51
|
Knights AJ, Fucikova J, Pasam A, Koernig S, Cebon J. Inhibitor of apoptosis protein (IAP) antagonists demonstrate divergent immunomodulatory properties in human immune subsets with implications for combination therapy. Cancer Immunol Immunother 2013; 62:321-35. [PMID: 22923192 PMCID: PMC11028923 DOI: 10.1007/s00262-012-1342-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 08/13/2012] [Indexed: 02/07/2023]
Abstract
Inhibitor of apoptosis proteins (IAPs) are critical in regulating apoptosis resistance in cancer. Antagonists of IAPs, such as LCL161, are in clinical development and show promise as anti-cancer agents for solid and hematological cancers, with preliminary data suggesting they may act as immunomodulators. IAP antagonists hypersensitize tumor cells to TNF-α-mediated apoptosis, an effect that may work in synergy with that of cancer vaccines. This study aimed to further investigate the immunomodulatory properties of LCL161 on human immune subsets. T lymphocytes treated with LCL161 demonstrated significantly enhanced cytokine secretion upon activation, with little effect on CD4 and CD8 T-cell survival or proliferation. LCL161 treatment of peripheral blood mononuclear cells significantly enhanced priming of naïve T cells with synthetic peptides in vitro. Myeloid dendritic cells underwent phenotypic maturation upon IAP antagonism and demonstrated a reduced capacity to cross-present a tumor antigen-based vaccine. These effects are potentially mediated through an observed activation of the canonical and non-canonical NF-κB pathways, following IAP antagonism with a resulting upregulation of anti-apoptotic molecules. In conclusion, this study demonstrated the immunomodulatory properties of antagonists at physiologically relevant concentrations and indicates their combination with immunotherapy requires further investigation.
Collapse
Affiliation(s)
- Ashley J Knights
- Ludwig Institute for Cancer Research Melbourne, Austin Branch, Austin Hospital, 145-163 Studley Road, Heidelberg, VIC, 3084, Australia.
| | | | | | | | | |
Collapse
|
52
|
Control of RelB during dendritic cell activation integrates canonical and noncanonical NF-κB pathways. Nat Immunol 2012; 13:1162-70. [PMID: 23086447 PMCID: PMC3634611 DOI: 10.1038/ni.2446] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 08/29/2012] [Indexed: 12/15/2022]
Abstract
The NF-κB protein RelB controls dendritic cell (DC) maturation and may be targeted therapeutically to manipulate T cell responses in disease. Here we report that RelB promoted DC activation not as the expected RelB-p52 effector of the non-canonical NF-κB pathway, but as a RelB-p50 dimer regulated by canonical IκBs, IκBα and IκBε. IκB control of RelB minimized spontaneous maturation but enabled rapid pathogen-responsive maturation. Computational modeling of the NF-κB signaling module identified control points of this unexpected cell-type-specific regulation. Fibroblasts that were engineered accordingly showed DC-like RelB control. Canonical pathway control of RelB regulated pathogen-responsive gene expression programs. This work illustrates the potential utility of systems analyses in guiding the development of combination therapeutics for modulating DC-dependent T cell responses.
Collapse
|
53
|
Zhang X, Beduhn M, Zheng X, Lian D, Chen D, Li R, Siu LKS, Marleau A, French PW, Ichim TE, Min WP. Induction of alloimmune tolerance in heart transplantation through gene silencing of TLR adaptors. Am J Transplant 2012; 12:2675-88. [PMID: 22823145 DOI: 10.1111/j.1600-6143.2012.04196.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Toll-like receptors (TLRs) activate biochemical pathways that evoke activation of innate immunity, which leads to dendritic cell (DC) maturation and initiation of adaptive immune responses that provoke allograft rejection. We aimed to prolong allograft survival by selectively inhibiting expression of the common adaptors of TLR signaling, namely MyD88 and TRIF, using siRNA. In vitro we demonstrated that blocking expression of MyD88 and TRIF led to reduced DC maturation. In vivo treatment of recipients with MyD88 and TRIF siRNA significantly prolonged allograft survival in the BALB/c > C57BL6 cardiac transplant model. Moreover, the combination of MyD88 and TRIF siRNA along with a low dose of rapamycin further extended the allograft survival (88.8 ± 7.1 days). Tissue histopathology demonstrated an overall reduction in lymphocyte interstitium infiltration, vascular obstruction and hemorrhage in mice treated with MyD88 and TRIF siRNA vector plus rapamycin. Furthermore, treatment was associated with an increase in the numbers of CD4(+) CD25(+) FoxP3(+) regulatory T cells and Th2 deviation. To our knowledge, this study is the first demonstration of prolonging the survival of allogeneic heart grafts through gene silencing of TLR signaling adaptors, highlighting the therapeutic potential of siRNA in clinical transplantation.
Collapse
Affiliation(s)
- X Zhang
- Departments of Surgery, Pathology, and Oncology, University of Western Ontario, London, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Moreau A, Varey E, Bériou G, Hill M, Bouchet-Delbos L, Segovia M, Cuturi MC. Tolerogenic dendritic cells and negative vaccination in transplantation: from rodents to clinical trials. Front Immunol 2012; 3:218. [PMID: 22908013 PMCID: PMC3414843 DOI: 10.3389/fimmu.2012.00218] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/06/2012] [Indexed: 12/12/2022] Open
Abstract
The use of immunosuppressive (IS) drugs to treat transplant recipients has markedly reduced the incidence of acute rejection and early graft loss. However, such treatments have numerous adverse side effects and fail to prevent chronic allograft dysfunction. In this context, therapies based on the adoptive transfer of regulatory cells are promising strategies to induce indefinite transplant survival. The use of tolerogenic dendritic cells (DC) has shown great potential, as preliminary experiments in rodents have demonstrated that administration of tolerogenic DC prolongs graft survival. Recipient DC, Donor DC, or Donor Ag-pulsed recipient DC have been used in preclinical studies and administration of these cells with suboptimal immunosuppression increases their tolerogenic potential. We have demonstrated that autologous unpulsed tolerogenic DC injected in the presence of suboptimal immunosuppression are able to induce Ag-specific allograft tolerance. We derived similar tolerogenic DC in different animal models (mice and non-human primates) and confirmed their protective abilities in vitro and in vivo. The mechanisms involved in the tolerance induced by autologous tolerogenic DC were also investigated. With the aim of using autologous DC in kidney transplant patients, we have developed and characterized tolerogenic monocyte-derived DC in humans. In this review, we will discuss the preclinical studies and describe our recent results from the generation and characterization of tolerogenic monocyte-derived DC in humans for a clinical application. We will also discuss the limits and difficulties in translating preclinical experiments to theclinic.
Collapse
|
55
|
Wu W, Shan J, Li Y, Luo L, Sun G, Zhou Y, Yang T, Xia M, Guo Y, Feng L. Adoptive transfusion of tolerance dendritic cells prolongs the survival of cardiac allograft: a systematic review of 44 basic studies in mice. J Evid Based Med 2012; 5:139-53. [PMID: 23672221 DOI: 10.1111/j.1756-5391.2012.01191.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Tolerogenic DCs (Tol-DCs), a group of cells with imDC phenotype, can stably induce T cells low-reactivity and immune tolerance. We systematically reviewed the adoptive transfusion of Tol-DCs induced by different ways to prolong cardiac allograft survival and its possible mechanism. METHOD MEDLINE (1966 to March 2011), EMbase (1980 to March 2011), and ISI (inception to March 2011) were searched for identification of relevant studies. We used allogeneic heart graft survival time as endpoint outcome to analyze the effect of adoptive transfusion of Tol-DC on cardiac allograft. By integrating studies' information, we summarized the mechanisms of Tol-DC in prolonging cardiac grafts. RESULTS Four methods were used to induce Tol-DC in all of the 44 included studies including gene-modified, drug-intervened, cytokine-induced, and other-derived (liver-derived & spleen-derived) DCs. The results showed that all types of Tol-DC can effectively prolong graft survival, and the average extension of graft survival time for each group was as follows: 22.02 ± 21.9 days (3.2 folds to control group) in the gene modified group, 25.94 ± 16.9 days (4.3 folds) in the drug-intervened groups, 9.00 ± 8.13 days (1.9 folds) in the cytokine-induced group, and 10.69 ± 9.94 days (2.1 folds) in the other-derived group. The main mechanisms of Tol-DCs to prolong graft survival were as follows: (1) induceT-cell hyporeactivity (detected by MLR); (2) reduce the effect of cytotoxic lymphocyte (CTL); (3) promote Th2 differentiation; (4) induce Treg; (5) induce chimerism. CONCLUSION For fully MHC mismatched allogeneic heart transplant recipients of inbred mouse, adoptive transfusion of Tol-DC, which can be gene-modified, drug-intervened, cytokine-induced, spleen-derived or liver-derived, can clearly prolong the survival of cardiac allograft or induce immune tolerance. Gene-modified and drug-induced Tol-DC can prolong graft survival most obviously. Having better reliability and stability than drug-induction, gene-modification is the best way to induce Tol-DCs at present. One-time intravenous infusion of 2 × 10(6) Tol-DC is a simple and feasible way to induce long-term graft survival. Multiple infusions will prolong it but increase the risk and cost. Adoptive transfusion of Tol-DC in conjunction with immunosuppressive agents may also prolong the graft survival time.
