Interleukin-32θ inhibits tumor-promoting effects of macrophage-secreted CCL18 in breast cancer

Abstract Background Tumor-associated macrophages can promote breast cancer metastasis by secreting cytokines and growth factors. Interleukin (IL)-32θ, a newly identified IL-32 isoform, was previously shown to down-regulate various proinflammatory factors of macrophages. Here, we report the presence...

Full description

Bibliographic Details
Main Authors: Thu-Huyen Pham, Yesol Bak, Taeho Kwon, Sae-Bom Kwon, Jae-Wook Oh, Jong-Hyung Park, Yang-Kyu Choi, Jin Tae Hong, Do-Young Yoon
Format: Article
Language:English
Published: BMC 2019-05-01
Series:Cell Communication and Signaling
Subjects:
Online Access:http://link.springer.com/article/10.1186/s12964-019-0374-y
id doaj-a4875e9cbde74092bd308f955d9d494d
record_format Article
spelling doaj-a4875e9cbde74092bd308f955d9d494d2020-11-25T03:23:36ZengBMCCell Communication and Signaling1478-811X2019-05-0117111410.1186/s12964-019-0374-yInterleukin-32θ inhibits tumor-promoting effects of macrophage-secreted CCL18 in breast cancerThu-Huyen Pham0Yesol Bak1Taeho Kwon2Sae-Bom Kwon3Jae-Wook Oh4Jong-Hyung Park5Yang-Kyu Choi6Jin Tae Hong7Do-Young Yoon8Department of Bioscience and Biotechnology, Konkuk UniversityDepartment of Bioscience and Biotechnology, Konkuk UniversityPrimate Resource Center, Division of Bioinfrastructure, Korea Research Institute of Bioscience and Biotechnology (KRIBB)Department of Bioscience and Biotechnology, Konkuk UniversityDepartment of Stem Cell and Regenerative Biotechnology, Konkuk UniversityDepartment of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk UniversityDepartment of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk UniversityCollege of Pharmacy and Medical Research Center, Chungbuk National UniversityDepartment of Bioscience and Biotechnology, Konkuk UniversityAbstract Background Tumor-associated macrophages can promote breast cancer metastasis by secreting cytokines and growth factors. Interleukin (IL)-32θ, a newly identified IL-32 isoform, was previously shown to down-regulate various proinflammatory factors of macrophages. Here, we report the presence of IL-32θ in breast cancer tissues and evaluate its effects on macrophage-regulated breast cancer metastasis. Methods RT-qPCR was used to analyze the mRNA expression of IL-32θ, Chemokine (C-C motif) ligand 18 (CCL18) in breast cancer tissues. In vitro cell-based experiments using IL-32θ-expressing MDA-MB-231 cells were conducted to examine the effects of IL-32θ on metastasis and its molecular signaling. In vivo xenograft, immunohistochemistry, and optical imaging models were generated to support in vitro and clinical findings. Results The clinical data displayed opposite expression patterns of CCL18 and IL-32θ mRNA in macrophage-infiltrated breast tumor tissues compared with those in the other tissues tested. In MDA-MB-231 cells, IL-32θ overexpression attenuated migration, invasion, tumor-promoting factors, and increased epithelial markers levels upon treatment with conditioned media from THP-1-derived macrophages. Additionally, IL-32θ expression in a xenograft model led to a remarkable decrease in tumor size and macrophage-stimulated tumor promotion. This inhibition was mediated through a direct interaction with protein kinase C-δ (PKCδ), subsequently eliminating the downstream factors STAT3 and NF-κB. Blocking CCL18 during co-culture of macrophages and breast cancer cells reduced the levels of breast cancer progression-related factors and PKCδ downstream signaling suggesting CCL18 as the main macrophage-secreted factors triggering the signaling pathway inhibited by IL-32θ. Conclusions Our findings demonstrate a novel role of IL-32θ as an intracellular modulator to suppress macrophage-promoted breast cancer progression by targeting CCL18-dependent signaling.http://link.springer.com/article/10.1186/s12964-019-0374-yIL-32θMacrophageBreast cancer metastasisCCL18PKCδSTAT3
collection DOAJ
language English
format Article
sources DOAJ
author Thu-Huyen Pham
Yesol Bak
Taeho Kwon
Sae-Bom Kwon
Jae-Wook Oh
Jong-Hyung Park
Yang-Kyu Choi
Jin Tae Hong
Do-Young Yoon
spellingShingle Thu-Huyen Pham
Yesol Bak
Taeho Kwon
Sae-Bom Kwon
