cells (DC) may stimulate myeloid DC to produce IL-12 resulting in classical T-cell activation or to produce IL-23 activating IL-17 producing T-cells (IL-23/IL-17 pathway). (iii) Adaptive immunity, which is based on antigen presenting cells, T-cells and B-cells and which is characterized by specificity and memory. In contrast to epithelial defense and innate-inflammatory immunity, adaptive immune functions provide slowly reacting protection. Recent improvements of our knowledge of dysregulated immune pathways associated with inflammatory skin diseases represent an important basis of novel immunomodulatory treatment modalities.
Collapse
Affiliation(s)
- T Meyer
- Institute for Immunology, Clinical Pathology and Molecular Medicine, Lademannbogen 61, 22339 Hamburg, Germany.
| | | | | |
Collapse
|
18
|
Maestri M, Rademacher J, Gaspari A, Lenti LM, Crespi S, Cansolino L, Novelli G, Agoglitta D, Maffeis F, Ferrario di Tor Vajana A, Oldani G, Dionigi P. Short-term cyclosporine therapy and cotransplantation of donor splenocytes: effects on graft rejection and survival rates in pigs subjected to renal transplantation. J Surg Res 2008; 150:100-9. [PMID: 18561953 DOI: 10.1016/j.jss.2008.01.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2007] [Revised: 01/10/2008] [Accepted: 01/24/2008] [Indexed: 10/22/2022]
Abstract
BACKGROUND Donor-specific allogeneic loading can prolong the survival of solid organ transplants by inducing a state known as acceptance. Several populations of cells are known to be involved in this process, but their exact roles have yet to be defined. The aim of this study was to assess the effects of portal-vein transfusion of donor-specific splenocytes (DST) after short-term cyclosporine A (CyA) therapy in pigs subjected to renal transplantation. METHODS Four groups of unrelated swine underwent renal transplantation with removal of the native kidneys. Antirejection protocols consisted in portal-vein DST (3 x 10(8) cells/kg) (Group 2, n = 7); intravenous CyA (9 mg/kg/d) on postoperative days 1-12 (Group 3, n = 14); and DST + CyA (as described above) (Group 4, n = 13). Results (through postoperative day 90) were compared with those obtained in untreated control recipients (Group 1, n = 7). RESULTS Compared with animals of Groups 1, 2, and 3, Group 4 recipients presented significantly longer survival (mean: 90 days, P < 0.01 in Kaplan-Meier analysis) and better renal function (P < 0.05). Graft histology revealed preserved parenchyma. CONCLUSION The role of spleen cells in the immune response has probably been underestimated. Cotransplantation of donor splenocytes seems to induce a certain degree of acceptance toward the renal allograft. The route of administration (portal-vein infusion in this study) may be crucial for developing favorable mechanisms of recognition.
Collapse
Affiliation(s)
- Marcello Maestri
- Laboratory of Experimental Surgery, Department of Surgical Sciences, University of Pavia, Pavia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Prolongation of Composite Tissue Allograft Survival by Immature Recipient Dendritic Cells Pulsed with Donor Antigen and Transient Low-Dose Immunosuppression. Plast Reconstr Surg 2008; 121:37-49. [DOI: 10.1097/01.prs.0000293754.55706.7f] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
20
|
Abstract
Technical aspects of rare-event detection are discussed in this article in a practical context, with two real-life examples. A growing number of flow cytometry-based assays depend on rare-event detection for basic science and clinical applications.
Collapse
Affiliation(s)
- Albert D Donnenberg
- University of Pittsburgh School of Medicine, Hillman Cancer Research Center Suite2.42c, 5117 Center Avenue, Pittsburgh, PA 15213-2582, USA.
| | | |
Collapse
|
21
|
Goldberg GL, Alpdogan O, Muriglan SJ, Hammett MV, Milton MK, Eng JM, Hubbard VM, Kochman A, Willis LM, Greenberg AS, Tjoe KH, Sutherland JS, Chidgey A, van den Brink MRM, Boyd RL. Enhanced immune reconstitution by sex steroid ablation following allogeneic hemopoietic stem cell transplantation. THE JOURNAL OF IMMUNOLOGY 2007; 178:7473-84. [PMID: 17513799 DOI: 10.4049/jimmunol.178.11.7473] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Delayed immune reconstitution in adult recipients of allogeneic hemopoietic stem cell transplantations (HSCT) is related to age-induced thymic atrophy. Overcoming this paucity of T cell function is a major goal of clinical research but in the context of allogeneic transplants, any strategy must not exacerbate graft-vs-host disease (GVHD) yet ideally retain graft-vs-tumor (GVT) effects. We have shown sex steroid ablation reverses thymic atrophy and enhances T cell recovery in aged animals and in congenic bone marrow (BM) transplant but the latter does not have the complications of allogeneic T cell reactivity. We have examined whether sex steroid ablation promoted hemopoietic and T cell recovery following allogeneic HSCT and whether this benefit was negated by enhanced GVHD. BM and thymic cell numbers were significantly increased at 14 and 28 days after HSCT in castrated mice compared with sham-castrated controls. In the thymus, the numbers of donor-derived thymocytes and dendritic cells were significantly increased after HSCT and castration; donor-derived BM precursors and developing B cells were also significantly increased. Importantly, despite restoring T cell function, sex steroid inhibition did not exacerbate the development of GVHD or ameliorate GVT activity. Finally, IL-7 treatment in combination with castration had an additive effect on thymic cellularity following HSCT. These results indicate that sex steroid ablation can profoundly enhance thymic and hemopoietic recovery following allogeneic HSCT without increasing GVHD and maintaining GVT.
