1
|
Karan S, Durán-Meza AL, Chapman A, Tanimoto C, Chan SK, Knobler CM, Gelbart WM, Steinmetz NF. In Vivo Delivery of Spherical and Cylindrical In Vitro Reconstituted Virus-like Particles Containing the Same Self-Amplifying mRNA. Mol Pharm 2024; 21:2727-2739. [PMID: 38709860 PMCID: PMC11250921 DOI: 10.1021/acs.molpharmaceut.3c01105] [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] [Indexed: 05/08/2024]
Abstract
The dramatic effectiveness of recent mRNA (mRNA)-based COVID vaccines delivered in lipid nanoparticles has highlighted the promise of mRNA therapeutics in general. In this report, we extend our earlier work on self-amplifying mRNAs delivered in spherical in vitro reconstituted virus-like particles (VLPs), and on drug delivery using cylindrical virus particles. In particular, we carry out separate in vitro assemblies of a self-amplifying mRNA gene in two different virus-like particles: one spherical, formed with the capsid protein of cowpea chlorotic mottle virus (CCMV), and the other cylindrical, formed from the capsid protein of tobacco mosaic virus (TMV). The mRNA gene is rendered self-amplifying by genetically fusing it to the RNA-dependent RNA polymerase (RdRp) of Nodamura virus, and the relative efficacies of cell uptake and downstream protein expression resulting from their CCMV- and TMV-packaged forms are compared directly. This comparison is carried out by their transfections into cells in culture: expressions of two self-amplifying genes, enhanced yellow fluorescent protein (EYFP) and Renilla luciferase (Luc), packaged alternately in CCMV and TMV VLPs, are quantified by fluorescence and chemiluminescence levels, respectively, and relative numbers of the delivered mRNAs are measured by quantitative real-time PCR. The cellular uptake of both forms of these VLPs is further confirmed by confocal microscopy of transfected cells. Finally, VLP-mediated delivery of the self-amplifying-mRNA in mice following footpad injection is shown by in vivo fluorescence imaging to result in robust expression of EYFP in the draining lymph nodes, suggesting the potential of these plant virus-like particles as a promising mRNA gene and vaccine delivery modality. These results establish that both CCMV and TMV VLPs can deliver their in vitro packaged mRNA genes to immune cells and that their self-amplifying forms significantly enhance in situ expression. Choice of one VLP (CCMV or TMV) over the other will depend on which geometry of nucleocapsid is self-assembled more efficiently for a given length and sequence of RNA, and suggests that these plant VLP gene delivery systems will prove useful in a wide variety of medical applications, both preventive and therapeutic.
Collapse
Affiliation(s)
- Sweta Karan
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Ana Luisa Durán-Meza
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Abigail Chapman
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Cheylene Tanimoto
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Soo Khim Chan
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
| | - Charles M Knobler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - William M Gelbart
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- UCLA Molecular Biology Institute, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Institute for Materials Discovery and Design, University of California San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Radiology, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
- Center for Engineering in Cancer, Institute for Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
2
|
Kwon S, Kwon M, Im S, Lee K, Lee H. mRNA vaccines: the most recent clinical applications of synthetic mRNA. Arch Pharm Res 2022; 45:245-262. [PMID: 35426547 PMCID: PMC9012156 DOI: 10.1007/s12272-022-01381-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/05/2022] [Indexed: 12/24/2022]
Abstract
Synthetic mRNA has been considered as an emerging biotherapeutic agent for the past decades. Recently, the SARS-CoV-2 pandemic has led to the first clinical use of synthetic mRNA. mRNA vaccines showed far surpassing influences on the public as compared to other vaccine platforms such as viral vector vaccines and recombinant protein vaccines. It allowed rapid development and production of vaccines that have never been achieved in history. Synthetic mRNA, called in vitro transcribed (IVT) mRNA, is the key component of mRNA vaccines. It has several advantages over conventional gene-expressing systems such as plasmid DNA and viral vectors. It can translate proteins in the cytoplasm by structurally resembling natural mRNA and exhibit various protein expression patterns depending on how it is engineered. Another advantage is that synthetic mRNA enables fast, scalable, and cost-effective production. Therefore, starting with the mRNA vaccine, synthetic mRNA is now in the spotlight as a promising new drug development agent. In this review, we will summarize the latest IVT mRNA technology such as new mRNA structures or large-scale production. In addition, the nature of the innate immunogenicity of IVT mRNA will be discussed along with its roles in the development of vaccines. Finally, the principles of the mRNA vaccine and the future direction of synthetic mRNA will be provided.
Collapse
Affiliation(s)
- Suji Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Minseon Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seongeun Im
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Kyuri Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
3
|
Sánchez-Álvarez E, Quiroga B, de Sequera P. Position statement of the Spanish Society of Nephrology on the SARS-CoV-2 vaccines. Nefrologia 2021; 41:412-416. [PMID: 34561208 PMCID: PMC8455244 DOI: 10.1016/j.nefroe.2021.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 11/17/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread worldwide over the last year causing more than one million deaths. Several treatments have tried to modify the natural history of the coronavirus disease 2019 (COVID-19) but only corticosteroids have demonstrated to be effective in moderate or severe affectation. In that situation, the development of vaccines for preventing the SARS-CoV-2 infection has focused the attention of the scientific community. At present, available messenger RNA-based technology vaccines have received the approval of local and international sanitary authorities. In this position statement, the Spanish Society of Nephrology wants to state that patients with chronic kidney disease and healthcare workers are at high-risk for contagion and complications of COVID-19 so they must have priority in the vaccine administration.
Collapse
Affiliation(s)
- Emilio Sánchez-Álvarez
- Servicio de Nefrología, Hospital Universitario de Cabueñes, Gijón, Spain; Junta Directiva de la Sociedad Española de Nefrología, Spain
| | - Borja Quiroga
- Servicio de Nefrología, Hospital Universitario de La Princesa, Madrid, Spain; Junta Directiva de la Sociedad Española de Nefrología, Spain.
| | - Patricia de Sequera
- Servicio de Nefrología, Hospital Universitario Infanta Leonor, Madrid, Spain; Junta Directiva de la Sociedad Española de Nefrología, Spain
| |
Collapse
|
4
|
Sánchez-Álvarez E, Quiroga B, de Sequera P. [Position statement of the Spanish Society of Nephrology on the SARS-CoV-2 vaccines]. Nefrologia 2021; 41:412-416. [PMID: 38620386 PMCID: PMC7775792 DOI: 10.1016/j.nefro.2020.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/17/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread worldwide over the last year causing more than one million deaths. Several treatments have tried to modify the natural history of the coronavirus disease 2019 (COVID-19) but only corticosteroids have demonstrated to be effective in moderate or severe affectation. In that situation, the development of vaccines for preventing the SARS-CoV-2 infection has focused the attention of the scientific community. At present, available messenger RNA-based technology vaccines have received the approval of local and international sanitary authorities. In this position statement, the Spanish Society of Nephrology wants to state that patients with chronic kidney disease and healthcare workers are at high-risk for contagion and complications of COVID-19 so they must have priority in the vaccine administration.
