1
|
Liu W, Yao Y, Meng J, Qian S, Han Y, Zhou L, Wang T, Chen Y, Chen L, Ye Z, Xu L, Zhang M, Qiu J, Han T, Liu X, Kuang C, Ding Z, Liu Z. Architecture-driven quantitative nanoscopy maps cytoskeleton remodeling. Proc Natl Acad Sci U S A 2024; 121:e2410688121. [PMID: 39374388 PMCID: PMC11494298 DOI: 10.1073/pnas.2410688121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/01/2024] [Indexed: 10/09/2024] Open
Abstract
Cytoskeleton remodeling which generates force and orchestrates signaling and trafficking to govern cell migration remains poorly understood, partly due to a lack of an investigation tool with high system flexibility, spatiotemporal resolution, and computational sensitivity. Herein, we developed a multimodal superresolution imaging system-based architecture-driven quantitative (ADQ) framework in spatiotemporal-angular hyperspace to enable both identification of the optimal imaging mode with well-balanced fidelity and phototoxicity and accurate postcharacterization of microtubule remodeling. In the ADQ framework, a pixel/voxel-wise metric reflecting heterogeneous intertubule alignment was proposed with improved sensitivity over previous efforts and further incorporated with temporal features to map dynamic microtubule rearrangements. The ADQ framework was verified by assessing microtubule remodeling in drug-induced (de)polymerization, lysosome transport, and migration. Different remodeling patterns from two migration modes were successfully revealed by the ADQ framework, with a front-rear polarization for individual directed migration and a contact site-centered polarization for cell-cell interaction-induced migration in an immune response model. Meanwhile, these migration modes were found to have consistent orientation changes, which exhibited the potential of predicting migration trajectory.
Collapse
Affiliation(s)
- Wenjie Liu
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
- Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, Zhejiang311200, China
- The Kavli Institute for Nanoscience Discovery, University of Oxford, OxfordOX1 3QU, United Kingdom
| | - Yushi Yao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
| | - Jia Meng
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Shuhao Qian
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Yubing Han
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Lingxi Zhou
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Tao Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
| | - Youhua Chen
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Lingmei Chen
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Zitong Ye
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Liang Xu
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Meng Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei430074, China
| | - Jianrong Qiu
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Tao Han
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Xu Liu
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
- Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, Zhejiang311200, China
| | - Cuifang Kuang
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
- Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, Zhejiang311200, China
| | - Zhihua Ding
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
- Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, Zhejiang311200, China
| | - Zhiyi Liu
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, Zhejiang310027, China
- Jiaxing Key Laboratory of Photonic Sensing & Intelligent Imaging, Intelligent Optics & Photonics Research Center, Jiaxing Research Institute Zhejiang University, Jiaxing314000, China
| |
Collapse
|
2
|
Immune infiltration could predict the efficacy of short-term radiotherapy in patients with cervical cancer. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 25:1353-1367. [PMID: 36510039 DOI: 10.1007/s12094-022-03033-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Radiotherapy is the main treatment for cervical cancer. It is usually applied alone or in combination with surgery and/or chemotherapy. To explore the association between immune microenvironment of cervical cancer and radiotherapy response, we collected 20 paired cervical cancer tumor samples before and after radiotherapy and partial clinical information. With paired-end RNA-seq, we quantified the immune infiltration and tumor purity of these samples, and obtained 6350 differentially expressed genes before and after radiotherapy. With the help of R language, the function enrichment analysis and 22 immune cells infiltration analysis were carried out. Moreover, we built a random forest model based on the immune microenvironment to predict the short-term efficacy of radiotherapy. We found that the effect of radiotherapy on the immune microenvironment of stage III and IV cervical cancer patients was weaker than that of stage I and II cervical cancer patients. Radiotherapy can significantly reduce the tumor purity and increase immune infiltration. The proportions of the immune infiltrating cells are predictive of the radiotherapy efficacy. In addition, the local mucositis caused by radiotherapy can improve the curative effect of radiotherapy.
Collapse
|
3
|
van der Vegt SA, Wang YJ, Polonchuk L, Wang K, Waters SL, Baker RE. A model-informed approach to assess the risk of immune checkpoint inhibitor-induced autoimmune myocarditis. Front Pharmacol 2022; 13:966180. [PMID: 36249751 PMCID: PMC9555336 DOI: 10.3389/fphar.2022.966180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs), as a novel immunotherapy, are designed to modulate the immune system to attack malignancies. Despite their promising benefits, immune-related adverse events (IRAEs) may occur, and incidences are bound to increase with surging demand of this class of drugs in treating cancer. Myocarditis, although rare compared to other IRAEs, has a significantly higher fatal frequency. Due to the overwhelming complexity of the immune system, this condition is not well understood, despite the significant research efforts devoted to it. To better understand the development and progression of autoimmune myocarditis and the roles of ICIs therein, we suggest a new approach: mathematical modelling. Mathematical modelling of myocarditis has enormous potential to determine which parts of the immune system are critical to the development and progression of the disease, and therefore warrant further investigation. We provide the immunological background needed to develop a mathematical model of this disease and review relevant existing models of immunology that serve as the mathematical inspiration needed to develop this field.