Collapse
Affiliation(s)
- Wenqiao Wu
- Key Laboratory of Transplant Engineering and Immunology of Health Ministry of China, Regenerative medical research center, West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Zheng X, Koropatnick J, Chen D, Velenosi T, Ling H, Zhang X, Jiang N, Navarro B, Ichim TE, Urquhart B, Min W. Silencing IDO in dendritic cells: a novel approach to enhance cancer immunotherapy in a murine breast cancer model. Int J Cancer 2012; 132:967-77. [PMID: 22870862 DOI: 10.1002/ijc.27710] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 06/14/2012] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapeutic agents (vaccines) in the form of antigen-loaded dendritic cells (DCs) reached an important milestone with the recent approval of Provenge, the first DC vaccine for treatment of prostate cancer. Although this heralds a new era of tumor immunotherapy, it also highlights the compelling need to optimize such DC-based therapies as they are increasingly tested and used to treat human patients. In this study we sought to augment and enhance the antitumor activity of a DC-based vaccine using siRNA to silence expression of immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO) in DCs. We report here that DCs loaded with tumor antigens, but with siRNA-silenced IDO expression, were introduced into 4T1 breast tumor-bearing mice, the treatment: (i) lengthened the time required for tumor onset, (ii) decreased tumor size compared to tumors grown for equal lengths of time in mice treated with antigen-loaded DCs without IDO silencing and (iii) reduced CD4(+) and CD8(+) T cell apoptosis. Furthermore, immunization with IDO-silenced DCs enhanced tumor antigen-specific T cell proliferation and CTL activity, and decreased numbers of CD4(+) CD25(+) Foxp3(+) T(reg). This study provides evidence to support silencing of immunosuppressive genes (IDO) as an effective strategy to enhance the efficacy of DC-based cancer immunotherapeutic.
Collapse
Affiliation(s)
- Xiufen Zheng
- Department of Surgery, University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Benson KF, Redman KA, Carter SG, Keller D, Farmer S, Endres JR, Jensen GS. Probiotic metabolites from Bacillus coagulans GanedenBC30 TM support maturation of antigen-presenting cells in vitro. World J Gastroenterol 2012; 18:1875-83. [PMID: 22563167 PMCID: PMC3337562 DOI: 10.3748/wjg.v18.i16.1875] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 12/20/2011] [Accepted: 04/01/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the effects of probiotic metabolites on maturation stage of antigen-presenting immune cells.
METHODS: Ganeden Bacillus coagulans 30 (GBC30) bacterial cultures in log phase were used to isolate the secreted metabolite (MET) fraction. A second fraction was made to generate a crude cell-wall-enriched fraction, by centrifugation and lysis, followed by washing. A preparation of MET was subjected to size exclusion centrifugation, generating three fractions: < 3 kDa, 3-30 kDa, and 30-200 kDa and activities were tested in comparison to crude MET and cell wall in primary cultures of human peripheral blood mononuclear cell (PBMC) as a source of antigen-presenting mononuclear phagocytes. The maturation status of mononuclear phagocytes was evaluated by staining with monoclonal antibodies towards CD14, CD16, CD80 and CD86 and analyzed by flow cytometry.
RESULTS: Treatment of PBMC with MET supported maturation of mononuclear phagocytes toward both macrophage and dendritic cell phenotypes. The biological activity unique to the metabolites included a reduction of CD14+ CD16+ pro-inflammatory cells, and this property was associated with the high molecular weight metabolite fraction. Changes were also seen for the dendritic cell maturation markers CD80 and CD86. On CD14dim cells, an increase in both CD80 and CD86 expression was seen, in contrast to a selective increase in CD86 expression on CD14bright cells. The co-expression of CD80 and CD86 indicates effective antigen presentation to T cells and support of T helper cell differentiation. The selective expression of CD86 in the absence of CD80 points to a role in generating T regulatory cells.
CONCLUSION: The data show that a primary mechanism of action of GBC30 metabolites involves support of more mature phenotypes of antigen-presenting cells, important for immunological decision-making.
Collapse
|
58
|
Regulation of dendritic cell development by GM-CSF: molecular control and implications for immune homeostasis and therapy. Blood 2012; 119:3383-93. [PMID: 22323450 DOI: 10.1182/blood-2011-11-370130] [Citation(s) in RCA: 256] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Dendritic cells (DCs) represent a small and heterogeneous fraction of the hematopoietic system, specialized in antigen capture, processing, and presentation. The different DC subsets act as sentinels throughout the body and perform a key role in the induction of immunogenic as well as tolerogenic immune responses. Because of their limited lifespan, continuous replenishment of DC is required. Whereas the importance of GM-CSF in regulating DC homeostasis has long been underestimated, this cytokine is currently considered a critical factor for DC development under both steady-state and inflammatory conditions. Regulation of cellular actions by GM-CSF depends on the activation of intracellular signaling modules, including JAK/STAT, MAPK, PI3K, and canonical NF-κB. By directing the activity of transcription factors and other cellular effector proteins, these pathways influence differentiation, survival and/or proliferation of uncommitted hematopoietic progenitors, and DC subset–specific precursors, thereby contributing to specific aspects of DC subset development. The specific intracellular events resulting from GM-CSF–induced signaling provide a molecular explanation for GM-CSF–dependent subset distribution as well as clues to the specific characteristics and functions of GM-CSF–differentiated DCs compared with DCs generated by fms-related tyrosine kinase 3 ligand. This knowledge can be used to identify therapeutic targets to improve GM-CSF–dependent DC-based strategies to regulate immunity.
Collapse
|
59
|
Rojas-Canales D, Krishnan R, Jessup CF, Coates PT. Early exposure of interferon-γ inhibits signal transducer and activator of transcription-6 signalling and nuclear factor κB activation in a short-term monocyte-derived dendritic cell culture promoting 'FAST' regulatory dendritic cells. Clin Exp Immunol 2012; 167:447-58. [PMID: 22288588 DOI: 10.1111/j.1365-2249.2011.04537.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Interferon (IFN)-γ is a cytokine with immunomodulatory properties, which has been shown previously to enhance the generation of tolerogenic dendritic cells (DC) when administered early ex vivo in 7-day monocyte-derived DC culture. To generate tolerogenic DC rapidly within 48 h, human monocytes were cultured for 24 h with interleukin (IL)-4 and granulocyte-macrophage colony-stimulating factor (GM-CSF) in the presence (IFN-γ-DC) or absence of IFN-γ (500 U/ml) (UT-DC). DC were matured for 24 h with TNF-α and prostaglandin E(2) (PGE(2) ). DC phenotype, signal transducer and activator of transcription-6 (STAT-6) phosphorylation and promotion of CD4(+) CD25(+) CD127(neg/low) forkhead box P3 (FoxP3)(hi) T cells were analysed by flow cytometry. DC nuclear factor (NF)-κB transcription factor reticuloendotheliosis viral oncogene homologue B (RELB) and IL-12p70 protein expression were also determined. Phenotypically, IFN-γ-DC displayed reduced DC maturation marker CD83 by 62% and co-stimulation molecules CD80 (26%) and CD86 (8%). IFN-γ treatment of monocytes inhibited intracellular STAT6, RELB nuclear translocation and IL-12p70 production. IFN-γ-DC increased the proportion of CD4(+) CD25(+) CD127(neg/low) foxp3(hi) T cells compared to UT-DC from 12 to 23%. IFN-γ-DC primed T cells inhibited antigen-specific, autologous naive T cell proliferation by 70% at a 1:1 naive T cells to IFN-γ-DC primed T cell ratio in suppression assays. In addition, we examined the reported paradoxical proinflammatory effects of IFN-γ and confirmed in this system that late IFN-γ exposure does not inhibit DC maturation marker expression. Early IFN-γ exposure is critical in promoting the generation of regulatory DC. Early IFN-γ modulated DC generated in 48 h are maturation arrested and promote the generation of antigen-specific regulatory T cells, which may be clinically applicable as a novel cellular therapy for allograft rejection.
Collapse
Affiliation(s)
- D Rojas-Canales
- Renal and Transplantation Immunobiology Laboratory, Hanson Institute, Royal Adelaide Hospital, Adelaide, SA, Australia
| | | | | | | |
Collapse
|
60
|
Li R, Zheng X, Popov I, Zhang X, Wang H, Suzuki M, Necochea-Campion RD, French PW, Chen D, Siu L, Koos D, Inman RD, Min WP. Gene silencing of IL-12 in dendritic cells inhibits autoimmune arthritis. J Transl Med 2012; 10:19. [PMID: 22289162 PMCID: PMC3293054 DOI: 10.1186/1479-5876-10-19] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/31/2012] [Indexed: 11/18/2022] Open
Abstract
Background We have previously demonstrated that immune modulation can be accomplished by administration of gene silenced dendritic cells (DC) using siRNA. In this study, we demonstrate the therapeutic utilization of shRNA-modified DC as an antigen-specific tolerogenic vaccine strategy for autoimmune arthritis. Methods A shRNA that specifically targets IL-12 p35 was designed and cloned into a plasmid vectors (IL-12 shRNA). Bone marrow-derived DC from DBA/1 mice were transfected with the IL-12 shRNA construct in vitro. Mice with collagen II (CII)-induced arthritis (CIA) were treated with the modified DCs expressing the shRNA. Recall response and disease progression were assessed. Results After gene silencing of IL-12 in DC, DC were shown to selectively inhibit T cell proliferation on recall responses and in an MLR. In murine CIA, we demonstrated that administration of IL-12 shRNA-expressing DC that were pulsed with CII inhibited progression of arthritis. The therapeutic effects were evidenced by decreased clinical scores, inhibition of inflammatory cell infiltration in the joint, and suppression of T cell and B cell responses to CII. Conclusion We demonstrate a novel tolerance-inducing protocol for the treatment of autoimmune inflammatory joint disease in which the target antigen is known, utilizing DNA-directed RNA interference.
