1
|
Ullah Khan F, Khongorzul P, Gris D, Amrani A. Stat5b/Ezh2 axis governs high PD-L1 expressing tolerogenic dendritic cell subset in autoimmune diabetes. Int Immunopharmacol 2024; 133:112166. [PMID: 38678673 DOI: 10.1016/j.intimp.2024.112166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that play an important role in inducing and maintaining immune tolerance. The altered distribution and/or function of DCs contributes to defective tolerance in autoimmune diseases such as type 1 diabetes (T1D). In human T1D and in NOD mouse models, DCs share some defects and are often described as less tolerogenic and excessively immunogenic. In the NOD mouse model, the autoimmune response is associated with a defect in the Stat5b signaling pathway. We have reported that expressing a constitutively active form of Stat5b in DCs of transgenic NOD mice (NOD.Stat5b-CA), re-established their tolerogenic function, restored autoimmune tolerance and conferred protection from diabetes. However, the role and molecular mechanisms of Stat5b signaling in regulating splenic conventional DCs tolerogenic signature remained unclear. In this study, we reported that, compared to immunogenic splenic DCs of NOD, splenic DCs of NOD.Stat5b-CA mice exhibited a tolerogenic profile marked by elevated PD-L1 and PD-L2 expression, reduced pro-inflammatory cytokine production, increased frequency of the cDC2 subset and decreased frequency of the cDC1 subset. This tolerogenic profile was associated with increased Ezh2 and IRF4 but decreased IRF8 expression. We also found an upregulation of PD-L1 in the cDC1 subset and high PD-L1 and PD-L2 expression in cDC2 of NOD.Stat5b-CA mice. Mechanistically, we demonstrated that Ezh2 plays an important role in the maintenance of high PD-L1 expression in cDC1 and cDC2 subsets and that Ezh2 inhibition resulted in PD-L1 but not PD-L2 downregulation which was more drastic in the cDC2 subset. Additionally, Ezh2 inhibition severely reduced the cDC2 subset and increased the cDC1 subset and Stat5b-CA.DC pro-inflammatory cytokine production. Together our data suggest that the Stat5b-Ezh2 axis is critical for the maintenance of tolerogenic high PD-L1-expressing cDC2 and autoimmune tolerance in NOD.Stat5b-CA mice.
Collapse
Affiliation(s)
- Farhan Ullah Khan
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, 3001, 12th Avenue North, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Puregmaa Khongorzul
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, 3001, 12th Avenue North, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Denis Gris
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, 3001, 12th Avenue North, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Abdelaziz Amrani
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, 3001, 12th Avenue North, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
2
|
Giannoukakis N. Tolerogenic dendritic cells in type 1 diabetes: no longer a concept. Front Immunol 2023; 14:1212641. [PMID: 37388741 PMCID: PMC10303908 DOI: 10.3389/fimmu.2023.1212641] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
Tolerogenic dendritic cells (tDC) arrest the progression of autoimmune-driven dysglycemia into clinical, insulin-requiring type 1 diabetes (T1D) and preserve a critical mass of β cells able to restore some degree of normoglycemia in new-onset clinical disease. The safety of tDC, generated ex vivo from peripheral blood leukocytes, has been demonstrated in phase I clinical studies. Accumulating evidence shows that tDC act via multiple layers of immune regulation arresting the action of pancreatic β cell-targeting effector lymphocytes. tDC share a number of phenotypes and mechanisms of action, independent of the method by which they are generated ex vivo. In the context of safety, this yields confidence that the time has come to test the best characterized tDC in phase II clinical trials in T1D, especially given that tDC are already being tested for other autoimmune conditions. The time is also now to refine purity markers and to "universalize" the methods by which tDC are generated. This review summarizes the current state of tDC therapy for T1D, presents points of intersection of the mechanisms of action that the different embodiments use to induce tolerance, and offers insights into outstanding matters to address as phase II studies are imminent. Finally, we present a proposal for co-administration and serially-alternating administration of tDC and T-regulatory cells (Tregs) as a synergistic and complementary approach to prevent and treat T1D.
Collapse
Affiliation(s)
- Nick Giannoukakis
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
3
|
The relationship between Schistosoma and glycolipid metabolism. Microb Pathog 2021; 159:105120. [PMID: 34358648 DOI: 10.1016/j.micpath.2021.105120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 02/02/2023]
Abstract
Diabetes and obesity have become the most popular metabolic diseases in the world. A large number of previous studies have shown that glucose and lipid metabolism disorder is an important risk factor and a main cause of diabetes and obesity. Schistosoma is a parasite transmitted by freshwater snails. It can induce a series of inflammatory and immune reactions after infecting the human body, causing schistosomiasis. However, in recent years, studies have found that Schistosoma infection or Schistosoma related products can improve or prevent some immune and inflammatory diseases, such as severe asthma, inflammatory bowel disease, diabetes and so on. Further experiments have also revealed that Schistosoma can promote the secretion of anti-inflammatory factors and regulate the glucose and lipid metabolism in the host body by polarizing immune cells such as T cells, B cells and dendritic cells (DCs). In this review, we summarize studies that investigated Schistosoma and Schistosoma-derived products and their relationship with glycolipid metabolism and related diseases, highlighting potential protective mechanisms.
