Please use this identifier to cite or link to this item: 10.3390/microorganisms9051073
Title: Reciprocal inhibition of immunogenic performance in mice of two potent dna immunogens targeting hcv-related liver cancer
Authors: Jansons, Juris
Skrastina, Dace
Kurlanda, Alisa
Petkov, Stefan
Avdoshina, Darya
Kuzmenko, Yulia
Krotova, Olga
Trofimova, Olga
Gordeychuk, Ilya
Sominskaya, Irina
Isaguliants, Maria
Institute of Microbiology and Virology
Keywords: Assays of reporter expression;CD4+ and CD8+ T cell response;Eukaryotic expression;Hepatitis C virus;Hepatocellular carcinoma;Immune suppression;Immunotherapy;Induction of type I interferons;Multi-component DNA vaccine;Nucleocapsid (core) protein;Telomerase reverse transcriptase;3.1 Basic medicine;3.2 Clinical medicine;3.3 Health sciences;1.1. Scientific article indexed in Web of Science and/or Scopus database;Microbiology;Microbiology (medical);Virology;SDG 3 - Good Health and Well-being
Issue Date: May-2021
Citation: Jansons , J , Skrastina , D , Kurlanda , A , Petkov , S , Avdoshina , D , Kuzmenko , Y , Krotova , O , Trofimova , O , Gordeychuk , I , Sominskaya , I & Isaguliants , M 2021 , ' Reciprocal inhibition of immunogenic performance in mice of two potent dna immunogens targeting hcv-related liver cancer ' , Microorganisms , vol. 9 , no. 5 , 1073 . https://doi.org/10.3390/microorganisms9051073
Abstract: Chronic HCV infection and associated liver cancer impose a heavy burden on the healthcare system. Direct acting antivirals eliminate HCV, unless it is drug resistant, and partially reverse liver disease, but they cannot cure HCV-related cancer. A possible remedy could be a multi-component immunotherapeutic vaccine targeting both HCV-infected and malignant cells, but also those not infected with HCV. To meet this need we developed a two-component DNA vaccine based on the highly conserved core protein of HCV to target HCV-infected cells, and a renowned tumor-associated antigen telomerase reverse transcriptase (TERT) based on the rat TERT, to target malignant cells. Their synthetic genes were expression-optimized, and HCV core was truncated after aa 152 (Core152opt) to delete the domain interfering with immunogenicity. Core152opt and TERT DNA were highly immunogenic in BALB/c mice, inducing IFN-γ/IL-2/TNF-α response of CD4+ and CD8+ T cells. Additionally, DNA-immunization with TERT enhanced cellular immune response against luciferase encoded by a co-delivered plasmid (Luc DNA). However, DNA-immunization with Core152opt and TERT mix resulted in abrogation of immune response against both components. A loss of bioluminescence signal after co-delivery of TERT and Luc DNA into mice indicated that TERT affects the in vivo expression of luciferase directed by the immediate early cytomegalovirus and interferon-β promoters. Panel of mutant TERT variants was created and tested for their expression effects. TERT with deleted N-terminal nucleoli localization signal and mutations abrogating telomerase activity still suppressed the IFN-β driven Luc expression, while the inactivated reverse transcriptase domain of TERT and its analogue, enzymatically active HIV-1 reverse transcriptase, exerted only weak suppressive effects, implying that suppression relied on the presence of the full-length/nearly full-length TERT, but not its enzymatic activity. The effect(s) could be due to interference of the ectopically expressed xenogeneic rat TERT with biogenesis of mRNA, ribosomes and protein translation in murine cells, affecting the expression of immunogens. HCV core can aggravate this effect, leading to early apoptosis of co-expressing cells, preventing the induction of immune response.
Description: Funding Information: This study was supported by the Latvian Science Council grants LZP-2018/2-0308 to M.I. and LZP-2020/2-0376 to J.J. and M.I., and FoBe grant of Karolinska Institutet 2018-02329 to S.P. M.I. and S.P. were partly supported by EU Twinning project VACTRAIN no. 692293, and M.I. Additionally by EAVI2020 no. 681137. Funding Information: Funding: This study was supported by the Latvian Science Council grants LZP-2018/2-0308 to M.I. and LZP-2020/2-0376 to J.J. and M.I., and FoBe grant of Karolinska Institutet 2018-02329 to S.P. M.I. and S.P. were partly supported by EU Twinning project VACTRAIN no. 692293, and M.I. Additionally by EAVI2020 no. 681137. Publisher Copyright: © 2021 by the authors. Licensee MDPI, Basel, Switzerland.
DOI: 10.3390/microorganisms9051073
Appears in Collections:Research outputs from Pure / Zinātniskās darbības rezultāti no ZDIS Pure



Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.