Collapse
Affiliation(s)
- Solveig A. van der Vegt
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
- *Correspondence: Solveig A. van der Vegt,
| | - Ying-Jie Wang
- Department of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre of Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Liudmila Polonchuk
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Ken Wang
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Sarah L. Waters
- Oxford Centre for Industrial and Applied Mathematics, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Ruth E. Baker
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Hu W, Wang Y, Chen J, Yu P, Tang F, Hu Z, Zhou J, Liu L, Qiu W, Ye Y, Jia Y, Zhou S, Long J, Zeng Z. Regulation of biomaterial implantation-induced fibrin deposition to immunological functions of dendritic cells. Mater Today Bio 2022; 14:100224. [PMID: 35252832 PMCID: PMC8894278 DOI: 10.1016/j.mtbio.2022.100224] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 11/04/2022] Open
Abstract
The performance of implanted biomaterials is largely determined by their interaction with the host immune system. As a fibrous-like 3D network, fibrin matrix formed at the interfaces of tissue and material, whose effects on dendritic cells (DCs) remain unknown. Here, a bone plates implantation model was developed to evaluate the fibrin matrix deposition and DCs recruitment in vivo. The DCs responses to fibrin matrix were further analyzed by a 2D and 3D fibrin matrix model in vitro. In vivo results indicated that large amount of fibrin matrix deposited on the interface between the tissue and bone plates, where DCs were recruited. Subsequent in vitro testing denoted that DCs underwent significant shape deformation and cytoskeleton reorganization, as well as mechanical property alteration. Furthermore, the immune function of imDCs and mDCs were negatively and positively regulated, respectively. The underlying mechano-immunology coupling mechanisms involved RhoA and CDC42 signaling pathways. These results suggested that fibrin plays a key role in regulating DCs immunological behaviors, providing a valuable immunomodulatory strategy for tissue healing, regeneration and implantation.
Collapse
|
5
|
Mathematical modelling of autoimmune myocarditis and the effects of immune checkpoint inhibitors. J Theor Biol 2022; 537:111002. [PMID: 35007511 DOI: 10.1016/j.jtbi.2021.111002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 12/26/2022]
Abstract
Autoimmune myocarditis is a rare, but frequently fatal, side effect of immune checkpoint inhibitors (ICIs), a class of cancer therapies. Despite extensive experimental work on the causes, development and progression of this disease, much still remains unknown about the importance of the different immunological pathways involved. We present a mathematical model of autoimmune myocarditis and the effects of ICIs on its development and progression to either resolution or chronic inflammation. From this, we gain a better understanding of the role of immune cells, cytokines and other components of the immune system in driving the cardiotoxicity of ICIs. We parameterise the model using existing data from the literature, and show that qualitative model behaviour is consistent with disease characteristics seen in patients in an ICI-free context. The bifurcation structures of the model show how the presence of ICIs increases the risk of developing autoimmune myocarditis. This predictive modelling approach is a first step towards determining treatment regimens that balance the benefits of treating cancer with the risk of developing autoimmune myocarditis.
Collapse
|
6
|
Immunostimulatory Potential of Extracellular Vesicles Isolated from an Edible Plant, Petasites japonicus, via the Induction of Murine Dendritic Cell Maturation. Int J Mol Sci 2021; 22:ijms221910634. [PMID: 34638974 PMCID: PMC8508627 DOI: 10.3390/ijms221910634] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) have recently been isolated from different plants. Plant-derived EVs have been proposed as potent therapeutics and drug-delivery nanoplatforms for delivering biomolecules, including proteins, RNAs, DNAs, and lipids. Herein, Petasites japonicus-derived EVs (PJ-EVs) were isolated through a series of centrifugation steps and characterized using dynamic light scattering and transmission electron microscopy. Immunomodulatory effects of PJ-EVs were assessed using dendritic cells (DCs). PJ-EVs exhibited a spherical morphology with an average size of 122.6 nm. They induced the maturation of DCs via an increase in the expression of surface molecules (CD80, CD86, MHC-I, and MHC-II), production of Th1-polarizing cytokines (TNF-α and IL-12p70), and antigen-presenting ability; however, they reduced the antigen-uptake ability. Furthermore, maturation of DCs induced by PJ-EVs was dependent on the activation and phosphorylation of MAPK and NF-κB signal pathways. Notably, PJ-EV-treated DCs strongly induced the proliferation and differentiation of naïve T cells toward Th1-type T cells and cytotoxic CD8+ T cells along with robust secretion of IFN-γ and IL-2. In conclusion, our study indicates that PJ-EVs can be potent immunostimulatory candidates with an ability of strongly inducing the maturation of DCs.
Collapse
|
7
|
Nüssing S, Trapani JA, Parish IA. Revisiting T Cell Tolerance as a Checkpoint Target for Cancer Immunotherapy. Front Immunol 2020; 11:589641. [PMID: 33072137 PMCID: PMC7538772 DOI: 10.3389/fimmu.2020.589641] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/04/2020] [Indexed: 12/30/2022] Open
Abstract
Immunotherapy has revolutionized the treatment of cancer. Nevertheless, the majority of patients do not respond to therapy, meaning a deeper understanding of tumor immune evasion strategies is required to boost treatment efficacy. The vast majority of immunotherapy studies have focused on how treatment reinvigorates exhausted CD8+ T cells within the tumor. In contrast, how therapies influence regulatory processes within the draining lymph node is less well studied. In particular, relatively little has been done to examine how tumors may exploit peripheral CD8+ T cell tolerance, an under-studied immune checkpoint that under normal circumstances prevents detrimental autoimmune disease by blocking the initiation of T cell responses. Here we review the therapeutic potential of blocking peripheral CD8+ T cell tolerance for the treatment of cancer. We first comprehensively review what has been learnt about the regulation of CD8+ T cell peripheral tolerance from the non-tumor models in which peripheral tolerance was first defined. We next consider how the tolerant state differs from other states of negative regulation, such as T cell exhaustion and senescence. Finally, we describe how tumors hijack the peripheral tolerance immune checkpoint to prevent anti-tumor immune responses, and argue that disruption of peripheral tolerance may contribute to both the anti-cancer efficacy and autoimmune side-effects of immunotherapy. Overall, we propose that a deeper understanding of peripheral tolerance will ultimately enable the development of more targeted and refined cancer immunotherapy approaches.
