1
|
Song C, Hu J, Liu Y, Tian Y, Zhu Y, Xi J, Cui M, Wang X, Zhang BZ, Fan L, Li Q. Vaccination-Route-Dependent Adjuvanticity of Antigen-Carrying Nanoparticles for Enhanced Vaccine Efficacy. Vaccines (Basel) 2024; 12:125. [PMID: 38400110 PMCID: PMC10892493 DOI: 10.3390/vaccines12020125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccination-route-dependent adjuvanticity was identified as being associated with the specific features of antigen-carrying nanoparticles (NPs) in the present work. Here, we demonstrated that the mechanical properties and the decomposability of NP adjuvants play key roles in determining the antigen accessibility and thus the overall vaccine efficacy in the immune system when different vaccination routes were employed. We showed that soft nano-vaccines were associated with more efficient antigen uptake when administering subcutaneous (S.C.) vaccination, while the slow decomposition of hard nano-vaccines promoted antigen uptake when intravenous (I.V.) vaccination was employed. In comparison to the clinically used aluminum (Alum) adjuvant, the NP adjuvants were found to stimulate both humoral and cellular immune responses efficiently, irrespective of the vaccination route. For vaccination via S.C. and I.V. alike, the NP-based vaccines show excellent protection for mice from Staphylococcus aureus (S. aureus) infection, and their survival rates are 100% after lethal challenge, being much superior to the clinically used Alum adjuvant.
Collapse
Affiliation(s)
- Chaojun Song
- School of Life Science, Northwestern Polytechnical University, 127th Youyi West Road, Xi’an 710072, China;
| | - Jinwei Hu
- Department of Pharmaceutical Chemistry and Analysis, School of Pharmacy, Airforce Medical University, 169th Changle West Road, Xi’an 710032, China; (J.H.); (Y.L.); (Y.Z.); (J.X.); (M.C.)
| | - Yutao Liu
- Department of Pharmaceutical Chemistry and Analysis, School of Pharmacy, Airforce Medical University, 169th Changle West Road, Xi’an 710032, China; (J.H.); (Y.L.); (Y.Z.); (J.X.); (M.C.)
| | - Yi Tian
- Department of Oncology, Airforce Medical Center of PLA, 30th Fu Cheng Road, Beijing 100142, China;
| | - Yupu Zhu
- Department of Pharmaceutical Chemistry and Analysis, School of Pharmacy, Airforce Medical University, 169th Changle West Road, Xi’an 710032, China; (J.H.); (Y.L.); (Y.Z.); (J.X.); (M.C.)
| | - Jiayue Xi
- Department of Pharmaceutical Chemistry and Analysis, School of Pharmacy, Airforce Medical University, 169th Changle West Road, Xi’an 710032, China; (J.H.); (Y.L.); (Y.Z.); (J.X.); (M.C.)
| | - Minxuan Cui
- Department of Pharmaceutical Chemistry and Analysis, School of Pharmacy, Airforce Medical University, 169th Changle West Road, Xi’an 710032, China; (J.H.); (Y.L.); (Y.Z.); (J.X.); (M.C.)
| | - Xiaolei Wang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China;
| | - Bao-Zhong Zhang
- Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
| | - Li Fan
- Department of Pharmaceutical Chemistry and Analysis, School of Pharmacy, Airforce Medical University, 169th Changle West Road, Xi’an 710032, China; (J.H.); (Y.L.); (Y.Z.); (J.X.); (M.C.)
| | - Quan Li
- Department of Physics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
2
|
Kelly JJ, Ankrom E, Thévenin D, Pires MM. Targeted Acidosis Mediated Delivery of Antigenic MHC-Binding Peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.562409. [PMID: 37904977 PMCID: PMC10614887 DOI: 10.1101/2023.10.18.562409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cytotoxic T lymphocytes are the primary effector immune cells responsible for protection against cancer, as they target peptide neoantigens presented through the major histocompatibility complex (MHC) on cancer cells, leading to cell death. Targeting peptide-MHC (pMHC) complexes offers a promising strategy for immunotherapy due to its specificity and effectiveness against cancer. In this work, we exploit the acidic tumor micro-environment to selectively deliver antigenic peptides to cancer cells using pH(low) insertion peptides (pHLIP). We demonstrated that the delivery of MHC binding peptides directly to the cytoplasm of melanoma cells resulted in the presentation of antigenic peptides on MHC, and subsequent activation of T cells. This work highlights the potential of pHLIP as a vehicle for targeted delivery of antigenic peptides and their presentation via MHC-bound complexes on cancer cell surfaces for activation of T cells with implications for enhancing anti-cancer immunotherapy.
Collapse
|
3
|
Malfanti A, Bausart M, Vanvarenberg K, Ucakar B, Préat V. Hyaluronic acid-antigens conjugates trigger potent immune response in both prophylactic and therapeutic immunization in a melanoma model. Drug Deliv Transl Res 2023; 13:2550-2567. [PMID: 37040031 DOI: 10.1007/s13346-023-01337-4] [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: 03/16/2023] [Indexed: 04/12/2023]
Abstract
Immunotherapy of advanced melanoma has encountered significant hurdles in terms of clinical efficacy. Here, we designed a clinically translatable hyaluronic acid (HA)-based vaccine delivering a combination of major histocompatibility complex (MHC) class I- and class II-restricted melanoma antigens (TRP2 and Gp100, respectively) conjugated to HA. HA-nanovaccine (HA-TRP2-Gp100 conjugate) exhibited tropism in the lymph nodes and promoted stimulation of the immune response (2.3-fold higher than the HA+TRP2+Gp100). HA-nanovaccine significantly delayed the growth of B16F10 melanoma and extended survival in both the prophylactic and therapeutic settings (median survival of 22 and 27, respectively, vs 17 days of the untreated group). Moreover, mice prophylactically treated with the HA-nanovaccine displayed significantly higher CD8+ and CD4+ T-cell/Treg ratios in both the spleen and tumor at day 16, suggesting that the HA-nanovaccine overcame the immunosuppressive tumor microenvironment. Superior infiltration of active CD4+ and CD8+ T cells was observed at the endpoint. This study supports the conclusion that HA potentiates the effect of a combination of MHC I and MHC II antigens via a potent immune response against melanoma.
Collapse
Affiliation(s)
- Alessio Malfanti
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium.
| | - Mathilde Bausart
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Kevin Vanvarenberg
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Bernard Ucakar
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, UCLouvain, Louvain Drug Research Institute, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium.
| |
Collapse
|
4
|
Hou Y, Chen M, Bian Y, Zheng X, Tong R, Sun X. Advanced subunit vaccine delivery technologies: From vaccine cascade obstacles to design strategies. Acta Pharm Sin B 2023; 13:3321-3338. [PMID: 37655334 PMCID: PMC10465871 DOI: 10.1016/j.apsb.2023.01.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/23/2022] [Accepted: 12/03/2022] [Indexed: 01/12/2023] Open
Abstract
Designing and manufacturing safe and effective vaccines is a crucial challenge for human health worldwide. Research on adjuvant-based subunit vaccines is increasingly being explored to meet clinical needs. Nevertheless, the adaptive immune responses of subunit vaccines are still unfavorable, which may partially be attributed to the immune cascade obstacles and unsatisfactory vaccine design. An extended understanding of the crosstalk between vaccine delivery strategies and immunological mechanisms could provide scientific insight to optimize antigen delivery and improve vaccination efficacy. In this review, we summarized the advanced subunit vaccine delivery technologies from the perspective of vaccine cascade obstacles after administration. The engineered subunit vaccines with lymph node and specific cell targeting ability, antigen cross-presentation, T cell activation properties, and tailorable antigen release patterns may achieve effective immune protection with high precision, efficiency, and stability. We hope this review can provide rational design principles and inspire the exploitation of future subunit vaccines.
Collapse
Affiliation(s)
- Yingying Hou
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Min Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xi Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Zahedipour F, Zamani P, Mashreghi M, Astaneh M, Sankian M, Amiri A, Jamialahmadi K, Jaafari MR. Nanoliposomal VEGF-R2 peptide vaccine acts as an effective therapeutic vaccine in a murine B16F10 model of melanoma. Cancer Nanotechnol 2023; 14:62. [PMID: 37333490 PMCID: PMC10264216 DOI: 10.1186/s12645-023-00213-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Abstract
Background The vascular endothelial growth factor receptor-2 (VEGFR-2) plays an important role in melanoma development and progression. Peptide vaccines have shown great potential in cancer immunotherapy by targeting VEGFR-2 as a tumor-associated antigen and boosting the immune response against both tumor cells and tumor endothelial cells. Despite this, the low efficiency of peptide vaccines has resulted in moderate therapeutic results in the majority of studies. Enhancing the delivery of peptide vaccines using nanoliposomes is an important strategy for improving the efficacy of peptide vaccines. In this regard, we designed VEGFR-2-derived peptides restricted to both mouse MHC I and human HLA-A*02:01 using immunoinformatic tools and selected three peptides representing the highest binding affinities. The peptides were encapsulated in nanoliposomal formulations using the film method plus bath sonication and characterized for their colloidal properties. Results The mean diameter of peptide-encapsulated liposomes was around 135 nm, zeta potential of - 17 mV, and encapsulation efficiency of approximately 70%. Then, vaccine formulations were injected subcutaneously in mice bearing B16F10-established melanoma tumors and their efficiency in triggering immunological, and anti-tumor responses was evaluated. Our results represented that one of our designed VEGFR-2 peptide nanoliposomal formulations (Lip-V1) substantially activated CD4+ (p < 0.0001) and CD8+ (P < 0.001) T cell responses and significantly boosted the production of IFN-γ (P < 0.0001) and IL-4 (P < 0.0001). Furthermore, this formulation led to a significant decrease in tumor volume (P < 0.0001) and enhanced survival (P < 0.05) in mice. Conclusion Our findings suggest that the nanoliposomal formulation containing VEGFR-2 peptides could be a promising therapeutic vaccination approach capable of eliciting strong antigen-specific immunologic and anti-tumor responses. Supplementary Information The online version contains supplementary material available at 10.1186/s12645-023-00213-7.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mashreghi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Astaneh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Rashid M, Omar M, Mohanta TK. FungiProteomeDB: a database for the molecular weight and isoelectric points of the fungal proteomes. Database (Oxford) 2023; 2023:7078806. [PMID: 36929177 PMCID: PMC10019025 DOI: 10.1093/database/baad004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 02/01/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
Proteins' molecular weight (MW) and isoelectric point (pI) are crucial for their subcellular localization and subsequent function. These are also useful in 2D gel electrophoresis, liquid chromatography-mass spectrometry and X-ray protein crystallography. Moreover, visualizations like a virtual 2D proteome map of pI vs. MW are worthwhile to discuss the proteome diversity among different species. Although the genome sequence data of the fungi kingdom improved enormously, the proteomic details have been poorly elaborated. Therefore, we have calculated the MW and pI of the fungi proteins and reported them in, FungiProteomeDB, an online database (DB) https://vision4research.com/fungidb/. We analyzed the proteome of 685 fungal species that contain 7 127 141 protein sequences. The DB provides an easy-to-use and efficient interface for various search options, summary statistics and virtual 2D proteome map visualizations. The MW and pI of a protein can be obtained by searching the name of a protein, a keyword or a list of accession numbers. It also allows querying protein sequences. The DB will be helpful in hypothesis formulation and in various biotechnological applications. Database URL https://vision4research.com/fungidb/.
