1
|
Zhu E, Yu J, Li YR, Ma F, Wang YC, Liu Y, Li M, Kim YJ, Zhu Y, Hahn Z, Zhou Y, Brown J, Zhang Y, Pelegrini M, Hsiai T, Yang L, Huang Y. Biomimetic cell stimulation with a graphene oxide antigen-presenting platform for developing T cell-based therapies. NATURE NANOTECHNOLOGY 2024:10.1038/s41565-024-01781-4. [PMID: 39313679 DOI: 10.1038/s41565-024-01781-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/08/2024] [Indexed: 09/25/2024]
Abstract
Chimeric antigen receptor (CAR)-engineered T cells represent a front-line therapy for cancers. However, the current CAR T cell manufacturing protocols do not adequately reproduce immunological synapse formation. Here, in response to this limitation, we have developed a flexible graphene oxide antigen-presenting platform (GO-APP) that anchors antibodies onto graphene oxide. By decorating anti-CD3 (αCD3) and anti-CD28 (αCD28) on graphene oxide (GO-APP3/28), we achieved remarkable T cell proliferation. In vitro interactions between GO-APP3/28 and T cells closely mimic the in vivo immunological synapses between antigen-presenting cells and T cells. This immunological synapse mimicry shows a high capacity for stimulating T cell proliferation while preserving their multifunctionality and high potency. Meanwhile, it enhances CAR gene-engineering efficiency, yielding a more than fivefold increase in CAR T cell production compared with the standard protocol. Notably, GO-APP3/28 stimulated appropriate autocrine interleukin-2 (IL-2) in T cells and overcame the in vitro reliance on external IL-2 supplementation, offering an opportunity to culture T cell-based products independent of IL-2 supplementation.
Collapse
Affiliation(s)
- Enbo Zhu
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiaji Yu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Feiyang Ma
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yu-Chen Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yang Liu
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Miao Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yu Jeong Kim
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zoe Hahn
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yang Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - James Brown
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yuchong Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matteo Pelegrini
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tzung Hsiai
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Yu Huang
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Hodapp SJ, Gravel N, Kannan N, Newton AC. Cancer-associated mutations in protein kinase C theta are loss-of-function. Biochem J 2024; 481:759-775. [PMID: 38752473 PMCID: PMC11346454 DOI: 10.1042/bcj20240148] [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: 04/02/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/11/2024]
Abstract
The Ca2+-independent, but diacylglycerol-regulated, novel protein kinase C (PKC) theta (θ) is highly expressed in hematopoietic cells where it participates in immune signaling and platelet function. Mounting evidence suggests that PKCθ may be involved in cancer, particularly blood cancers, breast cancer, and gastrointestinal stromal tumors, yet how to target this kinase (as an oncogene or as a tumor suppressor) has not been established. Here, we examine the effect of four cancer-associated mutations, R145H/C in the autoinhibitory pseudosubstrate, E161K in the regulatory C1A domain, and R635W in the regulatory C-terminal tail, on the cellular activity and stability of PKCθ. Live-cell imaging studies using the genetically-encoded fluorescence resonance energy transfer-based reporter for PKC activity, C kinase activity reporter 2 (CKAR2), revealed that the pseudosubstrate and C1A domain mutations impaired autoinhibition to increase basal signaling. This impaired autoinhibition resulted in decreased stability of the protein, consistent with the well-characterized behavior of Ca2+-regulated PKC isozymes wherein mutations that impair autoinhibition are paradoxically loss-of-function because the mutant protein is degraded. In marked contrast, the C-terminal tail mutation resulted in enhanced autoinhibition and enhanced stability. Thus, the examined mutations were loss-of-function by different mechanisms: mutations that impaired autoinhibition promoted the degradation of PKC, and those that enhanced autoinhibition stabilized an inactive PKC. Supporting a general loss-of-function of PKCθ in cancer, bioinformatics analysis revealed that protein levels of PKCθ are reduced in diverse cancers, including lung, renal, head and neck, and pancreatic. Our results reveal that PKCθ function is lost in cancer.
Collapse
Affiliation(s)
- Stefanie J. Hodapp
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Nathan Gravel
- Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, U.S.A
| | - Natarajan Kannan
- Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, U.S.A
| | - Alexandra C. Newton
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
| |
Collapse
|
3
|
Liu J, Zhang B, Zhang G, Shang D. Reprogramming of regulatory T cells in inflammatory tumor microenvironment: can it become immunotherapy turning point? Front Immunol 2024; 15:1345838. [PMID: 38449875 PMCID: PMC10915070 DOI: 10.3389/fimmu.2024.1345838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Overcoming the immunosuppressive tumor microenvironment and identifying widely used immunosuppressants with minimal side effects are two major challenges currently hampering cancer immunotherapy. Regulatory T cells (Tregs) are present in almost all cancer tissues and play an important role in preserving autoimmune tolerance and tissue homeostasis. The tumor inflammatory microenvironment causes the reprogramming of Tregs, resulting in the conversion of Tregs to immunosuppressive phenotypes. This process ultimately facilitates tumor immune escape or tumor progression. However, current systemic Treg depletion therapies may lead to severe autoimmune toxicity. Therefore, it is crucial to understand the mechanism of Treg reprogramming and develop immunotherapies that selectively target Tregs within tumors. This article provides a comprehensive review of the potential mechanisms involved in Treg cell reprogramming and explores the application of Treg cell immunotherapy. The interference with reprogramming pathways has shown promise in reducing the number of tumor-associated Tregs or impairing their function during immunotherapy, thereby improving anti-tumor immune responses. Furthermore, a deeper understanding of the mechanisms that drive Treg cell reprogramming could reveal new molecular targets for future treatments.
Collapse
Affiliation(s)
- Jinming Liu
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Biao Zhang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guolin Zhang
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
4
|
Zhang Z, Liu C, Wang M, Sun R, Yang Z, Hua Z, Wu Y, Wu M, Wang H, Qiu W, Yin H, Yang M. Treating solid tumors with TCR-based chimeric antigen receptor targeting extra domain B-containing fibronectin. J Immunother Cancer 2023; 11:e007199. [PMID: 37586774 PMCID: PMC10432677 DOI: 10.1136/jitc-2023-007199] [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] [Accepted: 07/27/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND The suppression of chimeric antigen receptor (CAR) T cells by the tumor microenvironment (TME) is a crucial obstacle in the T-cell-based treatment of solid tumors. Extra domain B (EDB)-fibronectin is an oncofetal antigen expressed on the endothelium layer of the neovasculature and cancer cells. Though recognized as a T cell therapy target, engineered CAR T cells thus far have failed to demonstrate satisfactory in vivo efficacy. In this study, we report that targeting EDB-fibronectin by redirected TCR-CAR T cells (rTCR-CAR) bypasses the suppressive TME for solid tumor treatment and sufficiently suppressed tumor growth.We generated EDB-targeting CAR by fusing single-chain variable fragment to CD3ε, resulting in rTCR-CAR. Human primary T cells and Jurkat cells were used to study the EDB-targeting T cells. Differences to the traditional second-generation CAR T cell in signaling, immune synapse formation, and T cell exhaustion were characterized. Cytotoxicity of the rTCR-CAR T cells was tested in vitro, and therapeutic efficacies were demonstrated using xenograft models. METHODS RESULTS: In the xenograft models, the rTCR-CAR T cells demonstrated in vivo efficacies superior to that based on traditional CAR design. A significant reduction in tumor vessel density was observed alongside tumor growth inhibition, extending even to tumor models established with EDB-negative cancer cells. The rTCR-CAR bound to immobilized EDB, and the binding led to immune synapse structures superior to that formed by second-generation CARs. By a mechanism similar to that for the conventional TCR complex, EDB-fibronectin activated the rTCR-CAR, resulting in rTCR-CAR T cells with low basal activation levels and increased in vivo expansion. CONCLUSION Our study has demonstrated the potential of rTCR-CAR T cells targeting the EDB-fibronectin as an anticancer therapeutic. Engineered to possess antiangiogenic and cytotoxic activities, the rTCR-CAR T cells showed therapeutic efficacies not impacted by the suppressive TMEs. These combined characteristics of a single therapeutic agent point to its potential to achieve sustained control of solid tumors.
Collapse
Affiliation(s)
- Zhijie Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chang Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Muhan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Rongcheng Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
- Jiangsu Cell Tech Medical Research Institute, Nanjing, Jiangsu, China
| | - Zhe Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Zhen Hua
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yushuang Wu
- Jiangsu Cell Tech Medical Research Institute, Nanjing, Jiangsu, China
| | - Mengting Wu
- Jiangsu Cell Tech Medical Research Institute, Nanjing, Jiangsu, China
| | - Hang Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wen Qiu
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongping Yin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Meijia Yang
- Jiangsu Cell Tech Medical Research Institute, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Shen DD, Bi YP, Pang JR, Zhao LJ, Zhao LF, Gao Y, Wang B, Liu HM, Liu Y, Wang N, Zheng YC, Liu HM. Generation, secretion and degradation of cancer immunotherapy target PD-L1. Cell Mol Life Sci 2022; 79:413. [PMID: 35819633 PMCID: PMC11073444 DOI: 10.1007/s00018-022-04431-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 06/06/2022] [Accepted: 06/14/2022] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy is a rapidly developing and effective method for the treatment of a variety of malignancies in recent years. As a significant immune checkpoint, programmed cell death 1 ligand 1 (PD-L1) and its receptor programmed cell death protein 1 (PD-1) play the most significant role in cancer immune escape and cancer immunotherapy. Though PD-L1 have become an important target for drug development and there have been various approved drugs and clinic trials targeting it, and various clinical response rate and adverse reactions prevent many patients from benefiting from it. In recent years, combination trials have become the main direction of PD-1/PD-L1 antibodies development. Here, we summarized PD-L1 biofunctions and key roles in various cancers along with the development of PD-L1 inhibitors. The regulators that are involved in controlling PD-L1 expression including post-translational modification, mRNA level regulation as well as degradation and exosome secretory pathway of PD-L1 were focused. This systematic summary may provide comprehensive understanding of different regulations on PD-L1 as well as a broad prospect for the search of the important regulator of PD-L1. The regulatory factors of PD-L1 can be potential targets for immunotherapy and increase strategies of immunotherapy in combination.