Collapse
Affiliation(s)
- Simone Nüssing
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Joseph A Trapani
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Ian A Parish
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
8
|
Hegde S, Krisnawan VE, Herzog BH, Zuo C, Breden MA, Knolhoff BL, Hogg GD, Tang JP, Baer JM, Mpoy C, Lee KB, Alexander KA, Rogers BE, Murphy KM, Hawkins WG, Fields RC, DeSelm CJ, Schwarz JK, DeNardo DG. Dendritic Cell Paucity Leads to Dysfunctional Immune Surveillance in Pancreatic Cancer. Cancer Cell 2020; 37:289-307.e9. [PMID: 32183949 PMCID: PMC7181337 DOI: 10.1016/j.ccell.2020.02.008] [Citation(s) in RCA: 261] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/04/2019] [Accepted: 02/14/2020] [Indexed: 12/26/2022]
Abstract
Here, we utilized spontaneous models of pancreatic and lung cancer to examine how neoantigenicity shapes tumor immunity and progression. As expected, neoantigen expression during lung adenocarcinoma development leads to T cell-mediated immunity and disease restraint. By contrast, neoantigen expression in pancreatic ductal adenocarcinoma (PDAC) results in exacerbation of a fibro-inflammatory microenvironment that drives disease progression and metastasis. Pathogenic TH17 responses are responsible for this neoantigen-induced tumor progression in PDAC. Underlying these divergent T cell responses in pancreas and lung cancer are differences in infiltrating conventional dendritic cells (cDCs). Overcoming cDC deficiency in early-stage PDAC leads to disease restraint, while restoration of cDC function in advanced PDAC restores tumor-restraining immunity and enhances responsiveness to radiation therapy.
Collapse
Affiliation(s)
- Samarth Hegde
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Varintra E Krisnawan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett H Herzog
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chong Zuo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marcus A Breden
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett L Knolhoff
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Graham D Hogg
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jack P Tang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John M Baer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cedric Mpoy
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kyung Bae Lee
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katherine A Alexander
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Buck E Rogers
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William G Hawkins
- Department of Surgery, Barnes-Jewish Hospital, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Ryan C Fields
- Department of Surgery, Barnes-Jewish Hospital, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Carl J DeSelm
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Julie K Schwarz
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - David G DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA.
| |
Collapse
|
9
|
Trypanosoma cruzi Mexican Strains Differentially Modulate Surface Markers and Cytokine Production in Bone Marrow-Derived Dendritic Cells from C57BL/6 and BALB/c Mice. Mediators Inflamm 2019; 2019:7214798. [PMID: 31636507 PMCID: PMC6766131 DOI: 10.1155/2019/7214798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/08/2019] [Accepted: 07/29/2019] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are a type of antigen-presenting cells that play an important role in the immune response against Trypanosoma cruzi, the causative agent of Chagas disease. In vitro and in vivo studies have shown that the modulation of these cells by this parasite can directly affect the innate and acquired immune response of the host in order to facilitate its biological cycle and the spreading of the species. Many studies show the mechanisms by which T. cruzi modulates DCs, but the interaction of these cells with the Mexican strains of T. cruzi such as Ninoa and INC5 has not yet been properly investigated. Here, we evaluated whether Ninoa and INC5 strains evaded the immunity of their hosts by modulating the biology and function of murine DCs. The CL-Brener strain was used as the reference strain. Herein, it was demonstrated that Ninoa was more infective toward bone marrow-derived dendritic cells (BMDCs) than INC5 and CL-Brener strains in both BMDCs of BALB/c and C57BL/6 mice. Mexican strains of T. cruzi induced different cytokine patterns. In BMDCs obtained from BALB/c mice, Ninoa strain led to the reduction in IL-6 and increased IL-10 production, while in C57BL/6 mice Ninoa strain considerably increased the productions of TNF-α and IL-10. Also, Ninoa and INC5 differentially modulated BMDC expressions of MHC-II, TLR2, and TLR4 in both BALB/c and C57BL/6 mice compared to Brazilian strain CL-Brener. These results indicate that T. cruzi Mexican strains differentially infect and modulate MHC-II, toll-like receptors, and cytokine production in DCs obtained from C57BL/6 and BALB/c mice, suggesting that these strains have developed particular modulatory strategies to disrupt DCs and, consequently, the host immune responses.
Collapse
|
10
|
Huang WZ, Hu WH, Wang Y, Chen J, Hu ZQ, Zhou J, Liu L, Qiu W, Tang FZ, Zhang SC, Ouyang Y, Ye YN, Xu GQ, Long JH, Zeng Z. A Mathematical Modelling of Initiation of Dendritic Cells-Induced T Cell Immune Response. Int J Biol Sci 2019; 15:1396-1403. [PMID: 31337970 PMCID: PMC6643141 DOI: 10.7150/ijbs.33412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/19/2019] [Indexed: 01/27/2023] Open
Abstract
Dendritic cells (DCs) are the most potent specialized antigen-presenting cells as now known, which play a crucial role in initiating and amplifying both the innate and adaptive immune responses. Immunologically, the motilities and T cell activation capabilities of DCs are closely related to the resulting immune responses. However, due to the complexity of the immune system, the dynamic changes in the number of cells during the peripheral tissue (e.g. skin and mucosa) immune response induced by DCs are still poorly understood. Therefore, this study simulated dynamic number changes of DCs and T cells in this process by constructing several ordinary differential equations and setting the initial conditions of the functions and parameters. The results showed that these equations could simulate dynamic numerical changes of DCs and T cells in peripheral tissue and lymph node, which was in accordance with the physiological conditions such as the duration of immune response, the proliferation rates and the motilities of DCs and T cells. This model provided a theoretical reference for studying the immunologic functions of DCs and practical guidance for the clinical DCs-based therapy against immune-related diseases.