Collapse
Affiliation(s)
- Emilio Sánchez-Álvarez
- Servicio de Nefrología, Hospital Universitario de Cabueñes, Gijón, España
- Junta Directiva de la Sociedad Española de Nefrología
| | - Borja Quiroga
- Servicio de Nefrología, Hospital Universitario de La Princesa, Madrid, España
- Junta Directiva de la Sociedad Española de Nefrología
| | - Patricia de Sequera
- Servicio de Nefrología, Hospital Universitario Infanta Leonor, Madrid, España
- Junta Directiva de la Sociedad Española de Nefrología
| |
Collapse
|
5
|
Lai X, Dreyer FS, Cantone M, Eberhardt M, Gerer KF, Jaitly T, Uebe S, Lischer C, Ekici A, Wittmann J, Jäck HM, Schaft N, Dörrie J, Vera J. Network- and systems-based re-engineering of dendritic cells with non-coding RNAs for cancer immunotherapy. Theranostics 2021; 11:1412-1428. [PMID: 33391542 PMCID: PMC7738891 DOI: 10.7150/thno.53092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that induce and regulate adaptive immunity by presenting antigens to T cells. Due to their coordinative role in adaptive immune responses, DCs have been used as cell-based therapeutic vaccination against cancer. The capacity of DCs to induce a therapeutic immune response can be enhanced by re-wiring of cellular signalling pathways with microRNAs (miRNAs). Methods: Since the activation and maturation of DCs is controlled by an interconnected signalling network, we deploy an approach that combines RNA sequencing data and systems biology methods to delineate miRNA-based strategies that enhance DC-elicited immune responses. Results: Through RNA sequencing of IKKβ-matured DCs that are currently being tested in a clinical trial on therapeutic anti-cancer vaccination, we identified 44 differentially expressed miRNAs. According to a network analysis, most of these miRNAs regulate targets that are linked to immune pathways, such as cytokine and interleukin signalling. We employed a network topology-oriented scoring model to rank the miRNAs, analysed their impact on immunogenic potency of DCs, and identified dozens of promising miRNA candidates, with miR-15a and miR-16 as the top ones. The results of our analysis are presented in a database that constitutes a tool to identify DC-relevant miRNA-gene interactions with therapeutic potential (https://www.synmirapy.net/dc-optimization). Conclusions: Our approach enables the systematic analysis and identification of functional miRNA-gene interactions that can be experimentally tested for improving DC immunogenic potency.
Collapse
Affiliation(s)
- Xin Lai
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| | - Florian S. Dreyer
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| | - Martina Cantone
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| | - Martin Eberhardt
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| | - Kerstin F. Gerer
- RNA Group, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| | - Tanushree Jaitly
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| | - Steffen Uebe
- Department of Human Genetics, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christopher Lischer
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| | - Arif Ekici
- Department of Human Genetics, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jürgen Wittmann
- Division of Molecular Immunology, Department of Medicine 3, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Medicine 3, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Niels Schaft
- RNA Group, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| | - Jan Dörrie
- RNA Group, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| | - Julio Vera
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center (CCC) Erlangen, Erlangen, Germany
| |
Collapse
|
6
|
Broos K, Van der Jeught K, Puttemans J, Goyvaerts C, Heirman C, Dewitte H, Verbeke R, Lentacker I, Thielemans K, Breckpot K. Particle-mediated Intravenous Delivery of Antigen mRNA Results in Strong Antigen-specific T-cell Responses Despite the Induction of Type I Interferon. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e326. [PMID: 27327138 PMCID: PMC5022130 DOI: 10.1038/mtna.2016.38] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/26/2016] [Indexed: 12/25/2022]
Abstract
Cancer vaccines based on mRNA are extensively studied. The fragile nature of mRNA has instigated research into carriers that can protect it from ribonucleases and as such enable its systemic use. However, carrier-mediated delivery of mRNA has been linked to production of type I interferon (IFN) that was reported to compromise the effectiveness of mRNA vaccines. In this study, we evaluated a cationic lipid for encapsulation of mRNA. The nanometer-sized, negatively charged lipid mRNA particles (LMPs) efficiently transfected dendritic cells and macrophages in vitro. Furthermore, i.v. delivery of LMPs resulted in rapid expression of the mRNA-encoded protein in spleen and liver, predominantly in CD11c(+) cells and to a minor extent in CD11b(+) cells. Intravenous immunization of mice with LMPs containing ovalbumin, human papilloma virus E7, and tyrosinase-related protein-2 mRNA, either combined or separately, elicited strong antigen-specific T-cell responses. We further showed the production of type I IFNs upon i.v. LMP delivery. Although this decreased the expression of the mRNA-encoded protein, it supported the induction of antigen-specific T-cell responses. These data question the current notion that type I IFNs hamper particle-mediated mRNA vaccines.
Collapse
Affiliation(s)
- Katrijn Broos
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kevin Van der Jeught
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Janik Puttemans
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Heleen Dewitte
- Laboratory for General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, Belgium
| | - Rein Verbeke
- Laboratory for General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, Belgium
| | - Ine Lentacker
- Laboratory for General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
7
|
Electroporated Antigen-Encoding mRNA Is Not a Danger Signal to Human Mature Monocyte-Derived Dendritic Cells. J Immunol Res 2015; 2015:952184. [PMID: 26824052 PMCID: PMC4707322 DOI: 10.1155/2015/952184] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/01/2015] [Indexed: 11/25/2022] Open
Abstract
For therapeutic cancer vaccination, the adoptive transfer of mRNA-electroporated dendritic cells (DCs) is frequently performed, usually with monocyte-derived, cytokine-matured DCs (moDCs). However, DCs are rich in danger-sensing receptors which could recognize the exogenously delivered mRNA and induce DC activation, hence influencing the DCs' immunogenicity. Therefore, we examined whether electroporation of mRNA with a proper cap and a poly-A tail of at least 64 adenosines had any influence on cocktail-matured moDCs. We used 16 different RNAs, encoding tumor antigens (MelanA, NRAS, BRAF, GNAQ, GNA11, and WT1), and variants thereof. None of those RNAs induced changes in the expression of CD25, CD40, CD83, CD86, and CD70 or the secretion of the cytokines IL-8, IL-6, and TNFα of more than 1.5-fold compared to the control condition, while an mRNA encoding an NF-κB-activation protein as positive control induced massive secretion of the cytokines. To determine whether mRNA electroporation had any effect on the whole transcriptome of the DCs, we performed microarray analyses of DCs of 6 different donors. None of 60,000 probes was significantly different between mock-electroporated DCs and MelanA-transfected DCs. Hence, we conclude that no transcriptional programs were induced within cocktail-matured DCs by electroporation of single tumor-antigen-encoding mRNAs.
Collapse
|
8
|
McCullough KC, Milona P, Thomann-Harwood L, Démoulins T, Englezou P, Suter R, Ruggli N. Self-Amplifying Replicon RNA Vaccine Delivery to Dendritic Cells by Synthetic Nanoparticles. Vaccines (Basel) 2014; 2:735-54. [PMID: 26344889 PMCID: PMC4494254 DOI: 10.3390/vaccines2040735] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/29/2014] [Accepted: 09/28/2014] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC) play essential roles determining efficacy of vaccine delivery with respect to immune defence development and regulation. This renders DCs important targets for vaccine delivery, particularly RNA vaccines. While delivery of interfering RNA oligonucleotides to the appropriate intracellular sites for RNA-interference has proven successful, the methodologies are identical for RNA vaccines, which require delivery to RNA translation sites. Delivery of mRNA has benefitted from application of cationic entities; these offer value following endocytosis of RNA, when cationic or amphipathic properties can promote endocytic vesicle membrane perturbation to facilitate cytosolic translocation. The present review presents how such advances are being applied to the delivery of a new form of RNA vaccine, replicons (RepRNA) carrying inserted foreign genes of interest encoding vaccine antigens. Approaches have been developed for delivery to DCs, leading to the translation of the RepRNA and encoded vaccine antigens both in vitro and in vivo. Potential mechanisms favouring efficient delivery leading to translation are discussed with respect to the DC endocytic machinery, showing the importance of cytosolic translocation from acidifying endocytic structures. The review relates the DC endocytic pathways to immune response induction, and the potential advantages for these self-replicating RNA vaccines in the near future.