Jae-Wook Oh
Jong-Hyung Park
Yang-Kyu Choi
Jin Tae Hong
Do-Young Yoon
Interleukin-32θ inhibits tumor-promoting effects of macrophage-secreted CCL18 in breast cancer
Cell Communication and Signaling
IL-32θ
Macrophage
Breast cancer metastasis
CCL18
PKCδ
STAT3
author_facet Thu-Huyen Pham
Yesol Bak
Taeho Kwon
Sae-Bom Kwon
Jae-Wook Oh
Jong-Hyung Park
Yang-Kyu Choi
Jin Tae Hong
Do-Young Yoon
author_sort Thu-Huyen Pham
title Interleukin-32θ inhibits tumor-promoting effects of macrophage-secreted CCL18 in breast cancer
title_short Interleukin-32θ inhibits tumor-promoting effects of macrophage-secreted CCL18 in breast cancer
title_full Interleukin-32θ inhibits tumor-promoting effects of macrophage-secreted CCL18 in breast cancer
title_fullStr Interleukin-32θ inhibits tumor-promoting effects of macrophage-secreted CCL18 in breast cancer
title_full_unstemmed Interleukin-32θ inhibits tumor-promoting effects of macrophage-secreted CCL18 in breast cancer
title_sort interleukin-32θ inhibits tumor-promoting effects of macrophage-secreted ccl18 in breast cancer
publisher BMC
series Cell Communication and Signaling
issn 1478-811X
publishDate 2019-05-01
description Abstract Background Tumor-associated macrophages can promote breast cancer metastasis by secreting cytokines and growth factors. Interleukin (IL)-32θ, a newly identified IL-32 isoform, was previously shown to down-regulate various proinflammatory factors of macrophages. Here, we report the presence of IL-32θ in breast cancer tissues and evaluate its effects on macrophage-regulated breast cancer metastasis. Methods RT-qPCR was used to analyze the mRNA expression of IL-32θ, Chemokine (C-C motif) ligand 18 (CCL18) in breast cancer tissues. In vitro cell-based experiments using IL-32θ-expressing MDA-MB-231 cells were conducted to examine the effects of IL-32θ on metastasis and its molecular signaling. In vivo xenograft, immunohistochemistry, and optical imaging models were generated to support in vitro and clinical findings. Results The clinical data displayed opposite expression patterns of CCL18 and IL-32θ mRNA in macrophage-infiltrated breast tumor tissues compared with those in the other tissues tested. In MDA-MB-231 cells, IL-32θ overexpression attenuated migration, invasion, tumor-promoting factors, and increased epithelial markers levels upon treatment with conditioned media from THP-1-derived macrophages. Additionally, IL-32θ expression in a xenograft model led to a remarkable decrease in tumor size and macrophage-stimulated tumor promotion. This inhibition was mediated through a direct interaction with protein kinase C-δ (PKCδ), subsequently eliminating the downstream factors STAT3 and NF-κB. Blocking CCL18 during co-culture of macrophages and breast cancer cells reduced the levels of breast cancer progression-related factors and PKCδ downstream signaling suggesting CCL18 as the main macrophage-secreted factors triggering the signaling pathway inhibited by IL-32θ. Conclusions Our findings demonstrate a novel role of IL-32θ as an intracellular modulator to suppress macrophage-promoted breast cancer progression by targeting CCL18-dependent signaling.
topic IL-32θ
Macrophage
Breast cancer metastasis
CCL18
PKCδ
STAT3
url http://link.springer.com/article/10.1186/s12964-019-0374-y
work_keys_str_mv AT thuhuyenpham interleukin32thinhibitstumorpromotingeffectsofmacrophagesecretedccl18inbreastcancer
AT yesolbak interleukin32thinhibitstumorpromotingeffectsofmacrophagesecretedccl18inbreastcancer
AT taehokwon interleukin32thinhibitstumorpromotingeffectsofmacrophagesecretedccl18inbreastcancer
AT saebomkwon interleukin32thinhibitstumorpromotingeffectsofmacrophagesecretedccl18inbreastcancer
AT jaewookoh interleukin32thinhibitstumorpromotingeffectsofmacrophagesecretedccl18inbreastcancer
AT jonghyungpark interleukin32thinhibitstumorpromotingeffectsofmacrophagesecretedccl18inbreastcancer
AT yangkyuchoi interleukin32thinhibitstumorpromotingeffectsofmacrophagesecretedccl18inbreastcancer
AT jintaehong interleukin32thinhibitstumorpromotingeffectsofmacrophagesecretedccl18inbreastcancer
AT doyoungyoon interleukin32thinhibitstumorpromotingeffectsofmacrophagesecretedccl18inbreastcancer
_version_ 1724605523374899200