Collapse
Affiliation(s)
- Rong Li
- Institute of Immunomodulation and Immunotherapy, Nanchang University Medical School, Nanchang, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Qiu T, Zhu HC, Liu XH, Dong WC, Weng XD, Hu CH, Kuang YL, Gao RH, Dan C, Tao T. Lentiviral-mediated shRNA against RelB induces the generation of tolerogenic dendritic cells. Int Immunopharmacol 2012; 12:501-9. [PMID: 22266276 DOI: 10.1016/j.intimp.2012.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 01/09/2012] [Accepted: 01/09/2012] [Indexed: 10/14/2022]
Abstract
OBJECTIVE Lentiviral-mediated shRNA against RelB was used to produce tolerogenic dendritic cells from murine bone marrow derived dendritic cells (BMDCs). METHOD RelB expression in the BMDCs was silenced by lentivirus carrying RelB shRNA. The apoptosis rate and surface markers of DCs were assessed by flow cytometry. IL-12,IL-10,TGF-β1 secreted by DCs and DNA binding capacity of NF-κB subunits in the nucleus were measured by ELISA, independently. MLR was used to analyze the capacity of DCs to inhibit immune response. RESULTS RelB expression was significantly inhibited in DCs following lentiviral mediated delivery of RelB specific shRNA. The RelB shRNA-DC produced lower IL-12 and higher IL-10 than mature dendritic cells (mDCs) and silencing control DCs. There was no difference in the apoptosis rate between shRNA RelB-DCs and mDCs. The expression levels of co-stimulatory molecules (CD80, CD86 and CD83) and MHC-II class molecule were lower in the RelB shRNA-DCs than in the mDCs and silencing control DCs. In addition, RelB shRNA also inhibited the RelB DNA binding capacity but had no effect on other NF-κB subunits. The shRNA RelB-DCs can significantly inhibit mixed lymphocyte reaction (MLR) and down-regulate Th1 cytokines and prompt the production of Th2 cytokines. CONCLUSION Our results indicate RelB shRNA transfection of DCs can induce the immature status, and produce tolerogenic DCs.
Collapse
Affiliation(s)
- Tao Qiu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Luo L, Li C, Wu W, Lu J, Shan J, Li S, Long D, Guo Y, Feng L, Li Y. Dendritic cells transduced with lentiviral vector targeting RelB gene using RNA interference induce hyporesponsiveness in memory CD4+ T cells and naïve CD4+ T cells. Cell Immunol 2012; 273:85-93. [DOI: 10.1016/j.cellimm.2011.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 11/03/2011] [Indexed: 10/15/2022]
|
63
|
Guindi C, Ménard M, Cloutier A, Gaudreau S, Besin G, Larivée P, McDonald PP, Dupuis G, Amrani A. Differential role of NF-κB, ERK1/2 and AP-1 in modulating the immunoregulatory functions of bone marrow-derived dendritic cells from NOD mice. Cell Immunol 2011; 272:259-68. [PMID: 22070873 DOI: 10.1016/j.cellimm.2011.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Revised: 09/29/2011] [Accepted: 10/05/2011] [Indexed: 10/16/2022]
Abstract
Tolerogenic dendritic cells represent a promising immunotherapy in autoimmunity. However, the molecular mechanisms that drive tolerogenic DCs functions are not well understood. We used GM-CSF or GM-CSF+IL-4 to generate tolerogenic (GM/DCs) and immunogenic (IL-4/DCs) BMDCs from NOD mice, respectively. GM/DCs were resistant to maturation, produced large amounts of IL-10 but not IL-12p70. GM/DCs displayed a reduced capacity to activate diabetogenic CD8(+) T-cells and were efficient to induce Tregs expansion and conversion. LPS stimulation triggered ERK1/2 activation that was sustained in GM/DCs but not in IL-4/DCs. ERK1/2 and AP-1 were involved in IL-10 production in GM/DCs but not in their resistance to maturation. Supershift analysis showed that NF-κB DNA binding complex contains p52 and p65 in GM/DCs, whereas it contains p52, p65 and RelB in IL-4/DCs. ChIP experiments revealed that p65 was recruited to IL-10 promoter following LPS stimulation of GM/DCs whereas its binding to IL-12p35 promoter was abolished. Our results suggest that immunoregulatory functions of GM/DCs are differentially regulated by ERK1/2, AP-1 and NF-κB pathways.
Collapse
Affiliation(s)
- Chantal Guindi
- Immunology Division, and Centre de Recherche Clinique Etienne LeBel, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Li C, Luo L, Lu J, Feng L, Shan J, Long D, Guo Y, Wu W, Li S, Li Y. A modified splint tubing technique for heterotopic heart transplantation in mouse. Transpl Immunol 2011; 25:82-7. [DOI: 10.1016/j.trim.2011.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 03/31/2011] [Accepted: 03/31/2011] [Indexed: 10/18/2022]
|
65
|
Astor TL. RNA interference, RSV, and lung transplantation: a promising future for siRNA therapeutics. Am J Respir Crit Care Med 2011; 183:427-8. [PMID: 21325081 DOI: 10.1164/rccm.201009-1488ed] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
66
|
Shan J, Feng L, Luo L, Wu W, Li C, Li S, Li Y. MicroRNAs: potential biomarker in organ transplantation. Transpl Immunol 2011; 24:210-5. [PMID: 21459143 DOI: 10.1016/j.trim.2011.03.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/10/2011] [Accepted: 03/26/2011] [Indexed: 02/05/2023]
Abstract
MicroRNAs (miRs) are non-coding RNAs that could regulate gene expression at the posttranscriptional level, and have been indicated to be involved in diverse biological processes. They are emerging as master regulator of immune response and may likely play a key role in transplant rejection process. The extensive and comprehensive use of miR microarrays has enabled the identification of miRs as potential biomarkers for transplantation; many miRs have been reported associated with transplant rejection. Here we reviewed the emerging data on transplant recipients' miRs expression pattern, and discussed the possible mechanism of how miRs regulate transplant immune response.
Collapse
Affiliation(s)
- Juan Shan
- Key Laboratory of Transplant Engineering and Immunology of Health Ministry of China, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China
| | | | | | | | | | | | | |
Collapse
|
67
|
Zhang ZX, Lian D, Huang X, Wang S, Sun H, Liu W, Garcia B, Min WP, Jevnikar AM. Adoptive transfer of DNT cells induces long-term cardiac allograft survival and augments recipient CD4(+)Foxp3(+) Treg cell accumulation. Transpl Immunol 2011; 24:119-126. [PMID: 21073952 DOI: 10.1016/j.trim.2010.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Revised: 11/03/2010] [Accepted: 11/04/2010] [Indexed: 01/26/2023]
Abstract
Regulatory T (Treg) cells play an important role in the regulation of immune responses but whether Treg will induce tolerance in transplant recipients in the clinic remains unknown. Our previous studies have shown that TCRαβ(+)CD3(+)CD4⁻CD8⁻NK1.1⁻ (double negative, DN) T cells suppress T cell responses and prolong allograft survival in a single locus MHC-mismatched mouse model. In this study, we investigated the role of DNT cells in a more robust, fully MHC-mismatched BALB/c to C57BL/6 transplantation model, which may be more clinically relevant. Adoptive transfer of DNT cells in combination with short-term rapamycin treatment (days 1-9) induced long-term heart allograft survival (101±31 vs. 39±13 days rapamycin alone, p<0.01). Furthermore adoptive transfer DNT cells augmented CD4+Foxp3+ Treg cells accumulation in transplant recipients while depletion of CD4(+) Treg cells by anti-CD25 inhibited the effect of DNT cells on long-term graft survival (48±12 days vs. 101±31 days, p<0.001). In conclusion, DNT cells combined with short-term immunosuppression can prolong allograft survival, which may be through the accumulation of CD4(+)Foxp3(+) Treg cells in the recipient. Our result suggests that allograft tolerance may require the co-existence of different type Treg cell phenotypes which are affected by current immunosuppression.
Collapse
Affiliation(s)
- Zhu-Xu Zhang
- The Multi-Organ Transplant Program, London Health Sciences Centre, London, Ontario, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Huang YL, Wang YZ, Chen JB, Wang F, Kang XP, Xia JJ, Lan TS, Xie BY, Ekberg H, Wang XM, Qi ZQ. Prevention of Acute and Chronic Allograft Rejection by Combinations of Tolerogenic Dendritic Cells. Scand J Immunol 2011; 73:91-101. [DOI: 10.1111/j.1365-3083.2010.02485.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
69
|
Bertin-Maghit S, Pang D, O'Sullivan B, Best S, Duggan E, Paul S, Thomas H, Kay TW, Harrison LC, Steptoe R, Thomas R. Interleukin-1β produced in response to islet autoantigen presentation differentiates T-helper 17 cells at the expense of regulatory T-cells: Implications for the timing of tolerizing immunotherapy. Diabetes 2011; 60:248-57. [PMID: 20980463 PMCID: PMC3012178 DOI: 10.2337/db10-0104] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The effectiveness of tolerizing immunotherapeutic strategies, such as anti-CD40L or dendritic cells (DCs), is greater when administered to young nonobese diabetic (NOD) mice than at peak insulitis. RelB(lo) DCs, generated in the presence of an nuclear factor-κB inhibitor, induce T-regulatory (Treg) cells and suppress inflammation in a model of rheumatoid arthritis. Interleukin (IL)-1β is overexpressed in humans and mice at risk of type 1 diabetes, dysregulates Treg cells, and accelerates diabetes in NOD mice. We investigated the relationship between IL-1β production and the response to RelB(lo) DCs in the prediabetic period. RESEARCH DESIGN AND METHODS We injected RelB(lo) DCs subcutaneously into 4- or 14-week-old NOD mice and tracked the incidence of diabetes and effect on Treg cell function. We measured the expression of proinflammatory cytokines by stimulated splenocytes and unstimulated islets from mice of different ages and strains and proliferative and cytokine responses of T effectors to Treg in vitro. RESULTS Tolerizing RelB(lo) DCs significantly inhibited diabetes progression when administered to 4-week-old but not 14-week-old mice. IL-1β production by NOD splenocytes and mRNA expression by islets increased from 6 to 16 weeks of age when major histocompatibility complex (MHC)-restricted islet antigen presentation to autoreactive T-cells occurred. IL-1 reduced the capacity of Treg cells to suppress effector cells and promoted their conversion to Th17 cells. RelB(lo) DCs exacerbated the IL-1-dependent decline in Treg function and promoted Th17 conversion. CONCLUSIONS IL-1β, generated by islet-autoreactive cells in MHC-susceptible mice, accelerates diabetes by differentiating Th17 at the expense of Treg. Tolerizing DC therapies can regulate islet autoantigen priming and prevent diabetes, but progression past the IL-1β/IL-17 checkpoint signals the need for other strategies.