Collapse
Affiliation(s)
- Gabrielle L Goldberg
- Department of Pathology and Immunology, Central and Eastern Clinical School, Monash University, Melbourne, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Löb S, Ebner S, Wagner S, Weinreich J, Schäfer R, Königsrainer A. Are Indoleamine-2,3-Dioxygenase Producing Human Dendritic Cells a Tool for Suppression of Allogeneic T-cell Responses? Transplantation 2007; 83:468-73. [PMID: 17318080 DOI: 10.1097/01.tp.0000251646.09702.ec] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Suppressive dendritic cells (DCs) are a promising tool for tolerance induction in transplantation. A human DCs subpopulation, which constitutively expresses indoleamine-2,3-dioxygenase (IDO), a molecule shown to prevent the rejection of fetus during pregnancy, has recently been described. This subset, characterized by nonadherence and CD123/CCR6 expression, exhibited sustained IDO production if exposed to interleukin (IL)-10. In the present work, we generated human nonadherent, CD123/CCR6 DCs secreting IL-10. METHODS Monocytes were separated by plastic adherence and differentiated to DCs in the presecence of IL-3 and IL-4. Expression of IDO was determined by reverse-transcriptase polymerase chain reaction and enzyme activity by reverse-phased high-performance liquid chromatography. Mixed lymphocyte cultures were performed with allogeneic nylon wool-purified T-cells. RESULTS Contradicting previous findings, CD123+/CCR6+ DCs did not express IDO. Maturation of these cells with inducer-cytokines up-regulated IDO, but the allogeneic T-cell stimulatory capacity of these DCs was even stronger than that of immature IDO DCs, and chemical abrogation of IDO activity did not increase T-cell proliferation. In parallel, we generated mature IDO DCs, but these cells also did not induce stronger T-cell stimulation than their IDO counterpart. CONCLUSIONS In conclusion, CD123/CCR6 DCs do not constitutively express IDO and "induced" IDO DCs, even coexpressing anti-inflammatory IL-10, do not suppress allogeneic T-cell responses.
Collapse
Affiliation(s)
- Stefan Löb
- Department of General-, Visceral- and Transplantation Surgery, Eberhard Karls University, Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|
23
|
Colson YL, Shinde Patil VR, Ildstad ST. Facilitating cells: Novel promoters of stem cell alloengraftment and donor-specific transplantation tolerance in the absence of GVHD. Crit Rev Oncol Hematol 2007; 61:26-43. [PMID: 17150368 DOI: 10.1016/j.critrevonc.2006.06.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2006] [Revised: 05/30/2006] [Accepted: 06/16/2006] [Indexed: 10/23/2022] Open
Abstract
Bone marrow transplantation (BMT) is the treatment of choice for many hematological malignancies and immunopathologies. Unfortunately, success is often impeded by engraftment failure and graft-versus-host disease (GVHD). A rare bone marrow population known as the facilitating cell (FC) has been identified which facilitates stem cell engraftment and circumvents these obstacles in murine experimental models. This review discusses the identification and characterization of this rare population and provides an emerging portrait of FC origin, ontogeny and function. The promotion of durable stem cell engraftment in MHC disparate recipients, GVHD inhibition and tolerance induction by the FC suggests that future therapies in hematopoietic cell transplantation and tolerance induction for solid organ transplants may be significantly improved through the application of FC transplantation.
Collapse
Affiliation(s)
- Yolonda L Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
24
|
Kukhetpitakwong R, Hahnvajanawong C, Homchampa P, Leelavatcharamas V, Satra J, Khunkitti W. Aspirin modified dendritic cells are potent inducers of allo-specific regulatory T-cells. Int Immunopharmacol 2006; 6:1729-35. [PMID: 16979128 DOI: 10.1016/j.intimp.2006.07.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2006] [Accepted: 07/11/2006] [Indexed: 01/04/2023]
Abstract
Pods of Acacia concinna (Leguminosae) contain several saponins. In this study, four saponin fractions which were acetone fraction (AAC), aqueous fraction (WAC), hydromethanolic fraction (HAC) and methanolic fraction (MAC) were generated and their haemolytic activities and surface activities were determined in comparison with quillaja saponin (QS). There were no significant differences between the haemolytic activities of MAC and QS. However, the surface tensions of MAC was significantly lower than QS (p < 0.001). Furthermore, the immunomodulatory effect and the adjuvant potential of MAC on the cellular and humoral immune response of BALB/c mice against ovalbumin were investigated. The splenocyte proliferations induced by MAC were significantly higher than QS at the concentrations of 200, 400, 800 and 1000 microg/ml (p < 0.05). BALB/c mice were immunized subcutaneously either with OVA 20 microg alone or with OVA 20 microg combining with QS (10 microg) or MAC (10 and 40 microg). Ten days after the second immunization, concanavalin A (Con A)-, pokeweed mitogen (PWM)-, and OVA-stimulated splenocyte proliferation and OVA-specific antibodies in serum were measured. The results suggested that MAC (40 microg) could activate T and B cells. In addition, OVA-specific IgG, IgG1 IgG2a and IgG2b antibody levels in serum were significantly enhanced by MAC (40 microg) as compared with OVA control group (p < 0.001). This finding suggested that MAC might be effect on Th1 and Th2 helper T cells. In conclusion, the results indicated that MAC at a dose of 40 microg could be used as vaccine adjuvant to increase immune responses.