Collapse
Affiliation(s)
- Dan-Dan Shen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment Zhengzhou China, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China
| | - Ya-Ping Bi
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China
| | - Jing-Ru Pang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China
| | - Li-Juan Zhao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Academy of Medical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China
| | - Long-Fei Zhao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China
| | - Ya Gao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China
| | - Bo Wang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China
| | - Hui-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China
| | - Ying Liu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ning Wang
- The School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yi-Chao Zheng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment Zhengzhou China, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Academy of Medical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China.
| | - Hong-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Academy of Medical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
6
|
Wu H, Cao R, Wen M, Xue H, OuYang B. Structural characterization of a dimerization interface in the CD28 transmembrane domain. Structure 2022; 30:803-812.e5. [PMID: 35397202 DOI: 10.1016/j.str.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/15/2022] [Accepted: 03/01/2022] [Indexed: 11/19/2022]
Abstract
CD28 has a crucial role in regulating immune responses by enhancing T cell activation and differentiation. Recent studies have shown that the transmembrane helix (TMH) of CD28 mediates receptor assembly and activity, but a structural characterization of TMH is still lacking. Here, we determined the dimeric helix-helix packing of CD28-TMH using nuclear magnetic resonance (NMR) technology. Unexpectedly, wild-type CD28-TMH alone forms stable tetramers in lipid bicelles instead of dimers. The NMR structure of the CD28-TMH C165F mutant reveals that a GxxxA motif, which is highly conserved in many dimeric assemblies, is located at the dimerization interface. Mutating G160 and A164 can disrupt the transmembrane helix assembly and reduces CD28 enhancement in cells. In contrast, a previously proposed YxxxxT motif does not affect the dimerization of full-length CD28, but it does affect CD28 activity. These results imply that the transmembrane domain of CD28 regulates the signaling transduction in a complicated manner.
Collapse
Affiliation(s)
- Hongyi Wu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruiyu Cao
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Maorong Wen
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongjuan Xue
- National Facility for Protein Science in Shanghai, ZhangJiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bo OuYang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
7
|
Abstract
Immune principles formulated by Jenner, Pasteur, and early immunologists served as fundamental propositions for vaccine discovery against many dreadful pathogens. However, decisive success in the form of an efficacious vaccine still eludes for diseases such as tuberculosis, leishmaniasis, and trypanosomiasis. Several antileishmanial vaccine trials have been undertaken in past decades incorporating live, attenuated, killed, or subunit vaccination, but the goal remains unmet. In light of the above facts, we have to reassess the principles of vaccination by dissecting factors associated with the hosts' immune response. This chapter discusses the pathogen-associated perturbations at various junctures during the generation of the immune response which inhibits antigenic processing, presentation, or remodels memory T cell repertoire. This can lead to ineffective priming or inappropriate activation of memory T cells during challenge infection. Thus, despite a protective primary response, vaccine failure can occur due to altered immune environments in the presence of pathogens.
Collapse
Affiliation(s)
| | - Sunil Kumar
- National Centre for Cell Science, Pune, Maharashtra, India
| | | | - Bhaskar Saha
- National Centre for Cell Science, Pune, Maharashtra, India.
- Trident Academy of Creative Technology, Bhubaneswar, Odisha, India.
| |
Collapse
|
8
|
Belardi B, Son S, Felce JH, Dustin ML, Fletcher DA. Cell-cell interfaces as specialized compartments directing cell function. Nat Rev Mol Cell Biol 2020; 21:750-764. [PMID: 33093672 DOI: 10.1038/s41580-020-00298-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
Cell-cell interfaces are found throughout multicellular organisms, from transient interactions between motile immune cells to long-lived cell-cell contacts in epithelia. Studies of immune cell interactions, epithelial cell barriers, neuronal contacts and sites of cell-cell fusion have identified a core set of features shared by cell-cell interfaces that critically control their function. Data from diverse cell types also show that cells actively and passively regulate the localization, strength, duration and cytoskeletal coupling of receptor interactions governing cell-cell signalling and physical connections between cells, indicating that cell-cell interfaces have a unique membrane organization that emerges from local molecular and cellular mechanics. In this Review, we discuss recent findings that support the emerging view of cell-cell interfaces as specialized compartments that biophysically constrain the arrangement and activity of their protein, lipid and glycan components. We also review how these biophysical features of cell-cell interfaces allow cells to respond with high selectivity and sensitivity to multiple inputs, serving as the basis for wide-ranging cellular functions. Finally, we consider how the unique properties of cell-cell interfaces present opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Brian Belardi
- Department of Bioengineering & Biophysics Program, UC Berkeley, Berkeley, CA, USA
| | - Sungmin Son
- Department of Bioengineering & Biophysics Program, UC Berkeley, Berkeley, CA, USA
| | | | | | - Daniel A Fletcher
- Department of Bioengineering & Biophysics Program, UC Berkeley, Berkeley, CA, USA.
- Division of Biological Systems & Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
9
|
Edelstein HI, Donahue PS, Muldoon JJ, Kang AK, Dolberg TB, Battaglia LM, Allchin ER, Hong M, Leonard JN. Elucidation and refinement of synthetic receptor mechanisms. Synth Biol (Oxf) 2020; 5:ysaa017. [PMID: 33392392 PMCID: PMC7759213 DOI: 10.1093/synbio/ysaa017] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/13/2020] [Accepted: 09/02/2020] [Indexed: 12/22/2022] Open
Abstract
Synthetic receptors are powerful tools for engineering mammalian cell-based devices. These biosensors enable cell-based therapies to perform complex tasks such as regulating therapeutic gene expression in response to sensing physiological cues. Although multiple synthetic receptor systems now exist, many aspects of receptor performance are poorly understood. In general, it would be useful to understand how receptor design choices influence performance characteristics. In this study, we examined the modular extracellular sensor architecture (MESA) and systematically evaluated previously unexamined design choices, yielding substantially improved receptors. A key finding that might extend to other receptor systems is that the choice of transmembrane domain (TMD) is important for generating high-performing receptors. To provide mechanistic insights, we adopted and employed a Förster resonance energy transfer-based assay to elucidate how TMDs affect receptor complex formation and connected these observations to functional performance. To build further insight into these phenomena, we developed a library of new MESA receptors that sense an expanded set of ligands. Based upon these explorations, we conclude that TMDs affect signaling primarily by modulating intracellular domain geometry. Finally, to guide the design of future receptors, we propose general principles for linking design choices to biophysical mechanisms and performance characteristics.
Collapse
Affiliation(s)
- Hailey I Edelstein
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Patrick S Donahue
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL 60208, USA
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joseph J Muldoon
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL 60208, USA
| | - Anthony K Kang
- Honors Program in Medical Education, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Program in Biological Sciences, Northwestern University, Evanston, IL, 60208, USA
| | - Taylor B Dolberg
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Lauren M Battaglia
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Everett R Allchin
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Mihe Hong
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Joshua N Leonard
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL 60208, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
10
|
Siokis A, Robert PA, Demetriou P, Dustin ML, Meyer-Hermann M. F-Actin-Driven CD28-CD80 Localization in the Immune Synapse. Cell Rep 2019; 24:1151-1162. [PMID: 30067972 DOI: 10.1016/j.celrep.2018.06.114] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 04/18/2018] [Accepted: 06/28/2018] [Indexed: 12/27/2022] Open
Abstract
During immunological synapse (IS) formation, T cell receptor (TCR) signaling complexes, integrins, and costimulatory molecules exhibit a particular spatial localization. Here, we develop an agent-based model for the IS formation based on TCR peptide-bound major histocompatibility complex (pMHC) and leukocyte-function-associated antigen 1 (LFA-1) intracellular activation molecule 1 (ICAM-1) dynamics, including CD28 binding to a costimulatory ligand, coupling of molecules to the centripetal actin flow, and size-based segregation (SBS). A radial gradient of LFA-1 in the peripheral supramolecular activation cluster (pSMAC) toward the central supramolecular activation cluster (cSMAC) emerged as a combined consequence of actin binding and diffusion and modified the positioning of other molecules. The simulations predict a mechanism of CD28 movement, according to which CD28-CD80 complexes passively follow TCR-pMHC microclusters. However, the characteristic CD28-CD80 localization in a ring pattern around the cSMAC only emerges with a particular CD28-actin coupling strength that induces a centripetal motion. These results have implications for the understanding of T cell activation and fate decisions.
Collapse
Affiliation(s)
- Anastasios Siokis
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany
| | - Philippe A Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany.
| | - Philippos Demetriou
- Kennedy Institute, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7FY, UK
| | - Michael L Dustin
- Kennedy Institute, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7FY, UK; Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany; Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig 38106, Germany.
| |
Collapse
|
11
|
Gilfillan CB, Wang C, Mohsen MO, Rufer N, Hebeisen M, Allard M, Verdeil G, Irvine DJ, Bachmann MF, Speiser DE. Murine CD8 T-cell functional avidity is stable in vivo but not in vitro: Independence from homologous prime/boost time interval and antigen density. Eur J Immunol 2019; 50:505-514. [PMID: 31785153 PMCID: PMC7187562 DOI: 10.1002/eji.201948355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/17/2019] [Accepted: 11/27/2019] [Indexed: 01/13/2023]
Abstract
It is known that for achieving high affinity antibody responses, vaccines must be optimized for antigen dose/density, and the prime/boost interval should be at least 4 weeks. Similar knowledge is lacking for generating high avidity T‐cell responses. The functional avidity (FA) of T cells, describing responsiveness to peptide, is associated with the quality of effector function and the protective capacity in vivo. Despite its importance, the FA is rarely determined in T‐cell vaccination studies. We addressed the question whether different time intervals for short‐term homologous vaccinations impact the FA of CD8 T‐cell responses. Four‐week instead of 2‐week intervals between priming and boosting with potent subunit vaccines in C57BL/6 mice did not improve FA. Equally, similar FA was observed after vaccination with virus‐like particles displaying low versus high antigen densities. Interestingly, FA was stable in vivo but not in vitro, depending on the antigen dose and the time interval since T‐cell activation, as observed in murine monoclonal T cells. Our findings suggest dynamic in vivo modulation for equal FA. We conclude that low antigen density vaccines or a minimal 4‐week prime/boost interval are not crucial for the T‐cell's FA, in contrast to antibody responses.