Collapse
Affiliation(s)
- Wen-zhu Huang
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Wen-hui Hu
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Yun Wang
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Department of Immunology, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Jin Chen
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Zu-quan Hu
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Jing Zhou
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Lina Liu
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Wei Qiu
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Fu-zhou Tang
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Shi-chao Zhang
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Yan Ouyang
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Yuan-nong Ye
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Department of Immunology, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Guo-qiang Xu
- Department of Immunology, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Jin-hua Long
- Department of Head & Neck, Affiliated Tumor Hospital, Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Zhu Zeng
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Department of Immunology, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, P.R. China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, P.R. China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, P.R. China
| |
Collapse
|
11
|
Miyazawa R, Murata N, Matsuura Y, Shibasaki Y, Yabu T, Nakanishi T. Peculiar Expression of CD3-Epsilon in Kidney of Ginbuna Crucian Carp. Front Immunol 2018; 9:1321. [PMID: 29951063 PMCID: PMC6008321 DOI: 10.3389/fimmu.2018.01321] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/28/2018] [Indexed: 12/29/2022] Open
Abstract
TCR/CD3 complex is composed of the disulfide-linked TCR-αβ heterodimer that recognizes the antigen as a peptide presented by the MHC, and non-covalently paired CD3γε- and δε-chains together with disulfide-linked ζ-chain homodimers. The CD3 chains play key roles in T cell development and T cell activation. In the present study, we found nor or extremely lower expression of CD3ε in head- and trunk-kidney lymphocytes by flow cytometric analysis, while CD3ε was expressed at the normal level in lymphocytes from thymus, spleen, intestine, gill, and peripheral blood. Furthermore, CD4-1+ and CD8α+ T cells from kidney express Zap-70, but not CD3ε, while the T cells from other tissues express both Zap-70 and CD3ε, although expression of CD3ε was low. Quantitative analysis of mRNA expression revealed that the expression level of T cell-related genes including tcrb, cd3ε, zap-70, and lck in CD4-1+ and CD8α+ T cells was not different between kidney and spleen. Western blot analysis showed that CD3ε band was detected in the cell lysates of spleen but not kidney. To be interested, CD3ε-positive cells greatly increased after 24 h in in vitro culture of kidney leukocytes. Furthermore, expression of CD3ε in both transferred kidney and spleen leukocytes was not detected or very low in kidney, while both leukocytes expressed CD3ε at normal level in spleen when kidney and spleen leukocytes were injected into the isogeneic recipient. Lower expression of CD3ε was also found in kidney T lymphocytes of goldfish and carp. These results indicate that kidney lymphocytes express no or lower level of CD3ε protein in the kidney, although the mRNA of the gene was expressed. Here, we discuss this phenomenon from the point of function of kidney as reservoir for T lymphocytes in teleost, which lacks lymph node and bone marrow.
Collapse
Affiliation(s)
| | - Norifumi Murata
- Department of Veterinary Medicine, Nihon University, Fujisawa, Japan
| | - Yuta Matsuura
- Research Center for Fish Diseases, National Research Institute of Aquaculture, Minami-ise, Japan
| | - Yasuhiro Shibasaki
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Takeshi Yabu
- Department of Applied Biological Science, Nihon University, Fujisawa, Japan
| | | |
Collapse
|
12
|
Porphyromonas gulae Activates Unprimed and Gamma Interferon-Primed Macrophages via the Pattern Recognition Receptors Toll-Like Receptor 2 (TLR2), TLR4, and NOD2. Infect Immun 2017. [PMID: 28630066 DOI: 10.1128/iai.00282-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porphyromonas gulae is an anaerobic, Gram-negative coccobacillus that has been associated with periodontal disease in companion animals. The aims of this study were to analyze the ligation of pattern recognition receptors by P. gulae and the subsequent activation of macrophages. Exposure of HEK cells transfected with Toll-like receptors (TLRs) or NOD-like receptors to P. gulae resulted in the ligation of TLR2, TLR4, and NOD2. The effects of this engagement of receptors were investigated by measuring the synthesis of nitric oxide (NO), CD86 expression, and inflammatory cytokine production by wild-type, TLR2-/-, and TLR4-/- macrophages. The addition of P. gulae to unprimed and gamma interferon (IFN-γ)-primed (M1 phenotype) macrophages significantly increased the surface expression of CD86, but only M1 macrophages produced nitric oxide. P. gulae-induced expression of CD86 on unprimed macrophages was dependent on both TLR2 and TLR4, but CD86 expression and NO production in M1 macrophages were only TLR2 dependent. P. gulae induced an increase in secretion of interleukin-1α (IL-1α), IL-1β, IL-6, IL-12p70, IL-13, tumor necrosis factor alpha (TNF-α), granulocyte colony-stimulating factor (G-CSF), monocyte chemoattractant protein 1 (MCP-1), and macrophage inflammatory protein 1α (MIP-1α) by M1 macrophages compared to that by unprimed controls. Among these cytokines, secretion of IL-6 and TNF-α by M1 macrophages was dependent on either TLR2 or TLR4. Our data indicate that TLR2 and TLR4 are important for P. gulae activation of unprimed macrophages and that activation and effector functions induced in M1 macrophages by P. gulae are mainly dependent on TLR2. In conclusion, P. gulae induces a strong TLR2-dependent inflammatory M1 macrophage response which may be important in establishing the chronic inflammation associated with periodontal disease in companion animals.