Collapse
|
4
|
Zou J, Thornton C, Chambers ES, Rosser EC, Ciurtin C. Exploring the Evidence for an Immunomodulatory Role of Vitamin D in Juvenile and Adult Rheumatic Disease. Front Immunol 2021; 11:616483. [PMID: 33679704 PMCID: PMC7930375 DOI: 10.3389/fimmu.2020.616483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
Vitamin D is synthesized in the skin following exposure to UVB radiation or is directly absorbed from the diet. Following hydroxylation in the liver and kidneys, vitamin D becomes its bioactive form, 1,25(OH)2D, which has been described to have potent immunomodulatory capacity. This review will focus on the effect of vitamin D in modulating the dysregulated immune system of autoimmune rheumatic diseases (ARD) patients across age, in particular in arthritis (rheumatoid arthritis and juvenile idiopathic arthritis), and systemic lupus erythematosus (with adult and juvenile onset). As well as delineating the impact of vitamin D on the innate and adaptive immune functions associated with each disease pathology, this review will also summarize and evaluate studies that link vitamin D status with disease prevalence, and supplementation studies that examine the potential benefits of vitamin D on disease outcomes. Exploring this evidence reveals that better designed randomized controlled studies are required to clarify the impact of vitamin D supplementation on ARD outcomes and general health. Considering the accessibility and affordability of vitamin D as a therapeutic option, there is a major unmet need for evidence-based treatment recommendations for the use of vitamin D in this patient population.
Collapse
Affiliation(s)
- Jiaqi Zou
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - Clare Thornton
- Department of Rheumatology (Metabolic Bone Diseases), University College London Hospital NHS Foundation Trust, London, United Kingdom
| | - Emma S Chambers
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Elizabeth C Rosser
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London and Great Ormond Street Hospitals, London, United Kingdom
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London and Great Ormond Street Hospitals, London, United Kingdom
| |
Collapse
|
5
|
Elucidating the Role of Ezh2 in Tolerogenic Function of NOD Bone Marrow-Derived Dendritic Cells Expressing Constitutively Active Stat5b. Int J Mol Sci 2020; 21:ijms21186453. [PMID: 32899608 PMCID: PMC7554732 DOI: 10.3390/ijms21186453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 08/27/2020] [Indexed: 12/28/2022] Open
Abstract
Tolerogenic dendritic cells (toDCs) are crucial to controlling the development of autoreactive T cell responses and the prevention of autoimmunity. We have reported that NOD.CD11cStat5b-CA transgenic mice expressing a constitutively active (CA) form of Stat5b under the control of a CD11c promoter are protected from diabetes and that Stat5b-CA-expressing DCs are tolerogenic and halt ongoing diabetes in NOD mice. However, the molecular mechanisms by which Stat5b-CA modulates DC tolerogenic function are not fully understood. Here, we used bone marrow-derived DCs (BMDCs) from NOD.CD11cStat5b-CA transgenic mice (Stat5b-CA.BMDCs) and found that Stat5b-CA.BMDCs displayed high levels of MHC class II, CD80, CD86, PD-L1, and PD-L2 and produced elevated amounts of TGFβ but low amounts of TNFα and IL-23. Stat5b-CA.BMDCs upregulated Irf4 and downregulated Irf8 genes and protein expression and promoted CD11c+CD11b+ DC2 subset differentiation. Interestingly, we found that the histone methyltransferase Ezh2 and Stat5b-CA bound gamma-interferon activated site (GAS) sequences in the Irf8 enhancer IRF8 transcription, whereas Stat5b but not Ezh2 bound GAS sequences in the Irf4 promoter to enhance IRF4 transcription. Injection of Stat5b-CA.BMDCs into prediabetic NOD mice halted progression of islet inflammation and protected against diabetes. Importantly, inhibition of Ezh2 in tolerogenic Stat5b-CA.BMDCs reduced their ability to prevent diabetes development in NOD recipient mice. Taken together, our data suggest that the active form of Stat5b induces tolerogenic DC function by modulating IRF4 and IRF8 expression through recruitment of Ezh2 and highlight the fundamental role of Ezh2 in Stat5b-mediated induction of tolerogenic DC function.
Collapse
|
6
|
Loretelli C, Assi E, Seelam AJ, Ben Nasr M, Fiorina P. Cell therapy for type 1 diabetes. Expert Opin Biol Ther 2020; 20:887-897. [PMID: 32299257 DOI: 10.1080/14712598.2020.1748596] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Type 1 diabetes (T1D) is a lifelong condition resulting from autoimmune destruction of insulin-producing β-cells. Islet or whole-pancreas transplantation is limited by the shortage of donors and need for chronic immune suppression. Novel strategies are needed to prevent β-cell loss and to rescue production of endogenous insulin. AREAS COVERED This review covers the latest advances in cell-based therapies for the treatment and prevention of T1D. Topics include adoptive transfer of cells with increased immunoregulatory potential for β-cell protection, and β-cell replacement strategies such as generation of insulin-producing β-like cells from unlimited sources. EXPERT OPINION Cell therapy provides an opportunity to prevent or reverse T1D. Adoptive transfer of autologous cells having enhanced immunomodulatory properties can suppress autoimmunity and preserve β-cells. Such therapies have been made possible by a combination of genome-editing techniques and transplantation of tolerogenic cells. In-vitro modified autologous hematopoietic stem cells and tolerogenic dendritic cells may protect endogenous and newly generated β-cells from a patient's autoimmune response without hampering immune surveillance for infectious agents and malignant cellular transformations. However, methods to generate cells that meet quality and safety standards for clinical applications require further refinement.