Collapse
Affiliation(s)
| | - Chensu Wang
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Mona O Mohsen
- Inselspital, Universitaetsklinik RIA, Immunologie, Bern, Switzerland.,Jenner Institute, University of Oxford, Oxford, UK
| | - Nathalie Rufer
- Department of Oncology, University of Lausanne, Switzerland.,Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | | | | | | | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Martin F Bachmann
- Inselspital, Universitaetsklinik RIA, Immunologie, Bern, Switzerland.,Jenner Institute, University of Oxford, Oxford, UK
| | - Daniel E Speiser
- Department of Oncology, University of Lausanne, Switzerland.,Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
12
|
Connolly A, Gagnon E. Electrostatic interactions: From immune receptor assembly to signaling. Immunol Rev 2019; 291:26-43. [DOI: 10.1111/imr.12769] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Audrey Connolly
- Institut de Recherche en Immunologie et Cancérologie/Institute for Research in Immunology and Cancer Montréal Québec Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine Université de Montréal Montréal Québec Canada
| | - Etienne Gagnon
- Institut de Recherche en Immunologie et Cancérologie/Institute for Research in Immunology and Cancer Montréal Québec Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine Université de Montréal Montréal Québec Canada
| |
Collapse
|
13
|
|
14
|
Salerno F, Guislain A, Freen-Van Heeren JJ, Nicolet BP, Young HA, Wolkers MC. Critical role of post-transcriptional regulation for IFN-γ in tumor-infiltrating T cells. Oncoimmunology 2018; 8:e1532762. [PMID: 30713785 DOI: 10.1080/2162402x.2018.1532762] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022] Open
Abstract
Protective T cell responses against tumors require the production of Interferon gamma (IFN-γ). However, tumor-infiltrating T cells (TILs) gradually lose their capacity to produce IFN-γ and therefore fail to clear malignant cells. Dissecting the underlying mechanisms that block cytokine production is thus key for improving T cell products. Here we show that although TILs express substantial levels of Ifng mRNA, post-transcriptional mechanisms impede the production of IFN-γ protein due to loss of mRNA stability. CD28 triggering, but not PD1 blocking antibodies, effectively restores the stability of Ifng mRNA. Intriguingly, TILs devoid of AU-rich elements within the 3'untranslated region maintain stabilized Ifng mRNA and produce more IFN-γ protein than wild-type TILs. This sustained IFN-γ production translates into effective suppression of tumor outgrowth, which is almost exclusively mediated by direct effects on the tumor cells. We therefore conclude that post-transcriptional mechanisms could be modulated to potentiate effective T cell therapies in cancer.
Collapse
Affiliation(s)
- Fiamma Salerno
- Department of Hematopoiesis, Sanquin Research/AMC Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Aurelie Guislain
- Department of Hematopoiesis, Sanquin Research/AMC Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Julian J Freen-Van Heeren
- Department of Hematopoiesis, Sanquin Research/AMC Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Benoit P Nicolet
- Department of Hematopoiesis, Sanquin Research/AMC Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Howard A Young
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, MD, USA
| | - Monika C Wolkers
- Department of Hematopoiesis, Sanquin Research/AMC Landsteiner Laboratory, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Benard E, Nunès JA, Limozin L, Sengupta K. T Cells on Engineered Substrates: The Impact of TCR Clustering Is Enhanced by LFA-1 Engagement. Front Immunol 2018; 9:2085. [PMID: 30279692 PMCID: PMC6154019 DOI: 10.3389/fimmu.2018.02085] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/23/2018] [Indexed: 12/15/2022] Open
Abstract
We created APC-mimetic synthetic substrates to study the impact of ligand clustering on T cell activation and spreading. The substrates exhibit antibodies directed against the TCR-complex in the form of a patterned array of sub micrometric dots surrounded by a fluid supported lipid bilayer (SLB) which may itself be functionalized with another bio-molecule. We show that for T cell adhesion mediated by T cell receptor (TCR) alone, in the patterned, but not in the corresponding homogeneous controls, the TCR, ZAP-70 and actin are present in the form of clusters or patches that co-localize with the ligand-dots. However, global cell scale parameters like cell area and actin distribution are only weakly impacted by ligand clustering. In presence of ICAM-1 - the ligand of the T cell integrin LFA-1 - on the SLB, the TCR is still clustered due to the patterning of its ligands, but now global parameters are also impacted. The actin organization changes to a peripheral ring, resembling the classical actin distribution seen on homogeneous substrates, the patterned membrane topography disappears and the membrane is flat, whereas the cell area increases significantly. These observations taken together point to a possible pivotal role for LFA-1 in amplifying the effect of TCR-clustering. No such effect is evident for co-engagement of CD28, affected via its ligand B7.2. Unlike on ICAM-1, on B7.2 cell spreading and actin organization are similar for homogeneous and patterned substrates. However, TCR and ZAP-70 clusters are still formed in the patterned case. These results indicate complementary role for LFA-1 and CD28 in the regulation and putative coupling of TCR micro-clusters to actin. The engineered substrates presented here clearly have the potential to act as platform for fundamental research in immune cell biology, as well as translational analyses in immunotherapy, for example to screen molecules for their role in T cell adhesion/activation.
Collapse
Affiliation(s)
| | - Jacques A Nunès
- CNRS, UMR7258, Centre de Recherche en Cancerologie de Marseille, Immunity and Cancer Team, Institut Paoli-Calmettes, Inserm, U1068, Aix-Marseille Université UM 105, Marseille, France
| | - Laurent Limozin
- LAI, CNRS UMR 7333, INSERM UMR 1067, Aix-Marseille Université, Marseille, France
| | - Kheya Sengupta
- CNRS, CINaM UMR 7325, Aix-Marseille Université, Marseille, France
| |
Collapse
|
16
|
Kinoshita M, Suzuki KG, Murata M, Matsumori N. Evidence of lipid rafts based on the partition and dynamic behavior of sphingomyelins. Chem Phys Lipids 2018; 215:84-95. [DOI: 10.1016/j.chemphyslip.2018.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/13/2018] [Accepted: 07/10/2018] [Indexed: 01/10/2023]
|
17
|
Finetti F, Baldari CT. The immunological synapse as a pharmacological target. Pharmacol Res 2018; 134:118-133. [PMID: 29898412 DOI: 10.1016/j.phrs.2018.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/25/2018] [Accepted: 06/07/2018] [Indexed: 12/29/2022]
Abstract
The development of T cell mediated immunity relies on the assembly of a highly specialized interface between T cell and antigen presenting cell (APC), known as the immunological synapse (IS). IS assembly is triggered when the T cell receptor (TCR) binds to specific peptide antigen presented in association to the major histocompatibility complex (MHC) by the APC, and is followed by the spatiotemporal dynamic redistribution of TCR, integrins, co-stimulatory receptors and signaling molecules, allowing for the fine-tuning and integration of the signals that lead to T cell activation. The knowledge acquired to date about the mechanisms of IS assembly underscores this structure as a robust pharmacological target. The activity of molecules involved in IS assembly and function can be targeted by specific compounds to modulate the immune response in a number of disorders, including cancers and autoimmune diseases, or in transplanted patients. Here, we will review the state-of-the art of the current therapies which exploit the IS to modulate the immune response.
Collapse
Affiliation(s)
- Francesca Finetti
- Department of Life Sciences, University of Siena, via A. Moro 2, Siena, 53100, Italy.
| | - Cosima T Baldari
- Department of Life Sciences, University of Siena, via A. Moro 2, Siena, 53100, Italy
| |
Collapse
|
18
|
Li J, Zhang Y, Zhang Y, Lü S, Miao Y, Yang J, Huang S, Ma X, Han L, Deng J, Fan F, Liu B, Huo Y, Xu Q, Chen C, Wang X, Feng J. GSNOR modulates hyperhomocysteinemia-induced T cell activation and atherosclerosis by switching Akt S-nitrosylation to phosphorylation. Redox Biol 2018; 17:386-399. [PMID: 29860106 PMCID: PMC6007174 DOI: 10.1016/j.redox.2018.04.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 04/24/2018] [Accepted: 04/28/2018] [Indexed: 01/25/2023] Open
Abstract
The adaptive immune system plays a critical role in hyperhomocysteinemia (HHcy)-accelerated atherosclerosis. Recent studies suggest that HHcy aggravates atherosclerosis with elevated oxidative stress and reduced S-nitrosylation level of redox-sensitive protein residues in the vasculature. However, whether and how S-nitrosylation contributes to T-cell-driven atherosclerosis remain unclear. In the present study, we report that HHcy reduced the level of protein S-nitrosylation in T cells by inducing S-nitrosoglutathione reductase (GSNOR), the key denitrosylase that catalyzes S-nitrosoglutathione (GSNO), which is the main restored form of nitric oxide in vivo. Consequently, secretion of inflammatory cytokines [interferon-γ (IFN-γ) and interleukin-2] and proliferation of T cells were increased. GSNOR knockout or GSNO stimulation rectified HHcy-induced inflammatory cytokine secretion and T-cell proliferation. Site-directed mutagenesis of Akt at Cys224 revealed that S-nitrosylation at this site was pivotal for the reduced phosphorylation at Akt Ser473, which led to impaired Akt signaling. Furthermore, on HHcy challenge, as compared with GSNOR+/+ApoE-/- littermate controls, GSNOR-/-ApoE-/- double knockout mice showed reduced T-cell activation with concurrent reduction of atherosclerosis. Adoptive transfer of GSNOR-/- T cells to ApoE-/- mice fed homocysteine (Hcy) decreased atherosclerosis, with fewer infiltrated T cells and macrophages in plaques. In patients with HHcy and coronary artery disease, the level of plasma Hcy was positively correlated with Gsnor expression in peripheral blood mononuclear cells and IFN-γ+ T cells but inversely correlated with the S-nitrosylation level in T cells. These data reveal that T cells are activated, in part via GSNOR-dependent Akt denitrosylation during HHcy-induced atherosclerosis. Thus, suppression of GSNOR in T cells may reduce the risk of atherosclerosis.
Collapse
Affiliation(s)
- Jing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing 100034, China
| | - Yuying Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Silin Lü
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Yutong Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Juan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Shenming Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Xiaolong Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Lulu Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Jiacheng Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Fangfang Fan
- Department of Cardiology, Peking University First Hospital, Beijing 100034, China
| | - Bo Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing 100034, China
| | - Qingbo Xu
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, UK
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China.
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 38 Xueyuan Road, Beijing 100191, China.
| |
Collapse
|
19
|
Xiong W, Chen Y, Kang X, Chen Z, Zheng P, Hsu YH, Jang JH, Qin L, Liu H, Dotti G, Liu D. Immunological Synapse Predicts Effectiveness of Chimeric Antigen Receptor Cells. Mol Ther 2018; 26:963-975. [PMID: 29503199 PMCID: PMC6080133 DOI: 10.1016/j.ymthe.2018.01.020] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 01/19/2018] [Accepted: 01/25/2018] [Indexed: 12/20/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T cell therapy has the potential to improve the overall survival of patients with malignancies by enhancing the effectiveness of CAR T cells. Precisely predicting the effectiveness of various CAR T cells represents one of today’s key unsolved problems in immunotherapy. Here, we predict the effectiveness of CAR-modified cells by evaluating the quality of the CAR-mediated immunological synapse (IS) by quantitation of F-actin, clustering of tumor antigen, polarization of lytic granules (LGs), and distribution of key signaling molecules within the IS. Long-term killing capability, but not secretion of conventional cytokines or standard 4-hr cytotoxicity, correlates positively with the quality of the IS in two different CAR T cells that share identical antigen specificity. Xenograft model data confirm that the quality of the IS in vitro correlates positively with performance of CAR-modified immune cells in vivo. Therefore, we propose that the quality of the IS predicts the effectiveness of CAR-modified immune cells, which provides a novel strategy to guide CAR therapy.