Collapse
|
13
|
Ackerknecht M, Gollmer K, Germann P, Ficht X, Abe J, Fukui Y, Swoger J, Ripoll J, Sharpe J, Stein JV. Antigen Availability and DOCK2-Driven Motility Govern CD4+ T Cell Interactions with Dendritic Cells In Vivo. THE JOURNAL OF IMMUNOLOGY 2017; 199:520-530. [DOI: 10.4049/jimmunol.1601148] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 05/09/2017] [Indexed: 01/07/2023]
|
14
|
Mathematical Models for Immunology: Current State of the Art and Future Research Directions. Bull Math Biol 2016; 78:2091-2134. [PMID: 27714570 PMCID: PMC5069344 DOI: 10.1007/s11538-016-0214-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 09/26/2016] [Indexed: 01/01/2023]
Abstract
The advances in genetics and biochemistry that have taken place over the last 10 years led to significant advances in experimental and clinical immunology. In turn, this has led to the development of new mathematical models to investigate qualitatively and quantitatively various open questions in immunology. In this study we present a review of some research areas in mathematical immunology that evolved over the last 10 years. To this end, we take a step-by-step approach in discussing a range of models derived to study the dynamics of both the innate and immune responses at the molecular, cellular and tissue scales. To emphasise the use of mathematics in modelling in this area, we also review some of the mathematical tools used to investigate these models. Finally, we discuss some future trends in both experimental immunology and mathematical immunology for the upcoming years.
Collapse
|
15
|
Immunomodulation by Trypanosoma cruzi: toward understanding the association of dendritic cells with infecting TcI and TcII populations. J Immunol Res 2014; 2014:962047. [PMID: 25371910 PMCID: PMC4211313 DOI: 10.1155/2014/962047] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/12/2014] [Accepted: 09/09/2014] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are major immune components, and depending on how these cells are modulated, the protective host immune response changes drastically. Trypanosoma cruzi is a parasite with high genetic variability and modulates DCs by interfering with their capacity for antigen recognition, migration, and maturation. Despite recent efforts, the association between DCs and T. cruzi I (TcI) and TcII populations is unknown. Herein, it was demonstrated that AQ1.7 and MUTUM TcI strains present low rates of invasion of bone marrow-derived DCs, whereas the 1849 and 2369 TcII strains present higher rates. Whereas the four strains similarly induced the expression of PD-L1, the production and expression of IL-10 and TLR-2, respectively, in DCs were differentially increased. The production of TNF-α, IL-12, IL-6, and CCL2 and the expression of CD40, CD80, MHC-II, CCR5, and CCR7 changed depending on the strain. The 2369 strain yielded the most remarkable results because greater invasion correlated with an increase in the levels of anti-inflammatory molecules IL-10 and PD-L1 but not with a change in the levels of TNF-α, MHC-II, or CD40 molecules. These results suggest that T. cruzi strains belonging to different populations have evolved specific evasion strategies that subvert DCs and consequently the host response.
Collapse
|
16
|
Didierlaurent AM, Collignon C, Bourguignon P, Wouters S, Fierens K, Fochesato M, Dendouga N, Langlet C, Malissen B, Lambrecht BN, Garçon N, Van Mechelen M, Morel S. Enhancement of adaptive immunity by the human vaccine adjuvant AS01 depends on activated dendritic cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:1920-30. [PMID: 25024381 DOI: 10.4049/jimmunol.1400948] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Adjuvant System AS01 is a liposome-based vaccine adjuvant containing 3-O-desacyl-4'-monophosphoryl lipid A and the saponin QS-21. AS01 has been selected for the clinical development of several candidate vaccines including the RTS,S malaria vaccine and the subunit glycoprotein E varicella zoster vaccine (both currently in phase III). Given the known immunostimulatory properties of MPL and QS-21, the objective of this study was to describe the early immune response parameters after immunization with an AS01-adjuvanted vaccine and to identify relationships with the vaccine-specific adaptive immune response. Cytokine production and innate immune cell recruitment occurred rapidly and transiently at the muscle injection site and draining lymph node postinjection, consistent with the rapid drainage of the vaccine components to the draining lymph node. The induction of Ag-specific Ab and T cell responses was dependent on the Ag being injected at the same time or within 24 h after AS01, suggesting that the early events occurring postinjection were required for these elevated adaptive responses. In the draining lymph node, after 24 h, the numbers of activated and Ag-loaded monocytes and MHCII(high) dendritic cells were higher after the injection of the AS01-adjuvanted vaccine than after Ag alone. However, only MHCII(high) dendritic cells appeared efficient at and necessary for direct Ag presentation to T cells. These data suggest that the ability of AS01 to improve adaptive immune responses, as has been demonstrated in clinical trials, is linked to a transient stimulation of the innate immune system leading to the generation of high number of efficient Ag-presenting dendritic cells.
Collapse
Affiliation(s)
| | | | | | | | - Kaat Fierens
- Vlaams Instituut voor Biotechnologie Inflammation Research Center, Ghent University, 9052 Ghent, Belgium; and
| | | | | | | | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, INSERM U1104, Centre National de la Recherche Scientifique Unité Mixte de Recherche 7280, 13288 Marseille cedex 9, France
| | - Bart N Lambrecht
- Vlaams Instituut voor Biotechnologie Inflammation Research Center, Ghent University, 9052 Ghent, Belgium; and
| | | | | | - Sandra Morel
- GlaxoSmithKline Vaccines, 1330 Rixensart, Belgium
| |
Collapse
|
17
|
Leuenberger T, Pfueller CF, Luessi F, Bendix I, Paterka M, Prozorovski T, Treue D, Luenstedt S, Herz J, Siffrin V, Infante-Duarte C, Zipp F, Waiczies S. Modulation of dendritic cell immunobiology via inhibition of 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase. PLoS One 2014; 9:e100871. [PMID: 25013913 PMCID: PMC4094470 DOI: 10.1371/journal.pone.0100871] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 05/31/2014] [Indexed: 12/03/2022] Open
Abstract
The maturation status of dendritic cells determines whether interacting T cells are activated or if they become tolerant. Previously we could induce T cell tolerance by applying a 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase inhibitor (HMGCRI) atorvastatin, which also modulates MHC class II expression and has therapeutic potential in autoimmune disease. Here, we aimed at elucidating the impact of this therapeutic strategy on T cell differentiation as a consequence of alterations in dendritic cell function. We investigated the effect of HMGCRI during differentiation of peripheral human monocytes and murine bone marrow precursors to immature DC in vitro and assessed their phenotype. To examine the stimulatory and tolerogenic capacity of these modulated immature dendritic cells, we measured proliferation and suppressive function of CD4+ T cells after stimulation with the modulated immature dendritic cells. We found that an HMGCRI, atorvastatin, prevents dendrite formation during the generation of immature dendritic cells. The modulated immature dendritic cells had a diminished capacity to take up and present antigen as well as to induce an immune response. Of note, the consequence was an increased capacity to differentiate naïve T cells towards a suppressor phenotype that is less sensitive to proinflammatory stimuli and can effectively inhibit the proliferation of T effector cells in vitro. Thus, manipulation of antigen-presenting cells by HMGCRI contributes to an attenuated immune response as shown by promotion of T cells with suppressive capacities.