Collapse
Affiliation(s)
- Cristian Loretelli
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy
| | - Emma Assi
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy
| | - Andy Joe Seelam
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy
| | - Moufida Ben Nasr
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy.,Nephrology Division, Boston Children's Hospital, Harvard Medical School , Boston, MA, USA
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy.,Nephrology Division, Boston Children's Hospital, Harvard Medical School , Boston, MA, USA.,Division of Endocrinology, ASST Fatebenefratelli-Sacco , Milan, Italy
| |
Collapse
|
7
|
Funda DP, Palová-Jelínková L, Goliáš J, Kroulíková Z, Fajstová A, Hudcovic T, Špíšek R. Optimal Tolerogenic Dendritic Cells in Type 1 Diabetes (T1D) Therapy: What Can We Learn From Non-obese Diabetic (NOD) Mouse Models? Front Immunol 2019; 10:967. [PMID: 31139178 PMCID: PMC6527741 DOI: 10.3389/fimmu.2019.00967] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/15/2019] [Indexed: 12/21/2022] Open
Abstract
Tolerogenic dendritic cells (tolDCs) are explored as a promising standalone or combination therapy in type 1 diabetes (T1D). The therapeutic application of tolDCs, including in human trials, has been tested also in other autoimmune diseases, however, T1D displays some unique features. In addition, unlike in several disease-induced animal models of autoimmune diseases, the prevalent animal model for T1D, the NOD mouse, develops diabetes spontaneously. This review compares evidence of various tolDCs approaches obtained from animal (mainly NOD) models of T1D with a focus on parameters of this cell-based therapy such as protocols of tolDC preparation, antigen-specific vs. unspecific approaches, doses of tolDCs and/or autoantigens, application schemes, application routes, the migration of tolDCs as well as their preventive, early pre-onset intervention or curative effects. This review also discusses perspectives of tolDC therapy and areas of preclinical research that are in need of better clarification in animal models in a quest for effective and optimal tolDC therapies of T1D in humans.
Collapse
Affiliation(s)
- David P Funda
- Institute of Microbiology of the Czech Academy of Sciences, v.v.i., Prague, Czechia
| | - Lenka Palová-Jelínková
- SOTIO a s., Prague, Czechia.,Department of Immunology, 2nd Medical School, Charles University, Prague, Czechia
| | - Jaroslav Goliáš
- Institute of Microbiology of the Czech Academy of Sciences, v.v.i., Prague, Czechia
| | - Zuzana Kroulíková
- Institute of Microbiology of the Czech Academy of Sciences, v.v.i., Prague, Czechia
| | - Alena Fajstová
- Institute of Microbiology of the Czech Academy of Sciences, v.v.i., Prague, Czechia
| | - Tomáš Hudcovic
- Institute of Microbiology of the Czech Academy of Sciences, v.v.i., Prague, Czechia
| | - Radek Špíšek
- SOTIO a s., Prague, Czechia.,Department of Immunology, 2nd Medical School, Charles University, Prague, Czechia
| |
Collapse
|
8
|
Xu N, Li J, Gao Y, Zhou N, Ma Q, Wu M, Zhang Y, Sun X, Xie J, Shen G, Yang M, Tu Q, Xu X, Zhu J, Tao J. Apoptotic cell-mimicking gold nanocages loaded with LXR agonist for attenuating the progression of murine systemic lupus erythematosus. Biomaterials 2019; 197:380-392. [DOI: 10.1016/j.biomaterials.2019.01.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 12/31/2018] [Accepted: 01/20/2019] [Indexed: 02/07/2023]
|
9
|
Phillips BE, Garciafigueroa Y, Engman C, Trucco M, Giannoukakis N. Tolerogenic Dendritic Cells and T-Regulatory Cells at the Clinical Trials Crossroad for the Treatment of Autoimmune Disease; Emphasis on Type 1 Diabetes Therapy. Front Immunol 2019; 10:148. [PMID: 30787930 PMCID: PMC6372505 DOI: 10.3389/fimmu.2019.00148] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/17/2019] [Indexed: 02/06/2023] Open
Abstract
Tolerogenic dendritic cells and T-regulatory cells are two immune cell populations with the potential to prevent the onset of clinical stage type 1 diabetes, and manage the beginning of underlying autoimmunity, at the time-at-onset and onwards. Initial phase I trials demonstrated that the administration of a number of these cell populations, generated ex vivo from peripheral blood leukocytes, was safe. Outcomes of some of these trials also suggested some level of autoimmunity regulation, by the increase in the numbers of regulatory cells at different points in a network of immune regulation in vivo. As these cell populations come to the cusp of pivotal phase II efficacy trials, a number of questions still need to be addressed. At least one mechanism of action needs to be verified as operational, and through this mechanism biomarkers predictive of the underlying autoimmunity need to be identified. Efficacy in the regulation of the underlying autoimmunity also need to be monitored. At the same time, the absence of a common phenotype core among the different dendritic cell and T-regulatory cell populations, that have completed phase I and early phase II trials, necessitates a better understanding of what makes these cells tolerogenic, especially if a uniform phenotypic core cannot be identified. Finally, the inter-relationship of tolerogenic dendritic cells and T-regulatory cells for survival, induction, and maintenance of a tolerogenic state that manages the underlying diabetes autoimmunity, raises the possibility to co-administer, or even to serially-administer tolerogenic dendritic cells together with T-regulatory cells as a cellular co-therapy, enabling the best possible outcome. This is currently a knowledge gap that this review aims to address.
Collapse
Affiliation(s)
- Brett Eugene Phillips
- Allegheny Health Network Institute of Cellular Therapeutics, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Yesica Garciafigueroa
- Allegheny Health Network Institute of Cellular Therapeutics, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Carl Engman
- Allegheny Health Network Institute of Cellular Therapeutics, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Massimo Trucco
- Allegheny Health Network Institute of Cellular Therapeutics, Allegheny General Hospital, Pittsburgh, PA, United States.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Nick Giannoukakis
- Allegheny Health Network Institute of Cellular Therapeutics, Allegheny General Hospital, Pittsburgh, PA, United States.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
10
|
Herbelet S, De Bleecker JL. Immune checkpoint failures in inflammatory myopathies: An overview. Autoimmun Rev 2018; 17:746-754. [PMID: 29885538 DOI: 10.1016/j.autrev.2018.01.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 01/18/2018] [Indexed: 12/21/2022]
Abstract
Dermatomyositis (DM), polymyositis (PM), inclusion body myositis (IBM), immune mediated necrotizing myopathy (IMNM) and overlap myositis (OM) are classified as inflammatory myopathies (IM) with involvement of autoimmune features such as autoreactive lymphocytes and autoantibodies. Autoimmunity can be defined as a loss in self-tolerance and attack of autoantigens by the immune system. Self-tolerance is achieved by a group of immune mechanisms occurring in central and periphal lymphoid organs and tissues, called immune checkpoints, that work in synergy to protect the body from harmful immune reactions. Autoimmune disorders appear when immune checkpoints fail. In this review, the different immune checkpoint failures are discussed in DM, PM, IBM and IMNM. Exploring research contribution in each of these immune checkpoints might help to highlight research perspectives in the field and obtain a more complete picture of IM disease pathology.