Collapse
MESH Headings
- Animals
- Antigens, CD19/immunology
- Antigens, Neoplasm/immunology
- Biomarkers
- Cell Line
- Cytokines/metabolism
- Cytotoxicity, Immunologic
- Disease Models, Animal
- Gene Expression
- Gene Order
- Genes, Reporter
- Genetic Vectors/genetics
- Humans
- Immunological Synapses/immunology
- Immunological Synapses/metabolism
- Immunotherapy, Adoptive/methods
- Mice
- Microscopy, Confocal
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Retroviridae/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transduction, Genetic
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Wei Xiong
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Yuhui Chen
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Xi Kang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Zhiying Chen
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; Xiangya Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, P.R. China
| | - Peilin Zheng
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Yi-Hsin Hsu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Joon Hee Jang
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Hao Liu
- Biostatistics Core of the Dan L. Duncan Cancer Center, Houston, TX 77030, USA
| | - Gianpietro Dotti
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Dongfang Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
20
|
Elizondo DM, Andargie TE, Kubhar DS, Gugssa A, Lipscomb MW. CD40-CD40L cross-talk drives fascin expression in dendritic cells for efficient antigen presentation to CD4+ T cells. Int Immunol 2018; 29:121-131. [PMID: 28369442 DOI: 10.1093/intimm/dxx013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 03/17/2017] [Indexed: 01/10/2023] Open
Abstract
Fascin is an actin-bundling protein that, among immune cells, is restricted to expression in dendritic cells (DCs). Previous reports have suggested that fascin plays an important role in governing DC antigen presentation to CD4+ T cells. However, no report has clearly linked the receptor-ligand engagement that can direct downstream regulation of fascin expression. In this study, bone marrow-derived DCs from wild-type versus CD40-knockout C57BL/6 mice were used to elucidate the mechanisms of fascin expression and activity upon CD40-CD40 ligand (CD40L) engagement. These investigations now show that CD40 engagement governs fascin expression in DCs to promote CD4+ T-cell cytokine production. Absence of CD40 signaling resulted in diminished fascin expression in DCs and was associated with impaired CD4+ T-cell responses. Furthermore, the study found that loss of CD40-CD40L engagement resulted in reduced DC-T-cell contacts. Rescue by ectopic fascin expression in CD40-deficient DCs was able to re-establish sustained contacts with T cells and restore cytokine production. Taken together, these results show that cross-talk through CD40-CD40L signaling drives elevated fascin expression in DCs to support acquisition of full T-cell responses.
Collapse
Affiliation(s)
- Diana M Elizondo
- Biology Department, Howard University, 415 College Street NW, Washington, DC 20059, USA
| | - Temesgen E Andargie
- Biology Department, Howard University, 415 College Street NW, Washington, DC 20059, USA
| | - Dineeta S Kubhar
- Biology Department, Howard University, 415 College Street NW, Washington, DC 20059, USA
| | - Ayele Gugssa
- Biology Department, Howard University, 415 College Street NW, Washington, DC 20059, USA
| | - Michael W Lipscomb
- Biology Department, Howard University, 415 College Street NW, Washington, DC 20059, USA
| |
Collapse
|
21
|
Lee K, Yu Y. Janus Nanoparticles for T Cell Activation: Clustering Ligands to Enhance Stimulation. J Mater Chem B 2017; 5:4410-4415. [PMID: 28966791 PMCID: PMC5617359 DOI: 10.1039/c7tb00150a] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The in vitro activation of T cells by synthetic particles is a promising technique for adoptive cancer immunotherapy. While it is known that cell-surface receptors form clusters during T cell activation, the use of clustered ligands on synthetic particles to modulate T cell response is a largely unexplored concept. Building upon our previous finding that T cells respond differently to various micro-sized patterns of ligands, we here investigate the effect of nano-sized ligand clusters on T cell activation. Two-faced Janus nanoparticles were fabricated to display ligands of different functions in spatially segregated clusters on single nanoparticles. Going beyond our earlier qualitative study, here we precisely quantified and controlled the surface density and the total amount of ligands on single nanoparticles. We show that nanoparticles with clustered ligands activate T cells to a greater level than ones uniformly coated with the same number of ligands. The enhanced effect is due to increased local surface density of ligands. The results demonstrate that the spatial arrangement of ligands on particles influences activation response of T cells and may be used as a new strategy to increase T cell stimulation in the presence of insufficient amount of stimuli. This fundamental study also represents an initial step in using nanoscale Janus particles for manipulating immune cell responses.
Collapse
Affiliation(s)
- Kwahun Lee
- Department of Chemistry, Indiana University, Bloomington, IN
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN
| |
Collapse
|
22
|
Maru S, Jin G, Schell TD, Lukacher AE. TCR stimulation strength is inversely associated with establishment of functional brain-resident memory CD8 T cells during persistent viral infection. PLoS Pathog 2017; 13:e1006318. [PMID: 28410427 PMCID: PMC5406018 DOI: 10.1371/journal.ppat.1006318] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/26/2017] [Accepted: 03/27/2017] [Indexed: 11/22/2022] Open
Abstract
Establishing functional tissue-resident memory (TRM) cells at sites of infection is a newfound objective of T cell vaccine design. To directly assess the impact of antigen stimulation strength on memory CD8 T cell formation and function during a persistent viral infection, we created a library of mouse polyomavirus (MuPyV) variants with substitutions in a subdominant CD8 T cell epitope that exhibit a broad range of efficiency in stimulating TCR transgenic CD8 T cells. By altering a subdominant epitope in a nonstructural viral protein and monitoring memory differentiation of donor monoclonal CD8 T cells in immunocompetent mice, we circumvented potentially confounding changes in viral infection levels, virus-associated inflammation, size of the immunodominant virus-specific CD8 T cell response, and shifts in TCR affinity that may accompany temporal recruitment of endogenous polyclonal cells. Using this strategy, we found that antigen stimulation strength was inversely associated with the function of memory CD8 T cells during a persistent viral infection. We further show that CD8 TRM cells recruited to the brain following systemic infection with viruses expressing epitopes with suboptimal stimulation strength respond more efficiently to challenge CNS infection with virus expressing cognate antigen. These data demonstrate that the strength of antigenic stimulation during recruitment of CD8 T cells influences the functional integrity of TRM cells in a persistent viral infection. Tissue-resident memory (TRM) cells are a subset of memory T cells that primarily reside in non-lymphoid tissues and serve as sentinels and effectors against secondary infections. TRM cells have been extensively characterized in mucosal barriers, but much less is known about this population in non-barrier sites such as the brain. In this study, we designed a novel strategy to evaluate the impact of T cell stimulation strength on the generation and functionality of memory CD8 T cells in both lymphoid and nonlymphoid tissues. Using a mouse polyomavirus (MuPyV) library expressing variants of a subdominant epitope recognized by TCR transgenic CD8 T cells, we found that systemic infection producing weaker responses during T cell priming was sufficient for recruitment of effector cells to the brain. Furthermore, lower stimulation conferred greater functionality to memory T cells in the spleen and to brain TRM cells. Our findings demonstrate that the strength of antigenic stimulation experienced by a naïve T cell early in infection is a determinant of memory functional integrity during viral persistence in a non-barrier organ.
Collapse
Affiliation(s)
- Saumya Maru
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Ge Jin
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Todd D. Schell
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
23
|
Hammink R, Mandal S, Eggermont LJ, Nooteboom M, Willems PHGM, Tel J, Rowan AE, Figdor CG, Blank KG. Controlling T-Cell Activation with Synthetic Dendritic Cells Using the Multivalency Effect. ACS OMEGA 2017; 2:937-945. [PMID: 28393131 PMCID: PMC5377267 DOI: 10.1021/acsomega.6b00436] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/09/2017] [Indexed: 05/19/2023]
Abstract
Artificial antigen-presenting cells (aAPCs) have recently gained a lot of attention. They efficiently activate T cells and serve as powerful replacements for dendritic cells in cancer immunotherapy. Focusing on a specific class of polymer-based aAPCs, so-called synthetic dendritic cells (sDCs), we have investigated the importance of multivalent binding on T-cell activation. Using antibody-functionalized sDCs, we have tested the influence of polymer length and antibody density. Increasing the multivalent character of the antibody-functionalized polymer lowered the effective concentration required for T-cell activation. This was evidenced for both early and late stages of activation. The most important effect observed was the significantly prolonged activation of the stimulated T cells, indicating that multivalent sDCs sustain T-cell signaling. Our results highlight the importance of multivalency for the design of aAPCs and will ultimately allow for better mimics of natural dendritic cells that can be used as vaccines in cancer treatment.
Collapse
Affiliation(s)
- Roel Hammink
- Department
of Molecular Materials, Institute for Molecules
and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Subhra Mandal
- Department of Tumor Immunology and Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Loek J. Eggermont
- Department of Tumor Immunology and Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Marco Nooteboom
- Department of Tumor Immunology and Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Peter H. G. M. Willems
- Department of Tumor Immunology and Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Jurjen Tel
- Department of Tumor Immunology and Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Alan E. Rowan
- Department
of Molecular Materials, Institute for Molecules
and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- E-mail: (A.E.R.)
| | - Carl G. Figdor
- Department of Tumor Immunology and Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
- E-mail: (C.G.F.)
| | - Kerstin G. Blank
- Department
of Molecular Materials, Institute for Molecules
and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- E-mail: (K.G.B.)
| |
Collapse
|
24
|
Wagner DH. Overlooked Mechanisms in Type 1 Diabetes Etiology: How Unique Costimulatory Molecules Contribute to Diabetogenesis. Front Endocrinol (Lausanne) 2017; 8:208. [PMID: 28878738 PMCID: PMC5572340 DOI: 10.3389/fendo.2017.00208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/08/2017] [Indexed: 01/16/2023] Open
Abstract
Type 1 Diabetes (T1D) develops when immune cells invade the pancreatic islets resulting in loss of insulin production in beta cells. T cells have been proven to be central players in that process. What is surprising, however, is that classic mechanisms of tolerance cannot explain diabetogenesis; alternate mechanisms must now be considered. T cell receptor (TCR) revision is the process whereby T cells in the periphery alter TCR expression, outside the safety-net of thymic selection pressures. This process results in an expanded T cell repertoire, capable of responding to a universe of pathogens, but limitations are that increased risk for autoimmune disease development occurs. Classic T cell costimulators including the CD28 family have long been thought to be the major drivers for full T cell activation. In actuality, CD28 and its family member counterparts, ICOS and CTLA-4, all drive regulatory responses. Inflammation is driven by CD40, not CD28. CD40 as a costimulus has been largely overlooked. When naïve T cells interact with antigen presenting cell CD154, the major ligand for CD40, is induced. This creates a milieu for T cell (CD40)-T cell (CD154) interaction, leading to inflammation. Finally, defined pathogenic effector cells including TH40 (CD4+CD40+) cells can express FOXP3 but are not Tregs. The cells loose FOXP3 to become pathogenic effector cells. Each of these mechanisms creates novel options to better understand diabetogenesis and create new therapeutic targets for T1D.