Collapse
Affiliation(s)
- Tina Leuenberger
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- Max Delbrueck Center for Molecular Medicine Berlin-Buch, Berlin, Germany
| | - Caspar F. Pfueller
- NeuroCure Clinical Research Center, Charité University Medicine Berlin, Berlin, Germany
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- * E-mail:
| | - Ivo Bendix
- Department of Pediatrics I/Neonatology, University Hospital Essen, Essen, Germany
| | - Magdalena Paterka
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- Max Delbrueck Center for Molecular Medicine Berlin-Buch, Berlin, Germany
| | - Timour Prozorovski
- Department of Neurology, Heinrich-Heine-University, Duesseldorf, Germany
| | - Denise Treue
- Institute of Pathology, Charité University Medicine Berlin, Berlin, Germany
| | - Sarah Luenstedt
- Max Delbrueck Center for Molecular Medicine Berlin-Buch, Berlin, Germany
| | - Josephine Herz
- Department of Pediatrics I/Neonatology, University Hospital Essen, Essen, Germany
| | - Volker Siffrin
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- Max Delbrueck Center for Molecular Medicine Berlin-Buch, Berlin, Germany
| | - Carmen Infante-Duarte
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- Max Delbrueck Center for Molecular Medicine Berlin-Buch, Berlin, Germany
| | - Sonia Waiczies
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
18
|
Fabrik I, Link M, Härtlova A, Dankova V, Rehulka P, Stulik J. Application of SILAC labeling to primary bone marrow-derived dendritic cells reveals extensive GM-CSF-dependent arginine metabolism. J Proteome Res 2013; 13:752-62. [PMID: 24308431 DOI: 10.1021/pr4007798] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although dendritic cells (DCs) control the priming of the adaptive immunity response, a comprehensive description of their behavior at the protein level is missing. The introduction of the quantitative proteomic technique of metabolic labeling (SILAC) into the field of DC research would therefore be highly beneficial. To achieve this, we applied SILAC labeling to primary bone marow-derived DCs (BMDCs). These cells combine both biological relevance and experimental feasibility, as their in vitro generation permits the use of (13)C/(15)N-labeled amino acids. Interestingly, BMDCs appear to exhibit a very active arginine metabolism. Using standard cultivation conditions, ∼20% of all protein-incorporated proline was a byproduct of heavy arginine degradation. In addition, the dissipation of (15)N from labeled arginine to the whole proteome was observed. The latter decreased the mass accuracy in MS and affected the natural isotopic distribution of peptides. SILAC-connected metabolic issues were shown to be enhanced by GM-CSF, which is used for the differentiation of DC progenitors. Modifications of the cultivation procedure suppressed the arginine-related effects, yielding cells with a proteome labeling efficiency of ≥90%. Importantly, BMDCs generated according to the new cultivation protocol preserved their resemblance to inflammatory DCs in vivo, as evidenced by their response to LPS treatment.
Collapse
Affiliation(s)
- Ivo Fabrik
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence , Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | | | | | | | | | | |
Collapse
|
19
|
Antigen availability determines CD8⁺ T cell-dendritic cell interaction kinetics and memory fate decisions. Immunity 2013; 39:496-507. [PMID: 24054328 DOI: 10.1016/j.immuni.2013.08.034] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 08/23/2013] [Indexed: 01/29/2023]
Abstract
T cells are activated by antigen (Ag)-bearing dendritic cells (DCs) in lymph nodes in three phases. The duration of the initial phase of transient, serial DC-T cell interactions is inversely correlated with Ag dose. The second phase, characterized by stable DC-T cell contacts, is believed to be necessary for full-fledged T cell activation. Here we have shown that this is not the case. CD8⁺ T cells interacting with DCs presenting low-dose, short-lived Ag did not transition to phase 2, whereas higher Ag dose yielded phase 2 transition. Both antigenic constellations promoted T cell proliferation and effector differentiation but yielded different transcriptome signatures at 12 hr and 24 hr. T cells that experienced phase 2 developed long-lived memory, whereas conditions without stable contacts yielded immunological amnesia. Thus, T cells make fate decisions within hours after Ag exposure, resulting in long-term memory or abortive effector responses, correlating with T cell-DCs interaction kinetics.