Collapse
Affiliation(s)
- Sandrine Herbelet
- Department of Neurology, Ghent University and Ghent University Hospital, C. Heymanslaan 10, 9000 Gent, Belgium.
| | - Jan L De Bleecker
- Department of Neurology, Ghent University and Ghent University Hospital, C. Heymanslaan 10, 9000 Gent, Belgium
| |
Collapse
|
11
|
Elizondo DM, Andargie TE, Yang D, Kacsinta AD, Lipscomb MW. Inhibition of Allograft Inflammatory Factor-1 in Dendritic Cells Restrains CD4 + T Cell Effector Responses and Induces CD25 +Foxp3 + T Regulatory Subsets. Front Immunol 2017; 8:1502. [PMID: 29167673 PMCID: PMC5682305 DOI: 10.3389/fimmu.2017.01502] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/25/2017] [Indexed: 11/13/2022] Open
Abstract
Allograft inflammatory factor-1 (AIF1) is a cytoplasmic scaffold protein shown to influence immune responses in macrophages and microglial cells. The protein contains Ca2+ binding EF-hand and PDZ interaction domains important for mediating intracellular signaling complexes. This study now reports that AIF1 is expressed in CD11c+ dendritic cells (DC) and silencing of expression restrains induction of antigen-specific CD4+ T cell effector responses. AIF1 knockdown in murine DC resulted in impaired T cell proliferation and skewed polarization away from T helper type 1 and 17 fates. In turn, there was a parallel expansion of IL-10-producing and CD25+Foxp3+ T regulatory subsets. These studies are the first to demonstrate that AIF1 expression in DC serves as a potent governor of cognate T cell responses and presents a novel target for engineering tolerogenic DC-based immunotherapies.
Collapse
Affiliation(s)
- Diana M Elizondo
- Department of Biology, Howard University, Washington, DC, United States
| | | | - Dazhi Yang
- Department of Biology, Howard University, Washington, DC, United States
| | - Apollo D Kacsinta
- Department of Cellular and Molecular Medicine, UCSD School of Medicine, La Jolla, CA, United States
| | | |
Collapse
|
12
|
Heller P, Hobernik D, Lächelt U, Schinnerer M, Weber B, Schmidt M, Wagner E, Bros M, Barz M. Combining reactive triblock copolymers with functional cross-linkers: A versatile pathway to disulfide stabilized-polyplex libraries and their application as pDNA vaccines. J Control Release 2017; 258:146-160. [DOI: 10.1016/j.jconrel.2017.05.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 05/02/2017] [Accepted: 05/10/2017] [Indexed: 02/06/2023]
|
13
|
Conti P, Lessiani G, Kritas SK, Ronconi G, Caraffa A, Theoharides TC. Mast cells emerge as mediators of atherosclerosis: Special emphasis on IL-37 inhibition. Tissue Cell 2017; 49:393-400. [PMID: 28420489 DOI: 10.1016/j.tice.2017.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 04/10/2017] [Accepted: 04/10/2017] [Indexed: 02/06/2023]
Abstract
In atherosclerosis lipoproteins stimulate the innate immune response, leading to the release of inflammatory cytokines and chemokines. Hypercholesterolemia may activate the synthesis and release of inflammatory cytokines such as IL-1, which induces TNF release in mast cells (MCs). IL-1 and IL-1 family members orchestrate a broadening list of inflammatory diseases, including atherosclerosis. MCs are implicated in the pathophysiology of several diseases including allergy and inflammation. Activated MCs, located perivascularly, contribute to inflammation in atherosclerosis by producing inflammatory cytokines. MC IL-1-activation leads to the immediate release of inflammatory chemical mediators and TNF, and late inflammatory compounds such as cytokines. MCs can be activated by exogenous cytokines, antigens, microbial products (LPS) and neurotransmitters and generate IL-1 beta, TNF and several other inflammatory cytokines/chemokines along with PGD2, leukotrienes, histamine and proteases. MCs activated with IL-1 induce selective release of IL-6 without degranulation. TNF emerges as one of the most potent inflammatory cytokines involved in the response due to LDL. Cytokines, such as IL-1, IL-6, IL-33 and TNF, are generated in the inflammatory sites by both macrophages and MCs, mediating atherosclerosis. IL-37 (IL-1 family member 7) binds IL-18Ra chain and acts by an intracellular mechanism down-regulating the expression of pro-inflammatory signals cJun, MAP kinase p38a, STAT transcription factors and p53. Blocking IL-1 with IL-37 alleviates the symptoms in patients with inflammatory diseases including arteriosclerosis. The impact of IL-37 on inflammatory cytokines mediating atherosclerosis is beneficial and protective. However, more studies are needed to better define this mechanism and the safety and tolerability of IL-37.