Collapse
Affiliation(s)
- Sebastien Bertin-Maghit
- The University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Dimeng Pang
- The University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Brendan O'Sullivan
- The University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Shannon Best
- The University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Emily Duggan
- The University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Sanjoy Paul
- Queensland Clinical Trials and Biostatistics Centre, School of Population Health, The University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Helen Thomas
- Islet Biology Laboratory, St. Vincent's Institute, Melbourne, Australia
| | - Thomas W.H. Kay
- Autoimmunity and Transplantation Division, Walter and Eliza Hall Institute, Melbourne, Australia
| | - Leonard C. Harrison
- Autoimmunity and Transplantation Division, Walter and Eliza Hall Institute, Melbourne, Australia
| | - Raymond Steptoe
- The University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- Corresponding author: Ranjeny Thomas,
| |
Collapse
|
70
|
Li F, Mahato RI. RNA interference for improving the outcome of islet transplantation. Adv Drug Deliv Rev 2011; 63:47-68. [PMID: 21156190 PMCID: PMC3065652 DOI: 10.1016/j.addr.2010.11.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 11/19/2010] [Accepted: 11/25/2010] [Indexed: 01/06/2023]
Abstract
Islet transplantation has the potential to cure type 1 diabetes. Despite recent therapeutic success, it is still not common because a large number of transplanted islets get damaged by multiple challenges including instant blood mediated inflammatory reaction, hypoxia/reperfusion injury, inflammatory cytokines, and immune rejection. RNA interference (RNAi) is a novel strategy to selectively degrade target mRNA. The use of RNAi technologies to downregulate the expression of harmful genes has the potential to improve the outcome of islet transplantation. The aim of this review is to gain a thorough understanding of biological obstacles to islet transplantation and discuss how to overcome these barriers using different RNAi technologies. This eventually will help improve islet survival and function post transplantation. Chemically synthesized small interferring RNA (siRNA), vector based short hairpin RNA (shRNA), and their critical design elements (such as sequences, promoters, and backbone) are discussed. The application of combinatorial RNAi in islet transplantation is also discussed. Last but not the least, several delivery strategies for enhanced gene silencing are discussed, including chemical modification of siRNA, complex formation, bioconjugation, and viral vectors.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38103
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38103
| |
Collapse
|
71
|
Critical role of transcription factor PU.1 in the expression of CD80 and CD86 on dendritic cells. Blood 2010; 117:2211-22. [PMID: 21119111 DOI: 10.1182/blood-2010-06-291898] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In this study, we investigated the role of a transcription factor, PU.1, in the regulation of CD80 and CD86 expression in dendritic cells (DCs). A chromatin immunoprecipitation assay revealed that PU.1 is constitutively bound to the CD80 and CD86 promoters in bone marrow-derived DCs. In addition, co-expression of PU.1 resulted in the transactivation of the CD80 and CD86 promoters in a reporter assay. The binding of PU.1 to cis-enhancing regions was confirmed by electromobility gel-shift assay. As expected, inhibition of PU.1 expression by short interfering RNA (siRNA) in bone marrow-derived DCs resulted in marked down-regulation of CD80 and CD86 expression. Moreover, overexpression of PU.1 in murine bone marrow-derived lineage-negative cells induced the expression of CD80 and CD86 in the absence of monocyte/DC-related growth factors and/or cytokines. Based on these results, we conclude that PU.1 is a critical factor for the expression of CD80 and CD86. We also found that subcutaneous injection of PU.1 siRNA or topical application of a cream-emulsified PU.1 siRNA efficiently inhibited murine contact hypersensitivity. Our results suggest that PU.1 is a potential target for the treatment of immune-related diseases.
Collapse
|
72
|
van de Laar L, van den Bosch A, van der Kooij SW, Janssen HLA, Coffer PJ, van Kooten C, Woltman AM. A nonredundant role for canonical NF-κB in human myeloid dendritic cell development and function. THE JOURNAL OF IMMUNOLOGY 2010; 185:7252-61. [PMID: 21076069 DOI: 10.4049/jimmunol.1000672] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The plastic role of dendritic cells (DCs) in the regulation of immune responses has made them interesting targets for immunotherapy, but also for pathogens or tumors to evade immunity. Functional alterations of DCs are often ascribed to manipulation of canonical NF-κB activity. However, though this pathway has been linked to murine myeloid DC biology, a detailed analysis of its importance in human myeloid DC differentiation, survival, maturation, and function is lacking. The myeloid DC subsets include interstitial DCs and Langerhans cells. In this study, we investigated the role of canonical NF-κB in human myeloid DCs generated from monocytes (monocyte-derived DCs [mo-DCs]) or CD34(+) progenitors (CD34-derived myeloid DCs [CD34-mDCs]). Inhibition of NF-κB activation during and after mo-DC, CD34-interstitial DC, or CD34-Langerhans cell differentiation resulted in apoptosis induction associated with caspase 3 activation and loss of mitochondrial transmembrane potential. Besides regulating survival, canonical NF-κB activity was required for the acquisition of a DC phenotype. Despite phenotypic differences, however, Ag uptake, costimulatory molecule and CCR7 expression, as well as T cell stimulatory capacity of cells generated under NF-κB inhibition were comparable to control DCs, indicating that canonical NF-κB activity during differentiation is redundant for the development of functional APCs. However, both mo-DC and CD34-mDC functionality were reduced by NF-κB inhibition during activation. In conclusion, canonical NF-κB activity is essential for the development and function of mo-DCs as well as CD34-mDCs. Insight into the role of this pathway may help in understanding how pathogens and tumors escape immunity and aid in developing novel treatment strategies aiming to interfere with human immune responses.
Collapse
Affiliation(s)
- Lianne van de Laar
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
73
|
Bros M, Dexheimer N, Besche V, Masri J, Trojandt S, Hövelmeyer N, Reissig S, Massoumi R, Grabbe S, Waisman A, Reske-Kunz AB. Mutated cylindromatosis gene affects the functional state of dendritic cells. Eur J Immunol 2010; 40:2848-57. [DOI: 10.1002/eji.200939285] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
74
|
Thomas R. The balancing act of autoimmunity: central and peripheral tolerance versus infection control. Int Rev Immunol 2010; 29:211-33. [PMID: 20367141 DOI: 10.3109/08830180903434219] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Genetic associations with autoimmune disease are enriched in immune response regulators. The immune system in individuals at genetic risk of autoimmunity must balance pressures on the innate and adaptive immune system, most notably infection control, with those of maintaining self-tolerance or controlling autoimmune inflammation. In spite of multiple tolerance mechanisms, inflammation becomes chronic in autoimmune disease, and complete resolution is difficult. This article proposes a perspective on the pathogenesis of autoimmunity-focusing on rheumatoid arthritis and type 1 diabetes-integrating clinical advances and animal models with the role that colonizing micro-organisms play in the balance between tolerance and autoimmunity.
Collapse
Affiliation(s)
- Ranjeny Thomas
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, University of Queensland, Princess Alexandra Hospital, Australia.
| |
Collapse
|
75
|
Zheng X, Suzuki M, Ichim TE, Zhang X, Sun H, Zhu F, Shunnar A, Garcia B, Inman RD, Min W. Treatment of autoimmune arthritis using RNA interference-modulated dendritic cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:6457-64. [PMID: 20435931 DOI: 10.4049/jimmunol.0901717] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Dendritic cells (DCs) have a dual ability to either stimulate or suppress immunity, which is primarily associated with the expression of costimulatory molecules. Ag-loaded DCs have shown encouraging clinical results for treating cancer and infectious diseases; however, the use of these cells as a means of suppressing immune responses is only recently being explored. Here, we describe the induction of RNA interference through administering short interfering RNA (siRNA) as a means of specifically generating tolerogenic DCs. Knockdown of CD40, CD80, and CD86, prior to loading DCs with the arthritogenic Ag collagen II, led to a population of cells that could effectively suppress onset of collagen-induced arthritis. Maximum benefits were observed when all three genes were concurrently silenced. Disease suppression was associated with inhibition of collagen II-specific Ab production and suppression of T cell recall responses. Downregulation of IL-2, IFN-gamma, TNF-alpha, and IL-17 and increased FoxP3(+) cells with regulatory activity were observed in collagen-induced arthritis mice treated with siRNA-transfected DCs. Collectively, these data support the use of ex vivo gene manipulation in DCs using siRNA to generate tailor-made tolerogenic vaccines for treating autoimmunity.