Collapse
|
7
|
Makandar AI, Jain M, Yuba E, Sethi G, Gupta RK. Canvassing Prospects of Glyco-Nanovaccines for Developing Cross-Presentation Mediated Anti-Tumor Immunotherapy. Vaccines (Basel) 2022; 10:vaccines10122049. [PMID: 36560459 PMCID: PMC9784904 DOI: 10.3390/vaccines10122049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
In view of the severe downsides of conventional cancer therapies, the quest of developing alternative strategies still remains of critical importance. In this regard, antigen cross-presentation, usually employed by dendritic cells (DCs), has been recognized as a potential solution to overcome the present impasse in anti-cancer therapeutic strategies. It has been established that an elevated cytotoxic T lymphocyte (CTL) response against cancer cells can be achieved by targeting receptors expressed on DCs with specific ligands. Glycans are known to serve as ligands for C-type lectin receptors (CLRs) expressed on DCs, and are also known to act as a tumor-associated antigen (TAA), and, thus, can be harnessed as a potential immunotherapeutic target. In this scenario, integrating the knowledge of cross-presentation and glycan-conjugated nanovaccines can help us to develop so called 'glyco-nanovaccines' (GNVs) for targeting DCs. Here, we briefly review and analyze the potential of GNVs as the next-generation anti-tumor immunotherapy. We have compared different antigen-presenting cells (APCs) for their ability to cross-present antigens and described the potential nanocarriers for tumor antigen cross-presentation. Further, we discuss the role of glycans in targeting of DCs, the immune response due to pathogens, and imitative approaches, along with parameters, strategies, and challenges involved in cross-presentation-based GNVs for cancer immunotherapy. It is known that the effectiveness of GNVs in eradicating tumors by inducing strong CTL response in the tumor microenvironment (TME) has been largely hindered by tumor glycosylation and the expression of different lectin receptors (such as galectins) by cancer cells. Tumor glycan signatures can be sensed by a variety of lectins expressed on immune cells and mediate the immune suppression which, in turn, facilitates immune evasion. Therefore, a sound understanding of the glycan language of cancer cells, and glycan-lectin interaction between the cancer cells and immune cells, would help in strategically designing the next-generation GNVs for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Amina I. Makandar
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| | - Mannat Jain
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
- Correspondence: (E.Y.); (G.S.); or (R.K.G.)
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Correspondence: (E.Y.); (G.S.); or (R.K.G.)
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
- Correspondence: (E.Y.); (G.S.); or (R.K.G.)
| |
Collapse
|
8
|
Shields NJ, Peyroux EM, Campbell K, Mehta S, Woolley AG, Counoupas C, Neumann S, Young SL. Calpains Released from Necrotic Tumor Cells Enhance Antigen Cross-Presentation to Activate CD8 +T Cells In Vitro. THE JOURNAL OF IMMUNOLOGY 2022; 209:1635-1651. [DOI: 10.4049/jimmunol.2100500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/19/2022] [Indexed: 11/07/2022]
|
9
|
Mohanta TK, Kamran MS, Omar M, Anwar W, Choi GS. PlantMWpIDB: a database for the molecular weight and isoelectric points of the plant proteomes. Sci Rep 2022; 12:7421. [PMID: 35523906 PMCID: PMC9076895 DOI: 10.1038/s41598-022-11077-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 04/11/2022] [Indexed: 01/14/2023] Open
Abstract
The molecular weight and isoelectric point of the proteins are very important parameters that control their subcellular localization and subsequent function. Although the genome sequence data of the plant kingdom improved enormously, the proteomic details have been poorly elaborated. Therefore, we have calculated the molecular weight and isoelectric point of the plant proteins and reported them in this database. A database, PlantMWpIDB, containing protein data from 342 plant proteomes was created to provide information on plant proteomes for hypothesis formulation in basic research and for biotechnological applications. The Molecular weight and isoelectric point (pI) are important molecular parameters of proteins that are useful when conducting protein studies involving 2D gel electrophoresis, liquid chromatography-mass spectrometry, and X-ray protein crystallography. PlantMWpIDB provides an easy-to-use and efficient interface for search options and generates a summary of basic protein parameters. The database represents a virtual 2D proteome map of plants, and the molecular weight and pI of a protein can be obtained by searching on the name of a protein, a keyword, or by a list of accession numbers. The PlantMWpIDB database also allows one to query protein sequences. The database can be found in the following link https://plantmwpidb.com/ . The individual 2D virtual proteome map of the plant kingdom will enable us to understand the proteome diversity between different species. Further, the molecular weight and isoelectric point of individual proteins can enable us to understand their functional significance in different species.
Collapse
Affiliation(s)
- Tapan Kumar Mohanta
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Oman.
| | - Muhammad Shahzad Kamran
- Department of Computer Science and IT, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Omar
- Department of Data Science, Faculty of Computing, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.,Department of Information and Communication Engineering, Yeungnam University, 214-1, Gyeongsan-si, 712-749, South Korea
| | - Waheed Anwar
- Department of Computer Science and IT, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Gyu Sang Choi
- Department of Information and Communication Engineering, Yeungnam University, 214-1, Gyeongsan-si, 712-749, South Korea.
| |
Collapse
|
10
|
Improving potency of Nanoliposomal AE36 peptide vaccine by adding CD4+ T cell helper epitope and MPL in TUBO breast cancer mice model. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
11
|
Lu Y, Shi Y, You J. Strategy and clinical application of up-regulating cross presentation by DCs in anti-tumor therapy. J Control Release 2021; 341:184-205. [PMID: 34774890 DOI: 10.1016/j.jconrel.2021.11.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/20/2022]
Abstract
The cross presentation of exogenous antigen (Ag) by dendritic cells (DCs) facilitates a diversified mode of T-cell activation, orchestrates specific humoral and cellular immunity, and contributes to an efficient anti-tumor immune response. DCs-mediated cross presentation is subject to both intrinsic and extrinsic factors, including the homing and phenotype of DCs, the spatiotemporal trafficking and degradation kinetics of Ag, and multiple microenvironmental clues, with many details largely unexplored. Here, we systemically review the current mechanistic understanding and regulation strategies of cross presentation by heterogeneous DC populations. We also provide insights into the future exploitation of DCs cross presentation for a better clinical efficacy in anti-tumor therapy.
Collapse
Affiliation(s)
- Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
12
|
Shi Y, Zhu C, Liu Y, Lu Y, Li X, Qin B, Luo Z, Luo L, Jiang M, Zhang J, Guan G, Zheng C, You J. A Vaccination with Boosted Cross Presentation by ER-Targeted Antigen Delivery for Anti-Tumor Immunotherapy. Adv Healthc Mater 2021; 10:e2001934. [PMID: 33502831 DOI: 10.1002/adhm.202001934] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/17/2020] [Indexed: 12/16/2022]
Abstract
Vaccination is a widely-accepted resort against the invasion or proliferation of bacteria, parasites, viruses, and even cancer, which accounts heavily on an active involvement of CD8+ T cells. As one of the pivotal strategies taken by dendritic cells (DCs) to promote the responsiveness of CD8+ T cells to exogenous antigens, cross presentation culminates in an elevated overall host defense against cancer or infection. However, the precise mechanisms regulating such a process remains elusive, and current attempts to fuel cross presentation usually fail to exert efficiency. Here, model antigen OVA-loaded, endoplasmic reticulum (ER)-targeting cationic liposome (OVA@lipoT) is developed and characterized with a booster effect on the activation and maturation of DCs. Moreover, OVA@lipoT pulsed DCs exhibit overwhelming superiority in triggering cytotoxic T lymphocyte response both in vivo and in vitro. Data reveal that lipoT alters the intracellular trafficking and presenting pathway of antigen, which promotes cross presentation and bears close relationship to the ER-associated degradation (ERAD). These results may drop a hint about the interconnectivity between cross presentation and ER-targeted antigen delivery, provide extra information to the understanding of ERAD-mediated cross priming, and even shed new light on the design and optimization of vaccines against currently intractable cancers or virus-infection.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Chunqi Zhu
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Yu Liu
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Yichao Lu
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Xiang Li
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Bing Qin
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Zhenyu Luo
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Lihua Luo
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Mengshi Jiang
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Junlei Zhang
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Guannan Guan
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| | - Cheng Zheng
- Zhejiang Institute for Food and Drug Control Zhejiang 310058 P. R. China
| | - Jian You
- College of Pharmaceutical Sciences Zhejiang University Zhejiang 310058 P. R. China
| |
Collapse
|
13
|
Halcrow PW, Geiger JD, Chen X. Overcoming Chemoresistance: Altering pH of Cellular Compartments by Chloroquine and Hydroxychloroquine. Front Cell Dev Biol 2021; 9:627639. [PMID: 33634129 PMCID: PMC7900406 DOI: 10.3389/fcell.2021.627639] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Resistance to the anti-cancer effects of chemotherapeutic agents (chemoresistance) is a major issue for people living with cancer and their providers. A diverse set of cellular and inter-organellar signaling changes have been implicated in chemoresistance, but it is still unclear what processes lead to chemoresistance and effective strategies to overcome chemoresistance are lacking. The anti-malaria drugs, chloroquine (CQ) and its derivative hydroxychloroquine (HCQ) are being used for the treatment of various cancers and CQ and HCQ are used in combination with chemotherapeutic drugs to enhance their anti-cancer effects. The widely accepted anti-cancer effect of CQ and HCQ is their ability to inhibit autophagic flux. As diprotic weak bases, CQ and HCQ preferentially accumulate in acidic organelles and neutralize their luminal pH. In addition, CQ and HCQ acidify the cytosolic and extracellular environments; processes implicated in tumorigenesis and cancer. Thus, the anti-cancer effects of CQ and HCQ extend beyond autophagy inhibition. The present review summarizes effects of CQ, HCQ and proton pump inhibitors on pH of various cellular compartments and discuss potential mechanisms underlying their pH-dependent anti-cancer effects. The mechanisms considered here include their ability to de-acidify lysosomes and inhibit autophagosome lysosome fusion, to de-acidify Golgi apparatus and secretory vesicles thus affecting secretion, and to acidify cytoplasm thus disturbing aerobic metabolism. Further, we review the ability of these agents to prevent chemotherapeutic drugs from accumulating in acidic organelles and altering their cytosolic concentrations.