Collapse
Affiliation(s)
| | - Panagiota Milona
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | | | - Thomas Démoulins
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | - Pavlos Englezou
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | - Rolf Suter
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | - Nicolas Ruggli
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| |
Collapse
|
9
|
McCullough KC, Bassi I, Milona P, Suter R, Thomann-Harwood L, Englezou P, Démoulins T, Ruggli N. Self-replicating Replicon-RNA Delivery to Dendritic Cells by Chitosan-nanoparticles for Translation In Vitro and In Vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e173. [PMID: 25004099 PMCID: PMC4121514 DOI: 10.1038/mtna.2014.24] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 05/20/2014] [Indexed: 02/06/2023]
Abstract
Self-amplifying replicon RNA (RepRNA) possesses high potential for increasing antigen load within dendritic cells (DCs). The major aim of the present work was to define how RepRNA delivered by biodegradable, chitosan-based nanoparticulate delivery vehicles (nanogel-alginate (NGA)) interacts with DCs, and whether this could lead to translation of the RepRNA in the DCs. Although studies employed virus replicon particles (VRPs), there are no reports on biodegradable, nanoparticulate vehicle delivery of RepRNA. VRP studies employed cytopathogenic agents, contrary to DC requirements—slow processing and antigen retention. We employed noncytopathogenic RepRNA with NGA, demonstrating for the first time the efficiency of RepRNA association with nanoparticles, NGA delivery to DCs, and RepRNA internalization by DCs. RepRNA accumulated in vesicular structures, with patterns typifying cytosolic release. This promoted RepRNA translation, in vitro and in vivo. Delivery and translation were RepRNA concentration-dependent, occurring in a kinetic manner. Including cationic lipids with chitosan during nanoparticle formation enhanced delivery and translation kinetics, but was not required for translation of immunogenic levels in vivo. This work describes for the first time the characteristics associated with chitosan-nanoparticle delivery of self-amplifying RepRNA to DCs, leading to translation of encoded foreign genes, namely influenza virus hemagglutinin and nucleoprotein.
Collapse
Affiliation(s)
| | - Isabelle Bassi
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Panagiota Milona
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Rolf Suter
- 1] Institute of Virology and Immunology, Mittelhäusern, Switzerland [2] Current address: Laboratory of Experimental Biophysics, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Pavlos Englezou
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Thomas Démoulins
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Nicolas Ruggli
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| |
Collapse
|
10
|
Pollard C, De Koker S, Saelens X, Vanham G, Grooten J. Challenges and advances towards the rational design of mRNA vaccines. Trends Mol Med 2013; 19:705-13. [PMID: 24138818 DOI: 10.1016/j.molmed.2013.09.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 09/12/2013] [Accepted: 09/17/2013] [Indexed: 01/01/2023]
Abstract
In recent years, mRNA vaccines have emerged as a safe and potent approach for the induction of cellular immune responses. Whereas initial studies were limited to the ex vivo loading of dendritic cells (DCs) with antigen-encoding mRNA, recent progress has led to the development of improved mRNA vaccines that enable direct in vivo targeting of DCs. Although preclinical studies demonstrated their potency in inducing antitumor immunity, several bottlenecks hinder the broader application of mRNA vaccines. In this review, we discuss the challenges associated with mRNA-based vaccination strategies, the technological advances that have been made to overcome these limitations, and the hurdles that remain to be tackled for the development of an optimal mRNA vaccine.
Collapse
Affiliation(s)
- Charlotte Pollard
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium; Laboratory of Molecular Immunology, Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.
| | | | | | | | | |
Collapse
|
11
|
Functional RNA delivery targeted to dendritic cells by synthetic nanoparticles. Ther Deliv 2012; 3:1077-99. [DOI: 10.4155/tde.12.90] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dendritic cells (DCs) are essential to many aspects of immune defense development and regulation. They provide important targets for prophylactic and therapeutic delivery. While protein delivery has had considerable success, RNA delivery is still expanding. Delivering RNA molecules for RNAi has shown particular success and there are reports on successful delivery of mRNA. Central, therein, is the application of cationic entities. Following endocytosis of the delivery vehicle for the RNA, cationic entities should promote vesicular membrane perturbation, facilitating cytosolic release. The present review explains the diversity of DC function in immune response development and control. Promotion of delivered RNA cytosolic release is discussed, relating to immunoprophylactic and therapeutic potential, and DC endocytic machinery is reviewed, showing how DC endocytic pathways influence the handling of internalized material. The potential advantages for application of replicating RNA are presented and discussed, in consideration of their value and development in the near future.
Collapse
|
12
|
Sharma R, Ghasparian A, Robinson JA, McCullough KC. Synthetic virus-like particles target dendritic cell lipid rafts for rapid endocytosis primarily but not exclusively by macropinocytosis. PLoS One 2012; 7:e43248. [PMID: 22905240 PMCID: PMC3419204 DOI: 10.1371/journal.pone.0043248] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/18/2012] [Indexed: 01/27/2023] Open
Abstract
DC employ several endocytic routes for processing antigens, driving forward adaptive immunity. Recent advances in synthetic biology have created small (20-30 nm) virus-like particles based on lipopeptides containing a virus-derived coiled coil sequence coupled to synthetic B- and T-cell epitope mimetics. These self-assembling SVLP efficiently induce adaptive immunity without requirement for adjuvant. We hypothesized that the characteristics of DC interaction with SVLP would elaborate on the roles of cell membrane and intracellular compartments in the handling of a virus-like entity known for its efficacy as a vaccine. DC rapidly bind SVLP within min, co-localised with CTB and CD9, but not caveolin-1. In contrast, internalisation is a relatively slow process, delivering SVLP into the cell periphery where they are maintained for a number of hrs in association with microtubules. Although there is early association with clathrin, this is no longer seen after 10 min. Association with EEA-1(+) early endosomes is also early, but proteolytic processing appears slow, the SVLP-vesicles remaining peripheral. Association with transferrin occurs rarely, and only in the periphery, possibly signifying translocation of some SVLP for delivery to B-lymphocytes. Most SVLP co-localise with high molecular weight dextran. Uptake of both is impaired with mature DC, but there remains a residual uptake of SVLP. These results imply that DC use multiple endocytic routes for SVLP uptake, dominated by caveolin-independent, lipid raft-mediated macropinocytosis. With most SVLP-containing vesicles being retained in the periphery, not always interacting with early endosomes, this relates to slow proteolytic degradation and antigen retention by DC. The present characterization allows for a definition of how DC handle virus-like particles showing efficacious immunogenicity, elements valuable for novel vaccine design in the future.
Collapse
Affiliation(s)
- Rajni Sharma
- Institute of Virology and Immunoprophylaxis, Mittelhäusern, Switzerland
| | - Arin Ghasparian
- Department of Chemistry, University of Zürich, Zürich, Switzerland
| | - John A. Robinson
- Department of Chemistry, University of Zürich, Zürich, Switzerland
| | | |
Collapse
|
13
|
Hüsser L, Alves MP, Ruggli N, Summerfield A. Identification of the role of RIG-I, MDA-5 and TLR3 in sensing RNA viruses in porcine epithelial cells using lentivirus-driven RNA interference. Virus Res 2011; 159:9-16. [PMID: 21539869 DOI: 10.1016/j.virusres.2011.04.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 04/12/2011] [Accepted: 04/14/2011] [Indexed: 12/24/2022]
Abstract
Pathogen recognition receptors are essential for antiviral host immune responses. These specialized receptors detect conserved viral compounds and induce type I interferons (IFN) and pro-inflammatory cytokines. Here we evaluated the contribution of RIG-I, MDA-5 and TLR3 to the recognition of classical swine fever (CSFV), foot-and-mouth disease virus (FMDV), vesicular stomatitis virus (VSV) and influenza A virus (IAV) to IFN-β responses in the porcine epithelial cell line PK-15. To this end, we identified porcine gene specific small interfering RNA sequences and employed a lentivirus (LV)-based system to deliver the corresponding short hairpin RNA. With this, gene knockdown cell lines were created and tested with regard to the knockdown levels over time and following IFN-β stimulation. During several passages of the transduced cells, the expression of both the reporter gene eGFP and the reduced RNA levels of the targeted gene were stable, although the latter was relatively variable. IFN-β induced IFN-responsive genes such as RIG-I, but the levels of the silenced cell line remained reduced compared to the control cells. Based on virus-induced IFN-β mRNA responses, our results indicate that in PK-15 cells FMDV-detection is solely mediated by MDA-5, whereas VSV and IAV are mainly detected by RIG-I with a minor contribution of MDA-5, and CSFV is sensed by MDA-5, RIG-I and TLR3.