Collapse
Affiliation(s)
- Xiufen Zheng
- Department of Surgery, University of Western Ontario, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Fang F, Wang Y, Li R, Zhao Y, Guo Y, Jiang M, Sun J, Ma Y, Ren Z, Tian Z, Wei F, Yang D, Xiao W. Transcription factor E2F1 suppresses dendritic cell maturation. THE JOURNAL OF IMMUNOLOGY 2010; 184:6084-91. [PMID: 20421650 DOI: 10.4049/jimmunol.0902561] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Transcription factor E2F1 has been largely studied as a promoter of S-phase transition in the cell cycle and as a regulator of apoptosis. Recently, E2F1 has been shown to regulate a wide range of genes in response to inflammatory stimulation of macrophages and to contribute to T cell activation in response to pathogens, implicating an extensive immunological role for E2F1. Dendritic cells (DCs) play critical roles as professional APCs in the development of immune responses. However, it is unclear whether E2F1 has any effect on DC phenotype or function. In this paper, we report that E2F1 acts as a suppressor of DC maturation. The level of E2F1 expression was transiently downregulated in the course of LPS-induced maturation of both human monocyte-derived DCs and a mouse DC cell line, DC2.4. Knockdown of E2F1 by small interfering RNA in DC2.4 cells resulted in both phenotypic and functional maturation, even without LPS treatment. Conversely, ectopic overexpression of E2F1 suppressed LPS-induced maturation of DC2.4 cells. Furthermore, knockdown of E2F1 caused the activation of several major signaling pathways known to be activated in the course of DC maturation, including Erk1/2, NF-kappaB, and PI3K/Akt, suggesting that E2F1 may be involved in regulating multiple signaling pathways in DCs. Finally, the alteration of phenotypic maturation by E2F1 was confirmed with bone marrow-derived DCs from E2F1 knockout mice. Overall, our data demonstrate for the first time that E2F1 is a critical regulator of DC maturation.
Collapse
Affiliation(s)
- Fang Fang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Hilkens CMU, Isaacs JD, Thomson AW. Development of dendritic cell-based immunotherapy for autoimmunity. Int Rev Immunol 2010; 29:156-83. [PMID: 20199240 DOI: 10.3109/08830180903281193] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Dendritic cells are professional antigen-presenting cells that maintain immune tolerance to self-antigens by deleting or controlling the pathogenicity of auto-reactive T-cells. Dendritic cell-based immunotherapies show great promise for the restoration of tolerance in autoimmune disease. Dendritic cells can be modified ex vivo to induce stable tolerogenic function and be used as cellular 'vaccines' or they can be targeted in vivo with sophisticated antigen delivery systems. Tolerogenic dendritic cells induce antigen-specific T-cell tolerance in vivo and have therapeutic effects in animal models of autoimmunity. The current challenge is to bring tolerogenic dendritic cell therapy to the clinic.
Collapse
Affiliation(s)
- Catharien M U Hilkens
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | | | | |
Collapse
|
78
|
Zheng X, Suzuki M, Zhang X, Ichim TE, Zhu F, Ling H, Shunnar A, Wang MH, Garcia B, Inman RD, Min WP. RNAi-mediated CD40-CD154 interruption promotes tolerance in autoimmune arthritis. Arthritis Res Ther 2010; 12:R13. [PMID: 20102615 PMCID: PMC2875641 DOI: 10.1186/ar2914] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 12/08/2009] [Accepted: 01/26/2010] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION We have previously demonstrated that ex vivo inhibition of costimulatory molecules on antigen-pulsed dendritic cells (DCs) can be useful for induction of antigen-specific immune deviation and suppression of autoimmune arthritis in the collagen induced arthritis (CIA) model. The current study evaluated a practical method of immune modulation through temporary systemic inhibition of the costimulatory molecule CD40. METHODS Mice with collagen II (CII)-induced arthritis (CIA) were administered siRNA targeting the CD40 molecule. Therapeutic effects were evaluated by clinical symptoms, histopathology, Ag-specific T cell and B cell immune responses. RESULTS Systemic administration of CD40-targeting siRNA can inhibit antigen-specific T cell response to collagen II, as well as prevent pathogenesis of disease in both a pre- and post-immunization manner in the CIA model. Disease amelioration was associated with suppression of Th1 cytokines, attenuation of antibody production, and upregulation of T regulatory cells. CONCLUSIONS These studies support the feasibility of transient gene silencing at a systemic level as a mechanism of resetting autoreactive immunity.
Collapse
Affiliation(s)
- Xiufen Zheng
- Departments of Surgery, Pathology, Microbiology and Immunology, University of Western Ontario, 1393 Western Road, London, Ontario, N6G 1G9, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Suzuki M, Zheng X, Zhang X, Ichim TE, Beduhn ME, Min W. Oligonucleotide based-strategies for allergy with special reference to siRNA. Expert Opin Biol Ther 2010; 9:441-50. [PMID: 19344281 DOI: 10.1517/14712590902841924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Allergic diseases are a significant global health care problem. Current pharmacological approaches address symptoms but do not alter the underlying immune dysregulation. Current allergen-specific immunotherapy has several drawbacks. Therefore, approaches that attenuate allergic responses safely and effectively at the level of upstream causative events are desirable. Oligonuleotide-based therapies [CpG DNA, antisense oligonucleotides, and small interfering RNA (siRNA)] are promising approaches. OBJECTIVE/METHODS We review developments in oligonucleotide-based therapies and the potential of siRNA for treating allergy. RESULTS/CONCLUSIONS Strategies with oligonucleotides basically aim to reduce T helper type 2 (Th2) responses. It is controversial whether the reduction of Th2 responses does, in fact, attenuate allergic diseases. Increased understanding of allergic mechanisms will enhance the efficacy of oligonucleotide-based therapy.
Collapse
Affiliation(s)
- Motohiko Suzuki
- University of Western Ontario, University Hospital C9 - 136, Department of Surgery, London, Ontario, N6A 5A5, Canada
| | | | | | | | | | | |
Collapse
|
80
|
Zheng X, Vladau C, Shunner A, Min WP. siRNA specific delivery system for targeting dendritic cells. Methods Mol Biol 2010; 623:173-188. [PMID: 20217551 DOI: 10.1007/978-1-60761-588-0_11] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
siRNA therapy offers immense potential for clinical application. Under physiological conditions, however, siRNA was demonstrated to have a short half-life. Additionally, it may also cause ubiquitous gene silencing as it does not possess a tissue-specific homing mechanism. Thus, the rate-limiting step in the emergence of siRNA as a potential therapeutic agent is the current lack of a safe and tissue- or cell-specific in vivo delivery system. Herein, we propose a novel, cell-specific method for the in vivo delivery of siRNA to dendritic cells (DCs) with the purpose of inducing immune modulation. CD40 siRNA was incorporated within the interior of 86 nm liposomes, which were decorated with surface-bound mAb NLDC-145 as a targeting mechanism. The siRNA encapsulation efficiency was determined to be approximately 7%. CD40 siRNA immunoliposomes (CD40 siILs) were able to specifically bind to DCs and silence CD40 expression in vitro. Furthermore, in vitro CD40-silenced DCs significantly inhibited the proliferation of alloreactive T cells in an MLR. Upon in vivo administration, siIL-encapsulated, Cy3-labeled siRNA exhibited moderate uptake by the liver at an early time point following administration with greater accumulation in the spleen at a later time point. In contrast, naked siRNA primarily accumulated in the kidney immediately after administration and circulated out in a short time period. To address in vivo gene silencing and immune modulation, mice were simultaneously immunized with KLH and subcutaneously injected with DC-specific CD40 siILs, siILs containing negative control siRNA, naked CD40 siRNA, or PBS. A second injection of CD40 siILs, or control treatments, followed 24 h later. Flow cytometry, reverse transcriptase PCR, and quantitative real-time PCR analysis of CD11c(+) DCs from mice treated with CD40 siILs demonstrated reduced expression of CD40, in comparison with control groups. CD11c(-) cells were also analyzed by flow cytometry, but no differences were observed between treatment groups. Furthermore, CD40 siIL-treated mice were found to have an increased proportion of Treg cells (CD4(+)CD25(+) FoxP3(+)), and DCs cells from these mice were able to inhibit T cell proliferation in an antigen-specific recall response. In summary, CD40 siILs were shown to specifically target and deliver CD40 siRNA to DCs, significantly reducing CD40 expression and resulting in DC-mediated immune modulation as well as generation of Treg cells. These findings highlight the therapeutic potential for siRNA-based and DC-mediated immunotherapy in the clinic. To the best of our knowledge, this is the first study to use siILs for targeted delivery of siRNA to DCs and for immune modulation.
Collapse
Affiliation(s)
- Xiufen Zheng
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | | | |
Collapse
|
81
|
Bortezomib Can Suppress Activation of Rapamycin-Resistant Memory T Cells Without Affecting Regulatory T-Cell Viability in Non-Human Primates. Transplantation 2009; 88:1349-59. [DOI: 10.1097/tp.0b013e3181bd7b3a] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
82
|
Zaborsky N, Brunner M, Wallner M, Himly M, Karl T, Schwarzenbacher R, Ferreira F, Achatz G. Antigen aggregation decides the fate of the allergic immune response. THE JOURNAL OF IMMUNOLOGY 2009; 184:725-35. [PMID: 19995902 DOI: 10.4049/jimmunol.0902080] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Previously, defined naturally occurring isoforms of allergenic proteins were classified as hypoallergens and therefore suggested as an agent for immunotherapy in the future. In this paper, we report for the first time the molecular background of hypoallergenicity by comparing the immunological behavior of hyperallergenic Betula verrucosa major Ag 1a (Bet v 1a) and hypoallergenic Bet v 1d, two isoforms of the major birch pollen allergen Betula verrucosa 1. Despite their cross-reactivity, Bet v 1a and Bet v 1d differ in their capacity to induce protective Ab responses in BALB/c mice. Both isoforms induced similar specific IgE levels, but only Bet v 1d expressed relevant titers of serum IgGs and IgAs. Interestingly, hypoallergenic Bet v 1d activated dendritic cells more efficiently, followed by the production of increased amounts of Th1- as well as Th2-type cytokines. Surprisingly, compared with Bet v 1a, Bet v 1d-immunized mice showed a decreased proliferation of regulatory T cells. Crystallographic studies and dynamic light scattering revealed that Bet v 1d demonstrated a high tendency to form disulfide-linked aggregates due to a serine to cysteine exchange at residue 113. We conclude that aggregation of Bet v 1d triggers the establishment of a protective Ab titer and supports a rationale for Bet v 1d being a promising candidate for specific immunotherapy of birch pollen allergy.