Collapse
Affiliation(s)
| | | | - Xuesong Chen
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| |
Collapse
|
14
|
Canton J, Blees H, Henry CM, Buck MD, Schulz O, Rogers NC, Childs E, Zelenay S, Rhys H, Domart MC, Collinson L, Alloatti A, Ellison CJ, Amigorena S, Papayannopoulos V, Thomas DC, Randow F, Reis e Sousa C. The receptor DNGR-1 signals for phagosomal rupture to promote cross-presentation of dead-cell-associated antigens. Nat Immunol 2021; 22:140-153. [PMID: 33349708 PMCID: PMC7116638 DOI: 10.1038/s41590-020-00824-x] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022]
Abstract
Type 1 conventional dendritic (cDC1) cells are necessary for cross-presentation of many viral and tumor antigens to CD8+ T cells. cDC1 cells can be identified in mice and humans by high expression of DNGR-1 (also known as CLEC9A), a receptor that binds dead-cell debris and facilitates XP of corpse-associated antigens. Here, we show that DNGR-1 is a dedicated XP receptor that signals upon ligand engagement to promote phagosomal rupture. This allows escape of phagosomal contents into the cytosol, where they access the endogenous major histocompatibility complex class I antigen processing pathway. The activity of DNGR-1 maps to its signaling domain, which activates SYK and NADPH oxidase to cause phagosomal damage even when spliced into a heterologous receptor and expressed in heterologous cells. Our data reveal the existence of innate immune receptors that couple ligand binding to endocytic vesicle damage to permit MHC class I antigen presentation of exogenous antigens and to regulate adaptive immunity.
Collapse
Affiliation(s)
- Johnathan Canton
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Hanna Blees
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Conor M Henry
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Michael D Buck
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Oliver Schulz
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Neil C Rogers
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Eleanor Childs
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Santiago Zelenay
- Cancer Inflammation and Immunity Group, CRUK Manchester Institute, The University of Manchester, Alderley Park, UK
| | - Hefin Rhys
- Flow Cytometry STP, The Francis Crick Institute, London, UK
| | | | - Lucy Collinson
- Electron Microscopy STP, The Francis Crick Institute, London, UK
| | - Andres Alloatti
- Centre de Recherche, INSERM U932, Institut Curie, Paris, France
| | - Cara J Ellison
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | | | | - David C Thomas
- Immunity and Inflammation, 9NC, Imperial College, London, UK
| | - Felix Randow
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
15
|
Laborde RJ, Ishimura ME, Abreu-Butin L, Nogueira CV, Grubaugh D, Cruz-Leal Y, Luzardo MC, Fernández A, Mesa C, Pazos F, Álvarez C, Alonso ME, Starnbach MN, Higgins DE, Fernández LE, Longo-Maugéri IM, Lanio ME. Sticholysins, pore-forming proteins from a marine anemone can induce maturation of dendritic cells through a TLR4 dependent-pathway. Mol Immunol 2021; 131:144-154. [PMID: 33422341 DOI: 10.1016/j.molimm.2020.12.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/30/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023]
Abstract
Sticholysins (Sts) I and II (StI and StII) are pore-forming proteins (PFPs), purified from the Caribbean Sea anemone Stichodactyla helianthus. StII encapsulated into liposomes induces a robust antigen-specific cytotoxic CD8+ T lymphocytes (CTL) response and in its free form the maturation of bone marrow-derived dendritic cells (BM-DCs). It is probable that the latter is partially supporting in part the immunomodulatory effect on the CTL response induced by StII-containing liposomes. In the present work, we demonstrate that the StII's ability of inducing maturation of BM-DCs is also shared by StI, an isoform of StII. Using heat-denatured Sts we observed a significant reduction in the up-regulation of maturation markers indicating that both PFP's ability to promote maturation of BM-DCs is dependent on their conformational characteristics. StII-mediated DC maturation was abrogated in BM-DCs from toll-like receptor (TLR) 4 and myeloid differentiation primary response gene 88 (MyD88)-knockout mice but not in cells from TLR2-knockout mice. Furthermore, the antigen-specific CTL response induced by StII-containing liposomes was reduced in TLR4-knockout mice. These results indicate that StII, and probably by extension StI, has the ability to induce maturation of DCs through a TLR4/MyD88-dependent pathway, and that this activation contributes to the CTL response generated by StII-containing liposomes.
Collapse
Affiliation(s)
- Rady J Laborde
- Laboratory of Toxins and Liposomes, Center for Protein Studies, Faculty of Biology, University of Havana (UH), Lab UH-CIM, Havana, 10400, Cuba.
| | - Mayari E Ishimura
- Discipline of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), 04023-062, São Paulo, Brazil.
| | - Lianne Abreu-Butin
- Discipline of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), 04023-062, São Paulo, Brazil
| | - Catarina V Nogueira
- Department of Microbiology and Immunobiology of Harvard Medical School, Harvard University, MA, USA.
| | - Daniel Grubaugh
- Department of Microbiology and Immunobiology of Harvard Medical School, Harvard University, MA, USA.
| | - Yoelys Cruz-Leal
- Laboratory of Toxins and Liposomes, Center for Protein Studies, Faculty of Biology, University of Havana (UH), Lab UH-CIM, Havana, 10400, Cuba.
| | - María C Luzardo
- Laboratory of Toxins and Liposomes, Center for Protein Studies, Faculty of Biology, University of Havana (UH), Lab UH-CIM, Havana, 10400, Cuba.
| | - Audry Fernández
- Immunobiology Division, Center of Molecular Immunology (CIM), Havana, 11600, Cuba.
| | - Circe Mesa
- Immunobiology Division, Center of Molecular Immunology (CIM), Havana, 11600, Cuba.
| | - Fabiola Pazos
- Laboratory of Toxins and Liposomes, Center for Protein Studies, Faculty of Biology, University of Havana (UH), Lab UH-CIM, Havana, 10400, Cuba.
| | - Carlos Álvarez
- Laboratory of Toxins and Liposomes, Center for Protein Studies, Faculty of Biology, University of Havana (UH), Lab UH-CIM, Havana, 10400, Cuba.
| | - María E Alonso
- Laboratory of Toxins and Liposomes, Center for Protein Studies, Faculty of Biology, University of Havana (UH), Lab UH-CIM, Havana, 10400, Cuba
| | - Michael N Starnbach
- Department of Microbiology and Immunobiology of Harvard Medical School, Harvard University, MA, USA.
| | - Darren E Higgins
- Department of Microbiology and Immunobiology of Harvard Medical School, Harvard University, MA, USA.
| | - Luis E Fernández
- Immunobiology Division, Center of Molecular Immunology (CIM), Havana, 11600, Cuba.
| | - Ieda M Longo-Maugéri
- Discipline of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), 04023-062, São Paulo, Brazil.
| | - María E Lanio
- Laboratory of Toxins and Liposomes, Center for Protein Studies, Faculty of Biology, University of Havana (UH), Lab UH-CIM, Havana, 10400, Cuba.
| |
Collapse
|
16
|
Khan N, Kumar N, Geiger JD. Possible therapeutic targets for SARS-CoV-2 infection and COVID-19. JOURNAL OF ALLERGY AND INFECTIOUS DISEASES 2021; 2:75-83. [PMID: 37564275 PMCID: PMC10414779 DOI: 10.46439/allergy.2.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
SARS-CoV-2 infection causes COVID-19, which has emerged as a health emergency worldwide. SARS-CoV-2 infects cells by binding to ACE2 receptors and enters into the cytoplasm following its escape from endolysosomes. Once in the cytoplasm, the virus replicates and eventually causes various pathological conditions including acute respiratory distress syndrome (ARDS) that is caused by pro-inflammatory cytokine storms. Thus, endolysosomes and cytokine storms are important therapeutic targets to suppress SARS-CoV-2 infection and COVID-19. Here, we discuss therapeutic targets of SARS-CoV-2 infection and available drugs that could be helpful in the suppression of the SARS-CoV-2 infection and pathological condition COVID-19. The urgency of the COVID-19 pandemic precludes the development of new drugs and increased focus on drug repurposing might provide the quickest way to finding effective medicines.
Collapse
Affiliation(s)
- Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | - Nirmal Kumar
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| |
Collapse
|
17
|
Khan N, Chen X, Geiger JD. Role of Endolysosomes in Severe Acute Respiratory Syndrome Coronavirus-2 Infection and Coronavirus Disease 2019 Pathogenesis: Implications for Potential Treatments. Front Pharmacol 2020; 11:595888. [PMID: 33324224 PMCID: PMC7723437 DOI: 10.3389/fphar.2020.595888] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is an enveloped, single-stranded RNA virus. Humans infected with SARS-CoV-2 develop a disease known as coronavirus disease 2019 (COVID-19) with symptoms and consequences including acute respiratory distress syndrome (ARDS), cardiovascular disorders, and death. SARS-CoV-2 appears to infect cells by first binding viral spike proteins with host protein angiotensin-converting enzyme 2 (ACE2) receptors; the virus is endocytosed following priming by transmembrane protease serine 2 (TMPRSS2). The process of virus entry into endosomes and its release from endolysosomes are key features of enveloped viruses. Thus, it is important to focus attention on the role of endolysosomes in SARS-CoV-2 infection. Indeed, coronaviruses are now known to hijack endocytic machinery to enter cells such that they can deliver their genome at replication sites without initiating host detection and immunological responses. Hence, endolysosomes might be good targets for developing therapeutic strategies against coronaviruses. Here, we focus attention on the involvement of endolysosomes in SARS-CoV-2 infection and COVID-19 pathogenesis. Further, we explore endolysosome-based therapeutic strategies to restrict SARS-CoV-2 infection and COVID-19 pathogenesis.
Collapse
Affiliation(s)
| | | | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
18
|
Engineering anti-cancer nanovaccine based on antigen cross-presentation. Biosci Rep 2020; 39:220729. [PMID: 31652460 PMCID: PMC6822533 DOI: 10.1042/bsr20193220] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 01/16/2023] Open
Abstract
Dendritic cells (DCs) present exogenous antigens on major histocompatibility complex (MHC) class I molecules, thereby activating CD8+ T cells, contributing to tumor elimination through a mechanism known as antigen cross-presentation. A variety of factors such as maturation state of DCs, co-stimulatory signals, T-cell microenvironment, antigen internalization routes and adjuvants regulate the process of DC-mediated antigen cross-presentation. Recently, the development of successful cancer immunotherapies may be attributed to the ability of DCs to cross-present tumor antigens. In this review article, we focus on the underlying mechanism of antigen cross-presentation and ways to improve antigen cross-presentation in different DC subsets. We have critically summarized the recent developments in the generation of novel nanovaccines for robust CD8+ T-cell response in cancer. In this context, we have reviewed nanocarriers that have been used for cancer immunotherapeutics based on antigen cross-presentation mechanism. Additionally, we have also expressed our views on the future applications of this mechanism in curing cancer.