Collapse
Affiliation(s)
- Ratiya Kukhetpitakwong
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | | | | | | | | | | |
Collapse
|
25
|
Buckland M, Jago CB, Fazekasova H, Scott K, Tan PH, George AJT, Lechler R, Lombardi G. Aspirin-treated human DCs up-regulate ILT-3 and induce hyporesponsiveness and regulatory activity in responder T cells. Am J Transplant 2006; 6:2046-59. [PMID: 16869801 DOI: 10.1111/j.1600-6143.2006.01450.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Mature dendritic cells (mDCs) are potent antigen presenting cells, but immature DCs (iDCs) have been shown to have reduced antigen stimulatory capacity. Different strategies have been investigated to augment the tolerogenic capacity of dendritic cells (DCs). We demonstrate that in aspirin-treated human DCs, there is reduced expression of CD1a, HLA-DR and CD86, up-regulation of ILT-3 expression and marginal increases in PDL-1. Aspirin-treated DCs are partially resistant to phenotypic changes following maturational stimuli, such as lipopolysaccharide (LPS) or TNFalpha, IL-1alpha and PGE2. Aspirin-treated DCs demonstrate normal endocytic function, but have a reduced ability to stimulate allogeneic T cells, which is comparable to iDCs. Furthermore, they induce hyporesponsiveness and regulatory activity in responder naïve and memory T cells; for naïve T cells this is achieved more quickly and efficiently than with iDCs. We investigated the mechanism of this regulatory activity and found that both cell-cell contact and inhibitory cytokine activity are involved, although no one cytokine predominates in importance. Blocking ILT-3 or IL-12 does not diminish the capacity of these DCs to induce regulation or Foxp3 expression on the regulatory T cells. Results demonstrate that aspirin-treated DCs display tolerogenic potential, which is of interest in their therapeutic potential in reducing chronic allograft rejection.
Collapse
Affiliation(s)
- M Buckland
- Immunoregulation Laboratories, Department of Nephrology and Transplantation, King's College London School of Medicine at Guy's, King's College and St. Thomas' Hospitals, London, SE1 9RT, UK
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Xiang M, Zhang CL, Zou XL, Cai XH, Peng JB. Roles of dendritic cells and regulatory T cells in autoantigen-induced murine immune tolerance model. Shijie Huaren Xiaohua Zazhi 2006; 14:687-692. [DOI: 10.11569/wcjd.v14.i7.687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the important roles of dendritic cells (DC) and CD4+CD25+ regulatory T cells in immune prevention against insulin dependent diabetes (IDDM) by autoantigen insulin administration.
METHODS: The model of IDDM was established by intraperitoneal injection of low-dose streptozotocin (STZ) 40 mg/kg per day for 5 consecutive days in Balb/c mice. The bovine insulin (100 μg) in incomplete Freund's adjuvant (IFA, emulsified 1∶1) was given subcutaneously to the mice weekly for 4 wk. The blood glucose was examined once a week and all the mice were killed after 5 wk. Pancreas tissues were collected for histopathological examination. DC precursor cells from bone marrow and lymphocytes from spleen were isolated. The phenotype of DC and CD4+ CD25+ regulatory T cells were analyzed by fluorescence activated cell sorter (FACS). DC-stimulated proliferation of lymphacytes was determined by allo-mixed lymphocyte reaction (aMLR).
RESULTS: The level of blood glucose was decreased significantly after insulin injection in comparison with that in the model control group (13.79 ± 2.71 mmol/L vs 20.98 ± 1.43 mmol/L, P < 0.05). Fewer lymphocytes infiltration was observed and pancreatic histological structure was intact. The surface marker CD11c on DC from bone marrow was decreased markedly in IDDM mice (26.4%) than that in normal mice (47.5%). DC differentiated abnormally, and the capacity of stimulating proliferation of allogeneic T cell was weakened as compared with that of normal mice (1.47 ± 0.01 vs 2.93 ± 0.01, P < 0.01, and 1.32 ± 0.01 vs 2.94 ± 0.02, P < 0.01, at DC/T ratios of 1∶10 and 1∶20, respectively). The percentage of CD4+CD25+ T cells were decreased to 1.43%, while it was 5.09% in normal mice. In contrast, blood glucose in mice given insulin subcutaneously was well controlled, and the amount of DC with CD11c was increased (50% approximately); the expression of CD86 and MHC-Ⅱ was low (26.6% and 28.8%, respectively) and MLR showed that DC capacity in stimulating T cell proliferation was lower than those from the normal mice, but higher than those from IDDM model mice (2.30 ± 0.06 and 2.17 ± 0.02, at DC/T ratios of 1∶10 and 1∶20, respectively); the percentage of CD4+CD25+ T cells from spleen was enhanced to 7.15%.
CONCLUSION: Subcutaneous administration of insulin can confer protection to mice against IDDM induced by STZ. The immune protection of autoantigen may be associated with the establishment of immune tolerance by improving the function of abnormal DC and promoting the production of CD4+CD25+ T cells in vivo.
Collapse
|
27
|
|
28
|
Mathers AR, Larregina AT. Professional antigen-presenting cells of the skin. Immunol Res 2006; 36:127-36. [PMID: 17337773 DOI: 10.1385/ir:36:1:127] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/06/2023]
Abstract
The skin functions as an important pro-inflammatory and immune organ. Accordingly, the epidermis and dermis are highly populated by dendritic cells (DC), which are potent antigen-presenting cells (APC) with important immunostimulatory and migratory activities. Whereas the biological characteristics and immunological functions of epidermal DC known as Langernahs cells (LC) have been the focus of intense research in the past, less is known regarding their dermal counterparts named dermal dendritic cells (DDC). Although it has been widely accepted that LC are the more relevant skin-resident APC, recent experimental evidence challenges this concept and proposes a different role for these important cell populations. In this article we compile recent scientific advances regarding the function of different skin-resident DC and we try to reconcile the new observations with the previously established paradigm.