Collapse
Affiliation(s)
- Pio Conti
- Immunology Division, Postgraduate Medical School, University of Chieti-Pescara, Viale Unità dell'Italia 73, 66013, Chieti, Italy.
| | - Gianfranco Lessiani
- Angiology Unit, Medicine and Geriatria, Villa Serena Hospital, Città Sant'Angelo, Italy
| | | | - Gianpaolo Ronconi
- Clinica dei Pazienti del Territorio, Policlinico Gemelli, Roma, Italy
| | | | - Theoharis C Theoharides
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
14
|
Adenovirus-Mediated CCR7 and BTLA Overexpression Enhances Immune Tolerance and Migration in Immature Dendritic Cells. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3519745. [PMID: 28393074 PMCID: PMC5368407 DOI: 10.1155/2017/3519745] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 02/20/2017] [Indexed: 01/08/2023]
Abstract
Our previous report revealed that immature dendritic cells (imDCs) with adenovirus-mediated CCR7 overexpression acquired an enhanced migratory ability but also exhibited the lower immune tolerance observed in more mature cells. In the present study, we aimed to investigate whether BTLA overexpression was sufficient to preserve immune tolerance in imDCs with exogenous CCR7 overexpression. Scanning electron microscopy and surface antigens analysis revealed that BTLA overexpression suppressed DC maturation, an effect further potentiated in CCR7 and BTLA cooverexpressing cells. Correspondingly, in vitro chemotaxis assays and mixed lymphocyte reactions demonstrated increased migratory potential and immune tolerance in CCR7 and BTLA coexpressing cells. Furthermore, CCR7 and BTLA cooverexpressed imDCs suppressed IFN-γ and IL-17 expression and promoted IL-4 and TGF-beta expression of lymphocyte, indicating an increase of T helper 2 (Th2) regulatory T cell (Treg). Thus, these data indicate that CCR7 and BTLA cooverexpression imparts an intermediate immune phenotype in imDCs when compared to that in CCR7- or BTLA-expressing counterparts that show a more immunocompetent or immunotolerant phenotype, respectively. All these results indicated that adenovirus-mediated CCR7 and BTLA overexpression could enhance immune tolerance and migration of imDCs. Our study provides a basis for further studies on imDCs in immune tolerance, with the goal of developing effective cellular immunotherapies for transplant recipients.
Collapse
|
15
|
Zerif E, Maalem A, Gaudreau S, Guindi C, Ramzan M, Véroneau S, Gris D, Stankova J, Rola-Pleszczynski M, Mourad W, Dupuis G, Amrani A. Constitutively active Stat5b signaling confers tolerogenic functions to dendritic cells of NOD mice and halts diabetes progression. J Autoimmun 2017; 76:63-74. [DOI: 10.1016/j.jaut.2016.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/30/2016] [Accepted: 09/04/2016] [Indexed: 12/23/2022]
|
16
|
Shen L, Krauthäuser S, Fischer K, Hobernik D, Abassi Y, Dzionek A, Nikolaev A, Voltz N, Diken M, Krummen M, Montermann E, Tubbe I, Lorenz S, Strand D, Schild H, Grabbe S, Bros M. Vaccination with trifunctional nanoparticles that address CD8 + dendritic cells inhibits growth of established melanoma. Nanomedicine (Lond) 2016; 11:2647-2662. [PMID: 27628310 DOI: 10.2217/nnm-2016-0174] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM We wanted to assess the potency of a trifunctional nanoparticle (NP) that targeted and activated CD8+ dendritic cells (DC) and delivered an antigen to induce antitumor responses. MATERIALS & METHODS The DC targeting and activating properties of ferrous NPs conjugated with immunostimulatory CpG-oligonucleotides, anti-DEC205 antibody and ovalbumin (OVA) as a model antigen to induce antigen-specific T-cell responses and antitumor responses were analyzed. RESULTS OVA-loaded NP conjugated with immunostimulatory CpG-oligonucleotides and anti-DEC205 antibody efficiently targeted and activated CD8+ DC in vivo, and induced strong OVA-specific T-cell activation. Vaccination of B16/OVA tumor-burdened mice with this NP formulation resulted in tumor growth arrest. CONCLUSION CD8+ DC-targeting trifunctional nanocarriers bear significant potential for antitumor immunotherapy.
Collapse
Affiliation(s)
- Limei Shen
- Department of Dermatology, University Medical Center Mainz, Germany
| | | | - Karl Fischer
- Institute for Physical Chemistry, Johannes Gutenberg-University Mainz, Germany
| | | | - Yasmin Abassi
- Department of Internal Medicine III, Division of Translational & Experimental Oncology, University Medical Center Mainz, Germany
| | | | - Alexej Nikolaev
- Institute for Molecular Medicine, University Medical Center Mainz, Germany
| | - Nicole Voltz
- Department of Dermatology, University Medical Center Mainz, Germany
| | - Mustafa Diken
- TRON-Translational Oncology at the University Medical Center of Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Mathias Krummen
- Department of Dermatology, University Medical Center Mainz, Germany
| | | | - Ingrid Tubbe
- Department of Dermatology, University Medical Center Mainz, Germany
| | - Steffen Lorenz
- Imaging Core Facility, University Medical Center Mainz, Germany
| | - Dennis Strand
- Imaging Core Facility, University Medical Center Mainz, Germany
| | - Hansjörg Schild
- Institute of Immunology, University Medical Center Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Germany
| |
Collapse
|
17
|
Yoo S, Ha SJ. Generation of Tolerogenic Dendritic Cells and Their Therapeutic Applications. Immune Netw 2016; 16:52-60. [PMID: 26937232 PMCID: PMC4770100 DOI: 10.4110/in.2016.16.1.52] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/05/2016] [Accepted: 02/07/2016] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells (APCs) that bridge innate and adaptive immune responses, thereby leading to immune activation. DCs have been known to recognize pathogen-associated molecular patterns such as lipopolysaccharides (LPS) and nucleic acids via their pattern recognition receptors, which trigger signaling of their maturation and effector functions. Furthermore, DCs take up and process antigens as a form of peptide loaded on the major histocompatibility complex (MHC) and present them to T cells, which are responsible for the adaptive immune response. Conversely, DCs can also play a role in inducing immune suppression under specific circumstances. From this perspective, the role of DCs is related to tolerance rather than immunity. Immunologists refer to these special DCs as tolerogenic DCs (tolDCs). However, the definition of tolDCs is controversial, and there is limited information on their development and characteristics. In this review, we discuss the current concept of tolDCs, cutting-edge methods for generating tolDCs in vitro, and future applications of tolDCs, including clinical use.