Collapse
Affiliation(s)
- David H. Wagner
- The Program in Integrated Immunology, Department of Medicine, Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: David H. Wagner Jr.,
| |
Collapse
|
25
|
Ohtsuka S, Ogawa S, Wakamatsu E, Abe R. Cell cycle arrest caused by MEK/ERK signaling is a mechanism for suppressing growth of antigen-hyperstimulated effector T cells. Int Immunol 2016; 28:547-557. [PMID: 27543653 DOI: 10.1093/intimm/dxw037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/17/2016] [Indexed: 12/17/2022] Open
Abstract
Suppression of T-cell growth is an important mechanism for establishment of self-tolerance and prevention of unwanted prolonged immune responses that may cause tissue damage. Although negative selection of potentially self-reactive T cells in the thymus as well as in peripheral tissues has been extensively investigated and well documented, regulatory mechanisms to dampen proliferation of antigen-specific effector T cells in response to antigen stimulation remain largely unknown. Thus, in this work, we focus on the identification of growth suppression mechanisms of antigen-specific effector T cells. In order to address this issue, we investigated the cellular and molecular events in growth suppression of an ovalbumin (OVA)-specific T-cell clone after stimulation with a wide range of OVA-peptide concentrations. We observed that while an optimal dose of peptide leads to cell cycle progression and proliferation, higher doses of peptide reduced cell growth, a phenomenon that was previously termed high-dose suppression. Our analysis of this phenomenon indicated that high-dose suppression is a consequence of cell cycle arrest, but not Fas-Fas ligand-dependent apoptosis or T-cell anergy, and that this growth arrest occurs in S phase, accompanied by reduced expression of CDK2 and cyclin A. Importantly, inhibition of MEK/ERK activation eliminated this growth suppression and cell cycle arrest, while it reduced the proliferative response to optimal antigenic stimulation. These results suggest that cell cycle arrest is the major mechanism regulating antigen-specific effector T-cell expansion, and that the MEK/ERK signaling pathway has both positive and negative effects, depending on the strength of antigenic stimulation.
Collapse
Affiliation(s)
- Shizuka Ohtsuka
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba 278-0022, Japan
| | - Shuhei Ogawa
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba 278-0022, Japan
| | - Ei Wakamatsu
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba 278-0022, Japan
| | - Ryo Abe
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba 278-0022, Japan
| |
Collapse
|
26
|
Hashimoto-Tane A, Saito T. Dynamic Regulation of TCR-Microclusters and the Microsynapse for T Cell Activation. Front Immunol 2016; 7:255. [PMID: 27446085 PMCID: PMC4923147 DOI: 10.3389/fimmu.2016.00255] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 06/15/2016] [Indexed: 11/13/2022] Open
Abstract
The interaction between a T cell and an antigen-presenting cell is the initiating event in T cell-mediated adaptive immunity. The Immunological Synapse (IS) is formed at the interface between these two cell types, and is the site where antigen (Ag)-specific recognition and activation are induced through the T cell receptor (TCR). This occurs at the center of the IS, and cell adhesion is supported through integrins in the area surrounding the TCR. Recently, this model has been revised based on data indicating that the initial Ag-specific activation signal is triggered prior to IS formation at TCR-microclusters (MCs), sites where TCR, kinases and adaptors of TCR proximal downstream signaling molecules accumulate as an activation signaling cluster. TCR-MCs then move into the center of the cell-cell interface to generate the cSMAC. This translocation of TCR-MCs is mediated initially by the actin cytoskeleton and then by dynein-induced movement along microtubules. The translocation of TCR-MCs and cSMAC formation is induced upon strong TCR stimulation through the assembly of a TCR-dynein super complex with microtubules. The Ag-specific activation signal is induced at TCR-MCs, but the adhesion signal is now shown to be induced by generating a "microsynapse," which is composed of a core of TCR-MCs and the surrounding adhesion ring of integrin and focal adhesion molecules. Since the microsynapse is critical for activation, particularly under weak TCR stimulation, this structure supports a weak TCR signal through a cell-cell adhesion signal. The microsynapse has a structure similar to the IS but on a micro-scale and regulates Ag-specific activation as well as cell-cell adhesion. We describe here the dynamic regulation of TCR-MCs, responsible for inducing Ag-specific activation signals, and the microsynapse, responsible for adhesion signals critical for cell-cell interactions, and their interrelationship.
Collapse
Affiliation(s)
- Akiko Hashimoto-Tane
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences , Yokohama , Japan
| | - Takashi Saito
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences , Yokohama , Japan
| |
Collapse
|
27
|
Comrie WA, Burkhardt JK. Action and Traction: Cytoskeletal Control of Receptor Triggering at the Immunological Synapse. Front Immunol 2016; 7:68. [PMID: 27014258 PMCID: PMC4779853 DOI: 10.3389/fimmu.2016.00068] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 02/12/2016] [Indexed: 01/03/2023] Open
Abstract
It is well known that F-actin dynamics drive the micron-scale cell shape changes required for migration and immunological synapse (IS) formation. In addition, recent evidence points to a more intimate role for the actin cytoskeleton in promoting T cell activation. Mechanotransduction, the conversion of mechanical input into intracellular biochemical changes, is thought to play a critical role in several aspects of immunoreceptor triggering and downstream signal transduction. Multiple molecules associated with signaling events at the IS have been shown to respond to physical force, including the TCR, costimulatory molecules, adhesion molecules, and several downstream adapters. In at least some cases, it is clear that the relevant forces are exerted by dynamics of the T cell actomyosin cytoskeleton. Interestingly, there is evidence that the cytoskeleton of the antigen-presenting cell also plays an active role in T cell activation, by countering the molecular forces exerted by the T cell at the IS. Since actin polymerization is itself driven by TCR and costimulatory signaling pathways, a complex relationship exists between actin dynamics and receptor activation. This review will focus on recent advances in our understanding of the mechanosensitive aspects of T cell activation, paying specific attention to how F-actin-directed forces applied from both sides of the IS fit into current models of receptor triggering and activation.
Collapse
Affiliation(s)
- William A Comrie
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| |
Collapse
|
28
|
Brzostek J, Gascoigne NRJ, Rybakin V. Cell Type-Specific Regulation of Immunological Synapse Dynamics by B7 Ligand Recognition. Front Immunol 2016; 7:24. [PMID: 26870040 PMCID: PMC4740375 DOI: 10.3389/fimmu.2016.00024] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/18/2016] [Indexed: 01/07/2023] Open
Abstract
B7 proteins CD80 (B7-1) and CD86 (B7-2) are expressed on most antigen-presenting cells and provide critical co-stimulatory or inhibitory input to T cells via their T-cell-expressed receptors: CD28 and CTLA-4. CD28 is expressed on effector T cells and regulatory T cells (Tregs), and CD28-dependent signals are required for optimum activation of effector T cell functions. CD28 ligation on effector T cells leads to formation of distinct molecular patterns and induction of cytoskeletal rearrangements at the immunological synapse (IS). CD28 plays a critical role in recruitment of protein kinase C (PKC)-θ to the effector T cell IS. CTLA-4 is constitutively expressed on the surface of Tregs, but it is expressed on effector T cells only after activation. As CTLA-4 binds to B7 proteins with significantly higher affinity than CD28, B7 ligand recognition by cells expressing both receptors leads to displacement of CD28 and PKC-θ from the IS. In Tregs, B7 ligand recognition leads to recruitment of CTLA-4 and PKC-η to the IS. CTLA-4 plays a role in regulation of T effector and Treg IS stability and cell motility. Due to their important roles in regulating T-cell-mediated responses, B7 receptors are emerging as important drug targets in oncology. In this review, we present an integrated summary of current knowledge about the role of B7 family receptor–ligand interactions in the regulation of spatial and temporal IS dynamics in effector and Tregs.
Collapse
Affiliation(s)
- Joanna Brzostek
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine and Immunology Programme, National University of Singapore , Singapore , Singapore
| | - Nicholas R J Gascoigne
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine and Immunology Programme, National University of Singapore , Singapore , Singapore
| | - Vasily Rybakin
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine and Immunology Programme, National University of Singapore, Singapore, Singapore; Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Lee JH, Dustin ML, Kam LC. A microfluidic platform reveals differential response of regulatory T cells to micropatterned costimulation arrays. Integr Biol (Camb) 2015; 7:1442-53. [PMID: 26400012 PMCID: PMC4630128 DOI: 10.1039/c5ib00215j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 09/10/2015] [Indexed: 12/28/2022]
Abstract
T cells are key mediators of adaptive immunity. However, the overall immune response is often directed by minor subpopulations of this heterogeneous family of cells, owing to specificity of activation and amplification of functional response. Knowledge of differences in signaling and function between T cell subtypes is far from complete, but is clearly needed for understanding and ultimately leveraging this branch of the adaptive immune response. This report investigates differences in cell response to micropatterned surfaces by conventional and regulatory T cells. Specifically, the ability of cells to respond to the microscale geometry of TCR/CD3 and CD28 engagement is made possible using a magnetic-microfluidic device that overcomes limitations in imaging efficiency associated with conventional microscopy equipment. This device can be readily assembled onto micropatterned surfaces while maintaining the activity of proteins and other biomolecules necessary for such studies. In operation, a target population of cells is tagged using paramagnetic beads, and then trapped in a divergent magnetic field within the chamber. Following washing, the target cells are released to interact with a designated surface. Characterization of this system with mouse CD4(+) T cells demonstrated a 50-fold increase in target-to-background cell purity, with an 80% collection efficiency. Applying this approach to CD4(+)CD25(+) regulatory T cells, it is then demonstrated that these rare cells respond less selectively to micro-scale features of anti-CD3 antibodies than CD4(+)CD25(-) conventional T cells, revealing a difference in balance between TCR/CD3 and LFA-1-based adhesion. PKC-θ localized to the distal pole of regulatory T cells, away from the cell-substrate interface, suggests a mechanism for differential regulation of TCR/LFA-1-based adhesion. Moreover, specificity of cell adhesion to anti-CD3 features was dependent on the relative position of anti-CD28 signaling within the cell-substrate interface, revealing an important role for coincidence of TCR and costimulatory pathway in triggering regulatory T cell function.