Collapse
Affiliation(s)
- Alicia R Mathers
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | |
Collapse
|
29
|
Clarke JH, Cha JY, Walsh MD, Harken AH, McCarter MD. Dendritic cells as therapeutic adjuncts in surgical disease. Surgery 2005; 138:844-50. [PMID: 16291384 DOI: 10.1016/j.surg.2005.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Accepted: 02/13/2005] [Indexed: 12/22/2022]
Affiliation(s)
- Jason H Clarke
- Department of Surgery, University of Colorado Health Sciences Center, Denver, CO 80262, USA
| | | | | | | | | |
Collapse
|
30
|
Zeyda M, Kirsch BM, Geyeregger R, Stuhlmeier KM, Zlabinger GJ, Hörl WH, Säemann MD, Stulnig TM. Inhibition of Human Dendritic Cell Maturation and Function by the Novel Immunosuppressant FK778. Transplantation 2005; 80:1105-11. [PMID: 16278593 DOI: 10.1097/01.tp.0000178301.19732.a1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND FK778, a derivative of the active leflunomide-metabolite, A77 1726, has been shown to be a powerful immunosuppressant in several transplantation models, particularly efficient in the prevention of chronic allograft rejection. However, the cellular and molecular mechanisms underlying these effects of FK778 have not been investigated yet in detail. Because dendritic cells (DCs) are the most potent antigen-presenting cells (APCs) and are essential for the initiation of immune responses including acute and chronic allograft rejection, we investigated whether FK778 affects this particular cell type. METHODS Allogeneic T cell stimulation by FK778-treated human monocyte-derived DCs was determined by mixed leukocyte cultures. Surface molecule expression was analyzed by flow-cytometric analysis and cytokine production by ELISA from culture supernatants. Activation of NF-kappaB in DCs was assessed by electrophoretic mobility shift assays. RESULTS Treatment of DCs with FK778 inhibited their potency to stimulate allogeneic T cells. In line, LPS- and CD40L-induced upregulation of DC surface activation markers and production of IL-12 was significantly inhibited, irrespective of whether cells were treated during or after the monocyte to DC differentiation period. The effects of FK778 on DCs were not reversible by exogenous uridine indicating that FK778 acts independently of its action as an inhibitor of pyrimidine synthesis. On the signaling level, activation of NF-kappaB, the essential transcription factor involved in DC maturation and function, was markedly inhibited by FK778. CONCLUSIONS Inhibition of activation and function of DCs as the central APCs may significantly contribute to the immunosuppressive profile of FK778 when applied after allogeneic organ transplantation.
Collapse
Affiliation(s)
- Maximilian Zeyda
- Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Tan PH, Sagoo P, Chan C, Yates JB, Campbell J, Beutelspacher SC, Foxwell BMJ, Lombardi G, George AJT. Inhibition of NF-kappa B and oxidative pathways in human dendritic cells by antioxidative vitamins generates regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:7633-44. [PMID: 15944264 DOI: 10.4049/jimmunol.174.12.7633] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Dendritic cells (DCs) are central to T cell immunity, and many strategies have been used to manipulate DCs to modify immune responses. We investigated the effects of antioxidants ascorbate (vitamin C) and alpha-tocopherol (vitamin E) on DC phenotype and function. Vitamins C and E are both antioxidants, and concurrent use results in a nonadditive activity. We have demonstrated that DC treated with these antioxidants are resistant to phenotypic and functional changes following stimulation with proinflammatory cytokines. Following treatment, the levels of intracellular oxygen radical species were reduced, and the protein kinase RNA-regulated, eukaryotic translation initiation factor 2alpha, NF-kappaB, protein kinase C, and p38 MAPK pathways could not be activated following inflammatory agent stimulation. We went on to show that allogeneic T cells (including CD4(+)CD45RO, CD4(+)CD45RA, and CD4(+)CD25(-) subsets) were anergized following exposure to vitamin-treated DCs, and secreted higher levels of Th2 cytokines and IL-10 than cells incubated with control DCs. These anergic T cells act as regulatory T cells in a contact-dependent manner that is not dependent on IL-4, IL-5, IL-10, IL-13, and TGF-beta. These data indicate that vitamin C- and E-treated DC might be useful for the induction of tolerance to allo- or autoantigens.
Collapse
Affiliation(s)
- Peng H Tan
- Department of Immunology, Division of Medicine, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chen X, Murakami T, Oppenheim JJ, Howard OMZ. Triptolide, a constituent of immunosuppressive Chinese herbal medicine, is a potent suppressor of dendritic-cell maturation and trafficking. Blood 2005; 106:2409-16. [PMID: 15956285 PMCID: PMC1569904 DOI: 10.1182/blood-2005-03-0854] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Triptolide (TPT) is a chemically defined, potent immunosuppressive compound isolated from an anti-inflammatory Chinese herbal medicine. TPT has been reported to inhibit autoimmunity, allograft rejection, and graft-versus-host disease (GVHD), and its efficacy was previously attributed to the suppression of T cells. Since dendritic cells (DCs) play a major role in the initiation of T-cell-mediated immunity, we studied the effects of TPT on the phenotype, function, and migration of human monocyte-derived DCs. TPT treatment, over a pharmacologic concentration range, inhibited the lipopolysaccharide (LPS)-induced phenotypic changes, characteristic of mature DCs and the production of interleukin-12p70 (IL-12p70). Consequently, the allostimulatory functions of DCs were impaired by TPT treatment. Furthermore, the calcium mobilization and chemotactic responses of LPS-stimulated DCs to secondary lymphoid tissue chemokine (SLC)/CC chemokine ligand 21 (CCL21) were significantly lower in TPT-treated than untreated DCs, in association with lower chemokine receptor 7 (CCR7) and higher CCR5 expression. Egress of Langerhans cells (LCs) from explanted mouse skin in response to macrophage inflammatory protein-3beta (MIP-3beta)/CCL19 was arrested by TPT. In vivo administration of TPT markedly inhibited hapten (fluorescein isothiocyanate [FITC])-stimulated migration of mouse skin LCs to the draining lymph nodes. These data provide new insight into the mechanism of action of TPT and indicate that the inhibition of maturation and trafficking of DCs by TPT contributes to its immunosuppressive effects.