Collapse
Affiliation(s)
- Seungbo Yoo
- System Immunology Laboratory, Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Sang-Jun Ha
- System Immunology Laboratory, Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
18
|
Abstract
As a common procedure in dentistry for replacing a missing tooth, allogenic tooth transplantation has encountered many difficulties in the clinical application because of immunological rejection. It is hypothesized that immature dendritic cell injection might be a potential alternative method to avoid or alleviate immunological rejection in allogenic tooth transplantation. To test this hypothesis, a mouse model of allogenic and autogeneic tooth transplantation was to established test the immunosuppressive effect of immature dendritic cells (imDCs) derived from donor bone marrows on transplant rejection in allogenic tooth transplantation. 2 × 10(6) imDCs generated with 50 U/ml GM-CSF were injected to each recipient mouse by two ways: tail vein injection 7 days before transplantation or regional dermal injection at day 0 and day 3 after transplantation. Groups of autogeneic tooth transplantation and allogenic tooth transplantation without any treatment were set as control groups. The effects were evaluated with histopathology and immunohistochemistry. We found there was no obvious rejection in autogeneic tooth transplantation group; tail intravenous injection group showed obviously alleviated rejection while local injection group and none-treatment allogenic tooth transplantation group both showed severe rejection. Our results suggested that the rejection of allogenic tooth transplantation could be alleviated by tail vein injection of donor bone marrow-derived imDCs though it could not be completely eliminated. The clinical application of imDCs in allogenic tooth transplantation still needs further deep research.
Collapse
|
19
|
Thymus-deriving natural regulatory T cell generation in vitro: role of the source of activation signals. Immunol Lett 2014; 162:199-209. [PMID: 25445615 DOI: 10.1016/j.imlet.2014.10.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/09/2014] [Accepted: 10/21/2014] [Indexed: 12/20/2022]
Abstract
In this research we have examined different sources of activation signals in order to optimize culture conditions for in vitro generation of thymus-deriving natural regulatory T cells (nTregs). We have established a novel model using JAWS II dendritic cell line of immature phenotype and compared it to commonly used methods for the generation of Tregs from peripheral lymphoid organs or blood T cells. In our model the first activation signal is provided by anti-CD3 monoclonal antibodies while the second is delivered by costimulatory molecules expressed on JAWS II cells. The presence of JAWS II cells co-cultured in vitro with unsorted thymocytes directly isolated from the thymus gland creates environment favoring SP CD4+ differentiation, provides the apoptotic cells clearance, maintains the survival of thymocytes and facilitate nTreg generation. Moreover the usage of immature dendritic cells stimuli enables to conduct research on agents affecting nTreg survival, proliferation and development in conditions of cell-to-cell contact of undifferentiated thymocytes with dendritic cells.
Collapse
|
20
|
Han Y, Chen Z, Yang Y, Jiang Z, Gu Y, Liu Y, Lin C, Pan Z, Yu Y, Jiang M, Zhou W, Cao X. Human CD14+ CTLA-4+ regulatory dendritic cells suppress T-cell response by cytotoxic T-lymphocyte antigen-4-dependent IL-10 and indoleamine-2,3-dioxygenase production in hepatocellular carcinoma. Hepatology 2014; 59:567-79. [PMID: 23960017 DOI: 10.1002/hep.26694] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 08/11/2013] [Indexed: 12/17/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide with limited therapeutic options. HCC-induced immunosuppression often leads to ineffectiveness of immuno-promoting therapies. Currently, suppressing the suppressors has become the potential strategy for cancer immunotherapy. So, figuring out the immunosuppressive mechanisms induced and employed by HCC will be helpful to the design and application of HCC immunotherapy. Here, we identified one new subset of human CD14(+) CTLA-4(+) regulatory dendritic cells (CD14(+) DCs) in HCC patients, representing ∼13% of peripheral blood mononuclear cells. CD14(+) DCs significantly suppress T-cell response in vitro through interleukin (IL)-10 and indoleamine-2,3-dioxygenase (IDO). Unexpectedly, CD14(+) DCs expressed high levels of cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed death-1, and CTLA-4 was found to be essential to IL-10 and IDO production. So, we identified a novel human tumor-induced regulatory DC subset, which suppresses antitumor immune response through CTLA-4-dependent IL-10 and IDO production, thus indicating the important role of nonregulatory T-cell-derived CTLA-4 in tumor-immune escape or immunosuppression. CONCLUSIONS These data outline one mechanism for HCC to induce systemic immunosuppression by expanding CD14(+) DCs, which may contribute to HCC progression. This adds new insight to the mechanism for HCC-induced immunosuppression and may also provide a previously unrecognized target of immunotherapy for HCC.
Collapse
Affiliation(s)
- Yanmei Han
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wu CS, Li YR, Chen JJW, Chen YC, Chu CL, Pan IH, Wu YS, Lin CC. Antihelminthic niclosamide modulates dendritic cells activation and function. Cell Immunol 2013; 288:15-23. [PMID: 24561310 PMCID: PMC7094312 DOI: 10.1016/j.cellimm.2013.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/04/2013] [Accepted: 12/11/2013] [Indexed: 02/06/2023]
Abstract
Effect of niclosamide on the activation of LPS-stimulated BMDCs was investigated. Niclosamide decreased ability to stimulate antigen specific T cell proliferation. Niclosamide attenuated hapten induced contact hypersensitivity (CHS) in vivo. Blocking the MAPK and NF-κB contribute to the inhibitory effect of niclosamide.