Collapse
|
19
|
Chen X, Geiger JD. Janus sword actions of chloroquine and hydroxychloroquine against COVID-19. Cell Signal 2020; 73:109706. [PMID: 32629149 PMCID: PMC7333634 DOI: 10.1016/j.cellsig.2020.109706] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Chloroquine (CQ) and its analogue hydroxychloroquine (HCQ) have been thrust into our everyday vernacular because some believe, based on very limited basic and clinical data, that they might be helpful in preventing and/or lessening the severity of the pandemic coronavirus disease 2019 (COVID-19). However, lacking is a temperance in enthusiasm for their possible use as well as sufficient perspective on their effects and side-effects. CQ and HCQ have well-known properties of being diprotic weak bases that preferentially accumulate in acidic organelles (endolysosomes and Golgi apparatus) and neutralize luminal pH of acidic organelles. These primary actions of CQ and HCQ are responsible for their anti-malarial effects; malaria parasites rely on acidic digestive vacuoles for survival. Similarly, de-acidification of endolysosomes and Golgi by CQ and HCQ may block severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) integration into host cells because SARS-CoV-2 may require an acidic environment for its entry and for its ability to bud and infect bystander cells. Further, de-acidification of endolysosomes and Golgi may underly the immunosuppressive effects of these two drugs. However, modern cell biology studies have shown clearly that de-acidification results in profound changes in the structure, function and cellular positioning of endolysosomes and Golgi, in signaling between these organelles and other subcellular organelles, and in fundamental cellular functions. Thus, studying the possible therapeutic effects of CQ and HCQ against COVID-19 must occur concurrent with studies of the extent to which these drugs affect organellar and cell biology. When comprehensively examined, a better understanding of the Janus sword actions of these and other drugs might yield better decisions and better outcomes.
Collapse
Affiliation(s)
- Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America.
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| |
Collapse
|
20
|
Kim Y, Kang S, Shin H, Kim T, Yu B, Kim J, Yoo D, Jon S. Sequential and Timely Combination of a Cancer Nanovaccine with Immune Checkpoint Blockade Effectively Inhibits Tumor Growth and Relapse. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202006117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Yujin Kim
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Sukmo Kang
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Hocheol Shin
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Taewoo Kim
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Byeongjun Yu
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Jinjoo Kim
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Dohyun Yoo
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| |
Collapse
|
21
|
Hu F, Yue H, Lu T, Ma G. Cytosolic delivery of HBsAg and enhanced cellular immunity by pH-responsive liposome. J Control Release 2020; 324:460-470. [DOI: 10.1016/j.jconrel.2020.05.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 01/10/2023]
|
22
|
Kozik P, Gros M, Itzhak DN, Joannas L, Heurtebise-Chrétien S, Krawczyk PA, Rodríguez-Silvestre P, Alloatti A, Magalhaes JG, Del Nery E, Borner GHH, Amigorena S. Small Molecule Enhancers of Endosome-to-Cytosol Import Augment Anti-tumor Immunity. Cell Rep 2020; 32:107905. [PMID: 32668257 PMCID: PMC7370168 DOI: 10.1016/j.celrep.2020.107905] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 05/15/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022] Open
Abstract
Cross-presentation of antigens by dendritic cells (DCs) is critical for initiation of anti-tumor immune responses. Yet, key steps involved in trafficking of antigens taken up by DCs remain incompletely understood. Here, we screen 700 US Food and Drug Administration (FDA)-approved drugs and identify 37 enhancers of antigen import from endolysosomes into the cytosol. To reveal their mechanism of action, we generate proteomic organellar maps of control and drug-treated DCs (focusing on two compounds, prazosin and tamoxifen). By combining organellar mapping, quantitative proteomics, and microscopy, we conclude that import enhancers undergo lysosomal trapping leading to membrane permeation and antigen release. Enhancing antigen import facilitates cross-presentation of soluble and cell-associated antigens. Systemic administration of prazosin leads to reduced growth of MC38 tumors and to a synergistic effect with checkpoint immunotherapy in a melanoma model. Thus, inefficient antigen import into the cytosol limits antigen cross-presentation, restraining the potency of anti-tumor immune responses and efficacy of checkpoint blockers.
Collapse
Affiliation(s)
- Patrycja Kozik
- INSERM U932, PSL Research University, Institut Curie, 75005 Paris, France; MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| | - Marine Gros
- INSERM U932, PSL Research University, Institut Curie, 75005 Paris, France
| | - Daniel N Itzhak
- Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Leonel Joannas
- INSERM U932, PSL Research University, Institut Curie, 75005 Paris, France
| | | | | | | | - Andrés Alloatti
- INSERM U932, PSL Research University, Institut Curie, 75005 Paris, France
| | | | - Elaine Del Nery
- Institut Curie, PSL Research University, Department of Translational Research-Biophenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), 75005 Paris, France
| | - Georg H H Borner
- Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | | |
Collapse
|
23
|
Kim Y, Kang S, Shin H, Kim T, Yu B, Kim J, Yoo D, Jon S. Sequential and Timely Combination of a Cancer Nanovaccine with Immune Checkpoint Blockade Effectively Inhibits Tumor Growth and Relapse. Angew Chem Int Ed Engl 2020; 59:14628-14638. [PMID: 32430981 DOI: 10.1002/anie.202006117] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 12/12/2022]
Abstract
We describe a small lipid nanoparticle (SLNP)-based nanovaccine platform and a new combination treatment regimen. Tumor antigen-displaying, CpG adjuvant-embedded SLNPs (OVAPEP -SLNP@CpG) were prepared from biocompatible phospholipids and a cationic cholesterol derivative. The resulting nanovaccine showed highly potent antitumor efficacy in both prophylactic and therapeutic E.G7 tumor models. However, this vaccine induced T cell exhaustion by elevating PD-L1 expression, leading to tumor recurrence. Thus, the nanovaccine was combined with simultaneous anti-PD-1 antibody treatment, but the therapeutic efficacy of this regimen was comparable to that of the nanovaccine alone. Finally, mice that showed a good therapeutic response after the first cycle of immunization with the nanovaccine underwent a second cycle together with anti-PD-1 therapy, resulting in suppression of tumor relapse. This suggests that the antitumor efficacy of combinations of nanovaccines with immune checkpoint blockade therapy is dependent on treatment sequence and the timing of each modality.
Collapse
Affiliation(s)
- Yujin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Sukmo Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Hocheol Shin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Taewoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Byeongjun Yu
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Jinjoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Dohyun Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| |
Collapse
|
24
|
Halpert MM, Konduri V, Liang D, Vazquez-Perez J, Hofferek CJ, Weldon SA, Baig Y, Vedula I, Levitt JM, Decker WK. MHC class I and II peptide homology regulates the cellular immune response. FASEB J 2020; 34:8082-8101. [PMID: 32298026 DOI: 10.1096/fj.201903002r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/18/2020] [Accepted: 03/31/2020] [Indexed: 12/24/2022]
Abstract
Mammalian immune responses are initiated by "danger" signals--immutable molecular structures known as PAMPs. When detected by fixed, germline encoded receptors, pathogen-associated molecular pattern (PAMPs) subsequently inform the polarization of downstream adaptive responses depending upon identity and localization of the PAMP. Here, we report the existence of a completely novel "PAMP" that is not a molecular structure but an antigenic pattern. This pattern--the incidence of peptide epitopes with stretches of 100% sequence identity bound to both dendritic cell (DC) major histocompatibility (MHC) class I and MHC class II--strongly induces TH 1 immune polarization and activation of the cellular immune response. Inherent in the existence of this PAMP is the concomitant existence of a molecular sensor complex with the ability to scan and compare amino acid sequence identities of bound class I and II peptides. We provide substantial evidence implicating the multienzyme aminoacyl-tRNA synthetase (mARS) complex and its AIMp1 structural component as the key constituents of this complex. The results demonstrate a wholly novel mechanism by which T-helper (TH ) polarization is governed and provide critical information for the design of vaccination strategies intended to provoke cell-mediated immunity.
Collapse
Affiliation(s)
- Matthew M Halpert
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Vanaja Konduri
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Dan Liang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | | | - Colby J Hofferek
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Scott A Weldon
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Yunyu Baig
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Indira Vedula
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan M Levitt
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.,Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - William K Decker
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
25
|
Nanoparticle mediated cancer immunotherapy. Semin Cancer Biol 2020; 69:307-324. [PMID: 32259643 DOI: 10.1016/j.semcancer.2020.03.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 03/09/2020] [Accepted: 03/23/2020] [Indexed: 12/18/2022]
Abstract
The versatility and nanoscale size have helped nanoparticles (NPs) improve the efficacy of conventional cancer immunotherapy and opened up exciting approaches to combat cancer. This review first outlines the tumor immune evasion and the defensive tumor microenvironment (TME) that hinders the activity of host immune system against tumor. Then, a detailed description on how the NP based strategies have helped improve the efficacy of conventional cancer vaccines and overcome the obstacles led by TME. Sustained and controlled drug delivery, enhanced cross presentation by immune cells, co-encapsulation of adjuvants, inhibition of immune checkpoints and intrinsic adjuvant like properties have aided NPs to improve the therapeutic efficacy of cancer vaccines. Also, NPs have been efficient modulators of TME. In this context, NPs facilitate better penetration of the chemotherapeutic drug by dissolution of the inhibitory meshwork formed by tumor associated cells, blood vessels, soluble mediators and extra cellular matrix in TME. NPs achieve this by suppression, modulation, or reprogramming of the immune cells and other mediators localised in TME. This review further summarizes the applications of NPs used to enhance the efficacy of cancer vaccines and modulate the TME to improve cancer immunotherapy. Finally, the hurdles faced in commercialization and translation to clinic have been discussed and intriguingly, NPs owe great potential to emerge as clinical formulations for cancer immunotherapy in near future.