Collapse
Affiliation(s)
- Joung-Hyun Lee
- Department of Biomedical Engineering, Columbia University in the City of New York, New York, USA.
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University in the City of New York, New York, USA.
| |
Collapse
|
30
|
Bandyopadhyay S, Chandel HS, Singh S, Roy S, Krishnasastry MV, Saha B. Counteractive functions are encrypted in the residues of CD154. Hum Immunol 2015; 76:673-80. [PMID: 26429321 DOI: 10.1016/j.humimm.2015.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/21/2015] [Accepted: 09/27/2015] [Indexed: 10/23/2022]
Abstract
CD40, as a single receptor that binds CD154 (CD40-ligand or CD40L), regulates counteractive effector functions such as production of pro- and anti-inflammatory cytokines. Therefore, we examined whether such dual messages are encrypted in CD40L. As such message encryption was never investigated, we hypothesized that mutation of certain amino acid residues should in principle enhance pro-inflammatory cytokine production whereas mutation of some others would enhance anti-inflammatory cytokine secretion. We mutated six such residues, which were previously showed to participate in CD40L function. Here, we report that the mutant CD154 129E→V was superior to the wild-type CD154 in killing of Leishmania donovani, induction of inducible nitric oxide synthase (iNOS) and production of IL-12 and relative phosphorylation of p38MAPK and ERK-1/2 in PBMC-derived macrophages. By contrast, 128S→V promoted L. donovani survival, reducing iNOS, but increasing IL-10 expression and predominant ERK-1/2 phosphorylation. The mutant 144G→V did not have significant effects. Other mutants (142E→V, 143K→A, 145Y→F) mimicked the wild-type CD154. Molecular dynamics simulation suggested that these mutations induced differential conformational changes in the CD40-CD154 complex. Therefore, assortment of the contrasting messages encrypted in a given ligand performing counteractive functions presents a novel fundamental biological principle that can be used for devising various therapies.
Collapse
Affiliation(s)
| | | | - Shailza Singh
- National Centre for Cell Science, Ganeshkhind, Pune 411 007, India
| | - Somenath Roy
- Department of Human Physiology with Community Health, Vidyasagar University, Midnapore 721102, India
| | | | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411 007, India.
| |
Collapse
|
31
|
So T, Nagashima H, Ishii N. TNF Receptor-Associated Factor (TRAF) Signaling Network in CD4 + T-Lymphocytes. TOHOKU J EXP MED 2015; 236:139-54. [DOI: 10.1620/tjem.236.139] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Takanori So
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine
| | - Hiroyuki Nagashima
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine
| |
Collapse
|
32
|
Baek J, Lee H, Hwang KW, Kim E, Min H. The association of CD28 polymorphism, rs3116496, with Cancer: A meta-analysis. Comput Biol Med 2014; 61:172-7. [PMID: 25534869 DOI: 10.1016/j.compbiomed.2014.11.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 11/25/2014] [Accepted: 11/29/2014] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To determine the relationship between CD28 polymorphisms, rs3116496, and cancer. DESIGN Meta-analysis. METHODS PubMed, EMBASE, Web of Science, and Cochrane library databases were searched to identify studies reporting the association between CD28 polymorphism and cancer. Two authors selected identified studies, extracted, and analyzed the data independently. RESULTS Individuals carrying a T allele (TT homozygotes and TT+TC heterozygotes) at rs3116496 had a lower incidence of cancer than carriers of a C allele. Subgroup analysis showed that this association held true for Asians, but not Europeans. CONCLUSION CD28 polymorphism, rs3116496, contributes to cancer susceptibility in the case of multiple cancers.
Collapse
Affiliation(s)
- Jihae Baek
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Heeyoung Lee
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Kwang Woo Hwang
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Eunyoung Kim
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Hyeyoung Min
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea.
| |
Collapse
|
33
|
Kwiatek JM, Hinde E, Gaus K. Microscopy approaches to investigate protein dynamics and lipid organization. Mol Membr Biol 2014; 31:141-51. [DOI: 10.3109/09687688.2014.937469] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
34
|
Abstract
T cell activation is a key event in the adaptive immune response and vital to the generation of both cellular and humoral immunity. Activation is required not only for effective CD4 T cell responses but also to provide help for B cells and the generation of cytotoxic T cell responses. Unsurprisingly, impaired T cell activation results in infectious pathology, whereas dysregulated activation can result in autoimmunity. The decision to activate is therefore tightly regulated and the CD28/CTLA-4 pathway represents this apical decision point at the molecular level. In particular, CTLA-4 (CD152) is an essential checkpoint control for autoimmunity; however, the molecular mechanism(s) by which CTLA-4 achieves its regulatory function are not well understood, especially how it functionally intersects with the CD28 pathway. In this chapter, we review the established molecular and cellular concepts relating to CD28 and CTLA-4 biology, and attempt to integrate these by discussing the transendocytosis of ligands as a new model of CTLA-4 function.
Collapse
Affiliation(s)
- Blagoje Soskic
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom
| | | | - Tiezheng Hou
- UCL Institute of Immunity and Transplantation, Royal Free Campus, London, United Kingdom
| | - David M Sansom
- UCL Institute of Immunity and Transplantation, Royal Free Campus, London, United Kingdom.
| |
Collapse
|
35
|
Deeg J, Axmann M, Matic J, Liapis A, Depoil D, Afrose J, Curado S, Dustin M, Spatz JP. T cell activation is determined by the number of presented antigens. NANO LETTERS 2013; 13:5619-26. [PMID: 24117051 PMCID: PMC3828117 DOI: 10.1021/nl403266t] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 10/05/2013] [Indexed: 05/18/2023]
Abstract
Antigen recognition is a key event during T cell activation. Here, we introduce nanopatterned antigen arrays that mimic the antigen presenting cell surface during T cell activation. The assessment of activation related events revealed the requirement of a minimal density of 90-140 stimulating major histocompatibility complex class II proteins (pMHC) molecules per μm(2). We demonstrate that these substrates induce T cell responses in a pMHC dose-dependent manner and that the number of presented pMHCs dominates over local pMHC density.
Collapse
Affiliation(s)
- Janosch Deeg
- Department
of New Materials and Biosystems, Max Planck
Institute for Intelligent Systems, Heisenbergstraße 3, D-70569 Stuttgart, Germany
- Department
of Biophysical Chemistry, University of
Heidelberg, INF 253, D-69120 Heidelberg, Germany
| | - Markus Axmann
- Department
of New Materials and Biosystems, Max Planck
Institute for Intelligent Systems, Heisenbergstraße 3, D-70569 Stuttgart, Germany
- Department
of Biophysical Chemistry, University of
Heidelberg, INF 253, D-69120 Heidelberg, Germany
| | - Jovana Matic
- Department
of New Materials and Biosystems, Max Planck
Institute for Intelligent Systems, Heisenbergstraße 3, D-70569 Stuttgart, Germany
- Department
of Biophysical Chemistry, University of
Heidelberg, INF 253, D-69120 Heidelberg, Germany
| | - Anastasia Liapis
- Skirball
Institute of Biomolecular Medicine and Department of Pathology, New York University School of Medicine, New York, New York 10016, United States
| | - David Depoil
- Skirball
Institute of Biomolecular Medicine and Department of Pathology, New York University School of Medicine, New York, New York 10016, United States
| | - Jehan Afrose
- Skirball
Institute of Biomolecular Medicine and Department of Pathology, New York University School of Medicine, New York, New York 10016, United States
| | - Silvia Curado
- Skirball
Institute of Biomolecular Medicine and Department of Pathology, New York University School of Medicine, New York, New York 10016, United States
| | - Michael
L. Dustin
- Skirball
Institute of Biomolecular Medicine and Department of Pathology, New York University School of Medicine, New York, New York 10016, United States
- Kennedy
Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology
and Musculoskeletal Sciences, University
of Oxford, Oxford, OX37FY, United Kingdom
| | - Joachim P. Spatz
- Department
of New Materials and Biosystems, Max Planck
Institute for Intelligent Systems, Heisenbergstraße 3, D-70569 Stuttgart, Germany
- Department
of Biophysical Chemistry, University of
Heidelberg, INF 253, D-69120 Heidelberg, Germany
| |
Collapse
|
36
|
Viganò S, Bellutti Enders F, Miconnet I, Cellerai C, Savoye AL, Rozot V, Perreau M, Faouzi M, Ohmiti K, Cavassini M, Bart PA, Pantaleo G, Harari A. Rapid perturbation in viremia levels drives increases in functional avidity of HIV-specific CD8 T cells. PLoS Pathog 2013; 9:e1003423. [PMID: 23853580 PMCID: PMC3701695 DOI: 10.1371/journal.ppat.1003423] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/30/2013] [Indexed: 01/16/2023] Open
Abstract
The factors determining the functional avidity and its relationship with the broad heterogeneity of antiviral T cell responses remain partially understood. We investigated HIV-specific CD8 T cell responses in 85 patients with primary HIV infection (PHI) or chronic (progressive and non-progressive) infection. The functional avidity of HIV-specific CD8 T cells was not different between patients with progressive and non-progressive chronic infection. However, it was significantly lower in PHI patients at the time of diagnosis of acute infection and after control of virus replication following one year of successful antiretroviral therapy. High-avidity HIV-specific CD8 T cells expressed lower levels of CD27 and CD28 and were enriched in cells with an exhausted phenotype, i.e. co-expressing PD-1/2B4/CD160. Of note, a significant increase in the functional avidity of HIV-specific CD8 T cells occurred in early-treated PHI patients experiencing a virus rebound after spontaneous treatment interruption. This increase in functional avidity was associated with the accumulation of PD-1/2B4/CD160 positive cells, loss of polyfunctionality and increased TCR renewal. The increased TCR renewal may provide the mechanistic basis for the generation of high-avidity HIV-specific CD8 T cells. These results provide insights on the relationships between functional avidity, viremia, T-cell exhaustion and TCR renewal of antiviral CD8 T cell responses. CD8 T cells directed against virus are complex and functionally heterogeneous. One relevant component of CD8 T cells is their functional avidity which reflects their sensitivity to cognate antigens, i.e. how prone T cells are to respond when they encounter low doses of antigens. In patients with chronic and established HIV infection, we observed that the sensitivity of HIV-specific CD8 T cells was not different between patients with progressive or non-progressive disease. In contrast, the sensitivity of HIV-specific CD8 T cells was significantly lower in patients with early and recent HIV infection. Furthermore, CD8 T cells of high avidity were preferentially associated with a state of functional impairment known as exhaustion. Of interest, some patients treated with antiretroviral therapy during acute infection spontaneously interrupted their treatment and experienced a rebound of virus. In these patients, the avidity of HIV-specific CD8 T cells increased and this increase was associated to stronger cell exhaustion and greater renewal of the population of antiviral CD8 T cells, thus potentially providing the mechanistic basis for the generation of high-avidity CD8 T cells. Overall, our data suggest that rapid perturbation in viremia levels drove increases in the functional avidity of HIV-specific CD8 T cells.