Dendritic cells (DCs) link the sensing of the environment by the innate immune system to the initiation of adaptive immune responses. Accordingly, DCs are considered to be a major target in the development of immunomodulating compounds. In this study, the effect of niclosamide, a Food and Drug Administration-approved antihelminthic drug, on the activation of lipopolysaccharide (LPS)-stimulated murine bone marrow-derived DCs was examined. Our experimental results show that niclosamide reduced the pro-inflammatory cytokine and chemokine expression of LPS-activated DCs. In addition, niclosamide also affected the expression of MHC and costimulatory molecules and influenced the ability of the cells to take up antigens. Therefore, in mixed cell cultures composed of syngeneic OVA-specific T cells and DCs, niclosamide-treated DCs showed a decreased ability to stimulate T cell proliferation and IFN-γ production. Furthermore, intravenous injection of niclosamide also attenuated contact hypersensitivity (CHS) in mice during sensitization with 2,4-dinitro-1-fluorobenzene. Blocking the LPS-induced activation of MAPK-ERK, JNK and NF-κB may contribute to the inhibitory effect of niclosamide on DC activation. Collectively, our findings suggest that niclosamide can manipulate the function of DCs. These results provide new insight into the immunopharmacological role of niclosamide and suggest that it may be useful for the treatment of chronic inflammatory disorders or DC-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Chieh-Shan Wu
- Department of Dermatology, Kaohsiung Veterans General Hospital, Taiwan, ROC; Department of Dermatology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Taiwan, ROC; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Taiwan, ROC
| | - Yi-Rong Li
- Institute of Biomedical Science, National Chung-Hsing University, Taichung, Taiwan, ROC
| | - Jeremy J W Chen
- Institute of Biomedical Science, National Chung-Hsing University, Taichung, Taiwan, ROC; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taiwan, ROC
| | - Ying-Che Chen
- Institute of Biomedical Science, National Chung-Hsing University, Taichung, Taiwan, ROC
| | - Chiang-Liang Chu
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - I-Hong Pan
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan, ROC
| | - Yu-Shan Wu
- Department of Chemistry, Tunghai University, Taichung, Taiwan, ROC
| | - Chi-Chen Lin
- Institute of Biomedical Science, National Chung-Hsing University, Taichung, Taiwan, ROC; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taiwan, ROC; Department of Medical Research and Education, Taichung Veterans General Hospital, Taichung, Taiwan, ROC; Division of Chest Medicine, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan, ROC.
| |
Collapse
|
22
|
Morel PA. Dendritic cell subsets in type 1 diabetes: friend or foe? Front Immunol 2013; 4:415. [PMID: 24367363 PMCID: PMC3853773 DOI: 10.3389/fimmu.2013.00415] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/13/2013] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes (T1D) is a T cell mediated autoimmune disease characterized by immune mediated destruction of the insulin-producing β cells in the islets of Langerhans. Dendritic cells (DC) have been implicated in the pathogenesis of T1D and are also used as immunotherapeutic agents. Plasmacytoid (p)DC have been shown to have both protective and pathogenic effects and a newly described merocytic DC population has been shown to break tolerance in the mouse model of T1D, the non-obese diabetic (NOD) mouse. We have used DC populations to prevent the onset of T1D in NOD mice and clinical trials of DC therapy in T1D diabetes have been initiated. In this review we will critically examine the recent published literature on the role of DC subsets in the induction and regulation of the autoimmune response in T1D.
Collapse
Affiliation(s)
- Penelope A Morel
- Department of Immunology, University of Pittsburgh , Pittsburgh, PA , USA
| |
Collapse
|
23
|
A trifunctional dextran-based nanovaccine targets and activates murine dendritic cells, and induces potent cellular and humoral immune responses in vivo. PLoS One 2013; 8:e80904. [PMID: 24339889 PMCID: PMC3855172 DOI: 10.1371/journal.pone.0080904] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 10/17/2013] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) constitute an attractive target for specific delivery of nanovaccines for immunotherapeutic applications. Here we tested nano-sized dextran (DEX) particles to serve as a DC-addressing nanocarrier platform. Non-functionalized DEX particles had no immunomodulatory effect on bone marrow (BM)-derived murine DCs in vitro. However, when adsorbed with ovalbumine (OVA), DEX particles were efficiently engulfed by BM-DCs in a mannose receptor-dependent manner. A DEX-based nanovaccine containing OVA and lipopolysaccharide (LPS) as a DC stimulus induced strong OVA peptide-specific CD4(+) and CD8(+) T cell proliferation both in vitro and upon systemic application in mice, as well as a robust OVA-specific humoral immune response (IgG1>IgG2a) in vivo. Accordingly, this nanovaccine also raised both a more pronounced delayed-type hypersensitivity response and a stronger induction of cytotoxic CD8(+) T cells than obtained upon administration of OVA and LPS in soluble form. Therefore, DEX-based nanoparticles constitute a potent, versatile and easy to prepare nanovaccine platform for immunotherapeutic approaches.