Collapse
Affiliation(s)
- Xin Chen
- Basic Research Program, Science Applications International Corporation-Frederick, Inc., Laboratory of Molecular Immunoregulation, National Cancer Institute-Frederick, Frederick, MD 21702-1201, USA.
| | | | | | | |
Collapse
|
33
|
Fugier-Vivier IJ, Rezzoug F, Huang Y, Graul-Layman AJ, Schanie CL, Xu H, Chilton PM, Ildstad ST. Plasmacytoid precursor dendritic cells facilitate allogeneic hematopoietic stem cell engraftment. ACTA ACUST UNITED AC 2005; 201:373-83. [PMID: 15699072 PMCID: PMC2213023 DOI: 10.1084/jem.20041399] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Bone marrow transplantation offers great promise for treating a number of disease states. However, the widespread application of this approach is dependent upon the development of less toxic methods to establish chimerism and avoid graft-versus-host disease (GVHD). CD8+/TCR− facilitating cells (FCs) have been shown to enhance engraftment of hematopoietic stem cells (HSCs) in allogeneic recipients without causing GVHD. In the present studies, we have identified the main subpopulation of FCs as plasmacytoid precursor dendritic cells (p-preDCs). FCs and p-preDCs share many phenotypic, morphological, and functional features: both produce IFN-α and TNF-α, both are activated by toll-like receptor (TLR)-9 ligand (CpG ODN) stimulation, and both expand and mature after Flt3 ligand (FL) treatment. FL-mobilized FCs, most of which express a preDC phenotype, significantly enhance engraftment of HSCs and induce donor-specific tolerance to skin allografts. However, p-preDCs alone or p-preDCs from the FC population facilitate HSC engraftment less efficiently than total FCs. Moreover, FCs depleted of preDCs completely fail to facilitate HSC engraftment. These results are the first to define a direct functional role for p-preDCs in HSC engraftment, and also suggest that p-preDCs need to be in a certain state of maturation/activation to be fully functional.
Collapse
|
34
|
Müller H, Hofer S, Kaneider N, Neuwirt H, Mosheimer B, Mayer G, Konwalinka G, Heufler C, Tiefenthaler M. The immunomodulator FTY720 interferes with effector functions of human monocyte-derived dendritic cells. Eur J Immunol 2005; 35:533-45. [PMID: 15657952 DOI: 10.1002/eji.200425556] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The potent immunomodulator FTY720 elicits immunosuppression via acting on sphingosine 1-phosphate receptors (S1PR), thereby leading to an entrapment of lymphocytes in the secondary lymphoid tissue. To elucidate the potential in vitro effects of this drug on human monocyte-derived DC, we used low nanomolar therapeutic concentrations of FTY720 and phosphorylated FTY720 (FTY720-P) and investigated their influence on DC surface marker expression, protein levels of S1PR and DC effector functions: antigen uptake, chemotaxis, cytokine production, allostimulatory and Th-priming capacity. We report that both FTY720 and FTY720-P reduce chemotaxis of immature and mature DC. Mature DC generated in the presence of FTY720 or FTY720-P showed an impaired immunostimmulatory capacity and reduced IL-12 but increased IL-10 production. T cells cultured in the presence of FTY720- or FTY720-P-treated DC showed an altered cytokine production profile indicating a shift from Th1 toward Th2 differentiation. In treated immature and mature DC, expression levels for two S1PR proteins, S1P1 and S1P4, were reduced. We conclude that in vitro treatment with FTY720 affects DC features that are essential for serving their role as antigen-presenting cells. This might represent a new aspect of the overall immunosuppressive action of FTY720 and makes DC potential targets of further sphingolipid-derived drugs.
Collapse
Affiliation(s)
- Hansgeorg Müller
- Department of Nephrology, Innsbruck Medical School, Innsbruck, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Larregina AT, Falo LD. Changing Paradigms in Cutaneous Immunology: Adapting with Dendritic Cells. J Invest Dermatol 2005; 124:1-12. [PMID: 15654947 DOI: 10.1111/j.1523-1747.2004.23554.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Adriana T Larregina
- Department of Dermatology, University of Pittsburgh School of Medicine, PA, USA
| | | |
Collapse
|
36
|
Juretic K, Strbo N, Crncic TB, Laskarin G, Rukavina D. An Insight into the Dendritic Cells at the Maternal-Fetal Interface. Am J Reprod Immunol 2004; 52:350-5. [PMID: 15663599 DOI: 10.1111/j.1600-0897.2004.00232.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The conditions that permit the genetically distinct fetus to survive and develop within the mother are among the most fascinating immunologic puzzles. The presence of dendritic cells in the maternal decidua pointed to a biologic role of antigen-presenting cells in maternal-fetal interaction. The method of study included recent findings on the lineage, maturity, phenotype and function of dendritic cells at the maternal-fetal interface. The increment of uterine dendritic cells occurs simultaneously with the decisive phase of gestation, when implantation takes place. Decidual dendritic cells of the first trimester pregnancy, with a phenotype characteristic of the mature myeloid lineage, express MHC class II, co-stimulatory and adhesion molecules, control Th1/Th2 balance and activate the proliferative response of autologous NK cells. Dendritic cells are specifically equipped to control immunity, to trigger immune response and also to maintain tolerance, avoiding the rejection of the conceptus by the maternal immune system.
Collapse
Affiliation(s)
- Koraljka Juretic
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, Brace Branchetta 20, Rijeka 51 000, Croatia
| | | | | | | | | |
Collapse
|
37
|
Abstract
Bone marrow-derived dendritic cells (DCs) are the most potent antigen-presenting cells capable of activating naïve T cells. Loading DCs ex vivo with tumor antigens can stimulate potent antitumor immunity in tumor-bearing mice. This review describes the use of mRNA-encoded tumor antigens as a form of antigen loaded onto DCs, including our early experience from clinical trials in urological cancers. Transfection of DCs with mRNA is simple and effective. Comparative studies suggest that mRNA transfection is superior to other antigen-loading techniques in generating immunopotent DCs. The ability to amplify RNA from microscopic amounts of tumor tissue extends the use of DC vaccination to virtually every cancer patient. The striking observation from two phase I clinical trials, in patients with prostate cancer immunized with prostate-specific antigen mRNA-transfected DCs and patients with renal cancer immunized with autologous tumor RNA-transfected DCs, was that the majority of patients exhibited a vaccine-induced T-cell response. Suggestive evidence of clinically related responses was seen in both the trials. Immunization with mRNA-transfected DCs is a promising strategy to stimulate potent antitumor immunity and could serve as a foundation for developing effective treatments for cancer.