Collapse
Affiliation(s)
- Selena Viganò
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Felicitas Bellutti Enders
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Isabelle Miconnet
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Cristina Cellerai
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Anne-Laure Savoye
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Virginie Rozot
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Mohamed Faouzi
- The Center of Clinical Epidemiology, Institut de Médecine Sociale et Préventive, Lausanne University Hospital, Lausanne, Switzerland
| | - Khalid Ohmiti
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
- The Center of Clinical Epidemiology, Institut de Médecine Sociale et Préventive, Lausanne University Hospital, Lausanne, Switzerland
| | - Matthias Cavassini
- Service of Infectious Diseases, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Pierre-Alexandre Bart
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne, Switzerland
| | - Alexandre Harari
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
37
|
Kong KF, Altman A. In and out of the bull's eye: protein kinase Cs in the immunological synapse. Trends Immunol 2013; 34:234-42. [PMID: 23428395 DOI: 10.1016/j.it.2013.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 12/29/2012] [Accepted: 01/02/2013] [Indexed: 01/24/2023]
Abstract
The immunological synapse (IS) formed between immune cells and antigen-presenting cells (APCs) provides a platform for signaling. Protein kinase C (PKC)θ localizes in the T cell IS within the central supramolecular activation cluster (cSMAC), where it associates with CD28 and mediates T cell receptor (TCR)/CD28 signals leading to effector T (Teff) cell activation. In regulatory T (Treg) cells, PKCθ is sequestered away from the IS, and inhibits suppressive function. Other PKCs localizing in the IS mediate additional functions in various immune cells. Further work is needed to identify mechanisms underlying PKC recruitment or exclusion at the IS, potential redundancy among IS-localized PKCs, and the relevance of PKC localization for IS dynamics and lymphocyte activation.
Collapse
Affiliation(s)
- Kok-Fai Kong
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | |
Collapse
|
38
|
Peterson EJ, Maltzman JS, Koretzky GA. T-cell activation and tolerance. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00036-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
39
|
Abraham L, Fackler OT. HIV-1 Nef: a multifaceted modulator of T cell receptor signaling. Cell Commun Signal 2012; 10:39. [PMID: 23227982 PMCID: PMC3534016 DOI: 10.1186/1478-811x-10-39] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 11/28/2012] [Indexed: 12/26/2022] Open
Abstract
Nef, an accessory protein of the Human Immunodeficiency Virus type 1 (HIV-1), is dispensable for viral replication in cell culture, but promotes virus replication and pathogenesis in the infected host. Acting as protein-interaction adaptor, HIV-1 Nef modulates numerous target cell activities including cell surface receptor expression, cytoskeletal remodeling, vesicular transport, and signal transduction. In infected T-lymphocytes, altering T-cell antigen receptor (TCR) signaling has long been recognized as one key function of the viral protein. However, reported effects of Nef range from inhibition to activation of this cascade. Recent advances in the field begin to explain these seemingly contradictory observations and suggest that Nef alters intracellular trafficking of TCR proximal machinery to disrupt plasma membrane bound TCR signaling while at the same time, the viral protein induces localized signal transduction at the trans-Golgi network. This review summarizes these new findings on how HIV-1 Nef reprograms TCR signalling output from a broad response to selective activation of the RAS-Erk pathway. We also discuss the implications of these alterations in the context of HIV-1 infection and in light of current concepts of TCR signal transduction.
Collapse
Affiliation(s)
- Libin Abraham
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, INF 324, Heidelberg, 69120, Germany.
| | | |
Collapse
|
40
|
Functional avidity: a measure to predict the efficacy of effector T cells? Clin Dev Immunol 2012; 2012:153863. [PMID: 23227083 PMCID: PMC3511839 DOI: 10.1155/2012/153863] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 10/22/2012] [Indexed: 01/30/2023]
Abstract
The functional avidity is determined by exposing T-cell populations in vitro to different amounts of cognate antigen. T-cells with high functional avidity respond to low antigen doses. This in vitro measure is thought to correlate well with the in vivo effector capacity of T-cells. We here present the multifaceted factors determining and influencing the functional avidity of T-cells. We outline how changes in the functional avidity can occur over the course of an infection. This process, known as avidity maturation, can occur despite the fact that T-cells express a fixed TCR. Furthermore, examples are provided illustrating the importance of generating T-cell populations that exhibit a high functional avidity when responding to an infection or tumors. Furthermore, we discuss whether criteria based on which we evaluate an effective T-cell response to acute infections can also be applied to chronic infections such as HIV. Finally, we also focus on observations that high-avidity T-cells show higher signs of exhaustion and facilitate the emergence of virus escape variants. The review summarizes our current understanding of how this may occur as well as how T-cells of different functional avidity contribute to antiviral and anti-tumor immunity. Enhancing our knowledge in this field is relevant for tumor immunotherapy and vaccines design.
Collapse
|
41
|
Perkey E, Miller RA, Garcia GG. Ex vivo enzymatic treatment of aged CD4 T cells restores cognate T cell helper function and enhances antibody production in mice. THE JOURNAL OF IMMUNOLOGY 2012; 189:5582-9. [PMID: 23136198 DOI: 10.4049/jimmunol.1200487] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Previous in vitro studies showed that CD4 T cells from old mice have defects in TCR signaling, immune synapse formation, activation, and proliferation. We reported that removing a specific set of surface glycoproteins by ex vivo treatment with O-sialoglycoprotein endopeptidase (OSGE) can reverse many aspects of the age-related decline in CD4 T cell function. However, the specific mechanism by which this process occurs remains unclear, and it is unknown whether this enzymatic treatment can also restore important aspects of adaptive immunity in vivo. By using an in vivo model of the immune response based on adoptive transfer of CD4 T cells from pigeon cytochrome C-specific transgenic H-2(k/k) TCR-Vα(11)Vβ(3) CD4(+) mice to syngeneic hosts, we demonstrate that aging diminishes CD28 costimulatory signals in CD4 T cells. These age-associated defects include changes in phosphorylation of AKT and expression of glucose transporter type I, inducible T cell costimulatory molecule, and CD40L, suggesting that the lack of CD28 costimulation contributes to age-dependent loss of CD4 function. All of these deficits can be reversed by ex vivo OSGE treatment. Blocking B7-CD28 interactions on T cells prevents OSGE-mediated restoration of T cell function, suggesting that changes in surface glycosylation, including CD28, may be responsible for the age-related costimulation decline. Finally, we show that the age-related decline in CD4 cognate helper function for IgG production and long-term humoral immunity can also be restored by OSGE treatment of CD4 T cells prior to adoptive transfer.
Collapse
Affiliation(s)
- Eric Perkey
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan College of Literature, Science and the Arts, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
42
|
Lim TS, Goh JKH, Mortellaro A, Lim CT, Hämmerling GJ, Ricciardi-Castagnoli P. CD80 and CD86 differentially regulate mechanical interactions of T-cells with antigen-presenting dendritic cells and B-cells. PLoS One 2012; 7:e45185. [PMID: 23024807 PMCID: PMC3443229 DOI: 10.1371/journal.pone.0045185] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/13/2012] [Indexed: 01/17/2023] Open
Abstract
Functional T-cell responses are initiated by physical interactions between T-cells and antigen-presenting cells (APCs), including dendritic cells (DCs) and B-cells. T-cells are activated more effectively by DCs than by B-cells, but little is known about the key molecular mechanisms that underpin the particular potency of DC in triggering T-cell responses. To better understand the influence of physical intercellular interactions on APC efficacy in activating T-cells, we used single cell force spectroscopy to characterize and compare the mechanical forces of interactions between DC:T-cells and B:T-cells. Following antigen stimulation, intercellular interactions of DC:T-cell conjugates were stronger than B:T-cell interactions. DCs induced higher levels of T-cell calcium mobilization and production of IL-2 and IFNγ than were elicited by B-cells, thus suggesting that tight intercellular contacts are important in providing mechanically stable environment to initiate T-cell activation. Blocking antibodies targeting surface co-stimulatory molecules CD80 or CD86 weakened intercellular interactions and dampen T-cell activation, highlighting the amplificatory roles of CD80/86 in regulating APC:T-cell interactions and T-cell functional activation. The variable strength of mechanical forces between DC:T-cells and B:T-cell interactions were not solely dependent on differential APC expression of CD80/86, since DCs were superior to B-cells in promoting strong interactions with T-cells even when CD80 and CD86 were inhibited. These data provide mechanical insights into the effects of co-stimulatory molecules in regulating APC:T-cell interactions.
Collapse
Affiliation(s)
- Tong Seng Lim
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- * E-mail: (PR-C); (TSL)
| | - James Kang Hao Goh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Alessandra Mortellaro
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Bioengineering & Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | - Günter J. Hämmerling
- Division of Molecular Immunology, German Cancer Research Center DKFZ, Heidelberg, Germany
| | - Paola Ricciardi-Castagnoli
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- * E-mail: (PR-C); (TSL)
| |
Collapse
|
43
|
Kusumi A, Fujiwara TK, Chadda R, Xie M, Tsunoyama TA, Kalay Z, Kasai RS, Suzuki KGN. Dynamic organizing principles of the plasma membrane that regulate signal transduction: commemorating the fortieth anniversary of Singer and Nicolson's fluid-mosaic model. Annu Rev Cell Dev Biol 2012; 28:215-50. [PMID: 22905956 DOI: 10.1146/annurev-cellbio-100809-151736] [Citation(s) in RCA: 284] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The recent rapid accumulation of knowledge on the dynamics and structure of the plasma membrane has prompted major modifications of the textbook fluid-mosaic model. However, because the new data have been obtained in a variety of research contexts using various biological paradigms, the impact of the critical conceptual modifications on biomedical research and development has been limited. In this review, we try to synthesize our current biological, chemical, and physical knowledge about the plasma membrane to provide new fundamental organizing principles of this structure that underlie every molecular mechanism that realizes its functions. Special attention is paid to signal transduction function and the dynamic aspect of the organizing principles. We propose that the cooperative action of the hierarchical three-tiered mesoscale (2-300 nm) domains--actin-membrane-skeleton induced compartments (40-300 nm), raft domains (2-20 nm), and dynamic protein complex domains (3-10 nm)--is critical for membrane function and distinguishes the plasma membrane from a classical Singer-Nicolson-type model.
