Cargando…

Interferon regulatory factor 1 priming of tumour-derived exosomes enhances the antitumour immune response

BACKGROUND: Tumour-derived exosomes (TEXs) have a potential for application in cancer vaccines. Whether TEXs after induction by interferon regulatory factor 1 (IRF-1) are capable of enhancing the antitumour response remains to be determined. METHODS: Exosomes released by tumour cells infected with I...

Descripción completa

Detalles Bibliográficos
Autores principales: Yang, Mu-qing, Du, Qiang, Varley, Patrick R, Goswami, Julie, Liang, Zhihai, Wang, Ronghua, Li, Hui, Stolz, Donna B, Geller, David A
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5765230/
https://www.ncbi.nlm.nih.gov/pubmed/29112686
http://dx.doi.org/10.1038/bjc.2017.389
_version_ 1783292192170704896
author Yang, Mu-qing
Du, Qiang
Varley, Patrick R
Goswami, Julie
Liang, Zhihai
Wang, Ronghua
Li, Hui
Stolz, Donna B
Geller, David A
author_facet Yang, Mu-qing
Du, Qiang
Varley, Patrick R
Goswami, Julie
Liang, Zhihai
Wang, Ronghua
Li, Hui
Stolz, Donna B
Geller, David A
author_sort Yang, Mu-qing
collection PubMed
description BACKGROUND: Tumour-derived exosomes (TEXs) have a potential for application in cancer vaccines. Whether TEXs after induction by interferon regulatory factor 1 (IRF-1) are capable of enhancing the antitumour response remains to be determined. METHODS: Exosomes released by tumour cells infected with IRF-1-expressing adenovirus (IRF-1-Exo) or treated with interferon-γ (IFN-Exo) were isolated via ultracentrifugation. The IRF-1 target proteins IL-15Rα and MHC class I (MHC-I) were analysed by western blot. Exosomes along with CpG adjuvant were injected into tumour models to assess the antitumour effects. Tumours were harvested for immunofluorescence staining. Splenocytes from tumour-bearing mice were co-cultured with tumour cells. The IFNγ-positive and granzyme B-positive CD8α+ splenocyte cells were quantified by flow cytometry. RESULTS: The IRF-1-Exo or IFN-Exo displayed increased IL-15Rα and MHC-I expression. Injection of IRF-1-Exo or IFN-Exo combined with CpG had improved antitumour effects in mice. This effect may be a result of increased infiltration of tumours by CD4+ and CD8α+ T cells. Antibody-mediated depletion of CD4+ or CD8+ T cells abrogated the antitumour effects. Splenocytes isolated from CpG+IRF-1-Exo-injected Hepa 1–6 tumour mice had increased IFNγ-positive and granzyme B-positive CD8+ cells after co-culturing with Hepa 1–6 cells as compared with MC38 cells. CONCLUSIONS: The IRF-1 priming of TEXs enhances antitumour immune response.
format Online
Article
Text
id pubmed-5765230
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-57652302019-01-01 Interferon regulatory factor 1 priming of tumour-derived exosomes enhances the antitumour immune response Yang, Mu-qing Du, Qiang Varley, Patrick R Goswami, Julie Liang, Zhihai Wang, Ronghua Li, Hui Stolz, Donna B Geller, David A Br J Cancer Translational Therapeutics BACKGROUND: Tumour-derived exosomes (TEXs) have a potential for application in cancer vaccines. Whether TEXs after induction by interferon regulatory factor 1 (IRF-1) are capable of enhancing the antitumour response remains to be determined. METHODS: Exosomes released by tumour cells infected with IRF-1-expressing adenovirus (IRF-1-Exo) or treated with interferon-γ (IFN-Exo) were isolated via ultracentrifugation. The IRF-1 target proteins IL-15Rα and MHC class I (MHC-I) were analysed by western blot. Exosomes along with CpG adjuvant were injected into tumour models to assess the antitumour effects. Tumours were harvested for immunofluorescence staining. Splenocytes from tumour-bearing mice were co-cultured with tumour cells. The IFNγ-positive and granzyme B-positive CD8α+ splenocyte cells were quantified by flow cytometry. RESULTS: The IRF-1-Exo or IFN-Exo displayed increased IL-15Rα and MHC-I expression. Injection of IRF-1-Exo or IFN-Exo combined with CpG had improved antitumour effects in mice. This effect may be a result of increased infiltration of tumours by CD4+ and CD8α+ T cells. Antibody-mediated depletion of CD4+ or CD8+ T cells abrogated the antitumour effects. Splenocytes isolated from CpG+IRF-1-Exo-injected Hepa 1–6 tumour mice had increased IFNγ-positive and granzyme B-positive CD8+ cells after co-culturing with Hepa 1–6 cells as compared with MC38 cells. CONCLUSIONS: The IRF-1 priming of TEXs enhances antitumour immune response. Nature Publishing Group 2018-01 2017-11-07 /pmc/articles/PMC5765230/ /pubmed/29112686 http://dx.doi.org/10.1038/bjc.2017.389 Text en Copyright © 2018 Cancer Research UK http://creativecommons.org/licenses/by-nc-sa/4.0/ From twelve months after its original publication, this work is licensed under the Creative Commons Attribution-NonCommercial-Share Alike 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/4.0/
spellingShingle Translational Therapeutics
Yang, Mu-qing
Du, Qiang
Varley, Patrick R
Goswami, Julie
Liang, Zhihai
Wang, Ronghua
Li, Hui
Stolz, Donna B
Geller, David A
Interferon regulatory factor 1 priming of tumour-derived exosomes enhances the antitumour immune response
title Interferon regulatory factor 1 priming of tumour-derived exosomes enhances the antitumour immune response
title_full Interferon regulatory factor 1 priming of tumour-derived exosomes enhances the antitumour immune response
title_fullStr Interferon regulatory factor 1 priming of tumour-derived exosomes enhances the antitumour immune response
title_full_unstemmed Interferon regulatory factor 1 priming of tumour-derived exosomes enhances the antitumour immune response
title_short Interferon regulatory factor 1 priming of tumour-derived exosomes enhances the antitumour immune response
title_sort interferon regulatory factor 1 priming of tumour-derived exosomes enhances the antitumour immune response
topic Translational Therapeutics
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5765230/
https://www.ncbi.nlm.nih.gov/pubmed/29112686
http://dx.doi.org/10.1038/bjc.2017.389
work_keys_str_mv AT yangmuqing interferonregulatoryfactor1primingoftumourderivedexosomesenhancestheantitumourimmuneresponse
AT duqiang interferonregulatoryfactor1primingoftumourderivedexosomesenhancestheantitumourimmuneresponse
AT varleypatrickr interferonregulatoryfactor1primingoftumourderivedexosomesenhancestheantitumourimmuneresponse
AT goswamijulie interferonregulatoryfactor1primingoftumourderivedexosomesenhancestheantitumourimmuneresponse
AT liangzhihai interferonregulatoryfactor1primingoftumourderivedexosomesenhancestheantitumourimmuneresponse
AT wangronghua interferonregulatoryfactor1primingoftumourderivedexosomesenhancestheantitumourimmuneresponse
AT lihui interferonregulatoryfactor1primingoftumourderivedexosomesenhancestheantitumourimmuneresponse
AT stolzdonnab interferonregulatoryfactor1primingoftumourderivedexosomesenhancestheantitumourimmuneresponse
AT gellerdavida interferonregulatoryfactor1primingoftumourderivedexosomesenhancestheantitumourimmuneresponse