1
|
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: 1.7] [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
|
2
|
Xiao Z, Puré E. Imaging of T-cell Responses in the Context of Cancer Immunotherapy. Cancer Immunol Res 2021; 9:490-502. [PMID: 33941536 DOI: 10.1158/2326-6066.cir-20-0678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/18/2020] [Accepted: 02/18/2021] [Indexed: 12/16/2022]
Abstract
Immunotherapy, which promotes the induction of cytotoxic T lymphocytes and enhances their infiltration into and function within tumors, is a rapidly expanding and evolving approach to treating cancer. However, many of the critical denominators for inducing effective anticancer immune responses remain unknown. Efforts are underway to develop comprehensive ex vivo assessments of the immune landscape of patients prior to and during response to immunotherapy. An important complementary approach to these efforts involves the development of noninvasive imaging approaches to detect immune targets, assess delivery of immune-based therapeutics, and evaluate responses to immunotherapy. Herein, we review the merits and limitations of various noninvasive imaging modalities (MRI, PET, and single-photon emission tomography) and discuss candidate targets for cellular and molecular imaging for visualization of T-cell responses at various stages along the cancer-immunity cycle in the context of immunotherapy. We also discuss the potential use of these imaging strategies in monitoring treatment responses and predicting prognosis for patients treated with immunotherapy.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
3
|
El-Barbry H, Capitao M, Barrin S, Amziani S, Pierre Paul P, Borreill S, Guilbert T, Donnadieu E, Niedergang F, Ouaaz F. Extracellular Release of Antigen by Dendritic Cell Regurgitation Promotes B Cell Activation through NF-κB/cRel. THE JOURNAL OF IMMUNOLOGY 2020; 205:608-618. [PMID: 32580933 DOI: 10.4049/jimmunol.1900394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 05/03/2020] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are professional APCs, which sample Ags in the periphery and migrate to the lymph node where they activate T cells. DCs can also present native Ag to B cells through interactions observed both in vitro and in vivo. However, the mechanisms of Ag transfer and B cell activation by DCs remain incompletely understood. In this study, we report that murine DCs are an important cell transporter of Ag from the periphery to the lymph node B cell zone and also potent inducers of B cell activation both in vivo and in vitro. Importantly, we highlight a novel extracellular mechanism of B cell activation by DCs. In this study, we demonstrate that Ag released upon DC regurgitation is sufficient to efficiently induce early B cell activation, which is BCR driven and mechanistically dependent on the nuclear accumulation of the transcription factor NF-κB/cRel. Thus, our study provides new mechanistic insights into Ag delivery and B cell activation modalities by DCs and a promising approach for targeting NF-κB/cRel pathway to modulate the DC-elicited B cell responses.
Collapse
Affiliation(s)
- Houssam El-Barbry
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| | - Marisa Capitao
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| | - Sarah Barrin
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| | - Samir Amziani
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| | - Pascal Pierre Paul
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| | - Susanna Borreill
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| | - Thomas Guilbert
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| | - Emmanuel Donnadieu
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| | - Florence Niedergang
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| | - Fatah Ouaaz
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR 8104, F-75014 Paris, France
| |
Collapse
|
4
|
Krekorian M, Fruhwirth GO, Srinivas M, Figdor CG, Heskamp S, Witney TH, Aarntzen EHJG. Imaging of T-cells and their responses during anti-cancer immunotherapy. Theranostics 2019; 9:7924-7947. [PMID: 31656546 PMCID: PMC6814447 DOI: 10.7150/thno.37924] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/30/2019] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy has proven to be an effective approach in a growing number of cancers. Despite durable clinical responses achieved with antibodies targeting immune checkpoint molecules, many patients do not respond. The common denominator for immunotherapies that have successfully been introduced in the clinic is their potential to induce or enhance infiltration of cytotoxic T-cells into the tumour. However, in clinical research the molecules, cells and processes involved in effective responses during immunotherapy remain largely obscure. Therefore, in vivo imaging technologies that interrogate T-cell responses in patients represent a powerful tool to boost further development of immunotherapy. This review comprises a comprehensive analysis of the in vivo imaging technologies that allow the characterisation of T-cell responses induced by anti-cancer immunotherapy, with emphasis on technologies that are clinically available or have high translational potential. Throughout we discuss their respective strengths and weaknesses, providing arguments for selecting the optimal imaging options for future research and patient management.
Collapse
Affiliation(s)
- Massis Krekorian
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Gilbert O Fruhwirth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Timothy H Witney
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Erik H J G Aarntzen
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
5
|
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: 0.3] [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
|
6
|
Moura Rosa P, Gopalakrishnan N, Ibrahim H, Haug M, Halaas Ø. The intercell dynamics of T cells and dendritic cells in a lymph node-on-a-chip flow device. LAB ON A CHIP 2016; 16:3728-40. [PMID: 27560793 DOI: 10.1039/c6lc00702c] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
T cells play a central role in immunity towards cancer and infectious diseases. T cell responses are initiated in the T cell zone of the lymph node (LN), where resident antigen-bearing dendritic cells (DCs) prime and activate antigen-specific T cells passing by. In the present study, we investigated the T cell : DC interaction in a microfluidic device to understand the intercellular dynamics and physiological conditions in the LN. We show random migration of antigen-specific T cells onto the antigen-presenting DC monolayer independent of the flow direction with a mean T cell : DC dwell time of 12.8 min and a mean velocity of 6 μm min(-1). Furthermore, we investigated the antigen specific vs. unspecific attachment and detachment of CD8(+) and CD4(+) T cells to DCs under varying shear stress. In our system, CD4(+) T cells showed long stable contacts with APCs, whereas CD8(+) T cells presented transient interactions with DCs. By varying the shear stress from 0.01 to 100 Dyn cm(-2), it was also evident that there was a much stronger attachment of antigen-specific than unspecific T cells to stationary DCs up to 1-12 Dyn cm(-2). The mechanical force of the cell : cell interaction associated with the pMHC-TCR match under controlled tangential shear force was estimated to be in the range of 0.25-4.8 nN. Finally, upon performing attachment & detachment tests, there was a steady accumulation of antigen specific CD8(+) T cells and CD4(+) T cells on DCs at low shear stresses, which were released at a stress of 12 Dyn cm(-2). This microphysiological model provides new possibilities to recreate a controlled mechanical force threshold of pMHC-TCR binding, allowing the investigation of intercellular signalling of immune synapses and therapeutic targets for immunotherapy.