Collapse
Affiliation(s)
- Akihiro Kusumi
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Suzuki KGN, Kasai RS, Hirosawa KM, Nemoto YL, Ishibashi M, Miwa Y, Fujiwara TK, Kusumi A. Transient GPI-anchored protein homodimers are units for raft organization and function. Nat Chem Biol 2012; 8:774-83. [DOI: 10.1038/nchembio.1028] [Citation(s) in RCA: 200] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 06/19/2012] [Indexed: 01/08/2023]
|
45
|
Abraham L, Bankhead P, Pan X, Engel U, Fackler OT. HIV-1 Nef limits communication between linker of activated T cells and SLP-76 to reduce formation of SLP-76-signaling microclusters following TCR stimulation. THE JOURNAL OF IMMUNOLOGY 2012; 189:1898-910. [PMID: 22802418 DOI: 10.4049/jimmunol.1200652] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Signal initiation by engagement of the TCR triggers actin rearrangements, receptor clustering, and dynamic organization of signaling complexes to elicit and sustain downstream signaling. Nef, a pathogenicity factor of HIV, disrupts early TCR signaling in target T cells. To define the mechanism underlying this Nef-mediated signal disruption, we employed quantitative single-cell microscopy following surface-mediated TCR stimulation that allows for dynamic visualization of distinct signaling complexes as microclusters (MCs). Despite marked inhibition of actin remodeling and cell spreading, the induction of MCs containing TCR-CD3 or ZAP70 was not affected significantly by Nef. However, Nef potently inhibited the subsequent formation of MCs positive for the signaling adaptor Src homology-2 domain-containing leukocyte protein of 76 kDa (SLP-76) to reduce MC density in Nef-expressing and HIV-1-infected T cells. Further analyses suggested that Nef prevents formation of SLP-76 MCs at the level of the upstream adaptor protein, linker of activated T cells (LAT), that couples ZAP70 to SLP-76. Nef did not disrupt pre-existing MCs positive for LAT. However, the presence of the viral protein prevented de novo recruitment of active LAT into MCs due to retargeting of LAT to an intracellular compartment. These modulations in MC formation and composition depended on Nef's ability to simultaneously disrupt both actin remodeling and subcellular localization of TCR-proximal machinery. Nef thus employs a dual mechanism to disturb early TCR signaling by limiting the communication between LAT and SLP-76 and preventing the dynamic formation of SLP-76-signaling MCs.
Collapse
Affiliation(s)
- Libin Abraham
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
46
|
Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. ACTA ACUST UNITED AC 2012; 209:1201-17. [PMID: 22641383 PMCID: PMC3371732 DOI: 10.1084/jem.20112741] [Citation(s) in RCA: 819] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
After encounter with its ligand, PD-1 translocates into TCR microclusters, where it transiently recruits SHP2 and suppresses phosphorylation of TCR signaling components and TCR-driven stop signals. Programmed cell death 1 (PD-1) is a negative costimulatory receptor critical for the suppression of T cell activation in vitro and in vivo. Single cell imaging elucidated a molecular mechanism of PD-1–mediated suppression. PD-1 becomes clustered with T cell receptors (TCRs) upon binding to its ligand PD-L1 and is transiently associated with the phosphatase SHP2 (Src homology 2 domain–containing tyrosine phosphatase 2). These negative costimulatory microclusters induce the dephosphorylation of the proximal TCR signaling molecules. This results in the suppression of T cell activation and blockade of the TCR-induced stop signal. In addition to PD-1 clustering, PD-1–TCR colocalization within microclusters is required for efficient PD-1–mediated suppression. This inhibitory mechanism also functions in PD-1hi T cells generated in vivo and can be overridden by a neutralizing anti–PD-L1 antibody. Therefore, PD-1 microcluster formation is important for regulation of T cell activation.
Collapse
Affiliation(s)
- Tadashi Yokosuka
- Laboratory for Cell Signaling, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa 230-0045, Japan.
| | | | | | | | | | | |
Collapse
|
47
|
Mechanisms behind functional avidity maturation in T cells. Clin Dev Immunol 2012; 2012:163453. [PMID: 22611418 PMCID: PMC3351025 DOI: 10.1155/2012/163453] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 01/26/2012] [Indexed: 12/22/2022]
Abstract
During an immune response antigen-primed B-cells increase their antigen responsiveness by affinity maturation mediated by somatic hypermutation of the genes encoding the antigen-specific B-cell receptor (BCR) and by selection of higher-affinity B cell clones. Unlike the BCR, the T-cell receptor (TCR) cannot undergo affinity maturation. Nevertheless, antigen-primed T cells significantly increase their antigen responsiveness compared to antigen-inexperienced (naïve) T cells in a process called functional avidity maturation. This paper covers studies that describe differences in T-cell antigen responsiveness during T-cell differentiation along with examples of the mechanisms behind functional avidity maturation in T cells.
Collapse
|
48
|
Lichtenfels R, Rappl G, Hombach AA, Recktenwald CV, Dressler SP, Abken H, Seliger B. A proteomic view at T cell costimulation. PLoS One 2012; 7:e32994. [PMID: 22539942 PMCID: PMC3335147 DOI: 10.1371/journal.pone.0032994] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 02/07/2012] [Indexed: 12/31/2022] Open
Abstract
The "two-signal paradigm" in T cell activation predicts that the cooperation of "signal 1," provided by the T cell receptor (TCR) through engagement of major histocompatility complex (MHC)-presented peptide, with "signal 2″ provided by costimulatory molecules, the prototype of which is CD28, is required to induce T cell effector functions. While the individual signalling pathways are well understood, little is known about global changes in the proteome pattern during TCR/CD28-mediated activation. Therefore, comparative 2-DE-based proteome analyses of CD3(+) CD69(-) resting T cells versus cells incubated with (i) the agonistic anti-CD3 antibody OKT3 mimicking signal 1 in absence or presence of IL-2 and/or with (ii) the agonistic antibody 15E8 triggering CD28-mediated signaling were performed. Differentially regulated spots were defined leading to the identification of proteins involved in the regulation of the metabolism, shaping and maintenance of the cytoskeleton and signal transduction. Representative members of the differentially expressed protein families, such as calmodulin (CALM), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), L-lactate dehydrogenase (LDH), Rho GDP-dissociation inhibitor 2 (GDIR2), and platelet basic protein (CXCL7), were independently verified by flow cytometry. Data provide a detailed map of individual protein alterations at the global proteome level in response to TCR/CD28-mediated T cell activation.
Collapse
Affiliation(s)
- Rudolf Lichtenfels
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Gunter Rappl
- Center for Molecular Medicine Cologne (CMMC) and Tumor Genetics Section, Clinic I Internal Medicine, University Cologne, Cologne, Germany
| | - Andreas A. Hombach
- Center for Molecular Medicine Cologne (CMMC) and Tumor Genetics Section, Clinic I Internal Medicine, University Cologne, Cologne, Germany
| | | | - Sven P. Dressler
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Hinrich Abken
- Center for Molecular Medicine Cologne (CMMC) and Tumor Genetics Section, Clinic I Internal Medicine, University Cologne, Cologne, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
49
|
Hsu CJ, Hsieh WT, Waldman A, Clarke F, Huseby ES, Burkhardt JK, Baumgart T. Ligand mobility modulates immunological synapse formation and T cell activation. PLoS One 2012; 7:e32398. [PMID: 22384241 PMCID: PMC3284572 DOI: 10.1371/journal.pone.0032398] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 01/30/2012] [Indexed: 12/11/2022] Open
Abstract
T cell receptor (TCR) engagement induces clustering and recruitment to the plasma membrane of many signaling molecules, including the protein tyrosine kinase zeta-chain associated protein of 70 kDa (ZAP70) and the adaptor SH2 domain-containing leukocyte protein of 76 kDa (SLP76). This molecular rearrangement results in formation of the immunological synapse (IS), a dynamic protein array that modulates T cell activation. The current study investigates the effects of apparent long-range ligand mobility on T cell signaling activity and IS formation. We formed stimulatory lipid bilayers on glass surfaces from binary lipid mixtures with varied composition, and characterized these surfaces with respect to diffusion coefficient and fluid connectivity. Stimulatory ligands coupled to these surfaces with similar density and orientation showed differences in their ability to activate T cells. On less mobile membranes, central supramolecular activation cluster (cSMAC) formation was delayed and the overall accumulation of CD3ζ at the IS was reduced. Analysis of signaling microcluster (MC) dynamics showed that ZAP70 MCs exhibited faster track velocity and longer trajectories as a function of increased ligand mobility, whereas movement of SLP76 MCs was relatively insensitive to this parameter. Actin retrograde flow was observed on all surfaces, but cell spreading and subsequent cytoskeletal contraction were more pronounced on mobile membranes. Finally, increased tyrosine phosphorylation and persistent elevation of intracellular Ca2+ were observed in cells stimulated on fluid membranes. These results point to ligand mobility as an important parameter in modulating T cell responses.
Collapse
Affiliation(s)
- Chih-Jung Hsu
- Department of Chemistry, The Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Wan-Ting Hsieh
- Department of Chemistry, The Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Abraham Waldman
- Department of Chemistry, The Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Fiona Clarke
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Eric S. Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Janis K. Burkhardt
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (TB); (JKB)
| | - Tobias Baumgart
- Department of Chemistry, The Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (TB); (JKB)
| |
Collapse
|
50
|
Kusumi A, Fujiwara TK, Morone N, Yoshida KJ, Chadda R, Xie M, Kasai RS, Suzuki KGN. Membrane mechanisms for signal transduction: the coupling of the meso-scale raft domains to membrane-skeleton-induced compartments and dynamic protein complexes. Semin Cell Dev Biol 2012; 23:126-44. [PMID: 22309841 DOI: 10.1016/j.semcdb.2012.01.018] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 01/24/2012] [Indexed: 01/09/2023]
Abstract
Virtually all biological membranes on earth share the basic structure of a two-dimensional liquid. Such universality and peculiarity are comparable to those of the double helical structure of DNA, strongly suggesting the possibility that the fundamental mechanisms for the various functions of the plasma membrane could essentially be understood by a set of simple organizing principles, developed during the course of evolution. As an initial effort toward the development of such understanding, in this review, we present the concept of the cooperative action of the hierarchical three-tiered meso-scale (2-300 nm) domains in the plasma membrane: (1) actin membrane-skeleton-induced compartments (40-300 nm), (2) raft domains (2-20 nm), and (3) dynamic protein complex domains (3-10nm). Special attention is paid to the concept of meso-scale domains, where both thermal fluctuations and weak cooperativity play critical roles, and the coupling of the raft domains to the membrane-skeleton-induced compartments as well as dynamic protein complexes. The three-tiered meso-domain architecture of the plasma membrane provides an excellent perspective for understanding the membrane mechanisms of signal transduction.
Collapse
Affiliation(s)
- Akihiro Kusumi
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8507, Japan.
| | | | | | | | | | | | | | | |
Collapse
|