Collapse
Affiliation(s)
- Eli Gilboa
- Department of Surgery, Center for Genetic and Cellular Therapies, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
38
|
Martinson J, Bae J, Klingemann HG, Tam Y. Activated platelets rapidly up-regulate CD40L expression and can effectively mature and activate autologous ex vivo differentiated DC. Cytotherapy 2004. [DOI: 10.1080/14653240410005249-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
39
|
Ramakrishna V, Treml JF, Vitale L, Connolly JE, O'Neill T, Smith PA, Jones CL, He LZ, Goldstein J, Wallace PK, Keler T, Endres MJ. Mannose receptor targeting of tumor antigen pmel17 to human dendritic cells directs anti-melanoma T cell responses via multiple HLA molecules. THE JOURNAL OF IMMUNOLOGY 2004; 172:2845-52. [PMID: 14978085 DOI: 10.4049/jimmunol.172.5.2845] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Targeting recycling endocytic receptors with specific Abs provides a means for introducing a variety of tumor-associated Ags into human dendritic cells (DCs), culminating in their efficient presentation to T cells. We have generated a human mAb (B11) against the mannose receptor that is rapidly internalized by DCs through receptor-mediated endocytosis. By genetically linking the melanoma Ag, pmel17, to Ab B11, we obtained the fully human fusion protein, B11-pmel17. Treatment of DCs with B11-pmel17 resulted in the presentation of pmel17 in the context of HLA class I and class II molecules. Thus, potent pmel17-specific T cells were cytotoxic toward gp100(+) HLA-matched melanoma targets, but not HLA-mismatched melanoma or gp100(-) nonmelanoma tumor lines. Importantly, competitive inhibition of lysis of an otherwise susceptible melanoma cell line by cold targets pulsed with known gp100 CD8 T cell epitopes as well as a dose-dependent proliferative response to Th epitopes demonstrates that DCs can process targeted Ag for activation of cytotoxic as well as helper arms of the immune response. Thus, the specific targeting of soluble exogenous tumor Ag to the DC mannose receptor directly contributes to the generation of multiple HLA-restricted Ag-specific T cell responses.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/pharmacology
- Antibody Specificity/genetics
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Binding Sites, Antibody/genetics
- Binding, Competitive/genetics
- Binding, Competitive/immunology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line, Transformed
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Epitopes, T-Lymphocyte/immunology
- Gene Targeting/methods
- HLA Antigens/metabolism
- HLA Antigens/physiology
- HLA-A2 Antigen/metabolism
- HLA-A2 Antigen/physiology
- HLA-D Antigens/physiology
- Humans
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Mannose Receptor
- Mannose-Binding Lectins/genetics
- Mannose-Binding Lectins/immunology
- Mannose-Binding Lectins/metabolism
- Melanoma/immunology
- Melanoma/prevention & control
- Membrane Glycoproteins/chemical synthesis
- Membrane Glycoproteins/immunology
- Mice
- Mice, Transgenic
- Molecular Sequence Data
- Neoplasm Proteins/chemical synthesis
- Neoplasm Proteins/immunology
- Proteins/genetics
- Proteins/immunology
- Proteins/physiology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Recombinant Fusion Proteins/chemical synthesis
- Recombinant Fusion Proteins/metabolism
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Helper-Inducer/immunology
- Transfection
- gp100 Melanoma Antigen
Collapse
|
40
|
Park JK, Cho K, Johnson J, Perez RV. Induction of MIP-1α in Kupffer cell by portal venous transfusion. Transpl Immunol 2004; 13:33-8. [PMID: 15203126 DOI: 10.1016/j.trim.2004.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2003] [Revised: 02/08/2004] [Accepted: 02/20/2004] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Previous studies have shown that portal venous transfusion (PVT) induces a state of immunosuppression, and Kupffer cells may be involved in the mechanism. OBJECTIVE This study was aimed to investigate the effect of PVT on Kupffer cell gene expression. MATERIALS AND METHODS Each BALB/C mouse was subjected to laparotomy and received one of five treatments: PVT, portal venous saline injection (PVS), inferior vena caval transfusion (IVCT), inferior vena caval saline injection (IVCS) or sham operation (S). The blood for PVT and IVCT was sampled from C57BL/6J mice. Kupffer cells were then isolated 1 or 24 h after each of the 5 treatments, for a total of 10 experimental groups (1-h PVT, PVS, IVCT, IVCS and S, and 24-h PVT, PVS, IVCT, IVCS and S) from BALB/C mice. To examine the effect of PVT on Kupffer cell gene expression, RT-PCR differential display was performed. RESULTS Increase in the expression of MIP-1alpha mRNA post PVT and IVCT was identified by differential display. PVT groups revealed higher levels of serum MIP-1alpha than any other groups. CONCLUSION These results suggest that MIP-1alpha may be involved in a cascade of signaling events associated with the PVT-mediated immunologic modulation in Kupffer cells.