Collapse
|
26
|
Lung P, Yang J, Li Q. Nanoparticle formulated vaccines: opportunities and challenges. NANOSCALE 2020; 12:5746-5763. [PMID: 32124894 DOI: 10.1039/c9nr08958f] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Vaccines harness the inherent properties of the immune system to prevent diseases or treat existing ones. Continuous efforts have been devoted to both gaining a mechanistic understanding of how the immune system operates and designing vaccines with high efficacies and effectiveness. Advancements in nanotechnology in recent years have generated unique opportunities to meet the daunting challenges associated with immunology and vaccine development. Firstly, nanoparticle formulated systems provide ideal model systems for studying the operation of the immune system, making it possible to systematically identify key factors and understand their roles in specific immune responses. Also, the versatile compositions/architectures of nanoparticle systems enable new strategies/novel platforms for developing vaccines with high efficacies and effectiveness. In this review, we discuss the advantages of nanoparticles and the challenges faced during vaccine development, through the framework of the immunological mechanisms of vaccination, with the aim of bridging the gap between immunology and materials science, which are both involved in vaccine design. The knowledge obtained provides general guidelines for future vaccine development.
Collapse
Affiliation(s)
- Pingsai Lung
- Department of Physics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | | | | |
Collapse
|
27
|
Baldin AV, Savvateeva LV, Bazhin AV, Zamyatnin AA. Dendritic Cells in Anticancer Vaccination: Rationale for Ex Vivo Loading or In Vivo Targeting. Cancers (Basel) 2020; 12:cancers12030590. [PMID: 32150821 PMCID: PMC7139354 DOI: 10.3390/cancers12030590] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) have shown great potential as a component or target in the landscape of cancer immunotherapy. Different in vivo and ex vivo strategies of DC vaccine generation with different outcomes have been proposed. Numerous clinical trials have demonstrated their efficacy and safety in cancer patients. However, there is no consensus regarding which DC-based vaccine generation method is preferable. A problem of result comparison between trials in which different DC-loading or -targeting approaches have been applied remains. The employment of different DC generation and maturation methods, antigens and administration routes from trial to trial also limits the objective comparison of DC vaccines. In the present review, we discuss different methods of DC vaccine generation. We conclude that standardized trial designs, treatment settings and outcome assessment criteria will help to determine which DC vaccine generation approach should be applied in certain cancer cases. This will result in a reduction in alternatives in the selection of preferable DC-based vaccine tactics in patient. Moreover, it has become clear that the application of a DC vaccine alone is not sufficient and combination immunotherapy with recent advances, such as immune checkpoint inhibitors, should be employed to achieve a better clinical response and outcome.
Collapse
Affiliation(s)
- Alexey V. Baldin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Lyudmila V. Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians University of Munich, 81377 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
- Belozersky Institute of Physico-Chemical Biology, Department of Cell Signaling, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +74-956-229-843
| |
Collapse
|
28
|
Varisli L, Cen O, Vlahopoulos S. Dissecting pharmacological effects of chloroquine in cancer treatment: interference with inflammatory signaling pathways. Immunology 2020; 159:257-278. [PMID: 31782148 PMCID: PMC7011648 DOI: 10.1111/imm.13160] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
Chloroquines are 4-aminoquinoline-based drugs mainly used to treat malaria. At pharmacological concentrations, they have significant effects on tissue homeostasis, targeting diverse signaling pathways in mammalian cells. A key target pathway is autophagy, which regulates macromolecule turnover in the cell. In addition to affecting cellular metabolism and bioenergetic flow equilibrium, autophagy plays a pivotal role at the interface between inflammation and cancer progression. Chloroquines consequently have critical effects in tissue metabolic activity and importantly, in key functions of the immune system. In this article, we will review the work addressing the role of chloroquines in the homeostasis of mammalian tissue, and the potential strengths and weaknesses concerning their use in cancer therapy.
Collapse
Affiliation(s)
- Lokman Varisli
- Union of Education and Science Workers (EGITIM SEN), Diyarbakir Branch, Diyarbakir, Turkey
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir, Turkey
| | - Osman Cen
- Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Natural Sciences, Joliet Jr College, Joliet, IL, USA
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
29
|
Saylor K, Gillam F, Lohneis T, Zhang C. Designs of Antigen Structure and Composition for Improved Protein-Based Vaccine Efficacy. Front Immunol 2020; 11:283. [PMID: 32153587 PMCID: PMC7050619 DOI: 10.3389/fimmu.2020.00283] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/04/2020] [Indexed: 12/19/2022] Open
Abstract
Today, vaccinologists have come to understand that the hallmark of any protective immune response is the antigen. However, it is not the whole antigen that dictates the immune response, but rather the various parts comprising the whole that are capable of influencing immunogenicity. Protein-based antigens hold particular importance within this structural approach to understanding immunity because, though different molecules can serve as antigens, only proteins are capable of inducing both cellular and humoral immunity. This fact, coupled with the versatility and customizability of proteins when considering vaccine design applications, makes protein-based vaccines (PBVs) one of today's most promising technologies for artificially inducing immunity. In this review, we follow the development of PBV technologies through time and discuss the antigen-specific receptors that are most critical to any immune response: pattern recognition receptors, B cell receptors, and T cell receptors. Knowledge of these receptors and their ligands has become exceptionally valuable in the field of vaccinology, where today it is possible to make drastic modifications to PBV structure, from primary to quaternary, in order to promote recognition of target epitopes, potentiate vaccine immunogenicity, and prevent antigen-associated complications. Additionally, these modifications have made it possible to control immune responses by modulating stability and targeting PBV to key immune cells. Consequently, careful consideration should be given to protein structure when designing PBVs in the future in order to potentiate PBV efficacy.
Collapse
Affiliation(s)
- Kyle Saylor
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, United States
| | - Frank Gillam
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, United States
- Locus Biosciences, Morrisville, NC, United States
| | - Taylor Lohneis
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, United States
- BioPharmaceutical Technology Department, GlaxoSmithKline, Rockville, MD, United States
| | - Chenming Zhang
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
30
|
Imai J, Ohashi S, Sakai T. Endoplasmic Reticulum-Associated Degradation-Dependent Processing in Cross-Presentation and Its Potential for Dendritic Cell Vaccinations: A Review. Pharmaceutics 2020; 12:pharmaceutics12020153. [PMID: 32070016 PMCID: PMC7076524 DOI: 10.3390/pharmaceutics12020153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 01/14/2023] Open
Abstract
While the success of dendritic cell (DC) vaccination largely depends on cross-presentation (CP) efficiency, the precise molecular mechanism of CP is not yet characterized. Recent research revealed that endoplasmic reticulum (ER)-associated degradation (ERAD), which was first identified as part of the protein quality control system in the ER, plays a pivotal role in the processing of extracellular proteins in CP. The discovery of ERAD-dependent processing strongly suggests that the properties of extracellular antigens are one of the keys to effective DC vaccination, in addition to DC subsets and the maturation of these cells. In this review, we address recent advances in CP, focusing on the molecular mechanisms of the ERAD-dependent processing of extracellular proteins. As ERAD itself and the ERAD-dependent processing in CP share cellular machinery, enhancing the recognition of extracellular proteins, such as the ERAD substrate, by ex vivo methods may serve to improve the efficacy of DC vaccination.
Collapse
Affiliation(s)
- Jun Imai
- Correspondence: ; Tel.: +81-27-352-1180
| | | | | |
Collapse
|
31
|
Imai J, Otani M, Sakai T. Distinct Subcellular Compartments of Dendritic Cells Used for Cross-Presentation. Int J Mol Sci 2019; 20:ijms20225606. [PMID: 31717517 PMCID: PMC6888166 DOI: 10.3390/ijms20225606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/29/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells (DCs) present exogenous protein-derived peptides on major histocompatibility complex class I molecules to prime naïve CD8+ T cells. This DC specific ability, called cross-presentation (CP), is important for the activation of cell-mediated immunity and the induction of self-tolerance. Recent research revealed that endoplasmic reticulum-associated degradation (ERAD), which was first identified as a part of the unfolded protein response—a quality control system in the ER—plays a pivotal role in the processing of exogenous proteins in CP. Moreover, DCs express a variety of immuno-modulatory molecules and cytokines to regulate T cell activation in response to the environment. Although both CP and immuno-modulation are indispensable, contrasting ER conditions are required for their correct activity. Since ERAD substrates are unfolded proteins, their accumulation may result in ER stress, impaired cell homeostasis, and eventually apoptosis. In contrast, activation of the unfolded protein response should be inhibited for DCs to express immuno-modulatory molecules and cytokines. Here, we review recent advances on antigen CP, focusing on intracellular transport routes for exogenous antigens and distinctive subcellular compartments involved in ERAD.
Collapse
Affiliation(s)
- Jun Imai
- Correspondence: ; Tel.: +81-27-352-1180
| | | | | |
Collapse
|
32
|
Kotsias F, Cebrian I, Alloatti A. Antigen processing and presentation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:69-121. [PMID: 31810556 DOI: 10.1016/bs.ircmb.2019.07.005] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells are at the center of immune responses. They are defined by their ability to sense the environment, take up and process antigen, migrate to secondary lymphoid organs, where they present antigens to the adaptive immune system. In particular, they present lipids and proteins from pathogens, which they encountered in peripheral tissues, to T cells in order to induce a specific effector immune response. These complex antigens need to be broken down into peptides of a certain length in association with Major Histocompatibility Complex (MHC) molecules. Presentation of MHC/antigen complexes alongside costimulatory molecules and secretion of proinflammatory cytokines will induce an appropriate immune response. This interaction between dendritic cells and T cells takes place at defined locations within secondary lymphoid organs. In this review, we discuss the current knowledge and recent advances on the cellular and molecular mechanisms that underlie antigen processing and the subsequent presentation to T lymphocytes.
Collapse
Affiliation(s)
- Fiorella Kotsias
- Cátedra de Virología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina; Instituto de Investigaciones en Producción Animal (INPA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Ignacio Cebrian
- Facultad de Ciencias Médicas, Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET/Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Andrés Alloatti
- Facultad de Ciencias Médicas, Instituto de Inmunología Clínica y Experimental de Rosario (IDICER)-CONICET/Universidad Nacional de Rosario, Rosario, Argentina.
| |
Collapse
|
33
|
Proteomic analysis of lipopolysaccharide activated human monocytes. Mol Immunol 2018; 103:257-269. [PMID: 30326359 DOI: 10.1016/j.molimm.2018.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/20/2018] [Accepted: 09/25/2018] [Indexed: 12/21/2022]
Abstract
Monocytes are key mediators of innate immunity and comprise an important cellular defence against invading pathogens. However, exaggerated or dysregulated monocyte activation can lead to severe immune-mediated pathology such as sepsis or chronic inflammatory diseases. Thus, detailed insight into the molecular mechanisms of monocyte activation is essential to understand monocyte-driven inflammatory pathologies. We therefore investigated the global protein changes in human monocytes during lipopolysaccharide (LPS) activation to mimic bacterial activation. Purified human monocytes were stimulated with LPS for 17 h and analyzed by state-of-the-art liquid chromatography tandem mass spectrometry (LC-MS/MS). The label-free quantitative proteome analysis identified 2746 quantifiable proteins of which 101 had a statistically significantly different abundance between LPS-stimulated cells and unstimulated controls. Additionally, 143 proteins were exclusively identified in either LPS stimulated cells or unstimulated controls. Functional annotation clustering demonstrated that LPS, most significantly, regulates proteasomal- and lysosomal proteins but in opposite directions. Thus, seven proteasome subunits were upregulated by LPS while 11 lysosomal proteins were downregulated. Both systems are critically involved in processing of proteins for antigen-presentation and together with LPS-induced regulation of CD74 and tapasin, our data suggest that LPS can skew monocytic antigen-presentation towards MHC class I rather than MHC class II. In summary, this study provides a sensitive high throughput protein analysis of LPS-induced monocyte activation and identifies several LPS-regulated proteins not previously described in the literature which can be used as a source for future studies.