Collapse
Affiliation(s)
- Patrícia Moura Rosa
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7489 Trondheim, Norway.
| | | | | | | | | |
Collapse
|
7
|
Ziraldo C, Gong C, Kirschner DE, Linderman JJ. Strategic Priming with Multiple Antigens can Yield Memory Cell Phenotypes Optimized for Infection with Mycobacterium tuberculosis: A Computational Study. Front Microbiol 2016; 6:1477. [PMID: 26779136 PMCID: PMC4701940 DOI: 10.3389/fmicb.2015.01477] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/08/2015] [Indexed: 12/16/2022] Open
Abstract
Lack of an effective vaccine results in 9 million new cases of tuberculosis (TB) every year and 1.8 million deaths worldwide. Although many infants are vaccinated at birth with BCG (an attenuated M. bovis), this does not prevent infection or development of TB after childhood. Immune responses necessary for prevention of infection or disease are still unknown, making development of effective vaccines against TB challenging. Several new vaccines are ready for human clinical trials, but these trials are difficult and expensive; especially challenging is determining the appropriate cellular response necessary for protection. The magnitude of an immune response is likely key to generating a successful vaccine. Characteristics such as numbers of central memory (CM) and effector memory (EM) T cells responsive to a diverse set of epitopes are also correlated with protection. Promising vaccines against TB contain mycobacterial subunit antigens (Ag) present during both active and latent infection. We hypothesize that protection against different key immunodominant antigens could require a vaccine that produces different levels of EM and CM for each Ag-specific memory population. We created a computational model to explore EM and CM values, and their ratio, within what we term Memory Design Space. Our model captures events involved in T cell priming within lymph nodes and tracks their circulation through blood to peripheral tissues. We used the model to test whether multiple Ag-specific memory cell populations could be generated with distinct locations within Memory Design Space at a specific time point post vaccination. Boosting can further shift memory populations to memory cell ratios unreachable by initial priming events. By strategically varying antigen load, properties of cellular interactions within the LN, and delivery parameters (e.g., number of boosts) of multi-subunit vaccines, we can generate multiple Ag-specific memory populations that cover a wide range of Memory Design Space. Given a set of desired characteristics for Ag-specific memory populations, we can use our model as a tool to predict vaccine formulations that will generate those populations.
Collapse
Affiliation(s)
- Cordelia Ziraldo
- Department of Chemical Engineering, University of Michigan, Ann ArborMI, USA; Department of Microbiology and Immunology, University of Michigan Medical School, Ann ArborMI, USA
| | - Chang Gong
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann ArborMI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann ArborMI, USA
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor MI, USA
| | - Jennifer J Linderman
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor MI, USA
| |
Collapse
|
8
|
Al Heialy S, Zeroual M, Farahnak S, McGovern T, Risse PA, Novali M, Lauzon AM, Roman HN, Martin JG. Nanotubes connect CD4+ T cells to airway smooth muscle cells: novel mechanism of T cell survival. THE JOURNAL OF IMMUNOLOGY 2015; 194:5626-34. [PMID: 25934863 DOI: 10.4049/jimmunol.1401718] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 04/11/2015] [Indexed: 11/19/2022]
Abstract
Contact between airway smooth muscle (ASM) cells and activated CD4(+) T cells, a key interaction in diseases such as asthma, triggers ASM cell proliferation and enhances T cell survival. We hypothesized that direct contact between ASM and CD4(+) T cells facilitated the transfer of anti-apoptotic proteins via nanotubes, resulting in increased survival of activated CD4(+) T cells. CD4(+) T cells, isolated from PBMCs of healthy subjects, when activated and cocultured with ASM cells for 24 h, formed nanotubes that were visualized by immunofluorescence and atomic force microscopy. Cell-to-cell transfer of the fluorescent dye calcein-AM confirmed cytoplasmic communication via nanotubes. Immunoreactive B cell lymphoma 2 (Bcl-2) and induced myeloid leukemia cell differentiation protein (Mcl-1), two major anti-apoptotic proteins, were present within the nanotubes. Downregulation of Mcl-1 by small interfering RNA in ASM cells significantly increased T cell apoptosis, whereas downregulation of Bcl-2 had no effect. Transfer of GFP-tagged Mcl-1 from ASM cells to CD4(+) T cells via the nanotubes confirmed directionality of transfer. In conclusion, activated T cells communicate with ASM cells via nanotube formation. Direct transfer of Mcl-1 from ASM to CD(+) T cells via nanotubes is involved in T cell survival. This study provides a novel mechanism of survival of CD4(+) T cells that is dependent on interaction with a structural cell.
Collapse
Affiliation(s)
- Saba Al Heialy
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Melissa Zeroual
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Soroor Farahnak
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Toby McGovern
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Paul-André Risse
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Mauro Novali
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Horia N Roman
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - James G Martin
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| |
Collapse
|
9
|
Souza-Silva F, Pereira BAS, Finkelstein LC, Zucolotto V, Caffarena ER, Alves CR. Dynamic identification of H2 epitopes from Leishmania (Leishmania) amazonensis cysteine proteinase B with potential immune activity during murine infection. J Mol Recognit 2014; 27:98-105. [PMID: 24436127 DOI: 10.1002/jmr.2334] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 10/09/2013] [Accepted: 10/12/2013] [Indexed: 11/11/2022]
Abstract
Peptides from the COOH-terminal extension of cysteine proteinase B from Leishmania (Leishmania) amazonensis (cyspep) can modulate immune responses in vertebrate hosts. With this hypothesis as base, we used the online analysis tool SYFPEITHI to predict seven epitopes from this region with potential to bind H2 proteins. We performed proliferation tests and quantified reactive T lymphocytes applying a cytometry analysis, using samples from draining lymph node of lesions from L. (L.) amazonensis-infected mice. To define reactivity of T cells, we used complexes of DimerX (H2 D(b):Ig and H2 L(d):Ig) and the putative epitopes. Additionally, we applied surface plasmon resonance to verify real time interactions between the putative epitopes and DimerX proteins. Five peptides induced blastogenesis in BALB/c cells, while only two presented the same property in C57BL/6 mouse cells. In addition, our data indicate the existence of CD8+ T lymphocyte populations able to recognize each tested peptide in both murine strains. We observed an overlapping of results between the peptides that induced lymphocyte proliferation and those capable of binding to the DimerX in the surface plasmon resonance assays thus indicating that using these recombinant proteins in biosensing analyses is a promising tool to study real time molecular interactions in the context of major histocompatibility complex epitopes. The data gathered in this study reinforce the hypothesis that cyspep-derived peptides are important factors in the murine host infection by L. (L.) amazonensis.
Collapse
Affiliation(s)
- Franklin Souza-Silva
- Laboratório de Biologia Molecular e Doenças Endêmicas, IOC-Fundação Oswaldo Cruz, Rio de Janeiro, CEP, 21040-360, Brazil
| | | | | | | | | | | |
Collapse
|
10
|
New insights into the crosstalk between Shigella and T lymphocytes. Trends Microbiol 2014; 22:192-8. [PMID: 24613405 DOI: 10.1016/j.tim.2014.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/03/2014] [Accepted: 02/05/2014] [Indexed: 01/22/2023]
Abstract
Subversion of host immune responses is the key infection strategy employed by most, if not all, human pathogens. Modulation of the host innate response by pathogens has been vastly documented. Yet, especially for bacterial infections, it was only recently that cells of the adaptive immune response were recognized as targets of bacterial weapons such as the type III secretion system (T3SS) and its effector proteins. In this review, we focus on the recent advances made in the understanding of how the enteroinvasive bacterium Shigella flexneri interferes with the host adaptive response by targeting T lymphocytes, especially their migration capacities.
Collapse
|
11
|
Hess Michelini R, Doedens AL, Goldrath AW, Hedrick SM. Differentiation of CD8 memory T cells depends on Foxo1. ACTA ACUST UNITED AC 2013; 210:1189-200. [PMID: 23712431 PMCID: PMC3674697 DOI: 10.1084/jem.20130392] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The transcription factor Foxo1 is required for the differentiation of memory CD8+ T cells, and its absence hinders clearance of secondary infections. The forkhead O transcription factors (FOXO) integrate a range of extracellular signals, including growth factor signaling, inflammation, oxidative stress, and nutrient availability, to substantially alter the program of gene expression and modulate cell survival, cell cycle progression, and many yet to be unraveled cell type–specific responses. Naive antigen-specific CD8+ T cells undergo a rapid expansion and arming of effector function within days of pathogen exposure. In addition, by the peak of expansion, they form precursors to memory T cells capable of self-renewal and indefinite survival. Using lymphocytic choriomeningitis virus Armstrong to probe the response to infection, we found that Foxo1−/− CD8+ T cells expand normally with no defects in effector differentiation, but continue to exhibit characteristics of effector T cells long after antigen clearance. The KLRG1lo CD8+ T cells that are normally enriched for memory-precursor cells retain Granzyme B and CD69 expression, and fail to up-regulate TCF7, EOMES, and other memory signature genes. As a correlate, Foxo1−/− CD8+ T cells were virtually unable to expand upon secondary infection. Collectively, these results demonstrate an intrinsic role for FOXO1 in establishing the post-effector memory program that is essential to forming long-lived memory cells capable of immune reactivation.