Collapse
Affiliation(s)
- Linda Hüsser
- Institute of Virology and Immunoprophylaxis (IVI), Sensemattstrasse 293, CH-3147 Mittelhäusern, Switzerland
| | | | | | | |
Collapse
|
14
|
Differential activation and maturation of two porcine DC populations following TLR ligand stimulation. Mol Immunol 2010; 47:2103-11. [DOI: 10.1016/j.molimm.2010.03.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 03/19/2010] [Accepted: 03/23/2010] [Indexed: 01/26/2023]
|
15
|
Smits EL, Anguille S, Cools N, Berneman ZN, Van Tendeloo VF. Dendritic Cell-Based Cancer Gene Therapy. Hum Gene Ther 2009; 20:1106-18. [DOI: 10.1089/hum.2009.145] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Evelien L.J.M. Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
- Center for Cell Therapy and Regenerative Medicine (CCTRM), Antwerp University Hospital, B-2650 Edegem (Antwerp), Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
| | - Zwi N. Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
- Center for Cell Therapy and Regenerative Medicine (CCTRM), Antwerp University Hospital, B-2650 Edegem (Antwerp), Belgium
| | - Viggo F.I. Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
- Center for Cell Therapy and Regenerative Medicine (CCTRM), Antwerp University Hospital, B-2650 Edegem (Antwerp), Belgium
| |
Collapse
|
16
|
Basta S, Gerber H, Schaub A, Summerfield A, McCullough KC. Cellular processes essential for African swine fever virus to infect and replicate in primary macrophages. Vet Microbiol 2009; 140:9-17. [PMID: 19632793 DOI: 10.1016/j.vetmic.2009.07.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 06/23/2009] [Accepted: 07/03/2009] [Indexed: 10/20/2022]
Abstract
The macrophage (Mø) is an essential immune cell for innate immunity. Such cells are targeted by African swine fever virus (ASFV). The early phases of infection with ASFV have been previously characterized in non-leukocyte cells such as Vero cells. Here, we report on several additional key parameters that ASFV utilizes during the infection of primary Mø. Related to virus infection, we established that receptor-mediated endocytosis of the virus by Mø is not the exclusive means of entry to infect the host cells. Analysis of the ensuing processes identified divalent cation-dependent activities to be particularly important, relating to the virus requirement for microtubule assembly needed for endocytic and endosomal processing. Actin-dependent endocytosis and endocytic flux involving microtubule activity are also implicated, pointing to entry via phagocytosis. Subsequently, the virus avoids terminal degradation by circumventing mature lysosome activities, including autophagosome-lysosome delivery. Nevertheless, the replicative cycle is apparently dependent on certain lysosomal functions, i.e. activities sensitive to propylamine are essential for the virus, whereas vinblastine- and leupeptin-sensitive functions only partially influence viral replication. The present work has identified cellular processes essential for ASFV to infect and replicate in the macrophage. These findings will improve our understanding of the cellular pathways employed by viruses infecting immune scavenger cells.
Collapse
Affiliation(s)
- Sameh Basta
- Department of Microbiology and Immunology, Queen's University, Kingston, ON, Canada.
| | | | | | | | | |
Collapse
|
17
|
McCullough KC, Summerfield A. Targeting the porcine immune system--particulate vaccines in the 21st century. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:394-409. [PMID: 18771683 PMCID: PMC7103233 DOI: 10.1016/j.dci.2008.07.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 07/11/2008] [Accepted: 07/11/2008] [Indexed: 05/15/2023]
Abstract
During the last decade, the propagation of immunological knowledge describing the critical role of dendritic cells (DC) in the induction of efficacious immune responses has promoted research and development of vaccines systematically targeting DC. Based on the promise for the rational design of vaccine platforms, the current review will provide an update on particle-based vaccines of both viral and synthetic origin, giving examples of recombinant virus carriers such as adenoviruses and biodegradable particulate carriers. The viral carriers carry pathogen-associated molecular patterns (PAMP), used by the original virus for targeting DC, and are particularly efficient and versatile gene delivery vectors. Efforts in the field of synthetic vaccine carriers are focussing on decorating the particle surface with ligands for DC receptors such as heparan sulphate glycosaminoglycan structures, integrins, Siglecs, galectins, C-type lectins and toll-like receptors. The emphasis of this review will be placed on targeting the porcine immune system, but reference will be made to advances with murine and human vaccine delivery systems where information on DC targeting is available.
Collapse
Affiliation(s)
- Kenneth C McCullough
- Institute of Virology and Immunoprophylaxis, Sensemattstrasse 293, CH-3147 Mittelhäusern, Switzerland.
| | | |
Collapse
|
18
|
Summerfield A, McCullough KC. The porcine dendritic cell family. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:299-309. [PMID: 18582937 PMCID: PMC7103208 DOI: 10.1016/j.dci.2008.05.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 05/05/2008] [Accepted: 05/05/2008] [Indexed: 05/07/2023]
Abstract
Considering the pivotal roles played by dendritic cells (DCs) in both innate and adaptive immune responses, advances in the field of porcine immunology DC biology have recently progressed rapidly. As with the more extensively studied murine and human DCs, porcine DC can be generated from bone marrow haematopoietic cells or monocytes, and have been analysed in various immunological and non-immunological tissues. Both conventional DC (cDC) and plasmacytoid DC (pDC) have been characterized. The function of porcine monocyte-derived DC has not only been characterized in terms of antigen presentation and lymphocyte activation, but also their response to various ligands of pattern recognition receptors. These have been characterized in terms of the induction of DC maturation and pro-inflammatory, Th1-like or Th2-like cytokines secretion. Porcine pDC most effectively sense virus infections and are characterized by their capacity to produce large quantities of IFN-alpha and the pro-inflammatory cytokines TNF-alpha, IL-6 and IL-12. As such, the DC family as a whole is a powerful ally in the host battle against pathogen attack. Nevertheless, DC in particular tissue environments or under particular stimuli can down-regulate immune response development. This is not only important for preventing over-activation of the immune system and also for ensuring tolerance against self or "friendly" substances including food components, but may also be used as a mechanism of pathogens to evade immune responses.
Collapse
Affiliation(s)
- Artur Summerfield
- Institute of Virology and Immunoprophylaxis, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland.
| | | |
Collapse
|
19
|
Ceppi M, Schmidt E, Pierre P. Genetic modification of murine dendritic cells by RNA transfection. Methods Mol Biol 2009; 531:145-156. [PMID: 19347316 DOI: 10.1007/978-1-59745-396-7_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The ability to manipulate in vitro cultured dendritic cells (DCs) by transfection represents an attractive strategy to load these antigen-presenting cells with genetic material encoding various immunogenic epitopes. The gene transfer approach can also be applied to DCs with the aim of expressing immunologically active molecules such as cytokines, costimulatory molecules, or simply to transiently express proteins to perform cell biology studies. Available gene transfer technologies for DCs include both viral and non-viral vector-based approaches. In this chapter, we describe non-viral strategies of RNA transfection. Special emphasis is given to murine bone-marrow-derived DCs, since gene transfer to human DCs has been extensively described in the literature, especially in the context of cancer immunotherapy and other clinical applications. Methods to deliver small interfering RNA (siRNA) to DCs are described as well. Finally, the potential of exogenously delivered RNA to activate DCs is discussed and some practical advice to avoid DC activation is described.
Collapse
Affiliation(s)
- Maurizio Ceppi
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerranée, Case 906, 13288, INSERM U631, CNRS, UMR6102, Marseille, France.
| | | | | |
Collapse
|
20
|
Abstract
Dendritic cells (DC) have profound abilities to induce and coordinate T-cell immunity. This makes them ideal biological agents for use in immunotherapeutic strategies to augment T-cell immunity to HIV infection. Current clinical trials are administering DC-HIV antigen preparations carried out ex vivo as proof of principle that DC immunotherapy is safe and efficacious in HIV-infected patients. These trials are largely dependent on preclinical studies that will provide knowledge and guidance about the types of DC, form of HIV antigen, method of DC maturation, route of DC administration, measures of anti-HIV immune function and ultimately control of HIV replication. Additionally, promising immunotherapy approaches are being developed based on targeting of DC with HIV antigens in vivo. The objective is to define a safe and effective strategy for enhancing control of HIV infection in patients undergoing antiretroviral therapy.