Collapse
|
24
|
Zhou F, Ciric B, Zhang GX, Rostami A. Immune tolerance induced by intravenous transfer of immature dendritic cells via up-regulating numbers of suppressive IL-10(+) IFN-γ(+)-producing CD4(+) T cells. Immunol Res 2013; 56:1-8. [PMID: 23292714 DOI: 10.1007/s12026-012-8382-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) regulate immunity and immune tolerance in vivo. However, the mechanisms of DC-mediated tolerance have not been fully elucidated. Here, we demonstrate that intravenous (i.v.) transfer of bone marrow-derived DCs pulsed with myelin oligodendrocyte glycoprotein (MOG) peptide blocks the development of experimental autoimmune encephalomyelitis in C57BL/6J mice. i.v. transfer of MOG-pulsed DCs leads to the down-regulation of the production of IL-17A and IFN-γ and up-regulation of IL-10 secretion. The development of regulatory T cells (Tregs) is facilitated via up-regulation of FoxP3 expression and production of IL-10. The number of suppressive CD4(+)IL-10(+)IFN-γ(+) T cells is also improved. The expression of OX40, CD154, and CD28 is down-regulated, but the expression of CD152, CD80, PD-1, ICOS, and BTLA is up-regulated on CD4(+) T cells after i.v. transfer of immature DCs. The expression of CCR4, CCR5, and CCR7 on CD4(+) T cells is also improved. Our results suggest that immature DCs may induce tolerance via facilitating the development of CD4(+)FoxP3(+) Tregs and suppressive CD4(+)IL-10(+)IFN-γ(+) T cells in vivo.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|
25
|
Teijeira A, Garasa S, Peláez R, Azpilikueta A, Ochoa C, Marré D, Rodrigues M, Alfaro C, Aubá C, Valitutti S, Melero I, Rouzaut A. Lymphatic endothelium forms integrin-engaging 3D structures during DC transit across inflamed lymphatic vessels. J Invest Dermatol 2013; 133:2276-85. [PMID: 23528818 DOI: 10.1038/jid.2013.152] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 02/18/2013] [Accepted: 03/08/2013] [Indexed: 11/09/2022]
Abstract
Dendritic cell (DC) transmigration across the lymphatic endothelium is critical for the initiation and sustenance of immune responses. Under noninflammatory conditions, DC transit across the lymphatic endothelial cell (LEC) has been shown to be integrin independent. In contrast, there is increasing evidence for the participation of integrins and their ligands in DC transit across lymphatic endothelium under inflammation. In this sense, we describe the formation of ICAM-1 (CD54)-enriched three-dimensional structures on LEC/DC contacts, as these DCs adhere to inflamed skin lymphatic vessels and transmigrate into them. In vitro imaging revealed that under inflammation ICAM-1 accumulated on microvilli projections surrounding 60% of adhered DCs. In contrast, these structures were scarcely formed in noninflammatory conditions. Furthermore, ICAM-1-enriched microvilli were important in promoting DC transendothelial migration and DC crawling over the LEC surface. Microvilli formation was dependent on the presence of β-integrins on the DC side and on integrin conformational affinity to ligand. Finally, we observed that LEC microvilli structures appeared in close vicinity of CCL21 depots and that their assembly was partially inhibited by CCL21-neutralizing antibodies. Therefore, under inflammatory conditions, integrin ligands form three-dimensional membrane projections around DCs. These structures offer docking sites for DC transit from the tissue toward the lymphatic vessel lumen.
Collapse
Affiliation(s)
- Alvaro Teijeira
- Department of Oncology, Center for Applied Medical Research, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Activation-induced FoxP3 expression regulates cytokine production in conventional T cells stimulated with autologous dendritic cells. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1583-92. [PMID: 22855393 DOI: 10.1128/cvi.00308-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A defining feature of dendritic cells (DCs) is their ability to induce the proliferation of autologous T cells in the absence of foreign antigen-a process termed the "autologous mixed leukocyte reaction" (AMLR). We report that equine monocyte-derived DCs, but not macrophages, are potent inducers of the AMLR. The response is contact dependent and major histocompatibility complex class II dependent and primarily involves CD3(+) CD4(+) CD8(-) T cells. Upon stimulation with DCs or the mitogen concanavalin A, a subset of the proliferating T cells expresses the regulatory T-cell (Treg) transcription factor FoxP3. Although many of these FoxP3(+) T cells are capable of producing the effector cytokines interleukin-4 (IL-4) and gamma interferon (IFN-γ), they are more likely to produce IL-10 and less likely to produce IFN-γ than equivalent FoxP3(-) cells. Therefore, FoxP3 expression is an inherent component of equine T cell activation and is associated with a more immunosuppressive cytokine profile. These results confirm that FoxP3 expression in the horse, in contrast to the mouse, is regulated similarly to FOXP3 expression in humans and provide evidence that FoxP3 expression by conventional T cells may help regulate the developing immune response.
Collapse
|
27
|
Lutz MB. Therapeutic potential of semi-mature dendritic cells for tolerance induction. Front Immunol 2012; 3:123. [PMID: 22629255 PMCID: PMC3355325 DOI: 10.3389/fimmu.2012.00123] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/30/2012] [Indexed: 12/23/2022] Open
Abstract
Dendritic cells (DCs) are major players in the control of adaptive tolerance and immunity. Therefore, their specific generation and adoptive transfer into patients or their in vivo targeting is attractive for clinical applications. While injections of mature immunogenic DCs are tested in clinical trials, tolerogenic DCs still are awaiting this step. Besides the tolerogenic potential of immature DCs, also semi-mature DCs can show tolerogenic activity but both types also bear unfavorable features. Optimal tolerogenic DCs, their molecular tool bar, and their use for specific diseases still have to be defined. Here, the usefulness of in vitro generated and adoptively transferred semi-mature DCs for tolerance induction is outlined. The in vivo targeting of semi-mature DCs as represented by steady state migratory DCs are discussed for treatment of autoimmune diseases and allergies. First clinical trials with transcutaneous allergen application may point to their therapeutic use in the future.
Collapse
Affiliation(s)
- Manfred B Lutz
- Institute of Virology and Immunobiology, University of Wuerzburg Wuerzburg, Germany
| |
Collapse
|