Collapse
Affiliation(s)
- Rodrigo Hess Michelini
- Molecular Biology Section, Division of Biological Sciences, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | | | | |
Collapse
|
12
|
Mueller SN. Effector T-cell responses in non-lymphoid tissues: insights from in vivo imaging. Immunol Cell Biol 2013; 91:290-6. [PMID: 23295362 DOI: 10.1038/icb.2012.75] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
T-cell responses are initiated within secondary lymphoid organs, and effector T-cells are released into the circulation where they home to inflamed tissues and mediate protective immune responses. Within non-lymphoid tissues, the types of cellular interactions and the dynamics that lead to clearance of infections are still relatively poorly understood. Here I review how imaging of effector T-cells within tissues has contributed to our understanding of immune responses, and examine some of the remaining questions that may benefit from in vivo imaging to reveal the intricacies of how immune cells function. A detailed understanding of the dynamics of T-cell responses within non-lymphoid tissues is important for the rational design of targeted therapies that influence key steps in disease progression.
Collapse
Affiliation(s)
- Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
13
|
Mostardinha P, de Abreu FV. Positive and negative selection, self-nonself discrimination and the roles of costimulation and anergy. Sci Rep 2012; 2:769. [PMID: 23101027 PMCID: PMC3480656 DOI: 10.1038/srep00769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 09/17/2012] [Indexed: 01/28/2023] Open
Abstract
It is still unclear whether the adaptive immune system can perform accurate self-nonself discrimination and what could influence its performance. Starting from simple cellular interaction rules we show that it is possible to achieve perfect self-nonself discrimination in a consistent framework provided positive and negative selection operate during repertoire education, and costimulation and anergy are also considered during T cell activation. In this theory T cell receptors diversity is required for cells to sense differently different peptides; positive selection is needed to guarantee maximal lymphocyte's interactivity and to allow negative selection to reduce conjugation lifetimes maximally; costimulation is necessary to signal that an antigen presenting cell established an uncommon rate of long lived conjugations when presenting foreign peptides; anergy is required to guarantee that these stable contacts involved different T cells and not always the same. These results suggest that accurate self-nonself discrimination can have shaped the adaptive immune system.
Collapse
Affiliation(s)
- P Mostardinha
- Universidade de Aveiro, Departamento de Física, 3810-193 Aveiro, Portugal
| | | |
Collapse
|
14
|
Aarntzen EHJG, Srinivas M, Radu CG, Punt CJA, Boerman OC, Figdor CG, Oyen WJG, de Vries IJM. In vivo imaging of therapy-induced anti-cancer immune responses in humans. Cell Mol Life Sci 2012; 70:2237-57. [PMID: 23052208 PMCID: PMC3676735 DOI: 10.1007/s00018-012-1159-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 08/27/2012] [Accepted: 09/03/2012] [Indexed: 12/16/2022]
Abstract
Immunotherapy aims to re-engage and revitalize the immune system in the fight against cancer. Research over the past decades has shown that the relationship between the immune system and human cancer is complex, highly dynamic, and variable between individuals. Considering the complexity, enormous effort and costs involved in optimizing immunotherapeutic approaches, clinically applicable tools to monitor therapy-induced immune responses in vivo are most warranted. However, the development of such tools is complicated by the fact that a developing immune response encompasses several body compartments, e.g., peripheral tissues, lymph nodes, lymphatic and vascular systems, as well as the tumor site itself. Moreover, the cells that comprise the immune system are not static but constantly circulate through the vascular and lymphatic system. Molecular imaging is considered the favorite candidate to fulfill this task. The progress in imaging technologies and modalities has provided a versatile toolbox to address these issues. This review focuses on the detection of therapy-induced anticancer immune responses in vivo and provides a comprehensive overview of clinically available imaging techniques as well as perspectives on future developments. In the discussion, we will focus on issues that specifically relate to imaging of the immune system and we will discuss the strengths and limitations of the current clinical imaging techniques. The last section provides future directions that we envision to be crucial for further development.
Collapse
Affiliation(s)
- Erik H J G Aarntzen
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Kobezda T, Ghassemi-Nejad S, Glant TT, Mikecz K. In vivo two-photon imaging of T cell motility in joint-draining lymph nodes in a mouse model of rheumatoid arthritis. Cell Immunol 2012; 278:158-65. [PMID: 23023071 DOI: 10.1016/j.cellimm.2012.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/22/2012] [Accepted: 08/24/2012] [Indexed: 01/27/2023]
Abstract
Recent imaging studies on intact lymph nodes (LNs) of naïve T cell receptor (TCR)-transgenic mice have reported that T cells reduce their motility upon contact with relevant antigen-presenting cells (APCs). Using in vivo two-photon imaging of T cells in joint-draining (JD) LNs, we examined whether similar changes in T cell motility are observed in wild type mice. Co-transfer of T cells from naïve mice and antigen-experienced T cells from mice with proteoglycan (PG)-induced arthritis into naïve or arthritic recipients resulted in prolonged interactions of antigen-experienced T cells with APCs upon intra-articular antigen (PG) injection, indicating that T cells from arthritic wild type mice recapitulate the motile behavior reported in naïve TCR-transgenic mice. However, naïve T cells also engaged in stable interactions with APCs in the JDLNs of arthritic recipients, suggesting an enhanced ability of APCs in the JDLNs of arthritic hosts to present antigen to either naïve or antigen-experienced T cells.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/pathology
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/pathology
- Cell Movement
- Disease Models, Animal
- Female
- Humans
- Injections, Intra-Articular
- Joints/immunology
- Joints/pathology
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- Microscopy, Fluorescence, Multiphoton
- Proteoglycans/isolation & purification
- Proteoglycans/pharmacology
- T-Lymphocytes/immunology
- T-Lymphocytes/physiology
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- Tamás Kobezda
- Section of Molecular Medicine, Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
16
|
Liou HLR, Myers JT, Barkauskas DS, Huang AY. Intravital imaging of the mouse popliteal lymph node. J Vis Exp 2012:3720. [PMID: 22349264 PMCID: PMC3369637 DOI: 10.3791/3720] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Lymph nodes (LNs) are secondary lymphoid organs, which are strategically located throughout the body to allow for trapping and presentation of foreign antigens from peripheral tissues to prime the adaptive immune response. Juxtaposed between innate and adaptive immune responses, the LN is an ideal site to study immune cell interactions. Lymphocytes (T cells, B cells and NK cells), dendritic cells (DCs), and macrophages comprise the bulk of bone marrow-derived cellular elements of the LN. These cells are strategically positioned in the LN to allow efficient surveillance of self antigens and potential foreign antigens. The process by which lymphocytes successfully encounter cognate antigens is a subject of intense investigation in recent years, and involves an integration of molecular contacts including antigen receptors, adhesion molecules, chemokines, and stromal structures such as the fibro-reticular network. Prior to the development of high-resolution real-time fluorescent in vivo imaging, investigators relied on static imaging, which only offers answers regarding morphology, position, and architecture. While these questions are fundamental in our understanding of immune cell behavior, the limitations intrinsic with this technique does not permit analysis to decipher lymphocyte trafficking and environmental clues that affect dynamic cell behavior. Recently, the development of intravital two-photon laser scanning microscopy (2P-LSM) has allowed investigators to view the dynamic movements and interactions of individual cells within live LNs in situ. In particular, we and others have applied this technique to image cellular behavior and interactions within the popliteal LN, where its compact, dense nature offers the advantage of multiplex data acquisition over a large tissue area with diverse tissue sub-structures. It is important to note that this technique offers added benefits over explanted tissue imaging techniques, which require disruption of blood, lymph flow, and ultimately the cellular dynamics of the system. Additionally, explanted tissues have a very limited window of time in which the tissue remains viable for imaging after explant. With proper hydration and monitoring of the animal's environmental conditions, the imaging time can be significantly extended with this intravital technique. Here, we present a detailed method of preparing mouse popliteal LN for the purpose of performing intravital imaging.