Collapse
Affiliation(s)
- C R Rinaldo
- Department of Infectious Diseases, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
21
|
Dell K, Klein C, Gissmann L. Comparison of DNA- and mRNA-Transfected Mouse Dendritic Cells as Potential Vaccines against the Human Papillomavirus Type 16 associated Oncoprotein E7. Antivir Ther 2008. [DOI: 10.1177/135965350801300409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Dendritic cells (DCs) mediate the generation of strong cytotoxic T-lymphocyte (CTL) responses by functioning in antigen presentation and exerting adjuvant properties. We compared several activation markers and parameters of biological activity of DNA- and mRNA-transfected DCs in vitro and in vivo. Methods CpG-matured, bone marrow derived C57BL/6 mouse DCs were electroporated either with enhanced green fluorescence protein (EGFP) or human papillomavirus type 16 (HPV16) E7 expression plasmids or in vitro transcribed mRNAs encoding for the codon-optimized E7 or a shuffled version thereof. Activation marker expression and antigen presentation was analysed by fluorescence-activated cell sorting. The migratory behaviour of transfected DCs were investigated by in vitro chemotaxis experiments and cytokine expression by ELISA. CTL-priming capacity of transfected DCs were determined by vaccination of mice. Results mRNA transfection produced a two- to fourfold increase of the activation markers CD40, CD80, CD86 and MHC I and MHC II molecules. Predominately antigen-expressing DCs migrated after mRNA transfection. Furthermore, mRNA-transfected DCs were capable of inducing a chemokine gradient. After maturation, electroporation and activation with soluble CD40 ligand and interferon-γ, DCs displayed a T-helper cell type 2 cytokine expression pattern. Nevertheless, E7-transfected DCs were able to prime E7-specific CTL responses in vivo. The highest E7-specific CTL frequencies were found in mice immunized with mRNA-transfected DCs. The in vitro expanded CTLs exerted functional E7-specific cytotoxic activity. Conclusions Genetically modified DCs are suitable vehicles for the induction of E7-specific CTL responses in mice and hence could help to eradicate HPV-associated lesions in humans.
Collapse
Affiliation(s)
- Kerstin Dell
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Corinna Klein
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lutz Gissmann
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
22
|
Dendritic cell internalization of foot-and-mouth disease virus: influence of heparan sulfate binding on virus uptake and induction of the immune response. J Virol 2008; 82:6379-94. [PMID: 18448534 DOI: 10.1128/jvi.00021-08] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dendritic cells (DC), which are essential for inducing and regulating immune defenses and responses, represent the critical target for vaccines against pathogens such as foot-and-mouth disease virus (FMDV). Although it is clear that FMDV enters epithelial cells via integrins, little is known about FMDV interaction with DC. Accordingly, DC internalization of FMDV antigen was analyzed by comparing vaccine virus dominated by heparan sulfate (HS)-binding variants with FMDV lacking HS-binding capacity. The internalization was most efficient with the HS-binding virus, employing diverse endocytic pathways. Moreover, internalization relied primarily on HS binding. Uptake of non-HS-binding virus by DC was considerably less efficient, so much so that it was often difficult to detect virus interacting with the DC. The HS-binding FMDV replicated in DC, albeit transiently, which was demonstrable by its sensitivity to cycloheximide treatment and the short duration of infectious virus production. There was no evidence that the non-HS-binding virus replicated in the DC. These observations on virus replication may be explained by the activities of viral RNA in the DC. When DC were transfected with infectious RNA, only 1% of the translated viral proteins were detected. Nevertheless, the transfected cells, and DC which had internalized live virus, did present antigen to lymphocytes, inducing an FMDV-specific immunoglobulin G response. These results demonstrate that DC internalization of FMDV is most efficient for vaccine virus with HS-binding capacity, but HS binding is not an exclusive requirement. Both non-HS-binding virus and infectious RNA interacting with DC induce specific immune responses, albeit less efficiently than HS-binding virus.
Collapse
|
23
|
Le Gallo M, Toutirais O, Montier T, Cabillic F, Bouet F, Delépine P, Lehn P, Jotereau F, Catros V. Activation of tumor-specific T cells by dendritic cells expressing the NY-ESO-1 antigen after transfection with the cationic lipophosphoramide KLN5. J Gene Med 2008; 10:628-36. [PMID: 18338820 DOI: 10.1002/jgm.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genetic modification of human monocyte-derived dendritic cells (DC) with cDNA sequences encoding tumor-associated antigens (TAA) is a promising strategy for cancer immunotherapy. The present study aimed to develop a nonviral gene transfer method based on the use of the cationic lipophosphoramide reagent, KLN-5, as an alternative to the commonly used viral vectors. METHODS First, the efficiency of KLN5 for gene transfection into DC was investigated using the green fluorescent protein (GFP) reporter gene. The highest transfection efficiency/cell viability ratio was determined by flow cytometry. Next, DC were transfected with a plasmid encoding NY-ESO-1, a TAA expressed in numerous cancers, according to the transfection protocol previously established with the GFP reporter. Transfected DC were then co-cultured with a CD8+ NY-ESO-1 specific HLA-A*02.01 T cell clone to control their ability to correctly process and present the corresponding epitope in the HLA-A*02.01 context. Finally, T cell activation was assessed via flow cytometry-based detection of interferon-gamma production. RESULTS An optimal KLN5/plasmid DNA ratio allowing both significant transgene expression and high viability of DC could be determined. Under the established experimental conditions, antigen processing and presentation of the immunodominant (SLLMWITQC(157-165)) epitope in the HLA-A*0201 context was demonstrated by activation of the NY-ESO-1-specific CD8+ T cell clone. CONCLUSIONS KLN5-based gene transfection into DC allows the efficient induction of TAA presentation and may thus represent a novel attractive nonviral approach for cancer vaccination.
Collapse
Affiliation(s)
- Matthieu Le Gallo
- Université de Rennes 1, Faculté de Médecine, UPRES 3891, Rennes, France
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Jamin A, Gorin S, Cariolet R, Le Potier MF, Kuntz-Simon G. Classical swine fever virus induces activation of plasmacytoid and conventional dendritic cells in tonsil, blood, and spleen of infected pigs. Vet Res 2007; 39:7. [PMID: 18073094 DOI: 10.1051/vetres:2007045] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2007] [Accepted: 08/21/2007] [Indexed: 11/14/2022] Open
Abstract
Classical swine fever virus (CSFV) compromises the host immune system, causing indirect leucopoenia and disruption of in vitro T cell stimulation capacity. In order to explore the potential role of dendritic cells (DC) in such phenomena, the activation of conventional DC (cDC) and plasmacytoid DC (pDC) in blood and secondary lymphoid organs of infected pigs was investigated in the early time course post-inoculation (pi), together with viral components dissemination and cytokine production in serum. Whereas CD11R1+CD172a+ cDC frequencies were markedly reduced in blood and spleen, analysis of CD4+CD172a+ pDC numbers revealed a rapid turn-over of this DC subset in tissues pi. Both subsets matured and were activated after infection, as demonstrated by down-regulation of CD1a, up-regulation of the co-stimulation molecule CD80/86 and expression of cytokines. cDC essentially expressed tumor necrosis factor alpha (TNF-alpha) and interleukin (IL)-10, whereas pDC produced alpha interferon (IFN-alpha) and IL-12. IFN-alpha and TNF-alpha productions revealed an enhancement of innate anti-viral immune responses. Detection of antigen activated B lymphocytes in tonsil T-cell areas at 72 h pi, subsequently to the transient translocation of the viral E2 protein within germinal centres at 48 h pi, indicates the initiation of humoral response. This response was also evidenced by an important IL-10 production in serum one week pi. IL-12 expression in organs, as well as transient detection of IL-18 and IFN-gamma in serum, reflected the initiation of cellular immune responses. However, the uncommonly high levels of TNF-alpha and IFN-alpha produced by DC and measured in serum early post-infection, together with IL-10 expression in spleen, could play a role in the disruption of immune system cells, either inducing apoptosis or impairing DC functionalities themselves.