Collapse
Affiliation(s)
- Nadja Zaborsky
- Department of Molecular Biology, Christian Doppler Laboratory for Allergy Diagnosis and Therapy, University of Salzburg, Salzburg, Austria
| | | | | | | | | | | | | | | |
Collapse
|
83
|
The RNA binding protein tristetraprolin influences the activation state of murine dendritic cells. Mol Immunol 2009; 47:1161-70. [PMID: 19945750 DOI: 10.1016/j.molimm.2009.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 09/15/2009] [Accepted: 11/03/2009] [Indexed: 01/07/2023]
Abstract
Dendritic cells (DCs) serve to maintain peripheral tolerance under steady state conditions. Upon triggering by activation signals they initiate strong immune responses. The activation of DCs is accompanied by a rapid upregulation of proinflammatory cytokines, which were shown in other cell types to be regulated by mechanisms at the transcriptional and posttranscriptional level. Tristetraprolin (TTP), an important RNA binding protein, is involved in the regulation of mRNA stability of such cytokines. In this study we analyzed the significance of TTP for mouse DCs, which were derived from TTP(-/-) and WT bone marrow progenitor cells (BM-DCs). Unstimulated BM-DCs of TTP(-/-) mice expressed lower levels of mRNAs encoding the costimulatory molecules CD40 and CD86 and surprisingly also the canonical TTP targets TNF-alpha and IL-10 as compared with WT DCs. On the protein level, both DC populations expressed comparable amounts of CD80 and CD86 and of either cytokine, but TTP(-/-) DCs expressed less MHCII than WT DCs. On the other hand, TTP(-/-) DCs displayed elevated expression of other TTP target mRNAs like IL-1beta, c-fos and Mkp-1. Stimulation of BM-DCs of either genotype with lipopolysaccharide resulted in a rapid upregulation to a comparable extent of all molecules monitored so far, except for c-fos mRNA. Subsequent mRNA decay analysis revealed gene-specific differences in mRNA stability, which was influenced by the presence of TTP and the activation state of the DCs. Unstimulated TTP(-/-) DCs exerted a markedly lower allogeneic T cell stimulatory potential than WT DCs. Moreover, TTP(-/-) DCs induced an altered cytokine pattern in cocultures of DCs and T cells. However, allogeneic T cells primed by unstimulated DCs of either genotype were equally refractory to restimulation and suppressed the proliferation of naive T cells to the same extent. Thus, the findings of this study lend support to the interpretation that without external stimulation antigen presenting activity in DCs in the presence of TTP is more pronounced than in its absence and that posttranscriptional regulation contributes to the control of gene expression in DCs.
Collapse
|
84
|
Pedersen CD, Fang JJ, Pedersen AE. A comparative study of transfection methods for RNA interference in bone marrow-derived murine dendritic cells. Scand J Immunol 2009; 70:447-56. [PMID: 19874549 DOI: 10.1111/j.1365-3083.2009.02320.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Selective gene silencing using RNA interference (RNAi) has been shown to be an efficient method for manipulation of cellular functions. In this study, we compare three previously established methods for transfection of murine bone marrow-derived DC (BM-DC). We tested the efficacy of electroporation with the Mouse Nucleofector kit((R)) from Amaxa Biosystems and lipid-based transfection methods using transfection reagents from Santa Cruz Biotechnology or Genlantis. To analyse the transfection efficacy we used FITC-conjugated siRNA as a positive control together with CD80 and CD86 specific siRNA. We show that electroporation using the Mouse Nucleofector kit((R)) from Amaxa Biosystems was not an efficient method to transfect BM-DC with siRNA in our hands. Transfection with Santa Cruz Biotechnology reagents resulted in up to 59% FITC-siRNA positive cells, but did not result in effective silencing of CD80 surface expression. In contrast, the most effective method was the lipid-based method using the siRNA transfection reagent GeneSilencer((R)) from Genlantis. This protocol resulted in up to 92% FITC-siRNA positive cells after 4 h which declined to 62% and 59% 24 and 48 h post-transfection, respectively. The transfected BM-DC remained CD11c positive, expressed high MHC class II and intermediate CD40 and were functional as APC. In conclusion, this protocol was effective for manipulation of murine BM-DC function through the use of specific siRNA and such methods can be important for the future study of DC-T cell interactions.
Collapse
Affiliation(s)
- C D Pedersen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen N, Denmark
| | | | | |
Collapse
|
85
|
Tas SW, Vervoordeldonk MJBM, Tak PP. Gene therapy targeting nuclear factor-kappaB: towards clinical application in inflammatory diseases and cancer. Curr Gene Ther 2009; 9:160-70. [PMID: 19519361 PMCID: PMC2864453 DOI: 10.2174/156652309788488569] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Nuclear factor (NF)-κB is regarded as one of the most important transcription factors and plays an essential role in the transcriptional activation of pro-inflammatory cytokines, cell proliferation and survival. NF-κB can be activated via two distinct NF-κB signal transduction pathways, the so-called canonical and non-canonical pathways, and has been demonstrated to play a key role in a wide range of inflammatory diseases and various types of cancer. Much effort has been put in strategies to inhibit NF-κB activation, for example by the development of pharmacological compounds that selectively inhibit NF-κB activity and therefore would be beneficial for immunotherapy of transplantation, autoimmune and allergic diseases, as well as an adjuvant approach in patients treated with chemotherapy for cancer. Gene therapy targeting NF-κB is a promising new strategy with the potential of long-term effects and has been explored in a wide variety of diseases, ranging from cancer to transplantation medicine and autoimmune diseases. In this review we discuss recent progress made in the development of NF-κB targeted gene therapy and the evolution towards clinical application.
Collapse
Affiliation(s)
- Sander W Tas
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
86
|
Suppression of ongoing experimental autoimmune myasthenia gravis by transfer of RelB-silenced bone marrow dentritic cells is associated with a change from a T helper Th17/Th1 to a Th2 and FoxP3+ regulatory T-cell profile. Inflamm Res 2009; 59:197-205. [PMID: 19768385 DOI: 10.1007/s00011-009-0087-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 07/15/2009] [Accepted: 08/19/2009] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To observe the therapeutic effect of RelB-silenced dendritic cells (DCs) in experimental autoimmune myasthenia gravis (EAMG), and further to investigate the mechanism of this immune system therapeutic. METHODS (1) RelB-silenced DCs and control DCs were prepared and the supernatants were collected for IL-12p70, IL-6, and IL-23 measurement by ELISA. (2) RelB-silenced DCs and control DCs were co-cultured with AChR-specific T cells, and the supernatant was collected to observe the IL-17, IFN-gamma, IL-4 production. (3) EAMG mice with clinical scores of 1 to 2 were collected and enrolled randomly into the RelB-silenced DC group or the control DC group. RelB-silenced DCs or an equal amount of control DCs were injected intravenously on days 0, 7, and 14 after enrollment. Clinical scores were evaluated every other day. Twenty days after allotment, serum from individual mice was collected to detect serum concentrations of anti-mouse AChR IgG, IgG1, IgG2b, and IgG2c. The splenocytes were isolated for analysis of lymphocyte proliferative responses, cytokine (IL-17, IFN-gamma, IL-4) production, and protein levels of RORgammat, T-bet, GATA-3, and FoxP3 (the special transcription factors of Th17, Th1, Th2, and Treg, respectively). RESULTS (1) RelB-silenced DCs produced significantly reduced amounts of IL-12p70, IL-6, and IL-23, as compared with control DCs. (2) RelB-silenced DCs spurred on the CD4(+) T cells from Th1/Th17 to the Th2 cell subset in the co-culture system. (3) Treatment with RelB-silenced DCs effectively reduced myasthenic symptoms and levels of serum anti-acetylcholine receptor autoantibody in mice with ongoing EAMG. Th17-related markers (RORgammat, IL-17) and Th1-related markers (T-bet, IFN-gamma) were downregulated, whereas Th2 markers (IL-4 and GATA3) and Treg marker (FoxP3) were upregulated. CONCLUSIONS RelB-silenced DCs were able to create a particular cytokine environment that was absent of inflammatory cytokines. RelB-silenced DCs provide a practical means to normalize the differentiation of the four T-cell subsets (Th17, Th1, Th2, and Treg) in vivo, and thus possess therapeutic potential in Th1/Th17-dominant autoimmune disorders such as myasthenia gravis.