Collapse
|
17
|
Systems biology approaches for understanding cellular mechanisms of immunity in lymph nodes during infection. J Theor Biol 2011; 287:160-70. [PMID: 21798267 DOI: 10.1016/j.jtbi.2011.06.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 06/30/2011] [Accepted: 06/30/2011] [Indexed: 12/20/2022]
Abstract
Adaptive immunity is initiated in secondary lymphoid tissues when naive T cells recognize foreign antigen presented as MHC-bound peptide on the surface of dendritic cells. Only a small fraction of T cells in the naive repertoire will express T cell receptors specific for a given epitope, but antigen recognition triggers T cell activation and proliferation, thus greatly expanding antigen-specific clones. Expanded T cells can serve a helper function for B cell responses or traffic to sites of infection to secrete cytokines or kill infected cells. Over the past decade, two-photon microscopy of lymphoid tissues has shed important light on T cell development, antigen recognition, cell trafficking and effector functions. These data have enabled the development of sophisticated quantitative and computational models that, in turn, have been used to test hypotheses in silico that would otherwise be impossible or difficult to explore experimentally. Here, we review these models and their principal findings and highlight remaining questions where modeling approaches are poised to advance our understanding of complex immunological systems.
Collapse
|
18
|
Corse E, Gottschalk RA, Allison JP. Strength of TCR-peptide/MHC interactions and in vivo T cell responses. THE JOURNAL OF IMMUNOLOGY 2011; 186:5039-45. [PMID: 21505216 DOI: 10.4049/jimmunol.1003650] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The TCR can detect subtle differences in the strength of interaction with peptide/MHC ligand and transmit this information to influence downstream events in T cell responses. Manipulation of the factor commonly referred to as TCR signal strength can be achieved by changing the amount or quality of peptide/MHC ligand. Recent work has enhanced our understanding of the many variables that contribute to the apparent cumulative strength of TCR stimulation during immunogenic and tolerogenic T cell responses. In this review, we consider data from in vitro studies in the context of in vivo immune responses and discuss in vivo consequences of manipulation of strength of TCR stimulation, including influences on T cell-APC interactions, the magnitude and quality of the T cell response, and the types of fate decisions made by peripheral T cells.
Collapse
Affiliation(s)
- Emily Corse
- Department of Immunology, Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | | | | |
Collapse
|
19
|
Daniel C, von Boehmer H. Extra-thymically induced regulatory T cells: do they have potential in disease prevention? Semin Immunol 2011; 23:410-7. [PMID: 21724411 DOI: 10.1016/j.smim.2011.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 06/07/2011] [Indexed: 01/07/2023]
Abstract
Fopx3(+) Treg safeguard against autoimmune diseases and immune pathology. The extrathymic conversion of naïve T cells into Foxp3(+) regulatory T cells can be achieved in vivo by the delivery of strong-agonist ligands under subimmunogenic conditions. Tolerogenic vaccination with strong-agonist mimetopes of self-antigen to promote self-antigen specific tolerance may represent the most specific and safest means of preventing autoimmunity. This review discusses the requirements for induction of dominant tolerance exerted by Foxp3(+) Tregs in autoimmunity with special emphasis on their impact to interfere with T1D. The future goals are the understanding of self-non-self discrimination at the cellular and molecular level, which should then enable investigators to develop clinical vaccination protocols that specifically interfere with unwanted immune responses.
Collapse
Affiliation(s)
- Carolin Daniel
- Laboratory of Lymphocyte Biology, Department of Cancer Immunology and AIDS, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | | |
Collapse
|
20
|
Daniel C, von Boehmer H. Extrathymic generation of regulatory T cells--chances and challenges for prevention of autoimmune disease. Adv Immunol 2011; 112:177-213. [PMID: 22118409 DOI: 10.1016/b978-0-12-387827-4.00005-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fopx3(+) expressing regulatory T cells (Tregs) function as an indispensable cellular constituent of the immune system by establishing and maintaining immunological self-tolerance. T cell receptor (TCR) ligands of high agonist activity, when applied in vivo under subimmunogenic conditions, convert naive but not activated T cells into stable Tregs expressing Foxp3. Tolerogenic vaccination with strong-agonist mimetopes of self-antigens may function as a safe and highly specific instrument in the prevention of autoimmune disease by promoting self-antigen-specific tolerance. In this review, we address the requirements for generation of dominant tolerance exerted by Foxp3(+) Tregs in autoimmune disease with special focus on type 1 diabetes (T1D). Further understanding of differentiation of T cells into Tregs at the cellular and molecular level will facilitate development of additional tolerogenic vaccination strategies that can be used in prevention as well as therapeutically to combat unwanted immunity.
Collapse
Affiliation(s)
- Carolin Daniel
- Department of Cancer Immunology and AIDS, Laboratory of Lymphocyte Biology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | | |
Collapse
|
21
|
Vilekar P, Awasthi V, Lagisetty P, King C, Shankar N, Awasthi S. In vivo trafficking and immunostimulatory potential of an intranasally-administered primary dendritic cell-based vaccine. BMC Immunol 2010; 11:60. [PMID: 21143974 PMCID: PMC3018378 DOI: 10.1186/1471-2172-11-60] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 12/10/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Coccidioidomycosis or Valley fever is caused by a highly virulent fungal pathogen: Coccidioides posadasii or immitis. Vaccine development against Coccidioides is of contemporary interest because a large number of relapses and clinical failures are reported with antifungal agents. An efficient Th1 response engenders protection. Thus, we have focused on developing a dendritic cell (DC)-based vaccine for coccidioidomycosis. In this study, we investigated the immunostimulatory characteristics of an intranasal primary DC-vaccine in BALB/c mouse strain that is most susceptible to coccidioidomycosis. The DCs were transfected nonvirally with Coccidioides-Ag2/PRA-cDNA. Expression of DC-markers, Ag2/PRA and cytokines were studied by flow cytometry, dot-immunoblotting and cytometric bead array methods, respectively. The T cell activation was studied by assessing the upregulation of activation markers in a DC-T cell co-culture assay. For trafficking, the DCs were co-transfected with a plasmid DNA encoding HSV1 thymidine kinase (TK) and administered intranasally into syngeneic mice. The trafficking and homing of TK-expressing DCs were monitored with positron emission tomography (PET) using 18F-FIAU probe. Based on the PET-probe accumulation in vaccinated mice, selected tissues were studied for antigen-specific response and T cell phenotypes using ELISPOT and flow cytometry, respectively. RESULTS We found that the primary DCs transfected with Coccidioides-Ag2/PRA-cDNA were of immature immunophenotype, expressed Ag2/PRA and activated naïve T cells. In PET images and subsequent biodistribution, intranasally-administered DCs were found to migrate in blood, lung and thymus; lymphocytes showed generation of T effector memory cell population (T(EM)) and IFN-γ release. CONCLUSIONS In conclusion, our results demonstrate that the intranasally-administered primary DC vaccine is capable of inducing Ag2/PRA-specific T cell response. Unique approaches utilized in our study represent an attractive and novel means of producing and evaluating an autologous DC-based vaccine.