Collapse
Affiliation(s)
- Agnès Jamin
- French Agency for Food Safety (AFSSA), Poultry and Swine Research Laboratory, Swine Virology Immunology Unit, BP 53, 22440 Ploufragan, France
| | | | | | | | | |
Collapse
|
25
|
Melhem NM, Liu XD, Boczkowski D, Gilboa E, Barratt-Boyes SM. Robust CD4+ and CD8+ T cell responses to SIV using mRNA-transfected DC expressing autologous viral Ag. Eur J Immunol 2007; 37:2164-73. [PMID: 17615585 DOI: 10.1002/eji.200636782] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A potentially powerful strategy for therapeutic HIV vaccination is the use of DC transfected with mRNA encoding autologous viral Ag, as epitopes presented by transfected DC would exactly reflect those expressed by infected cells in the individual. Using human and rhesus macaque monocyte-derived DC, we show that nucleofection is a superior method for mRNA transfection, resulting in high-level protein expression and DC maturation. DC transfected with SIV gag isolated from an infected monkey stimulated robust Ag-specific recall T cell responses of similar magnitude to those induced by peptide-pulsed PBMC that were predominantly CD8+ T cell mediated. Enhanced CD4+ T cell responses were stimulated when Gag was redirected into the lysosomal pathway via the targeting signal derived from lysosome-associated membrane protein-1 (LAMP-1). Rhesus DC transfected with lysosome-targeted gag encoding an escape mutation in an immunodominant CTL epitope stimulated CD4+ and CD8+ T cell responses of almost equivalent magnitude directed towards undefined epitopes outside of the mutated region. Finally, gag-transfected DC from SIV-infected monkeys stimulated significant Ag-specific recall T cell responses in an entirely autologous system. These findings demonstrate that mRNA-transfected DC expressing SIV Ag derived from infected monkeys stimulate broad and relevant T cell responses, supporting this approach for therapeutic HIV vaccine development.
Collapse
Affiliation(s)
- Nada M Melhem
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
26
|
Mockey M, Bourseau E, Chandrashekhar V, Chaudhuri A, Lafosse S, Le Cam E, Quesniaux VFJ, Ryffel B, Pichon C, Midoux P. mRNA-based cancer vaccine: prevention of B16 melanoma progression and metastasis by systemic injection of MART1 mRNA histidylated lipopolyplexes. Cancer Gene Ther 2007; 14:802-14. [PMID: 17589432 DOI: 10.1038/sj.cgt.7701072] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Immunization with mRNA encoding tumor antigen is an emerging vaccine strategy for cancer. In this paper, we demonstrate that mice receiving systemic injections of MART1 mRNA histidylated lipopolyplexes were specifically and significantly protected against B16F10 melanoma tumor progression. The originality of this work concerns the use of a new tumor antigen mRNA formulation as vaccine, which allows an efficient protection against the growth of a highly aggressive tumor model after its delivery by intravenous route. Synthetic melanoma-associated antigen MART1 mRNA was formulated with a polyethylene glycol (PEG)ylated derivative of histidylated polylysine and L-histidine-(N,N-di-n-hexadecylamine)ethylamide liposomes (termed histidylated lipopolyplexes). Lipopolyplexes comprised mRNA/polymer complexes encapsulated by liposomes. The tumor protective effect was induced with MART1 mRNA carrying a poly(A) tail length of 100 adenosines at an optimal dose of 12.5 microg per mouse. MART1 mRNA lipopolyplexes elicited a cellular immune response characterized by the production of interferon-gamma and the induction of cytotoxic T lymphocytes. Finally, the anti-B16 response was enhanced using a formulation containing both MART1 mRNA and MART1-LAMP1 mRNA encoding the antigen targeted to the major histocompatibility complex class II compartments by the lysosomal sorting signal of LAMP1 protein. Our results provide a basis for the development of mRNA histidylated lipopolyplexes for cancer vaccine.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Disease Progression
- Histidine/metabolism
- MART-1 Antigen
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Mice
- Microscopy, Electron, Transmission
- Neoplasm Metastasis/prevention & control
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- RNA, Messenger/administration & dosage
- RNA, Messenger/genetics
- T-Lymphocytes, Cytotoxic/immunology
- Transcription, Genetic
Collapse
Affiliation(s)
- M Mockey
- Centre de Biophysique Moléculaire CNRS UPR 4301, University of Orléans and INSERM, Orléans cedex 2, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Rautsi O, Lehmusvaara S, Salonen T, Häkkinen K, Sillanpää M, Hakkarainen T, Heikkinen S, Vähäkangas E, Ylä-Herttuala S, Hinkkanen A, Julkunen I, Wahlfors J, Pellinen R. Type I interferon response against viral and non-viral gene transfer in human tumor and primary cell lines. J Gene Med 2007; 9:122-35. [PMID: 17191255 DOI: 10.1002/jgm.997] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Type I interferon (IFN-alpha/beta) response is one of the major host defence mechanisms against viruses. Some recent reports suggest that IFNs may interfere with the efficacy of both non-viral and virus-vector-mediated therapeutic gene transfer. METHODS The type I IFN response upon different gene transfer methods in human tumor and primary cell lines was studied by analysing IFN-beta mRNA expression, secretion of type I IFNs and accumulation of IFN-alpha/beta-induced MxA protein (myxovirus resistance protein A). RESULTS Infection with avirulent Semliki Forest virus A7[74] induced MxA protein accumulation and increased the IFN-beta mRNA level, whereas none of the studied virus vectors (adenovirus, CRAd, lentivirus or AAV) induced IFN response. However, plasmid DNA induced the accumulation of MxA protein when transfected with several commercial transfection reagents. RNA transfection appeared to be an efficient inducer of type I IFN response: replicating alphaviral RNA, eukaryotic total RNA, or mRNA all induced both MxA protein accumulation and IFN-beta expression. siRNA transfection failed to induce MxA response. CONCLUSIONS The non-viral gene transfer methods have gained more interest in recent years due to their better safety profiles when compared to their viral counterparts. However, the efficiency of non-viral gene transfer is well below those reached by viral vector systems. The type I interferon response induced by non-viral methods may in part contribute to this inefficiency, while most currently used viral gene transfer vectors fail to induce or are able to suppress type I IFN response.
Collapse
Affiliation(s)
- Outi Rautsi
- A. I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine, University of Kuopio, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Alves MP, Neuhaus V, Guzylack-Piriou L, Ruggli N, McCullough KC, Summerfield A. Toll-like receptor 7 and MyD88 knockdown by lentivirus-mediated RNA interference to porcine dendritic cell subsets. Gene Ther 2007; 14:836-44. [PMID: 17330086 DOI: 10.1038/sj.gt.3302930] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sensing of viruses by dendritic cell (DC) pathogen recognition receptors (PRRs) represents a critical event during innate antiviral immune responses. Identification of these PRRs has often posed a problem due to difficulties in performing gene function studies in the naturally targeted hosts. Consequently, we developed a lentivirus (LV)-based strategy for specific gene knockdown in porcine DC. Short hairpin RNAs (shRNAs) were designed, targeting toll-like receptor 7 (TLR7) and the adaptor protein MyD88. As cellular targets, monocyte-derived DC (MoDC) and Flt3 ligand-induced DC (Flt3L-DC), DC precursors including monocytes and haematopoietic stem cells (HSCs) as well as plasmacytoid DCs (pDCs) were employed. Transduction efficiencies ranged from 40 to 95%. The LV-mediated shRNA delivery was functionally active, reducing TLR7 and MyD88 mRNA in MoDC and conventional Flt3L-DC, and blunting the responsiveness to TLR7 ligands in Flt3L-DC. Although infection of MoDC by the LV did neither influence MHC class II and CD80/86 expressions, nor cytokine responses, the infection of Flt3L-DC induced a phenotypic maturation. Furthermore, the interaction of the LV with pDC induced high levels of interferon-alpha. Taken together, these studies characterize the interaction of the LV with different DC subsets and demonstrate the suitability of LV-mediated small interfering RNA delivery for targeting PRR knockout for MoDC and conventional Flt3L-DC.