Collapse
|
20
|
Groom JR, Richmond J, Murooka TT, Sorensen EW, Sung JH, Bankert K, von Andrian UH, Moon JJ, Mempel TR, Luster AD. CXCR3 chemokine receptor-ligand interactions in the lymph node optimize CD4+ T helper 1 cell differentiation. Immunity 2012; 37:1091-103. [PMID: 23123063 DOI: 10.1016/j.immuni.2012.08.016] [Citation(s) in RCA: 334] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 08/06/2012] [Accepted: 08/29/2012] [Indexed: 12/24/2022]
Abstract
Differentiation of naive CD4(+) T cells into T helper (Th) cells is a defining event in adaptive immunity. The cytokines and transcription factors that control Th cell differentiation are understood, but it is not known how this process is orchestrated within lymph nodes (LNs). Here we have shown that the CXCR3 chemokine receptor was required for optimal generation of interferon-γ (IFN-γ)-secreting Th1 cells in vivo. By using a CXCR3 ligand reporter mouse, we found that stromal cells predominately expressed the chemokine ligand CXCL9 whereas hematopoietic cells expressed CXCL10 in LNs. Dendritic cell (DC)-derived CXCL10 facilitated T cell-DC interactions in LNs during T cell priming while both chemokines guided intranodal positioning of CD4(+) T cells to interfollicular and medullary zones. Thus, different chemokines acting on the same receptor can function locally to facilitate DC-T cell interactions and globally to influence intranodal positioning, and both functions contribute to Th1 cell differentiation.
Collapse
Affiliation(s)
- Joanna R Groom
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Manda K, Glasow A, Paape D, Hildebrandt G. Effects of ionizing radiation on the immune system with special emphasis on the interaction of dendritic and T cells. Front Oncol 2012; 2:102. [PMID: 22937525 PMCID: PMC3426842 DOI: 10.3389/fonc.2012.00102] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 07/31/2012] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs), as professional antigen-presenting cells, are members of the innate immune system and function as key players during the induction phase of adaptive immune responses. Uptake, processing, and presentation of antigens direct the outcome toward either tolerance or immunity. The cells of the immune system are among the most highly radiosensitive cells in the body. For high doses of ionizing radiation (HD-IR) both immune-suppressive effects after whole body irradiation and possible immune activation during tumor therapy were observed. On the other hand, the effects of low doses of ionizing radiation (LD-IR) on the immune system are controversial and seem to show high variability among different individuals and species. There are reports revealing that protracted LD-IR can result in radioresistance. But immune-suppressive effects of chronic LD-IR are also reported, including the killing or sensitizing of certain cell types. This article shall review the current knowledge of radiation-induced effects on the immune system, paying special attention to the interaction of DCs and T cells.
Collapse
Affiliation(s)
- Katrin Manda
- Department of Radiotherapy and Radiation Oncology, University of Rostock Rostock, Germany
| | | | | | | |
Collapse
|
22
|
Christensen D, Henriksen-Lacey M, Kamath AT, Lindenstrøm T, Korsholm KS, Christensen JP, Rochat AF, Lambert PH, Andersen P, Siegrist CA, Perrie Y, Agger EM. A cationic vaccine adjuvant based on a saturated quaternary ammonium lipid have different in vivo distribution kinetics and display a distinct CD4 T cell-inducing capacity compared to its unsaturated analog. J Control Release 2012; 160:468-76. [PMID: 22709414 DOI: 10.1016/j.jconrel.2012.03.016] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 03/15/2012] [Accepted: 03/18/2012] [Indexed: 10/28/2022]
Abstract
Adjuvants are often composed of different constituents that can be divided into two groups based on their primary activity: the delivery system which carries and presents the vaccine antigen to antigen-presenting cells, and the immunostimulator that activates and modulates the ensuing immune response. Herein, we have investigated the importance of the delivery system and in particular its physical characteristics by comparing the delivery properties of two lipids which differ only in the degree of saturation of the acyl chains, rendering the liposomes either rigid (DDA, dimethyldioctadecylammonium) or highly fluid (DODA, dimethyldioleoylammonium) at physiological temperature. We show that these delivery systems are remarkably different in their ability to prime a Th1-directed immune response with the rigid DDA-based liposomes inducing a response more than 100 times higher compared to that obtained with the fluid DODA-based liposomes. Upon injection with a vaccine antigen, DDA-based liposomes form a vaccine depot that results in a continuous attraction of antigen-presenting cells that engulf a high amount of adjuvant and are subsequently efficiently activated as measured by an elevated expression of the co-stimulatory molecules CD40 and CD86. In contrast, the fluid DODA-based liposomes are more rapidly removed from the site of injection resulting in a lower up-regulation of co-stimulatory CD40 and CD86 molecules on adjuvant-positive antigen-presenting cells. Additionally, the vaccine antigen is readily dissociated from the DODA-based liposomes leading to a population of antigen-presenting cells that are antigen-positive but adjuvant-negative and consequently are not activated. These studies demonstrate the importance of studying in vivo characteristics of the vaccine components and furthermore show that physicochemical properties of the delivery system have a major impact on the vaccine-induced immune response.
Collapse
Affiliation(s)
- Dennis Christensen
- Dept. Infectious Disease Immunology, Statens Serum Institut, Copenhagen S, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Zheng J, Jiang HY, Li J, Tang HC, Zhang XM, Wang XR, Du JT, Li HB, Xu G. MicroRNA-23b promotes tolerogenic properties of dendritic cells in vitro through inhibiting Notch1/NF-κB signalling pathways. Allergy 2012; 67:362-70. [PMID: 22229716 DOI: 10.1111/j.1398-9995.2011.02776.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2011] [Indexed: 12/31/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) are known to regulate the inflammatory response in various cell types. However, the ability of miRNAs to modulate dendritic cells (DCs) function for allergen immunotherapy is unclear. OBJECTIVE To assess the role of miR-23b in the regulation of ovalbumin (OVA)-induced DC differentiation and function and to investigate the related molecular mechanisms. METHODS Bone marrow-derived dendritic cells (BMDCs) were generated from murine bone marrow progenitor cells and subsequently stimulated with OVA to examine the profile of miRNA expression. After transfection with miR-23b reagents, DCs were evaluated for endocytic ability, surface marker expression, cytokine secretion and CD4+ T-cell differentiation. The possible roles of the Notch and NF-κB signalling pathways were also evaluated. Human monocyte-derived dendritic cells (MDDCs) were similarly evaluated as well. RESULTS Significant upregulation of miR-23b was observed in BMDCs pulsed with OVA. Following miR-23b transfection, BMDCs showed decreased OVA uptake, increased IL-10 production, decreased IL-12 production and an enhanced capacity to promote FoxP3+ CD4+ T regulatory cells (Tregs) differentiation. In addition, inactivation of the Notch1 and NF-κB signalling pathways were observed. Conversely, inhibition of miR-23b in BMDCs resulted in the opposite effects. In human MDDCs, miRNA23b transfection similarly increased IL-10 and decreased IL-12 production, and that treated human MDDCs induced increased FoxP3+ CD4+ T cells. CONCLUSION Our findings provide evidence that miR-23b is capable of inducing tolerogenic DC activity and Treg responses in vitro through the inhibition of the Notch1 and NF-κB signalling pathways; thus, miR-23b might represent a therapeutic target for the management of allergic diseases.