Collapse
Affiliation(s)
- Jong Kwon Park
- Department of Surgery, College of Medicine, Dankook University, Cheonan, Chungnam 330-714, South Korea.
| | | | | | | |
Collapse
|
41
|
Rutella S, Lemoli RM. Regulatory T cells and tolerogenic dendritic cells: from basic biology to clinical applications. Immunol Lett 2004; 94:11-26. [PMID: 15234530 DOI: 10.1016/j.imlet.2004.04.015] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2004] [Revised: 04/20/2004] [Accepted: 04/21/2004] [Indexed: 10/26/2022]
Abstract
The induction of antigen (Ag)-specific tolerance is critical for the prevention of autoimmunity and maintenance of immune homeostasis. Recently, attention has been focused on induction of active suppression by regulatory T cells (Treg cells). Also, a role for dendritic cells (DCs) in the promotion of peripheral tolerance has been demonstrated by several studies and is the subject of intensive investigation. It is currently believed that the maturation/activation state of DCs might be a control point for the induction of peripheral tolerance through modifications of the activation state of T cells. Several lines of experimental evidence suggest that unique DC subsets or discrete functional states of the DCs might be devoted to the promotion of Treg cell differentiation. The present review summarizes the emerging literature on the developmental origin and function of human Treg cells and tolerogenic/regulatory DCs. Furthermore, clinical implications of these studies for cellular therapy of immune-mediated pathologies are discussed.
Collapse
Affiliation(s)
- Sergio Rutella
- Laboratory of Immunology, Department of Hematology, Catholic University Medical School, Largo Agostino Gemelli, 8, 00168 Rome, Italy.
| | | |
Collapse
|
42
|
van Kooten C, Woltman AM. Dendritic cells as a target of immunosuppressive drugs. Transplant Rev (Orlando) 2004. [DOI: 10.1016/j.trre.2004.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
43
|
Elbe-Bürger A, Stingl G. [The role of dendritic cells in immunity. Potential clinical use]. Ann Dermatol Venereol 2004; 131:93-103. [PMID: 15041858 DOI: 10.1016/s0151-9638(04)93556-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- A Elbe-Bürger
- Département de Dermatologie, Division d'Immunologie, Allergie et Maladies Infectieuses, Université de l'Ecole Médicale de Vienne, Vienne Centre de Recherche Internationale, Autriche
| | | |
Collapse
|
44
|
Jonuleit H, Schmitt E. The regulatory T cell family: distinct subsets and their interrelations. THE JOURNAL OF IMMUNOLOGY 2004; 171:6323-7. [PMID: 14662827 DOI: 10.4049/jimmunol.171.12.6323] [Citation(s) in RCA: 292] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Helmut Jonuleit
- Department of Dermatology, Johannes Gutenberg-University, Mainz, Germany.
| | | |
Collapse
|
45
|
Hackstein H, Thomson AW. Dendritic cells: emerging pharmacological targets of immunosuppressive drugs. Immunology 2004; 4:24-34. [PMID: 14704765 PMCID: PMC1424061 DOI: 10.1038/nri1256] [Citation(s) in RCA: 433] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Immunosuppressive drugs have revolutionized organ transplantation and improved the therapeutic management of autoimmune diseases. The development of immunosuppressive drugs and understanding of their action traditionally has been focused on lymphocytes, but recent evidence indicates that these agents interfere with immune responses at the earliest stage, targeting key functions of dendritic cells (DCs). Here, we review our present understanding of how classical and new immunosuppressive agents interfere with DC development and function. This knowledge might provide a rational basis for the selection of immunosuppressive drugs in different clinical settings and for the generation of tolerogenic DCs in the laboratory.
Collapse
Affiliation(s)
- Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig University Giessen, Langhansstr. 7, D-35392 Giessen, Germany.
| | | |
Collapse
|
46
|
Morelli AE, Thomson AW. Dendritic cells: regulators of alloimmunity and opportunities for tolerance induction. Immunol Rev 2003; 196:125-46. [PMID: 14617202 DOI: 10.1046/j.1600-065x.2003.00079.x] [Citation(s) in RCA: 229] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dendritic cells (DCs) are uniquely well-equipped antigen-presenting cells (APCs) regarded classically as sentinels of the immune response, which induce and regulate T-cell reactivity. They play critical roles in central tolerance and in the maintenance of peripheral tolerance in the normal steady state. Following cell or organ transplantation, DCs present antigen to T cells via the direct or indirect pathways of allorecognition. These functions of DCs set in train the rejection response, but they also serve as potential targets for suppression of alloimmune reactivity and promotion of tolerance induction. Much evidence from various model systems now indicates that DCs can induce specific T-cell tolerance. Although underlying mechanisms have not been fully elucidated, the capacity to induce T-regulatory cells may be an important property of tolerogenic or regulatory DCs. Efforts to generate "designer" DCs with tolerogenic properties in the laboratory using specific cytokines, immunologic or pharmacologic reagents, or genetic engineering approaches have already met with some success. Alternatively, targeting of DCs in vivo (e.g. by infusion of apoptotic allogeneic cells) to take advantage of their inherent tolerogenicity has also demonstrated exciting potential. The remarkable heterogeneity and plasticity of these important APCs present additional challenges to optimizing DC-based therapies that may lead to improved tolerance-enhancing strategies in the clinic.
Collapse
Affiliation(s)
- Adrian E Morelli
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, W1544 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
47
|
Liang S, Horuzsko A. Mobilizing dendritic cells for tolerance by engagement of immune inhibitory receptors for HLA-G. Hum Immunol 2003; 64:1025-32. [PMID: 14602231 DOI: 10.1016/j.humimm.2003.08.348] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The presence of dendritic cells (DC) in the maternal decidua has pointed to a biologic role of antigen-presenting cell in maternal-fetal interaction. The expression of immune inhibitory receptors on DC opens the intriguing possibility that these types of receptors are directly involved in maturation/activation of DC and modulate their function. We show that the triggering of the murine inhibitory receptor paired immunoglobulin-like receptor-B by cross-linking or by human leukocyte antigen (HLA)-G tetramer resulted in the modulation of DC function and prolongation of allogeneic graft survival. In addition, we found that the engagement of human inhibitory receptor ILT4 by its natural ligand, HLA-G, alters maturation of human DC. In this study, we examined the mechanisms for the modulation of antigen-presenting cells by HLA-G. These findings have established an important link between HLA-G and immune inhibitory receptor regulation in vivo and in vitro, thereby placing HLA-G in the inhibitory pathway.