Collapse
|
87
|
Herringson TP, Altin JG. Convenient targeting of stealth siRNA-lipoplexes to cells with chelator lipid-anchored molecules. J Control Release 2009; 139:229-38. [PMID: 19595724 DOI: 10.1016/j.jconrel.2009.06.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/18/2009] [Accepted: 06/30/2009] [Indexed: 01/03/2023]
Abstract
A major obstacle for the use of siRNAs as novel therapeutics is the requirement for functional delivery to specific cells in vivo. siRNA delivery by cationic agents is generally non-specific and a convenient targeting strategy has been lacking. This work explored the potential for using the chelator lipid 3(nitrilotriacetic acid)-ditetradecylamine (NTA(3)-DTDA) with neutral stealth liposomes to target siRNA to cells. A novel method for incorporating siRNAs into lipoplexes was developed which utilised helper lipids and the ionisable lipid 1,2-dioleoyl-3-dimethylammonium-propane (DODAP). This approach results in an efficient (>50%) incorporation of siRNA into lipoplexes, which when incorporated with Ni-NTA(3)-DTDA and engrafted with a His-tagged form of murine CD4 can target siRNA to murine A20 B cells, in vitro. Also, siRNA-lipoplexes engrafted with His-tagged peptides that target receptors on HEK-293 cells, or the receptor for tumour necrosis factor alpha expressed on the murine dendritic cell line DC2.4, could target siRNA and silence the expression of enhanced green fluorescence protein (EGFP). siRNA-lipoplexes produced by this method are approximately 240 nm dia, exhibit low zeta-potential (-1 mV), and target cells in serum-containing media. The results show that NTA(3)-DTDA can be used to target siRNA-lipoplexes to cells, and could provide a convenient approach for targeting siRNA to cells in vivo for therapeutic applications.
Collapse
Affiliation(s)
- Thomas P Herringson
- Biochemistry and Molecular Biology, Research School of Biology, ANU College of Medicine, Biology and Environment, The Australian National University, Canberra, ACT, 0200, Australia
| | | |
Collapse
|
88
|
Boudier A, Aubert-Pouëssel A, Gérardin C, Devoisselle JM, Bégu S, Louis-Plence P, Quentin J, Jorgensen C. Tripartite siRNA micelles as controlled delivery systems for primary dendritic cells. Drug Dev Ind Pharm 2009; 35:950-8. [DOI: 10.1080/03639040802716653] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
89
|
Feng B, Chen G, Zheng X, Sun H, Zhang X, Zhang ZX, Xiang Y, Ichim TE, Garcia B, Luke P, Jevnikar AM, Min WP. Small interfering RNA targeting RelB protects against renal ischemia-reperfusion injury. Transplantation 2009; 87:1283-1289. [PMID: 19424026 DOI: 10.1097/tp.0b013e3181a1905e] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Nuclear factor kappaB (NF-kappaB) has been found to be critical to the pathogenesis of renal ischemia-reperfusion injury (IRI). Using small interfering RNA (siRNA) to silence the expression of RelB, a component of the transcription factors Rel/nuclear factor kappaB, may protect renal IRI. Here, we report an siRNA-based treatment of preventing IRI. METHODS Renal IRI was induced in mice by clamping the left renal pedicle for 25 or 35 min. The therapeutic effects of siRNA were evaluated in renal function, histologic examination, and overall survival after lethal IRI. RESULTS A single injection of RelB siRNA resulted in knockdown of renal RelB expression. In comparison with control mice, levels of blood urea nitrogen and serum creatinine were significantly decreased in mice treated with siRNA. Pathologic examination demonstrated that tissue injury caused by IRI was markedly reduced as a result of RelB siRNA treatment. Additionally, with RelB siRNA treatment, immunohistochemistry showed a significant attenuation of tumor necrosis factor-alpha expression. Furthermore, survival experiments revealed that more than 90% of control mice died from lethal IRI, whereas 80% of siRNA-pretreated mice survived until the end of the 8-day observation period. CONCLUSION Silencing RelB, using siRNA, can significantly attenuate IRI-induced renal dysfunction and protect mice against lethal kidney ischemia, highlighting the potential for siRNA-based clinical therapy.
Collapse
Affiliation(s)
- Biao Feng
- Department of Surgery, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Zhang L, Procuik M, Fang T, Kung SKP. Functional analysis of the quantitative expression of a costimulatory molecule on dendritic cells using lentiviral vector-mediated RNA interference. J Immunol Methods 2009; 344:87-97. [PMID: 19303417 DOI: 10.1016/j.jim.2009.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 03/05/2009] [Accepted: 03/10/2009] [Indexed: 12/13/2022]
Abstract
The increasing number of co-stimulatory molecules identified on dendritic cells (DC) to date highlights the complex regulation of co-stimulatory signals in T cell activation. We previously established a single lentiviral vector system to stably express short hairpin RNA (shRNA) to induce RNA interference (RNAi) in cell lines and primary T cells. We reasoned that the choice of shRNA target sequences in the lentiviral vector system would also allow us to regulate different levels of surface expression of a co-stimulatory molecule stably and reproducibly. In this study, we first demonstrated that lentiviral vectors delivered RNA interference in DC without functional impairments. We used CD40 as a target co-stimulatory molecule to demonstrate the feasibility of using lentiviral vectors in delivering different shRNA target sequences to genetically modify DC that expressed different levels of CD40. We provided functional data to further demonstrate that quantitative expression of CD40 on LPS-stimulated DC have different functional outcomes on Ag-specific T cell responses in vitro. Collectively, we developed a simple system that will allow us to examine functional significance(s) of the quantitative and/or qualitative expression of a single or multiple co-stimulatory molecule(s) on DC.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Immunology, University of Manitoba, Room 417, Apotex Center, 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E0T5
| | | | | | | |
Collapse
|
91
|
Zheng X, Vladau C, Zhang X, Suzuki M, Ichim TE, Zhang ZX, Li M, Carrier E, Garcia B, Jevnikar AM, Min WP. A novel in vivo siRNA delivery system specifically targeting dendritic cells and silencing CD40 genes for immunomodulation. Blood 2009; 113:2646-2654. [PMID: 19164600 DOI: 10.1182/blood-2008-04-151191] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Translation of small interfering RNA (siRNA)-based approaches into practical therapeutics is limited because of lack of an effective and cell-specific delivery system. Herein, we present a new method of selectively delivering siRNA to dendritic cells (DCs) in vivo using CD40 siRNA-containing immunoliposomes (siILs) that were decorated with DC-specific DEC-205 mAb. Administration of CD40 siILs resulted in DC-specific cell targeting in vitro and in vivo. On treatment with CD40 siILs, the expression of CD40 in DCs, as well allostimulatory activity was inhibited. In vivo administration resulted in selective siRNA uptake into immune organs and functional immune modulation as assessed using a model antigen. In conclusion, this is the first demonstration of DC-specific siRNA delivery and gene silencing in vivo, which highlights the potential of DC-mediated immune modulation and the feasibility of siRNA-based clinical therapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antigen Presentation/drug effects
- Antigens, CD/immunology
- Bone Marrow Cells/cytology
- CD40 Antigens/antagonists & inhibitors
- CD40 Antigens/biosynthesis
- CD40 Antigens/genetics
- CD40 Antigens/immunology
- Cells, Cultured/drug effects
- Cells, Cultured/immunology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Drug Delivery Systems
- Genetic Therapy/methods
- Immunoconjugates/administration & dosage
- Immunosuppression Therapy/methods
- Lectins, C-Type/immunology
- Liposomes
- Lymphocyte Activation/drug effects
- Lymphoid Tissue/drug effects
- Lymphoid Tissue/ultrastructure
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Minor Histocompatibility Antigens
- RNA Interference
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/pharmacology
- RNA, Small Interfering/therapeutic use
- Receptors, Cell Surface/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Xiufen Zheng
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Capini C, Jaturanpinyo M, Chang HI, Mutalik S, McNally A, Street S, Steptoe R, O'Sullivan B, Davies N, Thomas R. Antigen-Specific Suppression of Inflammatory Arthritis Using Liposomes. THE JOURNAL OF IMMUNOLOGY 2009; 182:3556-65. [DOI: 10.4049/jimmunol.0802972] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
93
|
Suzuki M, Zheng X, Zhang X, Ichim TE, Sun H, Kubo N, Beduhn M, Shunnar A, Garcia B, Min WP. Inhibition of allergic responses by CD40 gene silencing. Allergy 2009; 64:387-97. [PMID: 19175598 DOI: 10.1111/j.1398-9995.2008.01839.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Gene silencing using small interfering RNA (siRNA) is a potent method of specifically knocking down molecular targets. Small interfering RNA is therapeutically promising, however, treatment of allergic diseases with siRNA has not been explored in vivo. The aim of this study was to evaluate therapeutic effects of CD40 siRNA on inhibition of allergic responses. METHODS Mice sensitized with ovalbumin (OVA) and alum were treated with CD40 siRNA, scrambled siRNA, or phosphate buffer saline (PBS) alone, and then challenged intranasally with OVA. RESULTS A significant reduction in nasal allergic symptoms was observed in the CD40 siRNA treated OVA-allergic mice compared to the controls of scrambled siRNA and PBS alone, which is correlated with the decrease of local eosinophil accumulation. CD40 siRNA treatment knocked down CD40 expression on dendritic cells (DCs) in vivo and impaired their antigen presenting function. Treatment with CD40 siRNA resulted in inhibition of OVA-specific T cell response and decrease of interleukin-4 (IL-4), IL-5, and interferon-gamma production from T cells stimulated with OVA. Administration of CD40 siRNA also suppressed CD40 expression on B cells, resulting in down-regulation of OVA-specific immunoglobulin E (IgE), IgG1, and IgG2a levels. Additionally, increased regulatory T cells were observed in the CD40 siRNA treated mice. CONCLUSIONS The present study demonstrates a novel therapeutic use for siRNA in allergy. CD40 siRNA attenuated allergy through inhibition of DC and B cell functions and generation of regulatory T (Treg) cells.