Collapse
|
34
|
Sharma R, Dubey S, Mody N, Sharma G, Kushwah V, Jain S, Katare OP, Vyas SP. Release promoter-based systematically designed nanocomposite(s): a novel approach for site-specific delivery of tumor-associated antigen(s) (TAAs). ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:776-789. [DOI: 10.1080/21691401.2018.1469137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Rajeev Sharma
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, India
| | - Surabhi Dubey
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, India
| | - Nishi Mody
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, India
| | - Gajanand Sharma
- University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Studies, Panjab University, Chandigarh, India
| | - Varun Kushwah
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Om Prakash Katare
- University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Studies, Panjab University, Chandigarh, India
| | - Suresh P. Vyas
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, India
| |
Collapse
|
35
|
Liposome-based immunity-inducing systems for cancer immunotherapy. Mol Immunol 2018; 98:8-12. [DOI: 10.1016/j.molimm.2017.11.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/21/2017] [Accepted: 11/03/2017] [Indexed: 12/13/2022]
|
36
|
Roques P, Thiberville SD, Dupuis-Maguiraga L, Lum FM, Labadie K, Martinon F, Gras G, Lebon P, Ng LFP, de Lamballerie X, Le Grand R. Paradoxical Effect of Chloroquine Treatment in Enhancing Chikungunya Virus Infection. Viruses 2018; 10:v10050268. [PMID: 29772762 PMCID: PMC5977261 DOI: 10.3390/v10050268] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/11/2018] [Accepted: 05/12/2018] [Indexed: 01/07/2023] Open
Abstract
Since 2005, Chikungunya virus (CHIKV) re-emerged and caused numerous outbreaks in the world, and finally, was introduced into the Americas in 2013. The lack of CHIKV-specific therapies has led to the use of non-specific drugs. Chloroquine, which is commonly used to treat febrile illnesses in the tropics, has been shown to inhibit CHIKV replication in vitro. To assess the in vivo effect of chloroquine, two complementary studies were performed: (i) a prophylactic study in a non-human primate model (NHP); and (ii) a curative study "CuraChik", which was performed during the Reunion Island outbreak in 2006 in a human cohort. Clinical, biological, and immunological data were compared between treated and placebo groups. Acute CHIKV infection was exacerbated in NHPs treated with prophylactic administration of chloroquine. These NHPs displayed a higher viremia and slower viral clearance (p < 0.003). Magnitude of viremia was correlated to the type I IFN response (Rho = 0.8, p < 0.001) and severe lymphopenia (Rho = 0.8, p < 0.0001), while treatment led to a delay in both CHIKV-specific cellular and IgM responses (p < 0.02 and p = 0.04, respectively). In humans, chloroquine treatment did not affect viremia or clinical parameters during the acute stage of the disease (D1 to D14), but affected the levels of C-reactive Protein (CRP), IFNα, IL-6, and MCP1 over time (D1 to D16). Importantly, no positive effect could be detected on prevalence of persistent arthralgia at Day 300. Although inhibitory in vitro, chloroquine as a prophylactic treatment in NHPs enhances CHIKV replication and delays cellular and humoral response. In patients, curative chloroquine treatment during the acute phase decreases the levels of key cytokines, and thus may delay adaptive immune responses, as observed in NHPs, without any suppressive effect on peripheral viral load.
Collapse
Affiliation(s)
- Pierre Roques
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), Univ. Paris-Sud⁻INSERM U1184, CEA, 92265 Fontenay-aux-Roses, France.
| | - Simon-Djamel Thiberville
- IRD, INSERM U1207, EHESP French School of Public Health, UMR190, Aix-Marseille University, 13005 Marseille, France.
| | - Laurence Dupuis-Maguiraga
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), Univ. Paris-Sud⁻INSERM U1184, CEA, 92265 Fontenay-aux-Roses, France.
| | - Fok-Moon Lum
- Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis 138648, Singapore.
| | - Karine Labadie
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), Univ. Paris-Sud⁻INSERM U1184, CEA, 92265 Fontenay-aux-Roses, France.
| | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), Univ. Paris-Sud⁻INSERM U1184, CEA, 92265 Fontenay-aux-Roses, France.
| | - Gabriel Gras
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), Univ. Paris-Sud⁻INSERM U1184, CEA, 92265 Fontenay-aux-Roses, France.
| | - Pierre Lebon
- Service de Virologie, AP-HP, Hôpital Cochin, Paris Descartes University, 75014 Paris, France.
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis 138648, Singapore.
| | - Xavier de Lamballerie
- IRD, INSERM U1207, EHESP French School of Public Health, UMR190, Aix-Marseille University, 13005 Marseille, France.
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), Univ. Paris-Sud⁻INSERM U1184, CEA, 92265 Fontenay-aux-Roses, France.
| |
Collapse
|
37
|
Self-Amplifying Replicon RNA Delivery to Dendritic Cells by Cationic Lipids. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 12:118-134. [PMID: 30195751 PMCID: PMC6023837 DOI: 10.1016/j.omtn.2018.04.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 04/29/2018] [Accepted: 04/29/2018] [Indexed: 01/09/2023]
Abstract
Advances in RNA technology during the past two decades have led to the construction of replication-competent RNA, termed replicons, RepRNA, or self-amplifying mRNA, with high potential for vaccine applications. Cytosolic delivery is essential for their translation and self-replication, without infectious progeny generation, providing high levels of antigen expression for inducing humoral and cellular immunity. Synthetic nanoparticle-based delivery vehicles can both protect the RNA molecules and facilitate targeting of dendritic cells—critical for immune defense development. Several cationic lipids were assessed, with RepRNA generated from classical swine fever virus encoding nucleoprotein genes of influenza A virus. The non-cytopathogenic nature of the RNA allowed targeting to dendritic cells without destroying the cells—important for prolonged antigen production and presentation. Certain lipids were more effective at delivery and at promoting translation of RepRNA than others. Selection of particular lipids provided delivery to dendritic cells that resulted in translation, demonstrating that delivery efficiency could not guarantee translation. The observed translation in vitro was reproduced in vivo by inducing immune responses against the encoded influenza virus antigens. Cationic lipid-mediated delivery shows potential for promoting RepRNA vaccine delivery to dendritic cells, particularly when combined with additional delivery elements.
Collapse
|
38
|
Song W, Musetti SN, Huang L. Nanomaterials for cancer immunotherapy. Biomaterials 2017; 148:16-30. [PMID: 28961532 DOI: 10.1016/j.biomaterials.2017.09.017] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 09/07/2017] [Accepted: 09/17/2017] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is quickly growing to be the fourth most important cancer therapy, after surgery, radiation therapy, and chemotherapy. Immunotherapy is the most promising cancer management strategy because it orchestrates the body's own immune system to target and eradicate cancer cells, which may result in durable antitumor responses and reduce metastasis and recurrence more than traditional treatments. Nanomaterials hold great promise in further improving the efficiency of cancer immunotherapy - in many cases, they are even necessary for effective delivery. In this review, we briefly summarize the basic principles of cancer immunotherapy and explain why and where to apply nanomaterials in cancer immunotherapy, with special emphasis on cancer vaccines and tumor microenvironment modulation.
Collapse
Affiliation(s)
- Wantong Song
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Sara N Musetti
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
39
|
Pathangey LB, McCurry DB, Gendler SJ, Dominguez AL, Gorman JE, Pathangey G, Mihalik LA, Dang Y, Disis ML, Cohen PA. Surrogate in vitro activation of innate immunity synergizes with interleukin-7 to unleash rapid antigen-driven outgrowth of CD4+ and CD8+ human peripheral blood T-cells naturally recognizing MUC1, HER2/neu and other tumor-associated antigens. Oncotarget 2017; 8:10785-10808. [PMID: 27974697 PMCID: PMC5355224 DOI: 10.18632/oncotarget.13911] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/23/2016] [Indexed: 01/21/2023] Open
Abstract
Effective adoptive immunotherapy has proved elusive for many types of human cancer, often due to difficulties achieving robust expansion of natural tumor-specific T-cells from peripheral blood. We hypothesized that antigen-driven T-cell expansion might best be triggered in vitro by acute activation of innate immunity to mimic a life-threatening infection. Unfractionated peripheral blood mononuclear cells (PBMC) were subjected to a two-step culture, first synchronizing their exposure to exogenous antigens with aggressive surrogate activation of innate immunity, followed by γ-chain cytokine-modulated T-cell hyperexpansion. Step 1 exposure to GM-CSF plus paired Toll-like receptor agonists (resiquimod and LPS), stimulated abundant IL-12 and IL-23 secretion, as well as upregulated co-stimulatory molecules and CD11c expression within the myeloid (CD33+) subpopulation. Added synthetic long peptides (>20aa) derived from widely expressed oncoproteins (MUC1, HER2/neu and CMVpp65), were reliably presented to CD4+ T-cells and cross-presented to CD8+ T-cells. Both presentation and cross-presentation demonstrated proteasomal and Sec61 dependence that could bypass the endoplasmic reticulum. Step 2 exposure to exogenous IL-7 or IL-7+IL-2 produced selective and sustained expansion of both CD4+ and CD8+ peptide-specific T-cells with a predominant interferon-γ-producing T1-type, as well as the antigen-specific ability to lyse tumor targets. Other γ-chain cytokines and/or combinations were initially proliferogenic, but followed by a contractile phase not observed with IL-7 or IL-7+IL-2. Regulatory T-cells were minimally propagated under these culture conditions. This mechanistically rational culture sequence, effective even for unvaccinated donors, enables rapid preparation of T-cells recognizing tumor-associated antigens expressed by the majority of human cancers, including pancreatic cancers, breast cancers and glioblastomas.