Collapse
Affiliation(s)
- M P Alves
- Institute of Virology and Immunoprophylaxis, Mittelhäusern, Switzerland
| | | | | | | | | | | |
Collapse
|
29
|
Jamin A, Gorin S, Le Potier MF, Kuntz-Simon G. Characterization of conventional and plasmacytoid dendritic cells in swine secondary lymphoid organs and blood. Vet Immunol Immunopathol 2006; 114:224-37. [PMID: 16978709 DOI: 10.1016/j.vetimm.2006.08.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 06/29/2006] [Accepted: 08/07/2006] [Indexed: 12/26/2022]
Abstract
Dendritic cells (DCs) act as antigen presenting cells that bridge innate and adaptive immune systems with the unique capacity to initiate primary T-cell responses and efficiently stimulate memory responses. In pig, little information is available about these cells in secondary lymphoid organs, the place where T cell activation usually occurs. As increased knowledge on DC is a necessary prerequisite to further understand their role in response to microbial infection or in protection after vaccination, we investigated the DC types that would be present in tonsil, spleen and non-subcutaneous lymph nodes in the steady state. One population was composed of CD172a(+)CD11R1(+)CD1(+/-)CD80/86(+/-) cells and would correspond to conventional DCs (cDC), while the other one was composed of CD172a(+)CD4(+)CD1(+/-)CD80/86(+/-) cells and would correspond to plasmacytoid DCs (pDC). These subsets were also detected in blood but spleen was the tissue with the higher frequency of such DCs. In lymphoid organs, most of cDC and pDC were in an immature status, as revealed by the low percentage of cells expressing the co-stimulatory molecule CD80/86. However, expression of that marker by 5% of DCs in organs and up to 15% in blood, together with lower expression of CD1a and expression of CD208, would indicate a partial activation and/or semi-maturation. Interestingly, 8% of tonsil pDC and 15% of blood pDC were shown to secrete IFN-alpha, while 18-20% of cDC expressed TNF-alpha in these tissues. Both cell types also expressed IL-12 and IL-10 in the steady state. Measurements of IFN-alpha, TNF-alpha, IL-12 and IL-10 levels in serum confirmed their production within immune homeostasis, whereas IL-6, IL-18 and IFN-gamma could not be detected. Altogether, these data complete knowledge on porcine immune system cells and will be a useful tool for further in vivo studies on porcine DC role in peripheral tolerance induction and in immune responses to pathogens.
Collapse
Affiliation(s)
- Agnès Jamin
- French Agency for Food Safety (AFSSA), Poultry and Swine Research Laboratory, Swine Virology Immunology Unit, Ploufragan, France
| | | | | | | |
Collapse
|
30
|
Samuel MA, Whitby K, Keller BC, Marri A, Barchet W, Williams BRG, Silverman RH, Gale M, Diamond MS. PKR and RNase L contribute to protection against lethal West Nile Virus infection by controlling early viral spread in the periphery and replication in neurons. J Virol 2006; 80:7009-19. [PMID: 16809306 PMCID: PMC1489062 DOI: 10.1128/jvi.00489-06] [Citation(s) in RCA: 187] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
West Nile virus (WNV) is a neurotropic, mosquito-borne flavivirus that can cause lethal meningoencephalitis. Type I interferon (IFN) plays a critical role in controlling WNV replication, spread, and tropism. In this study, we begin to examine the effector mechanisms by which type I IFN inhibits WNV infection. Mice lacking both the interferon-induced, double-stranded-RNA-activated protein kinase (PKR) and the endoribonuclease of the 2',5'-oligoadenylate synthetase-RNase L system (PKR(-/-) x RL(-/-)) were highly susceptible to subcutaneous WNV infection, with a 90% mortality rate compared to the 30% mortality rate observed in congenic wild-type mice. PKR(-/-) x RL(-/-) mice had increased viral loads in their draining lymph nodes, sera, and spleens, which led to early viral entry into the central nervous system (CNS) and higher viral burden in neuronal tissues. Although mice lacking RNase L showed a higher CNS viral burden and an increased mortality, they were less susceptible than the PKR(-/-) x RL(-/-) mice; thus, we also infer an antiviral role for PKR in the control of WNV infection. Notably, a deficiency in both PKR and RNase L resulted in a decreased ability of type I IFN to inhibit WNV in primary macrophages and cortical neurons. In contrast, the peripheral neurons of the superior cervical ganglia of PKR(-/-) x RL(-/-) mice showed no deficiency in the IFN-mediated inhibition of WNV. Our data suggest that PKR and RNase L contribute to IFN-mediated protection in a cell-restricted manner and control WNV infection in peripheral tissues and some neuronal subtypes.
Collapse
Affiliation(s)
- Melanie A Samuel
- Department of Molecular Microbiology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Guzylack-Piriou L, Piersma S, McCullough K, Summerfield A. Role of natural interferon-producing cells and T lymphocytes in porcine monocyte-derived dendritic cell maturation. Immunology 2006; 118:78-87. [PMID: 16630025 PMCID: PMC1782262 DOI: 10.1111/j.1365-2567.2006.02343.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Maturation of dendritic cells (DC) is a key immunological process regulating immune responses to pathogens and vaccines, as well as tolerance and autoimmune processes. Consequently, the regulation of DC maturation should reflect these multifaceted immunological processes. In the present study, we have defined the role of particular cytokines, Toll-like receptor (TLR) ligands and T lymphocytes in the porcine monocyte-derived DC (MoDC). Interferon-α (IFN-α) alone was a poor inducer of MoDC maturation, but in association with tumour necrosis factor-α (TNF-α), or TLR ligands such as lipopolysaccharide and polyinosinic-polycytidylic acid I:C, an up-regulation of major histocompatibility complex II and CD80/86 expression was noted, along with reduced endocytic activity. In contrast, TNF-α alone or in combination with the TLR ligands was a poor inducer of DC maturation, but co-operated with T-lymphocytes in the presence of antigen to induce DC maturation. Natural interferon producing cells (NIPC, or plasmacytoid DCs) represent a danger-recognition system of the immune defences, and can respond to viruses not otherwise recognized as posing a danger. Indeed, MoDC did not respond to transmissible gastroenteritis virus (TGEV), whereas NIPC produced high levels of IFN-α and TNF-α after TGEV stimulation. Moreover, supernatants from the stimulated NIPC induced maturation in MoDCs. These matured MoDCs displayed an enhanced ability to present antigen to and thus stimulate T cells. Taken together, the present work demonstrates that maturation of MoDC not only results from TLR signalling, but can require co-operation with various cell types – principally NIPC and activated T cells – which would reflect the particular immunological situation.
Collapse
|
32
|
Kelly KM, Zhuang H, Nacionales DC, Scumpia PO, Lyons R, Akaogi J, Lee P, Williams B, Yamamoto M, Akira S, Satoh M, Reeves WH. “Endogenous adjuvant” activity of the RNA components of lupus autoantigens Sm/RNP and Ro 60. ACTA ACUST UNITED AC 2006; 54:1557-67. [PMID: 16645989 DOI: 10.1002/art.21819] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Most lupus patients produce autoantibodies against small ribonucleoproteins such as Sm/RNP and Ro 60 (containing U1 and Y1-Y5 RNAs, respectively). We undertook this study to investigate whether the RNA components of these antigens, which contain extensive tracts of single- and double-stranded RNA, signatures of viral infection, activate innate immunity. METHODS U1 and Y RNAs were affinity purified from K562 cells. Murine bone marrow-derived dendritic cells (DCs), human HEK 293 cells, and murine RAW264.7 cells were stimulated with U1 RNA and other known Toll-like receptor (TLR) ligands. Expression of the interferon (IFN)-inducible gene Mx1 and other genes was quantified using real-time polymerase chain reaction, and cytokine production was measured by enzyme-linked immunosorbent assay. DC maturation was assessed using flow cytometry. RESULTS Purified U1 and Y1-Y5 RNAs and synthetic stem-loop II of U1 RNA stimulated type I IFN (IFN-I) production by cell lines and murine bone marrow-derived DCs and promoted DC maturation (CD86 expression). U1 RNA-stimulated, but not TLR-3 ligand-stimulated, IFN-I was blocked by bafilomycin A1, indicating that immunostimulation by U1 RNA requires endosomal acidification. Myeloid differentiation factor 88-deficient cells responded poorly to U1 RNA, suggesting that an endosomal TLR, probably TLR-7, mediates the stimulatory effects of U1 RNA. U1 RNA-induced IFN-I and interleukin-6 production also were protein kinase R (PKR) dependent (abrogated by 2-aminopurine and greatly reduced in PKR-/- cells). CONCLUSION We conclude that the RNA components of the Ro 60 (Y1-Y5 RNA) and Sm/RNP (U1 RNA) small ribonucleoproteins act as endogenous adjuvants that could play a role in the pathogenesis of autoimmunity by stimulating DC maturation and IFN-I production.