Collapse
Affiliation(s)
- Prachi Vilekar
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, OK 73117, USA
| | | | | | | | | | | |
Collapse
|
22
|
Corse E, Gottschalk RA, Krogsgaard M, Allison JP. Attenuated T cell responses to a high-potency ligand in vivo. PLoS Biol 2010; 8. [PMID: 20856903 PMCID: PMC2939023 DOI: 10.1371/journal.pbio.1000481] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 08/03/2010] [Indexed: 11/18/2022] Open
Abstract
According to this study, the strongest T cell receptor ligands in vitro do not necessarily induce the strongest T cell responses in vivo, suggesting that vaccine designers may need to reconsider their strategies. αβ T cell receptor (TCR) recognition of foreign peptides bound to major histocompatibility complex (pMHC) molecules on the surface of antigen presenting cells is a key event in the initiation of adaptive cellular immunity. In vitro, high-affinity binding and/or long-lived interactions between TCRs and pMHC correlate with high-potency T cell activation. However, less is known about the influence of TCR/pMHC interaction parameters on T cell responses in vivo. We studied the influence of TCR/pMHC binding characteristics on in vivo T cell immunity by tracking CD4+ T cell activation, effector, and memory responses to immunization with peptides exhibiting a range of TCR/pMHC half-lives and in vitro T cell activation potencies. Contrary to predictions from in vitro studies, we found that optimal in vivo T cell responses occur to ligands with intermediate TCR/pMHC half-lives. The diminished in vivo responses we observed to the ligand exhibiting the longest TCR/pMHC half-life were associated with attenuation of intracellular signaling, expansion, and function over a broad range of time points. Our results reveal a level of control over T cell activation in vivo not recapitulated in in vitro assays and highlight the importance of considering in vivo efficacy of TCR ligands as part of vaccine design. As an important part of immune system, T cells fight infections by recognizing signs of foreign invaders. A molecule on the surface of these cells—called the T cell receptor—recognizes and binds to protein components (peptides) from bacteria, viruses, and other pathogens that are displayed on the surface of other cells. The T cells can then use this information to orchestrate the fight against infection. Vaccination involves injecting into the body foreign peptides that mimic a pathogen, therefore tricking it into raising a T cell response against that pathogen that will be protective in the event of a real infection. We studied T cell responses in mice injected with one of several peptides to which the T cell receptor binds more or less strongly. Contrary to expectations, we found that the peptide that interacted most strongly with the T cell receptor did not provoke the strongest T cell response. This may be nature's way of preventing harmful inflammatory damage due to excessively strong T cell activation. Our work shows that peptides that bind the T cell receptor with medium strength may be best to use for vaccines. Current vaccine strategies seeking to design peptides that bind with maximum strength to the T cell receptor may need to be reconsidered in light of our findings.
Collapse
Affiliation(s)
- Emily Corse
- Department of Immunology, Howard Hughes Medical Institute and Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Rachel A. Gottschalk
- Department of Immunology, Howard Hughes Medical Institute and Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, New York, United States of America
| | - Michelle Krogsgaard
- Department of Pathology and New York University (NYU) Cancer Institute, NYU School of Medicine, New York, New York, United States of America
| | - James P. Allison
- Department of Immunology, Howard Hughes Medical Institute and Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
23
|
Villa CE, Caccia M, Sironi L, D'Alfonso L, Collini M, Rivolta I, Miserocchi G, Gorletta T, Zanoni I, Granucci F, Chirico G. Accumulative difference image protocol for particle tracking in fluorescence microscopy tested in mouse lymphonodes. PLoS One 2010; 5:e12216. [PMID: 20808918 PMCID: PMC2923183 DOI: 10.1371/journal.pone.0012216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 07/14/2010] [Indexed: 11/18/2022] Open
Abstract
The basic research in cell biology and in medical sciences makes large use of imaging tools mainly based on confocal fluorescence and, more recently, on non-linear excitation microscopy. Substantially the aim is the recognition of selected targets in the image and their tracking in time. We have developed a particle tracking algorithm optimized for low signal/noise images with a minimum set of requirements on the target size and with no a priori knowledge of the type of motion. The image segmentation, based on a combination of size sensitive filters, does not rely on edge detection and is tailored for targets acquired at low resolution as in most of the in-vivo studies. The particle tracking is performed by building, from a stack of Accumulative Difference Images, a single 2D image in which the motion of the whole set of the particles is coded in time by a color level. This algorithm, tested here on solid-lipid nanoparticles diffusing within cells and on lymphocytes diffusing in lymphonodes, appears to be particularly useful for the cellular and the in-vivo microscopy image processing in which few a priori assumption on the type, the extent and the variability of particle motions, can be done.
Collapse
Affiliation(s)
- Carlo E. Villa
- Dipartimento di Fisica, Università degli Studi di Milano Bicocca, Milano, Italy
| | - Michele Caccia
- Dipartimento di Fisica, Università degli Studi di Milano Bicocca, Milano, Italy
| | - Laura Sironi
- Dipartimento di Fisica, Università degli Studi di Milano Bicocca, Milano, Italy
| | - Laura D'Alfonso
- Dipartimento di Fisica, Università degli Studi di Milano Bicocca, Milano, Italy
| | - Maddalena Collini
- Dipartimento di Fisica, Università degli Studi di Milano Bicocca, Milano, Italy
| | - Ilaria Rivolta
- Dipartimento di Medicina Sperimentale, Università degli Studi di Milano Bicocca, Monza, Italy
| | - Giuseppe Miserocchi
- Dipartimento di Medicina Sperimentale, Università degli Studi di Milano Bicocca, Monza, Italy
| | - Tatiana Gorletta
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano Bicocca, Milano, Italy
| | - Ivan Zanoni
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano Bicocca, Milano, Italy
| | - Francesca Granucci
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano Bicocca, Milano, Italy
| | - Giuseppe Chirico
- Dipartimento di Fisica, Università degli Studi di Milano Bicocca, Milano, Italy
- * E-mail:
| |
Collapse
|
24
|
Jin GB, Moore AJ, Head JL, Neumiller JJ, Lawrence BP. Aryl hydrocarbon receptor activation reduces dendritic cell function during influenza virus infection. Toxicol Sci 2010; 116:514-22. [PMID: 20498003 DOI: 10.1093/toxsci/kfq153] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
It has long been known that activation of the aryl hydrocarbon receptor (AhR) by ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) suppresses T cell-dependent immune responses; however, the underlying cellular targets and mechanism remain unclear. We have previously shown that AhR activation by TCDD reduces the proliferation and differentiation of influenza virus-specific CD8(+) T cells through an indirect mechanism; suggesting that accessory cells are critical AhR targets during infection. Respiratory dendritic cells (DCs) capture antigen, migrate to lymph nodes, and play a key role in activating naive CD8(+) T cells during respiratory virus infection. Herein, we report an examination of how AhR activation alters DCs in the lung and affects their trafficking to and function in the mediastinal lymph nodes (MLN) during infection with influenza virus. We show that AhR activation impairs lung DC migration and reduces the ability of DCs isolated from the MLN to activate naive CD8(+) T cells. Using novel AhR mutant mice, in which the AhR protein lacks its DNA-binding domain, we show that the suppressive effects of TCDD require that the activated AhR complex binds to DNA. These new findings suggest that AhR activation by chemicals from our environment impacts DC function to stimulate naive CD8(+) T cells and that immunoregulatory genes within DCs are critical targets of AhR. Moreover, our results reinforce the idea that environmental signals and AhR ligands may contribute to differential susceptibilities and responses to respiratory infection.