Collapse
Affiliation(s)
- Latha B Pathangey
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Dustin B McCurry
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Sandra J Gendler
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Ana L Dominguez
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Jessica E Gorman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Girish Pathangey
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Laurie A Mihalik
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Yushe Dang
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA, USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA, USA
| | - Peter A Cohen
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
40
|
Cruz FM, Colbert JD, Merino E, Kriegsman BA, Rock KL. The Biology and Underlying Mechanisms of Cross-Presentation of Exogenous Antigens on MHC-I Molecules. Annu Rev Immunol 2017; 35:149-176. [PMID: 28125356 PMCID: PMC5508990 DOI: 10.1146/annurev-immunol-041015-055254] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
To monitor the health of cells, the immune system tasks antigen-presenting cells with gathering antigens from other cells and bringing them to CD8 T cells in the form of peptides bound to MHC-I molecules. Most cells would be unable to perform this function because they use their MHC-I molecules to exclusively present peptides derived from the cell's own proteins. However, the immune system evolved mechanisms for dendritic cells and some other phagocytes to sample and present antigens from the extracellular milieu on MHC-I through a process called cross-presentation. How this important task is accomplished, its role in health and disease, and its potential for exploitation are the subject of this review.
Collapse
Affiliation(s)
- Freidrich M Cruz
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts 01655; , , , ,
| | - Jeff D Colbert
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts 01655; , , , ,
| | - Elena Merino
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts 01655; , , , ,
| | - Barry A Kriegsman
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts 01655; , , , ,
| | - Kenneth L Rock
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts 01655; , , , ,
| |
Collapse
|
41
|
Reuben A, Chung JW, Lapointe R, Santos MM. The hemochromatosis protein HFE 20 years later: An emerging role in antigen presentation and in the immune system. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:218-232. [PMID: 28474781 PMCID: PMC5569368 DOI: 10.1002/iid3.158] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/30/2017] [Accepted: 02/10/2017] [Indexed: 12/13/2022]
Abstract
Introduction Since its discovery, the hemochromatosis protein HFE has been primarily defined by its role in iron metabolism and homeostasis, and its involvement in the genetic disease termed hereditary hemochromatosis (HH). While HH patients are typically afflicted by dysregulated iron levels, many are also affected by several immune defects and increased incidence of autoimmune diseases that have thereby implicated HFE in the immune response. Growing evidence has supported an immunological role for HFE with recent studies describing HFE specifically as it relates to MHC I antigen presentation. Methods/Results Here, we present a comprehensive overview of the relationship between iron metabolism, HFE, and the immune system to better understand the origin and cause of immune defects in HH patients. We further describe the role of HFE in MHC I antigen presentation and its potential to impair autoimmune responses in homeostatic conditions, a mechanism which may be exploited by tumors to evade immune surveillance. Conclusion Overall, this increased understanding of the role of HFE in the immune response sets the stage for better treatment and management of HH and other iron‐related diseases, as well as of the immune defects related to this condition.
Collapse
Affiliation(s)
- Alexandre Reuben
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médicine, Université de Montréal, Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Jacqueline W Chung
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Réjean Lapointe
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médicine, Université de Montréal, Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Manuela M Santos
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médicine, Université de Montréal, Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada
| |
Collapse
|
42
|
Du F, Liu YG, Scott EA. Immunotheranostic Polymersomes Modularly Assembled from Tetrablock and Diblock Copolymers with Oxidation-Responsive Fluorescence. Cell Mol Bioeng 2017; 10:357-370. [PMID: 28989540 DOI: 10.1007/s12195-017-0486-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Intracellular delivery is a key step for many applications in medicine and for investigations into cellular function. This is particularly true for immunotherapy, which often requires controlled delivery of antigen and adjuvants to the cytoplasm of immune cells. Due to the complex responses generated by the stimulation of diverse immune cell populations, it is critical to monitor which cells are targeted during treatment. To address this issue, we have engineered an immunotheranostic polymersome delivery system that fluorescently marks immune cells following intracellular delivery. METHODS N-(3-bromopropyl)phthalimide end-capped poly(ethylene glycol)-bl-poly(propylene sulfide) (PEG-PPS-PI) was synthesized by anionic ring opening polymerization and linked with PEG-PPS-NH2 via a perylene bisimide (PBI) bridge to form a tetrablock copolymer (PEG-PPS-PBI-PPS-PEG). Block copolymers were assembled into polymersomes by thin film hydration in phosphate buffered saline and characterized by dynamic light scattering, cryogenic electron microscopy and fluorescence spectroscopy. Polymersomes were injected subcutaneously into the backs of mice, and draining lymph nodes were extracted for flow cytometric analysis of cellular uptake and disassembly. RESULTS Modular self-assembly of tetrablock / diblock copolymers in aqueous solutions induced π-π stacking of the PBI linker that both red-shifted and quenched the PBI fluorescence. Reactive oxygen species within the endosomes of phagocytic immune cell populations oxidized the PPS blocks, which disassembled the polymersomes for dequenching and shifting of the PBI fluorescence from 640 nm to 550 nm emission. Lymph node resident macrophages and dendritic cells were found to increase in 550 nm emission over the course of 3 days by flow cytometry. CONCLUSIONS Immunotheranostic polymersomes present a versatile platform to probe the contributions of specific cell populations during the elicitation of controlled immune responses. Flanking PBI with two oxidation-sensitive hydrophobic PPS blocks enhanced π stacking and introduced a mechanism for disrupting π-π interactions to shift PBI fluorescence in response to oxidative conditions. Shifts from red (640 nm) to green (550 nm) fluorescence occurred in the presence of physiologically relevant concentrations of reactive oxygen species and could be observed within phagocytic cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Fanfan Du
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA.,Simpson Querrey Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yu-Gang Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA
| | - Evan Alexander Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA.,Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA.,Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois, USA.,Simpson Querrey Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
43
|
Kroger CJ, Wang B, Tisch R. Temporal increase in thymocyte negative selection parallels enhanced thymic SIRPα + DC function. Eur J Immunol 2016; 46:2352-2362. [PMID: 27501268 DOI: 10.1002/eji.201646354] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/24/2016] [Accepted: 08/04/2016] [Indexed: 01/24/2023]
Abstract
Dysregulation of negative selection contributes to T-cell-mediated autoimmunity, such as type 1 diabetes. The events regulating thymic negative selection, however, are ill defined. Work by our group and others suggest that negative selection is inefficient early in ontogeny and increases with age. This study examines temporal changes in negative selection and the thymic DC compartment. Peptide-induced thymocyte deletion in vivo was reduced in newborn versus 4-week-old NOD mice, despite a similar sensitivity of the respective thymocytes to apoptosis induction. The temporal increase in negative selection corresponded with an elevated capacity of thymic antigen-presenting cells to stimulate T cells, along with altered subset composition and function of resident DC. The frequency of signal regulatory protein α+ (SIRPα+ ) and plasmacytoid DCs was increased concomitant with a decrease in CD8α+ DC in 4-week-old NOD thymi. Importantly, 4-week-old versus newborn thymic SIRPα+ DC exhibited increased antigen processing and presentation via the MHC class II but not class I pathway, coupled with an enhanced T-cell stimulatory capacity not seen in thymic plasmacytoid DC and CD8α+ DC. These findings indicate that the efficiency of thymic DC-mediated negative selection is limited early after birth, and increases with age paralleling expansion of functionally superior thymic SIRPα+ DC.
Collapse
Affiliation(s)
- Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Bo Wang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Roland Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
44
|
Kourjian G, Rucevic M, Berberich MJ, Dinter J, Wambua D, Boucau J, Le Gall S. HIV Protease Inhibitor-Induced Cathepsin Modulation Alters Antigen Processing and Cross-Presentation. THE JOURNAL OF IMMUNOLOGY 2016; 196:3595-607. [PMID: 27009491 DOI: 10.4049/jimmunol.1600055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/01/2016] [Indexed: 02/06/2023]
Abstract
Immune recognition by T cells relies on the presentation of pathogen-derived peptides by infected cells, but the persistence of chronic infections calls for new approaches to modulate immune recognition. Ag cross-presentation, the process by which pathogen Ags are internalized, degraded, and presented by MHC class I, is crucial to prime CD8 T cell responses. The original degradation of Ags is performed by pH-dependent endolysosomal cathepsins. In this article, we show that HIV protease inhibitors (PIs) prescribed to HIV-infected persons variably modulate cathepsin activities in human APCs, dendritic cells and macrophages, and CD4 T cells, three cell subsets infected by HIV. Two HIV PIs acted in two complementary ways on cathepsin hydrolytic activities: directly on cathepsins and indirectly on their regulators by inhibiting Akt kinase activities, reducing NADPH oxidase 2 activation, and lowering phagolysosomal reactive oxygen species production and pH, which led to enhanced cathepsin activities. HIV PIs modified endolysosomal degradation and epitope production of proteins from HIV and other pathogens in a sequence-dependent manner. They altered cross-presentation of Ags by dendritic cells to epitope-specific T cells and T cell-mediated killing. HIV PI-induced modulation of Ag processing partly changed the MHC self-peptidome displayed by primary human cells. This first identification, to our knowledge, of prescription drugs modifying the regulation of cathepsin activities and the MHC-peptidome may provide an alternate therapeutic approach to modulate immune recognition in immune disease beyond HIV.
Collapse
Affiliation(s)
| | | | | | - Jens Dinter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - Daniel Wambua
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - Julie Boucau
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - Sylvie Le Gall
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| |
Collapse
|
45
|
Targeting dendritic cells: a promising strategy to improve vaccine effectiveness. Clin Transl Immunology 2016; 5:e66. [PMID: 27217957 PMCID: PMC4815026 DOI: 10.1038/cti.2016.6] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell (DC) targeting is a novel strategy to enhance vaccination efficacy. This approach is based on the in situ delivery of antigen via antibodies that are specific for endocytic receptors expressed at the surface of DCs. Here we review the complexity of the DC subsets and the antigen presentation pathways that need to be considered in the settings of DC targeting. We also summarize current knowledge about antigen delivery to DCs via DEC-205, Clec9A and Clec12A, receptor targets that strongly enhance cellular and humoral immune responses. Finally, we discuss the intracellular trafficking criteria of the targeted receptors that may impact their effectiveness as DC targets.