Collapse
|
33
|
Bauhofer O, Summerfield A, McCullough KC, Ruggli N. Role of double-stranded RNA and Npro of classical swine fever virus in the activation of monocyte-derived dendritic cells. Virology 2005; 343:93-105. [PMID: 16154171 DOI: 10.1016/j.virol.2005.08.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Revised: 06/25/2005] [Accepted: 08/12/2005] [Indexed: 11/18/2022]
Abstract
Classical swine fever virus (CSFV) is a noncytopathogenic (ncp) positive-sense RNA virus that replicates in myeloid cells including macrophages and dendritic cells (DC). The virus does not induce type I interferon (IFN-alpha/beta), which in macrophages has been related to the presence of the viral Npro gene. In the present work, the role of viral double-stranded (ds)RNA and Npro in the virus-host cell interaction has been analyzed. Higher levels of detectable dsRNA were produced by a genetically engineered cytopathogenic (cp) CSFV compared with ncp CSFV, and cp CSFV induced IFN-alpha/beta in PK-15 cells. With DC, there was only a small difference in the levels of dsRNA between the cp and ncp viruses, and no IFN-alpha/beta was produced. However, the cp virus induced a higher degree of DC maturation, in terms of CD80/86 and MHC II expression. Npro deletion mutants induced an increase in DC maturation and IFN-alpha/beta production-for both ncp and cp viruses-despite reduced replication efficiency in the DC. Deletion of Npro did not influence dsRNA levels, indicating that the interference was downstream of dsRNA turnover regulation. In conclusion, the capacity of CSFV to replicate in myeloid DC, and prevent IFN-alpha/beta induction and DC maturation, requires both regulated dsRNA levels and the presence of viral Npro.
Collapse
Affiliation(s)
- Oliver Bauhofer
- Institute of Virology and Immunoprophylaxis (IVI), Sensemattstrasse 293, CH-3147 Mittelhäusern, Switzerland.
| | | | | | | |
Collapse
|
34
|
Ceppi M, de Bruin MGM, Seuberlich T, Balmelli C, Pascolo S, Ruggli N, Wienhold D, Tratschin JD, McCullough KC, Summerfield A. Identification of classical swine fever virus protein E2 as a target for cytotoxic T cells by using mRNA-transfected antigen-presenting cells. J Gen Virol 2005; 86:2525-2534. [PMID: 16099911 DOI: 10.1099/vir.0.80907-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Vaccination of pigs against Classical swine fever virus (CSFV) by using live-virus vaccines induces early protection before detectable humoral immune responses. Immunological analyses indicate that this is associated with T-cell activation, underlining the importance of targeting cytotoxic T-lymphocyte (CTL) responses for vaccine improvement. Antigen-presenting cells (APCs) transfected with mRNA encoding structural protein E2 or non-structural viral proteins NS3-NS4A were used to identify viral genes encoding CTL epitopes. Monocyte-derived dendritic cells (DCs) and fibrocytes served as the APCs. In vitro translation of the mRNA and microscopic analysis of transfected cells demonstrated that E2 and NS3-NS4A could be identified. APCs transfected with either of the mRNA molecules restimulated CSFV-specific T cells to produce gamma interferon and specific cytotoxic activity against CSFV-infected target cells. The presence of CTL epitopes on E2 was confirmed by using d/d-haplotype MAX cells expressing E2 constitutively as target cells in d/d-haplotype CTL assays. A potent CTL activity against E2 was detected early (1-3 weeks) after CSFV challenge. This work corroborates the existence of CTL epitopes within the non-structural protein domain NS3-NS4A of CSFV. Furthermore, epitopes on the E2 protein can also now be classified as targets for CTLs, having important implications for vaccine design, especially subunit vaccines. As for the use of mRNA-transfected APCs, this represents a simple and efficient method to identify viral genes encoding CTL epitopes in outbred populations.
Collapse
Affiliation(s)
- M Ceppi
- Institute of Virology and Immunoprophylaxis (IVI), Postfach, CH-3147 Mittelhäusern, Switzerland
| | | | - T Seuberlich
- Institute of Virology and Immunoprophylaxis (IVI), Postfach, CH-3147 Mittelhäusern, Switzerland
| | - C Balmelli
- Institute of Virology and Immunoprophylaxis (IVI), Postfach, CH-3147 Mittelhäusern, Switzerland
| | - S Pascolo
- Organic Chemistry, Auf der Morgenstelle 18, D-72076 Tübingen, Germany
| | - N Ruggli
- Institute of Virology and Immunoprophylaxis (IVI), Postfach, CH-3147 Mittelhäusern, Switzerland
| | - D Wienhold
- Friedrich-Loeffler-Institut, Paul-Ehrlich-Straße 28, D-72076 Tübingen, Germany
| | - J D Tratschin
- Institute of Virology and Immunoprophylaxis (IVI), Postfach, CH-3147 Mittelhäusern, Switzerland
| | - K C McCullough
- Institute of Virology and Immunoprophylaxis (IVI), Postfach, CH-3147 Mittelhäusern, Switzerland
| | - A Summerfield
- Institute of Virology and Immunoprophylaxis (IVI), Postfach, CH-3147 Mittelhäusern, Switzerland
| |
Collapse
|
35
|
Geiger C, Regn S, Weinzierl A, Noessner E, Schendel DJ. A generic RNA-pulsed dendritic cell vaccine strategy for renal cell carcinoma. J Transl Med 2005; 3:29. [PMID: 16045799 PMCID: PMC1188079 DOI: 10.1186/1479-5876-3-29] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Accepted: 07/26/2005] [Indexed: 11/18/2022] Open
Abstract
We present a generic dendritic cell (DC) vaccine strategy for patients with renal cell carcinoma (RCC) based on the use of RNA as a source of multiplex tumor-associated antigens (TAAs). Instead of preparing RNA from tumor tissue of each individual RCC patient, we propose to substitute RNA prepared from a well characterized highly immunogenic RCC cell line (RCC-26 tumor cells) as a generic source of TAAs for loading of DCs. We demonstrate here that efficient RNA transfer can be achieved using lipofection of immature DCs, which are subsequently matured with a cytokine cocktail to express high levels of MHC and costimulatory molecules as well as the chemokine receptor CCR7. Neither RNA itself nor the lipid component impacted on the phenotype or the cytokine secretion of mature DCs. Following RNA loading, DCs derived from HLA-A2-positive donors were able to activate effector-memory cytotoxic T lymphocytes (CTLs) specific for a TAA ligand expressed by the RCC-26 cell line. CTL responses to RNA-loaded DCs reached levels comparable to those stimulated directly by the RCC-26 tumor cells. Furthermore, DCs expressing tumor cell RNA primed naïve T cells, yielding T cell lines with cytotoxicity and cytokine secretion after contact with RCC tumor cells. RCC-26 cell lines are available as good manufacturing practice (GMP)-certified reagents enabling this source of RNA to be easily standardized and adapted for clinical testing. In addition, well defined immune monitoring tools, including the use of RNA expressing B cell lines, are available. Thus, this DC vaccine strategy can be directly compared with an ongoing gene therapy trial using genetically-engineered variants of the RCC-26 cell line as vaccines for RCC patients with metastatic disease.
Collapse
Affiliation(s)
- Christiane Geiger
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| | - Sybille Regn
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| | - Andreas Weinzierl
- Institute of Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Elfriede Noessner
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| | - Dolores J Schendel
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| |
Collapse
|