Collapse
Affiliation(s)
- Guang-Bi Jin
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | | | | | |
Collapse
|
25
|
Fooksman DR, Vardhana S, Vasiliver-Shamis G, Liese J, Blair DA, Waite J, Sacristán C, Victora GD, Zanin-Zhorov A, Dustin ML. Functional anatomy of T cell activation and synapse formation. Annu Rev Immunol 2010; 28:79-105. [PMID: 19968559 DOI: 10.1146/annurev-immunol-030409-101308] [Citation(s) in RCA: 354] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cell activation and function require a structured engagement of antigen-presenting cells. These cell contacts are characterized by two distinct dynamics in vivo: transient contacts resulting from promigratory junctions called immunological kinapses or prolonged contacts from stable junctions called immunological synapses. Kinapses operate in the steady state to allow referencing to self-peptide-MHC (pMHC) and searching for pathogen-derived pMHC. Synapses are induced by T cell receptor (TCR) interactions with agonist pMHC under specific conditions and correlate with robust immune responses that generate effector and memory T cells. High-resolution imaging has revealed that the synapse is highly coordinated, integrating cell adhesion, TCR recognition of pMHC complexes, and an array of activating and inhibitory ligands to promote or prevent T cell signaling. In this review, we examine the molecular components, geometry, and timing underlying kinapses and synapses. We integrate recent molecular and physiological data to provide a synthesis and suggest ways forward.
Collapse
Affiliation(s)
- David R Fooksman
- Department of Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, 10016, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Adenovirus-mediated LIGHT gene modification in murine B-cell lymphoma elicits a potent antitumor effect. Cell Mol Immunol 2010; 7:296-305. [PMID: 20418899 DOI: 10.1038/cmi.2010.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Here, we investigated the antitumor effect of adenovirus-mediated gene transfer of LIGHT, the tumor-necrosis factor (TNF) superfamily member also known as TNFSF14, in the murine A20 B-cell lymphoma. LIGHT gene modification resulted in upregulated expression of Fas and the accessory molecule--intercellular adhesion molecule-1 (ICAM-1) on A20 cells and led to enhanced A20 cell apoptosis. LIGHT-modified A20 cells effectively stimulated the proliferation of T lymphocytes and interferon (IFN)-gamma production in vitro. Immunization of BALB/c mice with a LIGHT-modified A20 cell vaccine efficiently elicited protective immunity against challenge with the parental tumor cell line. Adenovirus-mediated gene transfer of LIGHT by intratumoral injection exerted a very potent antitumor effect against pre-existing A20 cell lymphoma in BALB/c mice. This adenovirus-mediated LIGHT therapy induced substantial splenic natural killer (NK) and cytotoxic T lymphocyte (CTL) activity, enhanced tumor infiltration by inflammatory cells and increased chemokine expression of CC chemokine ligand 21 (CCL21), IFN-inducible protein-10 (IP-10) and monokine induced by IFN-gamma (Mig) from tumor tissues. Thus, adenovirus-mediated LIGHT therapy might have potential utility for the prevention and treatment of B-cell lymphoma.
Collapse
|
27
|
Beattie L, Peltan A, Maroof A, Kirby A, Brown N, Coles M, Smith DF, Kaye PM. Dynamic imaging of experimental Leishmania donovani-induced hepatic granulomas detects Kupffer cell-restricted antigen presentation to antigen-specific CD8 T cells. PLoS Pathog 2010; 6:e1000805. [PMID: 20300603 PMCID: PMC2837408 DOI: 10.1371/journal.ppat.1000805] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Accepted: 02/03/2010] [Indexed: 01/16/2023] Open
Abstract
Kupffer cells (KCs) represent the major phagocytic population within the liver and provide an intracellular niche for the survival of a number of important human pathogens. Although KCs have been extensively studied in vitro, little is known of their in vivo response to infection and their capacity to directly interact with antigen-specific CD8+ T cells. Here, using a combination of approaches including whole mount and thin section confocal microscopy, adoptive cell transfer and intra-vital 2-photon microscopy, we demonstrate that KCs represent the only detectable population of mononuclear phagocytes within granulomas induced by Leishmania donovani infection that are capable of presenting parasite-derived peptide to effector CD8+ T cells. This restriction of antigen presentation to KCs within the Leishmania granuloma has important implications for the identification of new candidate vaccine antigens and for the design of novel immuno-therapeutic interventions. Leishmania donovani is a protozoan parasite that causes severe disease in humans with associated pathology in the spleen and liver. In experimental models of L. donovani infection, the hepatic response to infection is characterised by the presence of a focal mononuclear cell-rich inflammatory response (a granuloma) surrounding cells infected with intracellular amastigotes. Granulomas provide focus to the ensuing immune response, helping to contain parasite dissemination and providing the major effector site responsible for parasites elimination from the liver. Although granulomas are believed to form around infected resident liver macrophages (Kupffer cells), the role of these cells in intra-granuloma antigen presentation is currently unknown. As CD8+ T cells have been shown to play an important role in hepatic resistance to L. donovani following natural infection, vaccination and during immunotherapy, we asked which cells within the granuloma microenvironment serve as targets for antigen recognition by effector CD8+ T cells. Here we provide evidence that the heavily infected mononuclear cell core of the granuloma is composed almost entirely of Kupffer cells, many having migrated from the surrounding sinusoids. Furthermore, by intra-vital 2-photon microscopy, we show that only Kupffer cells laden with intracellular amastigotes are able to form long-lasting antigen-specific interactions with CD8+ T cells within the granuloma microenvironment. These data have important implications for the understanding of how granulomas function to limit infection and may have important implications for the development of vaccines to Leishmania that are designed to induce CD8+ T cell responses.
Collapse
Affiliation(s)
- Lynette Beattie
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Adam Peltan
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Asher Maroof
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Alun Kirby
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Najmeeyah Brown
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Mark Coles
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Deborah F. Smith
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Paul M. Kaye
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
- * E-mail:
| |
Collapse
|
28
|
Azar GA, Lemaître F, Robey EA, Bousso P. Subcellular dynamics of T cell immunological synapses and kinapses in lymph nodes. Proc Natl Acad Sci U S A 2010; 107:3675-80. [PMID: 20133676 PMCID: PMC2840513 DOI: 10.1073/pnas.0905901107] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In vitro studies have revealed that T cell activation occurs during the formation of either dynamic or stable interactions with antigen-presenting cells (APC), and the respective cell junctions have been referred to as immunological kinapses and synapses. However, the relevance and molecular dynamics of kinapses and synapses remain to be established in vivo. Using two-photon imaging, we tracked the distribution of LAT-EGFP molecules during antigen recognition by activated CD4(+) T cells in lymph nodes. At steady state, LAT-EGFP molecules were preferentially found at the uropod of rapidly migrating T cells. In contrast to naïve T cells that fully stopped upon systemic antigen delivery, recently activated T cells decelerated and formed kinapses, characterized by continuous extension of membrane protrusions and by the absence of persistent LAT-EGFP clustering. On the other hand, activated CD4(+) T cells formed stable immunological synapses with antigen-loaded B cells and displayed sustained accumulation of LAT-EGFP fluorescence at the contact zone. Our results show that the state of T cell activation and the type of APC largely influence T cell-APC contact dynamics in lymph nodes. Furthermore, we provide a dynamic look at immunological kinapses and synapses in lymph nodes and suggest the existence of distinct patterns of LAT redistribution during antigen recognition.