Collapse
Affiliation(s)
- Siyuan Liang
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, GA 30912, USA
| | | |
Collapse
|
48
|
Kaneko K, Morelli AE, Wang Z, Thomson AW. Alloantigen presentation by ethylcarbodiimide-treated dendritic cells induces T cell hyporesponsiveness, and prolongs organ graft survival. Clin Immunol 2003; 108:190-8. [PMID: 14499242 DOI: 10.1016/s1521-6616(03)00141-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Ethylcarbodiimide (ECDI) couples soluble antigens (Ag) to lymphoid cells bestowing tolerizing potential. We examined whether ECDI-treated allogeneic dendritic cells (DC) could promote Ag-specific T cell unresponsiveness and prolong graft survival. Exposure of murine myeloid DC to ECDI did not affect surface immunophenotype but reduced their ability to cluster with T cells, enhanced their apoptotic death, and markedly reduced their allostimulatory activity. Anti-donor proliferative and cytotoxic T cell responses of mice primed with ECDI-treated DC were markedly inhibited. Secretion of both Th1 (IFNgamma) and Th2 cytokines (IL-5, IL-10) was suppressed. Cardiac allograft survival in mice preconditioned with a single injection of ECDI-DC was prolonged significantly. These results indicate that ECDI-treated DC promote T cell unresponsiveness to donor alloAgs and prolong transplant survival. The effects are not associated with sparing of Th2 responses, but may reflect inhibitory effects of apoptotic donor DC on host immune reactivity.
Collapse
Affiliation(s)
- Katsuhiko Kaneko
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
49
|
Commeren DL, Van Soest PL, Karimi K, Löwenberg B, Cornelissen JJ, Braakman E. Paradoxical effects of interleukin-10 on the maturation of murine myeloid dendritic cells. Immunology 2003; 110:188-96. [PMID: 14511232 PMCID: PMC1783045 DOI: 10.1046/j.1365-2567.2003.01730.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The immunoregulatory cytokine, interleukin-10 (IL-10), has been shown to inhibit the maturation of human myeloid dendritic cells (DC). In the present study, we demonstrate that IL-10 has paradoxical effects on the maturation of murine myeloid bone marrow-derived DC. On the one hand, IL-10 inhibits the maturation of murine myeloid DC. The addition of IL-10 to granulocyte-macrophage colony-stimulating factor (GM-CSF)-supported murine BM-derived DC cultures reduced the frequency of major histocompatibility complex (MHC) class IIbright cells. These IL-10-pretreated DC have a reduced capacity to stimulate T cells in an allogeneic mixed leucocyte reaction. On the other hand, however, and in contrast to the effects of IL-10 on human DC, we found that the addition of IL-10 from the initiation of the culture onwards induced an up-regulation of the expression of the costimulatory molecules CD40, CD80 and CD86 on murine myeloid DC, as compared to DC generated with GM-CSF only. Moreover, a subpopulation of IL-10-pretreated MHC class IIdim DC lacked the capacity to take up dextran-fluorescein isothiocyanate (FITC), a feature of DC maturation. Taken together, our data demonstrate that the generation of murine myeloid DC in the presence of IL-10 results in a population of incompletely matured MHC class IIdim CD80+ CD86+ DC. These DC lack T-cell stimulatory capacity, suggesting a role for IL-10 in conferring tolerogenic properties on murine myeloid DC.
Collapse
|
50
|
Dong X, Craig T, Xing N, Bachman LA, Paya CV, Weih F, McKean DJ, Kumar R, Griffin MD. Direct transcriptional regulation of RelB by 1alpha,25-dihydroxyvitamin D3 and its analogs: physiologic and therapeutic implications for dendritic cell function. J Biol Chem 2003; 278:49378-85. [PMID: 14507914 DOI: 10.1074/jbc.m308448200] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The nuclear factor-kappaB (NF-kappaB) protein RelB plays a unique role in dendritic cell (DC) function and, as such, is an important regulator of antigen presentation and immune regulation. In this study, inhibition of RelB expression in DCs exposed to an analog of the active form of vitamin D3 (1alpha,25-dihydroxyvitamin D3 (1alpha,25-(OH)2D3)) was observed and shown to be mediated by the vitamin D receptor (VDR). Potential vitamin D response elements were identified within promoter regions of human and mouse relB genes. In gel shift experiments, these motifs specifically bound VDR.retinoid X receptor-alpha complexes. Reporter assays confirmed that transcriptional activity of human and mouse relB promoters was inhibited by 1alpha,25-(OH)2D3 agonists in a DC-derived cell line. The inhibition was abolished by mutagenesis of the putative vitamin D response elements and was enhanced by overexpression of VDR. Mutagenesis of NF-kappaB response elements within the relB promoter did not affect the magnitude of 1alpha,25-(OH)2D3 analog-mediated inhibition, ruling out an indirect effect on NF-kappaB signaling. Glucocorticoid caused additional inhibition of relB promoter activity when combined with the 1alpha,25-(OH)2D3 analog. This effect was dependent on the integrity of the NF-kappaB response elements, suggesting separate regulatory mechanisms for the two steroid pathways on this promoter. We conclude that relB is a direct target for 1alpha,25-(OH)2D3-mediated negative transcriptional regulation via binding of VDR.retinoid X receptor-alpha to discrete DNA motifs. This mechanism has important implications for the inhibitory effect of 1alpha,25-(OH)2D3 on DC maturation and for the potential immunotherapeutic use of 1alpha,25-(OH)2D3 analogs alone or combined with other agents.
Collapse
Affiliation(s)
- Xiangyang Dong
- Department of Internal Medicine, Mayo Clinic Foundation, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|