Collapse
|
46
|
Functional Specialty of CD40 and Dendritic Cell Surface Lectins for Exogenous Antigen Presentation to CD8(+) and CD4(+) T Cells. EBioMedicine 2016; 5:46-58. [PMID: 27077111 PMCID: PMC4816850 DOI: 10.1016/j.ebiom.2016.01.029] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 11/25/2022] Open
Abstract
Dendritic cells (DCs) are major antigen-presenting cells that can efficiently prime and cross-prime antigen-specific T cells. Delivering antigen to DCs via surface receptors is thus an appealing strategy to evoke cellular immunity. Nonetheless, which DC surface receptor to target to yield the optimal CD8+ and CD4+ T cell responses remains elusive. Herein, we report the superiority of CD40 over 9 different lectins and scavenger receptors at evoking antigen-specific CD8+ T cell responses. However, lectins (e.g., LOX-1 and Dectin-1) were more efficient than CD40 at eliciting CD4+ T cell responses. Common and distinct patterns of subcellular and intracellular localization of receptor-bound αCD40, αLOX-1 and αDectin-1 further support their functional specialization at enhancing antigen presentation to either CD8+ or CD4+ T cells. Lastly, we demonstrate that antigen targeting to CD40 can evoke potent antigen-specific CD8+ T cell responses in human CD40 transgenic mice. This study provides fundamental information for the rational design of vaccines against cancers and viral infections. Antigen delivery to DCs via CD40 is more efficient than through nine other receptors at eliciting CD8 T+ cell response. Antigen delivery via lectins (e.g., LOX-1 and Dectin-1) is more efficient than CD40 at eliciting CD4+ T cell responses.
The success of an immunotherapeutic vaccine for cancer is largely dependent on its ability to evoke potent cellular immunity. Although targeting antigens to dendritic cells (DCs) has been known to be an efficient strategy to evoke cellular immunity, which targeted receptors yield the optimal cellular immunity remained elusive. We report that targeting CD40, compared to 9 other DC receptors, results in the greatest levels of CD8+ cytotoxic T cell responses, while targeting lectins results in enhanced CD4+ helper T cell responses. The findings of this study will assist us in the rational design of immunotherapeutic vaccines against cancers.
Collapse
Key Words
- ANOVA, analysis of variance
- AP, alkaline phosphatase
- APC, antigen-presenting cells
- CD, cluster of differentiation
- CD40
- CFSE, carboxyfluorescein succinimidyl ester
- CTL, cytotoxic T lymphocyte
- Coh, cohesin
- Cross-presentation
- DC, dendritic cell
- Dendritic cell
- Doc, dockerin
- EEA1, early endosome antigen 1
- ELISA, enzyme-linked immunosorbent assay
- ELISpot, enzyme-linked immunospot
- Flu.M1, influenza virus matrix protein 1
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HA1, hemagglutinin subunit 1
- HLA, human leukocyte antigen
- HPV, human papillomavirus
- HRP, horseradish peroxidase
- IFN, interferon
- IL, interleukin
- JaCoP, Just another Colocalization Plugin
- LAMP-1, lysosomal-associated membrane protein 1
- Lectins
- MART-1, melanoma antigen recognized by T cells 1
- MHC, major histocompatibility complex
- Mo-DC, monocyte-derived dendritic cell
- NHP, non-human primate
- NP, nucleoprotein
- PBMC, peripheral blood mononuclear cells
- PBS, phosphate-buffered saline
- PSA, prostate specific antigen
- Poly(I:C), polyinosinic:polycytidylic acid
- TLR, toll-like receptor
- TMB, 3,3′,5,5′-tetramethylbenzidine
- TNF, tumor necrosis factor
- Vaccine
- hCD40Tg, human CD40 transgenic
- i.p., intraperitoneal(ly)
- mAb, monoclonal antibody
- mDC, myeloid dendritic cell
- pDC, plasmacytoid dendritic cell
- s.c., subcutaneous(ly)
Collapse
|
47
|
Abstract
The spondyloarthropathies comprise ankylosing spondylitis (AS), reactive arthritis, psoriatic arthritis (PsA) and arthritis associated with inflammatory bowel disease. In this Perspectives article, we describe how Behçet disease and several clinically distinct spondyloarthropathies-all associated with MHC class I (MHC-I) alleles such as HLA-B(*)51, HLA-C(*)0602 and HLA-B(*)27 and epistatic ERAP-1 interactions-have a shared immunopathogenetic basis. As a unifying concept, we propose that barrier dysfunction in environmentally exposed organs such as the skin, and aberrant innate immune reactions at sites of mechanical stress, can often trigger secondary adaptive immune CD8(+) T-cell responses with prominent neutrophilic inflammation that culminate in exacerbation and recurrence of these diseases. Of note, these 'MHC-I-opathies' show a differential immunopathology, probably reflecting antigenic differences within target tissues: HLA-B(*)51 is linked to ocular and mucocutaneous disease but not gut involvement, and HLA-C(*)0602 is linked to type I psoriasis but not scalp or nail disease.
Collapse
|
48
|
Gutiérrez-Martínez E, Planès R, Anselmi G, Reynolds M, Menezes S, Adiko AC, Saveanu L, Guermonprez P. Cross-Presentation of Cell-Associated Antigens by MHC Class I in Dendritic Cell Subsets. Front Immunol 2015; 6:363. [PMID: 26236315 PMCID: PMC4505393 DOI: 10.3389/fimmu.2015.00363] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/05/2015] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) have the unique ability to pick up dead cells carrying antigens in tissue and migrate to the lymph nodes where they can cross-present cell-associated antigens by MHC class I to CD8+ T cells. There is strong in vivo evidence that the mouse XCR1+ DCs subset acts as a key player in this process. The intracellular processes underlying cross-presentation remain controversial and several pathways have been proposed. Indeed, a wide number of studies have addressed the cellular process of cross-presentation in vitro using a variety of sources of antigen and antigen-presenting cells. Here, we review the in vivo and in vitro evidence supporting the current mechanistic models and disscuss their physiological relevance to the cross-presentation of cell-associated antigens by DCs subsets.
Collapse
Affiliation(s)
- Enric Gutiérrez-Martínez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Remi Planès
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Giorgio Anselmi
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Matthew Reynolds
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Shinelle Menezes
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Aimé Cézaire Adiko
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Loredana Saveanu
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Pierre Guermonprez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| |
Collapse
|
49
|
Allan ERO, Yates RM. Redundancy between Cysteine Cathepsins in Murine Experimental Autoimmune Encephalomyelitis. PLoS One 2015; 10:e0128945. [PMID: 26075905 PMCID: PMC4468166 DOI: 10.1371/journal.pone.0128945] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 05/01/2015] [Indexed: 11/18/2022] Open
Abstract
The cysteine cathepsins B, S, and L are functionally linked to antigen processing, and hence to autoimmune disorders such as multiple sclerosis. Stemming from several studies that demonstrate that mice can be protected from experimental autoimmune encephalomyelitis (EAE) through the pharmacologic inhibition of cysteine cathepsins, it has been suggested that targeting these enzymes in multiple sclerosis may be of therapeutic benefit. Utilizing mice deficient in cysteine cathepsins both individually and in combination, we found that the myelin-associated antigen myelin oligodendrocyte glycoprotein (MOG) was efficiently processed and presented by macrophages to CD4+ T cells in the individual absence of cathepsin B, S or L. Similarly, mice deficient in cathepsin B or S were susceptible to MOG-induced EAE and displayed clinical progression and immune infiltration into the CNS, similar to their wild-type counterparts. Owing to a previously described CD4+ T cell deficiency in mice deficient in cathepsin L, such mice were protected from EAE. When multiple cysteine cathepsins were simultaneously inhibited via genetic deletion of both cathepsins B and S, or by a cathepsin inhibitor (LHVS), MHC-II surface expression, MOG antigen presentation and EAE were attenuated or prevented. This study demonstrates the functional redundancy between cathepsin B, S and L in EAE, and suggests that the inhibition of multiple cysteine cathepsins may be needed to modulate autoimmune disorders such as multiple sclerosis.
Collapse
Affiliation(s)
- Euan Ramsay Orr Allan
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Alberta, Canada
| | - Robin Michael Yates
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
50
|
Garcia-Castillo MD, Tran T, Bobard A, Renard HF, Rathjen SJ, Dransart E, Stechmann B, Lamaze C, Lord M, Cintrat JC, Enninga J, Tartour E, Johannes L. Retrograde transport is not required for cytosolic translocation of the B-subunit of Shiga toxin. J Cell Sci 2015; 128:2373-87. [PMID: 25977475 DOI: 10.1242/jcs.169383] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/07/2015] [Indexed: 01/13/2023] Open
Abstract
Antigen-presenting cells have the remarkable capacity to transfer exogenous antigens to the cytosol for processing by proteasomes and subsequent presentation on major histocompatibility complex class-I (MHC-I) molecules, a process termed cross-presentation. This is the target of biomedical approaches that aim to trigger a therapeutic immune response. The receptor-binding B-subunit of Shiga toxin (STxB) has been developed as an antigen delivery tool for such immunotherapy applications. In this study, we have analyzed pathways and trafficking factors that are involved in this process. A covalent conjugate between STxB and saporin was generated to quantitatively sample the membrane translocation step to the cytosol in differentiated monocyte-derived THP-1 cells. We have found that retrograde trafficking to the Golgi complex was not required for STxB-saporin translocation to the cytosol or for STxB-dependent antigen cross-presentation. Depletion of endosomal Rab7 inhibited, and lowering membrane cholesterol levels favored STxB-saporin translocation. Interestingly, experiments with reducible and non-reducible linker-arm-STxB conjugates led to the conclusion that after translocation, STxB remains associated with the cytosolic membrane leaflet. In summary, we report new facets of the endosomal escape process bearing relevance to antigen cross-presentation.
Collapse
Affiliation(s)
- Maria Daniela Garcia-Castillo
- Institut Curie, PSL Research University, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris Cedex 05 75248, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| | - Thi Tran
- INSERM U970, PARCC Université Paris Descartes Sorbonne Paris Cité, Paris 75006, France Hôpital Européen Georges-Pompidou, AP-HP, Service d'Immunologie Biologique, Paris Cedex 15 75908, France
| | - Alexandre Bobard
- Dynamique des Interactions Hôte Pathogène, Institut Pasteur, Paris Cedex 15 75724, France
| | - Henri-François Renard
- Institut Curie, PSL Research University, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris Cedex 05 75248, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| | - Stefan J Rathjen
- Institut Curie, PSL Research University, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris Cedex 05 75248, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| | - Estelle Dransart
- Institut Curie, PSL Research University, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris Cedex 05 75248, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| | - Bahne Stechmann
- Institut Curie, PSL Research University, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris Cedex 05 75248, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| | - Christophe Lamaze
- CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France Institut Curie - Centre de Recherche, Membrane Dynamics and Mechanics of Intracellular Signaling Group, 26 rue d'Ulm, Paris Cedex 05 75248, France
| | - Mike Lord
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | | | - Jost Enninga
- Dynamique des Interactions Hôte Pathogène, Institut Pasteur, Paris Cedex 15 75724, France
| | - Eric Tartour
- INSERM U970, PARCC Université Paris Descartes Sorbonne Paris Cité, Paris 75006, France Hôpital Européen Georges-Pompidou, AP-HP, Service d'Immunologie Biologique, Paris Cedex 15 75908, France
| | - Ludger Johannes
- Institut Curie, PSL Research University, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris Cedex 05 75248, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| |
Collapse
|