Cargando…

Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide

BACKGROUND: Tumor-associated macrophages (TAMs) are the major component of tumor-infiltrating immune cells. Macrophages are broadly categorized as M1 or M2 types, and TAMs have been shown to express an M2-like phenotype. TAMs promote tumor progression and contribute to resistance to chemotherapies....

Descripción completa

Detalles Bibliográficos
Autores principales: Lee, Chanju, Jeong, Hyunju, Bae, Younghyeon, Shin, Kyungmoon, Kang, Sinwoo, Kim, Hwikyung, Oh, Jayoung, Bae, Hyunsu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6555931/
https://www.ncbi.nlm.nih.gov/pubmed/31174610
http://dx.doi.org/10.1186/s40425-019-0610-4
_version_ 1783425238313205760
author Lee, Chanju
Jeong, Hyunju
Bae, Younghyeon
Shin, Kyungmoon
Kang, Sinwoo
Kim, Hwikyung
Oh, Jayoung
Bae, Hyunsu
author_facet Lee, Chanju
Jeong, Hyunju
Bae, Younghyeon
Shin, Kyungmoon
Kang, Sinwoo
Kim, Hwikyung
Oh, Jayoung
Bae, Hyunsu
author_sort Lee, Chanju
collection PubMed
description BACKGROUND: Tumor-associated macrophages (TAMs) are the major component of tumor-infiltrating immune cells. Macrophages are broadly categorized as M1 or M2 types, and TAMs have been shown to express an M2-like phenotype. TAMs promote tumor progression and contribute to resistance to chemotherapies. Therefore, M2-like TAMs are potential targets for the cancer immunotherapy. In this study, we targeted M2-like TAMs using a hybrid peptide, MEL-dKLA, composed of melittin (MEL), which binds preferentially to M2-like TAMs, and the pro-apoptotic peptide d (KLAKLAK)(2) (dKLA), which induces mitochondrial death after cell membrane penetration. METHODS: The M1 or M2-differentiated RAW264.7 cells were used for mitochondrial colocalization and apoptosis test in vitro. For in vivo study, the murine Lewis lung carcinoma cells were inoculated subcutaneously in the right flank of mouse. The dKLA, MEL and MEL-dKLA peptides were intraperitoneally injected at 175 nmol/kg every 3 days. Flow cytometry analysis of tumor-associated macrophages and immunofluorescence staining were performed to investigate the immunotherapeutic effects of MEL-dKLA. RESULTS: We showed that MEL-dKLA induced selective cell death of M2 macrophages in vitro, whereas MEL did not disrupt the mitochondrial membrane. We also showed that MEL-dKLA selectively targeted M2-like TAMs without affecting other leukocytes, such as T cells and dendritic cells, in vivo. These features resulted in lower tumor growth rates, tumor weights, and angiogenesis in vivo. Importantly, although both MEL and MEL-dKLA reduced numbers of CD206(+) M2-like TAMs in tumors, only MEL-dKLA induced apoptosis in CD206(+) M2-like TAMs, and MEL did not induce cell death. CONCLUSION: Taken together, our study demonstrated that MEL-dKLA could be used to target M2-like TAMs as a promising cancer therapeutic agent.
format Online
Article
Text
id pubmed-6555931
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-65559312019-06-10 Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide Lee, Chanju Jeong, Hyunju Bae, Younghyeon Shin, Kyungmoon Kang, Sinwoo Kim, Hwikyung Oh, Jayoung Bae, Hyunsu J Immunother Cancer Research Article BACKGROUND: Tumor-associated macrophages (TAMs) are the major component of tumor-infiltrating immune cells. Macrophages are broadly categorized as M1 or M2 types, and TAMs have been shown to express an M2-like phenotype. TAMs promote tumor progression and contribute to resistance to chemotherapies. Therefore, M2-like TAMs are potential targets for the cancer immunotherapy. In this study, we targeted M2-like TAMs using a hybrid peptide, MEL-dKLA, composed of melittin (MEL), which binds preferentially to M2-like TAMs, and the pro-apoptotic peptide d (KLAKLAK)(2) (dKLA), which induces mitochondrial death after cell membrane penetration. METHODS: The M1 or M2-differentiated RAW264.7 cells were used for mitochondrial colocalization and apoptosis test in vitro. For in vivo study, the murine Lewis lung carcinoma cells were inoculated subcutaneously in the right flank of mouse. The dKLA, MEL and MEL-dKLA peptides were intraperitoneally injected at 175 nmol/kg every 3 days. Flow cytometry analysis of tumor-associated macrophages and immunofluorescence staining were performed to investigate the immunotherapeutic effects of MEL-dKLA. RESULTS: We showed that MEL-dKLA induced selective cell death of M2 macrophages in vitro, whereas MEL did not disrupt the mitochondrial membrane. We also showed that MEL-dKLA selectively targeted M2-like TAMs without affecting other leukocytes, such as T cells and dendritic cells, in vivo. These features resulted in lower tumor growth rates, tumor weights, and angiogenesis in vivo. Importantly, although both MEL and MEL-dKLA reduced numbers of CD206(+) M2-like TAMs in tumors, only MEL-dKLA induced apoptosis in CD206(+) M2-like TAMs, and MEL did not induce cell death. CONCLUSION: Taken together, our study demonstrated that MEL-dKLA could be used to target M2-like TAMs as a promising cancer therapeutic agent. BioMed Central 2019-06-07 /pmc/articles/PMC6555931/ /pubmed/31174610 http://dx.doi.org/10.1186/s40425-019-0610-4 Text en © The Author(s). 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Lee, Chanju
Jeong, Hyunju
Bae, Younghyeon
Shin, Kyungmoon
Kang, Sinwoo
Kim, Hwikyung
Oh, Jayoung
Bae, Hyunsu
Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide
title Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide
title_full Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide
title_fullStr Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide
title_full_unstemmed Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide
title_short Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide
title_sort targeting of m2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6555931/
https://www.ncbi.nlm.nih.gov/pubmed/31174610
http://dx.doi.org/10.1186/s40425-019-0610-4
work_keys_str_mv AT leechanju targetingofm2liketumorassociatedmacrophageswithamelittinbasedproapoptoticpeptide
AT jeonghyunju targetingofm2liketumorassociatedmacrophageswithamelittinbasedproapoptoticpeptide
AT baeyounghyeon targetingofm2liketumorassociatedmacrophageswithamelittinbasedproapoptoticpeptide
AT shinkyungmoon targetingofm2liketumorassociatedmacrophageswithamelittinbasedproapoptoticpeptide
AT kangsinwoo targetingofm2liketumorassociatedmacrophageswithamelittinbasedproapoptoticpeptide
AT kimhwikyung targetingofm2liketumorassociatedmacrophageswithamelittinbasedproapoptoticpeptide
AT ohjayoung targetingofm2liketumorassociatedmacrophageswithamelittinbasedproapoptoticpeptide
AT baehyunsu targetingofm2liketumorassociatedmacrophageswithamelittinbasedproapoptoticpeptide