Collapse
Affiliation(s)
- Georges A. Azar
- G5 Dynamiques des Réponses Immunes, Institut Pasteur, 75015 Paris, France
- Institut National de la Santé et de la Recherche Médicale U668, Equipe Avenir, 75015 Paris, France; and
| | - Fabrice Lemaître
- G5 Dynamiques des Réponses Immunes, Institut Pasteur, 75015 Paris, France
- Institut National de la Santé et de la Recherche Médicale U668, Equipe Avenir, 75015 Paris, France; and
| | - Ellen A. Robey
- Department of Molecular and Cell Biology, Life Sciences Addition, University of California, Berkeley, CA 94720
| | - Philippe Bousso
- G5 Dynamiques des Réponses Immunes, Institut Pasteur, 75015 Paris, France
- Institut National de la Santé et de la Recherche Médicale U668, Equipe Avenir, 75015 Paris, France; and
| |
Collapse
|
29
|
Linderman JJ, Riggs T, Pande M, Miller M, Marino S, Kirschner DE. Characterizing the dynamics of CD4+ T cell priming within a lymph node. THE JOURNAL OF IMMUNOLOGY 2010; 184:2873-85. [PMID: 20154206 DOI: 10.4049/jimmunol.0903117] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Generating adaptive immunity postinfection or immunization requires physical interaction within a lymph node T zone between Ag-bearing dendritic cells (DCs) and rare cognate T cells. Many fundamental questions remain regarding the dynamics of DC-CD4+ T cell interactions leading to priming. For example, it is not known how the production of primed CD4+ T cells relates to the numbers of cognate T cells, Ag-bearing DCs, or peptide-MHCII level on the DC. To address these questions, we developed an agent-based model of a lymph node to examine the relationships among cognate T cell frequency, DC density, parameters characterizing DC-T cell interactions, and the output of primed T cells. We found that the output of primed CD4+ T cells is linearly related to cognate frequency, but nonlinearly related to the number of Ag-bearing DCs present during infection. This addresses the applicability of two photon microscopy studies to understanding actual infection dynamics, because these types of experiments increase the cognate frequency by orders of magnitude compared with physiologic levels. We found a trade-off between the quantity of peptide-major histocompatibility class II on the surface of individual DCs and number of Ag-bearing DCs present in the lymph node in contributing to the production of primed CD4+ T cells. Interestingly, peptide-major histocompatibility class II t(1/2) plays a minor, although still significant, role in determining CD4+ T cell priming, unlike the primary role that has been suggested for CD8+ T cell priming. Finally, we identify several pathogen-targeted mechanisms that, if altered in their efficiency, can significantly effect the generation of primed CD4+ T cells.
Collapse
Affiliation(s)
- Jennifer J Linderman
- Department of Chemical Engineering, Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
During infection, Toll-like receptor agonists induce natural killer (NK)-cell activation by stimulating dendritic cells (DCs) to produce cytokines and transpresent IL-15 to NK cells. Yet the cellular dynamics underlying NK-cell activation by DCs in secondary lymphoid organs are largely unknown. Here, we have visualized NK-cell activation using mice in which NK cells and DCs express different fluorescent proteins. In response to poly I:C or lipopolysaccharide, NK cells maintained a vigorous migratory behavior, establishing multiple short contacts with maturing DCs. Furthermore, mature antigen-loaded DCs that made long-lived interactions with T cells formed short-lived contacts with NK cells. The different behaviors of T cells and NK cells during activation was correlated with distinct calcium responses upon interaction with DCs. That NK cells become activated while remaining motile may constitute an efficient strategy for sampling local concentrations of cytokines around DCs in secondary lymphoid tissues.
Collapse
|
31
|
Schaeffer M, Han SJ, Chtanova T, van Dooren GG, Herzmark P, Chen Y, Roysam B, Striepen B, Robey EA. Dynamic imaging of T cell-parasite interactions in the brains of mice chronically infected with Toxoplasma gondii. THE JOURNAL OF IMMUNOLOGY 2009; 182:6379-93. [PMID: 19414791 DOI: 10.4049/jimmunol.0804307] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The intracellular parasite Toxoplasma gondii can establish persistent infection in the brain of a mammalian host, a standoff that involves the active participation of host CD8 T cells to control infection. CD8 T cells generally protect against intracellular pathogens by local delivery of effector molecules upon recognition of specific pathogen Ags on invaded host cells. However, the interactions between CD8 T cells, T. gondii, and APCs in the brain have not yet been examined. In this study we have used a mouse infection model in conjunction with two-photon microscopy of living brain tissue and confocal microscopy of fixed brain sections to examine the interactions between CD8 T cells, parasites, and APCs from chronically infected mice. We found that Ag-specific CD8 T cells were recruited to the brains of infected mice and persisted there in the presence of ongoing Ag recognition. Cerebral CD8 T cells made transient contacts with granuloma-like structures containing parasites and with individual CD11b(+) APCs, including some that did not contain parasites. In contrast, T cells ignored intact Ag-bearing cysts and did not contact astrocytes or neurons, including neurons containing parasites or cysts. Our data represent the first direct observation of the dynamics of T cell-parasite interactions within living tissue and provide a new perspective for understanding immune responses to persistent pathogens in the brain.
Collapse
Affiliation(s)
- Marie Schaeffer
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kirschner DE, Linderman JJ. Mathematical and computational approaches can complement experimental studies of host-pathogen interactions. Cell Microbiol 2009; 11:531-9. [PMID: 19134115 DOI: 10.1111/j.1462-5822.2008.01281.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In addition to traditional and novel experimental approaches to study host-pathogen interactions, mathematical and computer modelling have recently been applied to address open questions in this area. These modelling tools not only offer an additional avenue for exploring disease dynamics at multiple biological scales, but also complement and extend knowledge gained via experimental tools. In this review, we outline four examples where modelling has complemented current experimental techniques in a way that can or has already pushed our knowledge of host-pathogen dynamics forward. Two of the modelling approaches presented go hand in hand with articles in this issue exploring fluorescence resonance energy transfer and two-photon intravital microscopy. Two others explore virtual or 'in silico' deletion and depletion as well as a new method to understand and guide studies in genetic epidemiology. In each of these examples, the complementary nature of modelling and experiment is discussed. We further note that multi-scale modelling may allow us to integrate information across length (molecular, cellular, tissue, organism, population) and time (e.g. seconds to lifetimes). In sum, when combined, these compatible approaches offer new opportunities for understanding host-pathogen interactions.