Collapse
Affiliation(s)
- M Suzuki
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Handbook of experimental pharmacology "dendritic cells": the use of dexamethasone in the induction of tolerogenic DCs. Handb Exp Pharmacol 2009:233-49. [PMID: 19031029 DOI: 10.1007/978-3-540-71029-5_11] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dendritic cells (DCs) have a central role in immune regulation, ranging from tolerance induction to the induction of specific immune responses. DCs serve as an essential link between innate and adaptive immunity. This broad range of powerful immune stimulatory as well as regulatory functions has made DCs as targets for vaccine development strategies. One approach to promote the tolerogenicity of DCs is to suppress their maturation by pharmacological agents, including glucocorticoids (GCs). In the present chapter we will review GCs used in vitro with cultured DCs, applied in vivo, or used to generate tolerogenic DCs for cellular therapy.
Collapse
|
95
|
Karimi MH, Ebadi P, Pourfathollah AA, Soheili ZS, Samiee S, Ataee Z, Tabei SZ, Moazzeni SM. Immune modulation through RNA interference-mediated silencing of CD40 in dendritic cells. Cell Immunol 2009; 259:74-81. [DOI: 10.1016/j.cellimm.2009.05.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2009] [Revised: 05/12/2009] [Accepted: 05/22/2009] [Indexed: 11/30/2022]
|
96
|
Sklavos MM, Tse HM, Piganelli JD. Redox modulation inhibits CD8 T cell effector function. Free Radic Biol Med 2008; 45:1477-86. [PMID: 18805480 DOI: 10.1016/j.freeradbiomed.2008.08.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 07/29/2008] [Accepted: 08/25/2008] [Indexed: 10/21/2022]
Abstract
The evolutionary preservation of reactive oxygen species in innate immunity underscores the important roles these constituents play in immune cell activity and as signaling intermediates. In an effort to exploit these pathways to achieve control of aberrant immune activation we demonstrate that modulation of redox status suppresses cell proliferation and production of IL-2, IFN-gamma, TNF-alpha, and IL-17 in two robust CD8 T-cell-dependent in vitro mouse models: (1) response to alloantigen in an mixed leukocyte reaction and (2) CD8 T cell receptor transgenic OT-1 response to cognate peptide (SIINFEKL). To correlate these findings with cytotoxic lymphocyte (CTL) function we performed cytotoxicity assays and found that redox modulation diminishes the ability of alloantigen-specific and antigen-specific OT-1 CTLs to kill their corresponding antigen-expressing target cells. To further examine the mechanisms of redox-mediated repression of CTL target cell lysis, we analyzed the expression of the effector molecules IFN-gamma, perforin, and granzyme B and the degranulation marker CD107a (LAMP-1). In both models, redox modulation reduced the expression of these effector components by at least fivefold. These results demonstrate that redox modulation quells the CD8 T cell response to alloantigen and the T cell receptor transgenic CD8 T cell response to its cognate antigen by inhibiting proliferation, proinflammatory cytokine synthesis, and CTL effector mechanisms.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Cell Proliferation/drug effects
- Disease Models, Animal
- Interferon-gamma/biosynthesis
- Interleukin-17/biosynthesis
- Interleukin-2/biosynthesis
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Oxidation-Reduction
- Peptide Fragments/pharmacology
- Reactive Oxygen Species/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/drug effects
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Tumor Necrosis Factor-alpha/biosynthesis
Collapse
Affiliation(s)
- Martha M Sklavos
- Diabetes Institute, Division of Immunogenetics, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
97
|
Anderson AE, Swan DJ, Sayers BL, Harry RA, Patterson AM, von Delwig A, Robinson JH, Isaacs JD, Hilkens CMU. LPS activation is required for migratory activity and antigen presentation by tolerogenic dendritic cells. J Leukoc Biol 2008; 85:243-50. [PMID: 18971286 PMCID: PMC2700018 DOI: 10.1189/jlb.0608374] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Autoimmune pathologies are caused by a breakdown in self-tolerance. Tolerogenic dendritic cells (tolDC) are a promising immunotherapeutic tool for restoring self-tolerance in an antigen-specific manner. Studies about tolDC have focused largely on generating stable maturation-resistant DC, but few have fully addressed questions about the antigen-presenting and migratory capacities of these cells, prerequisites for successful immunotherapy. Here, we investigated whether human tolDC, generated with dexamethasone and the active form of vitamin D3, maintained their tolerogenic function upon activation with LPS (LPS-tolDC), while acquiring the ability to present exogenous autoantigen and to migrate in response to the CCR7 ligand CCL19. LPS activation led to important changes in the tolDC phenotype and function. LPS-tolDC, but not tolDC, expressed the chemokine receptor CCR7 and migrated in response to CCL19. Furthermore, LPS-tolDC were superior to tolDC in their ability to present type II collagen, a candidate autoantigen in rheumatoid arthritis. tolDC and LPS-tolDC had low stimulatory capacity for allogeneic, naïve T cells and skewed T cell polarization toward an anti-inflammatory phenotype, although LPS-tolDC induced significantly higher levels of IL-10 production by T cells. Our finding that LPS activation is essential for inducing migratory and antigen-presenting activity in tolDC is important for optimizing their therapeutic potential.
Collapse
Affiliation(s)
- Amy E Anderson
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Suzuki M, Zheng X, Zhang X, Li M, Vladau C, Ichim TE, Sun H, Min LR, Garcia B, Min WP. Novel vaccination for allergy through gene silencing of CD40 using small interfering RNA. THE JOURNAL OF IMMUNOLOGY 2008; 180:8461-9. [PMID: 18523314 DOI: 10.4049/jimmunol.180.12.8461] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Small interfering RNA (siRNA) is a potent means of inducing gene-specific silencing. Gene silencing strategies using siRNA have demonstrated therapeutic benefits in animal models of various diseases, and are currently in clinical trials. However, the utility of gene silencing as a treatment for allergic diseases has not yet been reported. In this study, we report a novel therapy for allergy through gene silencing of CD40, a critical costimulatory molecule and a key factor in allergic immune responses. Silencing CD40 resulted in generation of immunoregulatory dendritic cells (DCs). Administration of CD40 siRNA remarkably reduced nasal allergic symptoms and local eosinophil accumulation in the OVA-induced allergic mice. The OVA-specific T cell response was inhibited after the CD40 siRNA treatment. Additionally, anti-OVA specific IgE and production of IL-4 and IL-5 of T cells stimulated by OVA were significantly decreased in CD40 siRNA-treated mice. Furthermore, we demonstrated that the therapeutic effects by CD40 siRNA were associated with impaired Ag-presenting functions of DCs and B cells, and generation of regulatory T cells. The present study highlights a therapeutic potential of siRNA-based treatment for allergic diseases.
Collapse
Affiliation(s)
- Motohiko Suzuki
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Abstract
Cardiac allograft vasculopathy (CAV) continues to limit the long-term success of cardiac transplantation. Recent insights have underscored the fact that innate and adaptive immune responses are involved in the pathogenesis of CAV. Vascular lesions are the result of cumulative endothelial injuries induced both by alloimmune responses and by nonspecific insults (including ischemia-reperfusion injury, viral infections, and metabolic disorders) in the context of impaired repair mechanisms. Intravascular ultrasound is the most sensitive method for detection of CAV, and progressive intimal thickening in the first posttransplant year identifies patients at high risk for future cardiovascular events. Encouraging results with regard to the detection of CAV by noninvasive methods should be an incentive to apply routine noninvasive imaging during mid- to long-term follow-up. Improved immunosuppressive drugs, including mycophenolate mofetil and proliferation signal inhibitors, as well as statins (in part via immunomodulation), have beneficial effects on CAV progression, although there is still a need to confirm the impact of vasodilators in improving outcome after heart transplantation. Coronary revascularization for CAV is only palliative, with no long-term survival benefit. Three main strategies for CAV prevention are currently under investigation: inhibition of growth factors and cytokines, cell therapy, and tolerance induction. However, because individual responses to an allograft change over time, assays to monitor the recipient's immune response and individualized methods for therapeutic immune modulation are clearly needed.
Collapse
Affiliation(s)
- Daniel Schmauss
- Medizinische Klinik und Poliklinik I, University Hospital Munich-Grosshadern, Marchioninistrasse 15, 81377 Munich, Germany
| | | |
Collapse
|
100
|
Ichim TE, Zheng X, Suzuki M, Kubo N, Zhang X, Min LR, Beduhn ME, Riordan NH, Inman RD, Min WP. Antigen-specific therapy of rheumatoid arthritis. Expert Opin Biol Ther 2008; 8:191-9. [PMID: 18194075 DOI: 10.1517/14712598.8.2.191] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Immunotherapy offers the promise of antigen-specific suppression of pathological immune responses in conditions such as autoimmunity and organ transplantation. Substantial advances have been made in recent years in terms of understanding basic immunological mechanisms of autoreactivity, as well as clinically implementing immune-based therapies that are antigen nonspecific. OBJECTIVE To provide an integrated overview of the current state of the art in terms of antigen-specific tolerance induction, as well as to predict future directions for the field. METHODS Examples of successes and failures of antigen-specific immunotherapy were sought. Particular attention was paid to the well-established collagen II-induced model of arthritis. RESULTS/CONCLUSIONS Previous failures of antigen-specific immunotherapy were associated with lack of identification of clinically relevant antigens, as well as inappropriate tolerogenic methodologies. The advances in proteomics combined with novel gene-specific immune modulatory techniques place today's translational researchers in a unique position to tackle the problem of antigen-specific immunotherapeutic protocols.
Collapse
Affiliation(s)
- Thomas E Ichim
- University of Western Ontario, Departments of Surgery, Pathology, Microbiology & Immunology, 339 Windermere Road, University Hospital C9-136, London, Ontario, N6A 5A5, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|