Cargando…

Cytoplasmic delivery of siRNA using human-derived membrane penetration-enhancing peptide

BACKGROUND: Although protein-based methods using cell-penetrating peptides such as TAT have been expected to provide an alternative approach to siRNA delivery, the low efficiency of endosomal escape of siRNA/protein complexes taken up into cells by endocytosis remains a problem. Here, to overcome th...

Descripción completa

Detalles Bibliográficos
Autores principales: Nakamura, Momoko, Fujiwara, Kei, Doi, Nobuhide
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9615171/
https://www.ncbi.nlm.nih.gov/pubmed/36303212
http://dx.doi.org/10.1186/s12951-022-01667-4
_version_ 1784820360116436992
author Nakamura, Momoko
Fujiwara, Kei
Doi, Nobuhide
author_facet Nakamura, Momoko
Fujiwara, Kei
Doi, Nobuhide
author_sort Nakamura, Momoko
collection PubMed
description BACKGROUND: Although protein-based methods using cell-penetrating peptides such as TAT have been expected to provide an alternative approach to siRNA delivery, the low efficiency of endosomal escape of siRNA/protein complexes taken up into cells by endocytosis remains a problem. Here, to overcome this problem, we adopted the membrane penetration-enhancing peptide S19 from human syncytin 1 previously identified in our laboratory. RESULTS: We prepared fusion proteins in which the S19 and TAT peptides were fused to the viral RNA-binding domains (RBDs) as carrier proteins, added the RBD-S19-TAT/siRNA complex to human cultured cells, and investigated the cytoplasmic delivery of the complex and the knockdown efficiency of target genes. We found that the intracellular uptake of the RBD-S19-TAT/siRNA complex was increased compared to that of the RBD-TAT/siRNA complex, and the expression level of the target mRNA was decreased. Because siRNA must dissociate from RBD and bind to Argonaute 2 (Ago2) to form the RNA-induced silencing complex (RISC) after the protein/siRNA complex is delivered into the cytoplasm, a dilemma arises: stronger binding between RBD and siRNA increases intracellular uptake but makes RISC formation more difficult. Thus, we next prepared fusion proteins in which the S19 and TAT peptides were fused with Ago2 instead of RBD and found that the efficiencies of siRNA delivery and knockdown obtained using TAT-S19-Ago2 were higher than those using TAT-Ago2. In addition, we found that the smallest RISC delivery induced faster knockdown than traditional siRNA lipofection, probably due to the decreased time required for RISC formation in the cytoplasm. CONCLUSION: These results indicated that S19 and TAT-fused siRNA-binding proteins, especially Ago2, should be useful for the rapid and efficient delivery of siRNA without the addition of any endosome-disrupting agent. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-022-01667-4.
format Online
Article
Text
id pubmed-9615171
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-96151712022-10-29 Cytoplasmic delivery of siRNA using human-derived membrane penetration-enhancing peptide Nakamura, Momoko Fujiwara, Kei Doi, Nobuhide J Nanobiotechnology Research BACKGROUND: Although protein-based methods using cell-penetrating peptides such as TAT have been expected to provide an alternative approach to siRNA delivery, the low efficiency of endosomal escape of siRNA/protein complexes taken up into cells by endocytosis remains a problem. Here, to overcome this problem, we adopted the membrane penetration-enhancing peptide S19 from human syncytin 1 previously identified in our laboratory. RESULTS: We prepared fusion proteins in which the S19 and TAT peptides were fused to the viral RNA-binding domains (RBDs) as carrier proteins, added the RBD-S19-TAT/siRNA complex to human cultured cells, and investigated the cytoplasmic delivery of the complex and the knockdown efficiency of target genes. We found that the intracellular uptake of the RBD-S19-TAT/siRNA complex was increased compared to that of the RBD-TAT/siRNA complex, and the expression level of the target mRNA was decreased. Because siRNA must dissociate from RBD and bind to Argonaute 2 (Ago2) to form the RNA-induced silencing complex (RISC) after the protein/siRNA complex is delivered into the cytoplasm, a dilemma arises: stronger binding between RBD and siRNA increases intracellular uptake but makes RISC formation more difficult. Thus, we next prepared fusion proteins in which the S19 and TAT peptides were fused with Ago2 instead of RBD and found that the efficiencies of siRNA delivery and knockdown obtained using TAT-S19-Ago2 were higher than those using TAT-Ago2. In addition, we found that the smallest RISC delivery induced faster knockdown than traditional siRNA lipofection, probably due to the decreased time required for RISC formation in the cytoplasm. CONCLUSION: These results indicated that S19 and TAT-fused siRNA-binding proteins, especially Ago2, should be useful for the rapid and efficient delivery of siRNA without the addition of any endosome-disrupting agent. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-022-01667-4. BioMed Central 2022-10-27 /pmc/articles/PMC9615171/ /pubmed/36303212 http://dx.doi.org/10.1186/s12951-022-01667-4 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Nakamura, Momoko
Fujiwara, Kei
Doi, Nobuhide
Cytoplasmic delivery of siRNA using human-derived membrane penetration-enhancing peptide
title Cytoplasmic delivery of siRNA using human-derived membrane penetration-enhancing peptide
title_full Cytoplasmic delivery of siRNA using human-derived membrane penetration-enhancing peptide
title_fullStr Cytoplasmic delivery of siRNA using human-derived membrane penetration-enhancing peptide
title_full_unstemmed Cytoplasmic delivery of siRNA using human-derived membrane penetration-enhancing peptide
title_short Cytoplasmic delivery of siRNA using human-derived membrane penetration-enhancing peptide
title_sort cytoplasmic delivery of sirna using human-derived membrane penetration-enhancing peptide
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9615171/
https://www.ncbi.nlm.nih.gov/pubmed/36303212
http://dx.doi.org/10.1186/s12951-022-01667-4
work_keys_str_mv AT nakamuramomoko cytoplasmicdeliveryofsirnausinghumanderivedmembranepenetrationenhancingpeptide
AT fujiwarakei cytoplasmicdeliveryofsirnausinghumanderivedmembranepenetrationenhancingpeptide
AT doinobuhide cytoplasmicdeliveryofsirnausinghumanderivedmembranepenetrationenhancingpeptide