Collapse
Affiliation(s)
- Denise E Kirschner
- Department of Microbiology and Immunology, 6730 Medical Science Bldg. II, University of Michigan Medical School, Ann Arbor, MI, USA.
| | | |
Collapse
|
33
|
Swirski FK, Weissleder R, Pittet MJ. Heterogeneous in vivo behavior of monocyte subsets in atherosclerosis. Arterioscler Thromb Vasc Biol 2009; 29:1424-32. [PMID: 19372462 DOI: 10.1161/atvbaha.108.180521] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Monocytes and macrophages play active roles in atherosclerosis, a chronic inflammatory disease that is a leading cause of death in the developed world. The prevailing paradigm states that, during human atherogenesis, monocytes accumulate in the arterial intima and differentiate into macrophages, which then ingest oxidized lipoproteins, secrete a diverse array of proinflammatory mediators, and eventually become foam cells, the key constituents of a vulnerable plaque. Yet monocytes are heterogeneous. In the mouse, one subset (Ly-6C(hi)) promotes inflammation, expands in hypercholesterolemic conditions, and selectively gives rise to macrophages in atheromata. A different subset (Ly-6C(lo)) attenuates inflammation and promotes angiogenesis and granulation tissue formation in models of tissue injury, but its role in atherosclerosis is largely unknown. In the human, monocyte heterogeneity is preserved but it is still unresolved how subsets correspond functionally. The contradistinctive properties of these cells suggest commitment for specific function before infiltrating tissue. Such commitment argues for discriminate targeting of deleterious subsets while sparing host defense and repair mechanisms. In addition to advancing our understanding of atherosclerosis, the ability to target and image monocyte subsets would allow us to evaluate drugs designed to selectively inhibit monocyte subset recruitment or function, and to stratify patients at risk for developing complications such as myocardial infarction or stroke. In this review we summarize recent advances of our understanding of the behavioral heterogeneity of monocytes during disease progression and outline emerging molecular imaging approaches to address key questions in the field.
Collapse
Affiliation(s)
- Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA.
| | | | | |
Collapse
|
34
|
de Abreu FV, Mostardinha P. Maximal frustration as an immunological principle. J R Soc Interface 2009; 6:321-34. [PMID: 18715844 PMCID: PMC2659376 DOI: 10.1098/rsif.2008.0280] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A fundamental problem in immunology is that of understanding how the immune system selects promptly which cells to kill without harming the body. This problem poses an apparent paradox. Strong reactivity against pathogens seems incompatible with perfect tolerance towards self. We propose a different view on cellular reactivity to overcome this paradox: effector functions should be seen as the outcome of cellular decisions which can be in conflict with other cells' decisions. We argue that if cellular systems are frustrated, then extensive cross-reactivity among the elements in the system can decrease the reactivity of the system as a whole and induce perfect tolerance. Using numerical and mathematical analyses, we discuss two simple models that perform optimal pathogenic detection with no autoimmunity if cells are maximally frustrated. This study strongly suggests that a principle of maximal frustration could be used to build artificial immune systems. It would be interesting to test this principle in the real adaptive immune system.
Collapse
Affiliation(s)
- F Vistulo de Abreu
- Departamento de Física, Universidade de Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal.
| | | |
Collapse
|
35
|
Kirschner DE, Linderman JJ. Mathematical and computational approaches can complement experimental studies of host-pathogen interactions. Cell Microbiol 2009. [DOI: 10.1111/j.1462-5822.2009.01281.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Abstract
A functional immune system depends on the appropriate activation of lymphocytes following antigen encounter. In this Review, we summarize studies that have used high-resolution imaging approaches to visualize antigen presentation to B cells in secondary lymphoid organs. These studies illustrate that encounters of B cells with antigen in these organs can be facilitated by diffusion of the antigen or by the presentation of antigen by macrophages, dendritic cells and follicular dendritic cells. We describe cell-surface molecules that might be important in mediating antigen presentation to B cells and also highlight the key role of B cells themselves in antigen transport. Data obtained from the studies discussed here highlight the predominance, importance and variety of the cell-mediated processes that are involved in presenting antigen to B cells in vivo.
Collapse
|
37
|
Worbs T, Förster R. T cell migration dynamics within lymph nodes during steady state: an overview of extracellular and intracellular factors influencing the basal intranodal T cell motility. Curr Top Microbiol Immunol 2009; 334:71-105. [PMID: 19521682 DOI: 10.1007/978-3-540-93864-4_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Naive T lymphocytes continuously recirculate through secondary lymphoid organs such as lymph nodes until they are eventually activated by recognizing cognate peptide/MHC-complexes on the surface of antigen-protecting cells. The intranodal T cell migration behavior leading to these crucial--and potentially rare--encounters during the induction of an adaptive immune response could not be directly addressed until, in 2002, the use of two-photon microscopy also allowed the visualization of cellular dynamics deep within intact lymph nodes. Since then, numerous studies have confirmed that, by default, naive T cells are extremely motile, scanning the paracortical T cell zone for cognate antigen by means of an apparent random walk. This review attempts to summarize the current knowledge of factors influencing the basal migration behavior of naive T lymphocytes within lymph nodes during steady state. Extracellular cues, such as the motility-promoting influence of CCR7 ligands and the role of integrins during interstitial migration, as well as intracellular signaling pathways involved in T cell motility, will be discussed. Particular emphasis is placed on structural features of the lymph node environment orchestrating T cell migration, namely the framework of fibroblastic reticular cells serving as migration "highways." Finally, new approaches to simulate the cellular dynamics within lymph nodes in silico by means of mathematical modeling will be reviewed.
Collapse
Affiliation(s)
- Tim Worbs
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany.
| | | |
Collapse
|
38
|
Junt T, Scandella E, Ludewig B. Form follows function: lymphoid tissue microarchitecture in antimicrobial immune defence. Nat Rev Immunol 2008; 8:764-75. [PMID: 18825130 DOI: 10.1038/nri2414] [Citation(s) in RCA: 209] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Secondary lymphoid organs (SLOs) are tissues that facilitate the induction of adaptive immune responses. These organs capture pathogens to limit their spread throughout the body, bring antigen-presenting cells into productive contact with their cognate lymphocytes and provide niches for the differentiation of immune effector cells. Therefore, the microanatomy of SLOs defines the ability of an organism to respond to pathogens. SLO microarchitecture is, at the same time, extremely adaptable to environmental changes. In this Review, we discuss recent insights into the function and plasticity of the SLO microenvironment with regards to antimicrobial immune defence.
Collapse
Affiliation(s)
- Tobias Junt
- Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland.
| | | | | |
Collapse
|
39
|
Mempel TR, Bauer CA. Intravital imaging of CD8+ T cell function in cancer. Clin Exp Metastasis 2008; 26:311-27. [PMID: 18665448 DOI: 10.1007/s10585-008-9196-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 07/14/2008] [Indexed: 12/11/2022]
Abstract
Recent technological advances in photonics are making intravital microscopy (IVM) an increasingly powerful approach for the mechanistic exploration of biological processes in the physiological context of complex native tissue environments. Direct, dynamic and multiparametric visualization of immune cell behavior in living animals at cellular and subcellular resolution has already proved its utility in auditing basic immunological concepts established through conventional approaches and has also generated new hypotheses that can conversely be complemented and refined by traditional experimental methods. The insight that outgrowing tumors must not necessarily have evaded recognition by the adaptive immune system, but can escape rejection by actively inducing a state of immunological tolerance calls for a detailed investigation of the cellular and molecular mechanisms by which the anti-cancer response is subverted. Along with molecular imaging techniques that provide dynamic information at the population level, IVM can be expected to make a critical contribution to this effort by allowing the observation of immune cell behavior in vivo at single cell-resolution. We review here how IVM-based investigation can help to clarify the role of cytotoxic T lymphocytes (CTL) in the immune response against cancer and identify the ways by which their function might be impaired through tolerogenic mechanisms.
Collapse
Affiliation(s)
- Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | | |
Collapse
|