Collapse
Affiliation(s)
- J. Zheng
- Otorhinolaryngology Hospital; the First Affiliated Hospital of Sun Yat-sen University; Otorhinolaryngology Institute of Sun Yat-sen University; Guangzhou; China
| | - H.-Y. Jiang
- Otorhinolaryngology Hospital; the First Affiliated Hospital of Sun Yat-sen University; Otorhinolaryngology Institute of Sun Yat-sen University; Guangzhou; China
| | - J. Li
- Otorhinolaryngology Hospital; the First Affiliated Hospital of Sun Yat-sen University; Otorhinolaryngology Institute of Sun Yat-sen University; Guangzhou; China
| | - H.-C. Tang
- Otorhinolaryngology Hospital; the First Affiliated Hospital of Sun Yat-sen University; Otorhinolaryngology Institute of Sun Yat-sen University; Guangzhou; China
| | - X.-M. Zhang
- Otorhinolaryngology Hospital; the First Affiliated Hospital of Sun Yat-sen University; Otorhinolaryngology Institute of Sun Yat-sen University; Guangzhou; China
| | - X.-R. Wang
- Otorhinolaryngology Hospital; the First Affiliated Hospital of Sun Yat-sen University; Otorhinolaryngology Institute of Sun Yat-sen University; Guangzhou; China
| | - J.-T. Du
- Otorhinolaryngology Hospital; the First Affiliated Hospital of Sun Yat-sen University; Otorhinolaryngology Institute of Sun Yat-sen University; Guangzhou; China
| | - H.-B. Li
- Otorhinolaryngology Hospital; the First Affiliated Hospital of Sun Yat-sen University; Otorhinolaryngology Institute of Sun Yat-sen University; Guangzhou; China
| | - G. Xu
- Otorhinolaryngology Hospital; the First Affiliated Hospital of Sun Yat-sen University; Otorhinolaryngology Institute of Sun Yat-sen University; Guangzhou; China
| |
Collapse
|
24
|
Nano-characterization of Jagged-1-educated dendritic cells. Open Life Sci 2011. [DOI: 10.2478/s11535-011-0063-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractJagged-1-educated dendritic cells (DCs) are of distinct phenotypes different from Lipopolysaccharide (LPS)-educated DCs. However, to date, little is known as to nano-features of Jagged-1-educated DCs. In this study, nanostructure of Jagged-1-educated DCs was observed through atomic force microscopy. Our results showed that the volume, surface area and width of Jagged-1-educated DCs, and the number of protrusion, pseudopodia and lamellapodia on the surface of Jagged-1-educated DCs were significantly more than those of GM-CSF-induced DCs, but less than those of LPS-educated DCs. Compared with GM-CSF-induced DCs, the roughness on the surface of Jagged-1-educated DCs was greatly increased, similar to LPS-educated DCs, but the particle size and number of them on the membrane were markedly less than the latter. The change of DC nanostructure caused by Jagged-1 was abrogated with N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycinet-butyl ester, a γ-secretase inhibitor of Notch signaling pathway. Compared to LPS-educated DCs, Jagged-1-educated DCs highly expressed NICD, Deltex-1 and Hes-1 proteins, moderately produced IL-12, but not IFN-γ, and the level of CD40 molecules on the surface of them was much lower, suggesting that Jagged-1-educated DCs are in a semi-mature status and may have unique functions distinguished from LPS-educated DCs.
Collapse
|
25
|
Xing F, Wang J, Hu M, Yu Y, Chen G, Liu J. Comparison of immature and mature bone marrow-derived dendritic cells by atomic force microscopy. NANOSCALE RESEARCH LETTERS 2011; 6:455. [PMID: 21762525 PMCID: PMC3211875 DOI: 10.1186/1556-276x-6-455] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 07/16/2011] [Indexed: 05/31/2023]
Abstract
A comparative study of immature and mature bone marrow-derived dendritic cells (BMDCs) was first performed through an atomic force microscope (AFM) to clarify differences of their nanostructure and adhesion force. AFM images revealed that the immature BMDCs treated by granulocyte macrophage-colony stimulating factor plus IL-4 mainly appeared round with smooth surface, whereas the mature BMDCs induced by lipopolysaccharide displayed an irregular shape with numerous pseudopodia or lamellapodia and ruffles on the cell membrane besides becoming larger, flatter, and longer. AFM quantitative analysis further showed that the surface roughness of the mature BMDCs greatly increased and that the adhesion force of them was fourfold more than that of the immature BMDCs. The nano-features of the mature BMDCs were supported by a high level of IL-12 produced from the mature BMDCs and high expression of MHC-II on the surface of them. These findings provide a new insight into the nanostructure of the immature and mature BMDCs.
Collapse
Affiliation(s)
- Feiyue Xing
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Jiongkun Wang
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Mingqian Hu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yu Yu
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Guoliang Chen
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Jing Liu
- Department of Stomatology, Jinan University, Guangzhou 510632, China
| |
Collapse
|