Skip to main content
Top
Published in: Cellular Oncology 5/2019

Open Access 01-10-2019 | Thyroid Cancer | Original paper

Interplay between thyroid cancer cells and macrophages: effects on IL-32 mediated cell death and thyroid cancer cell migration

Authors: Yvette J. E. Sloot, Katrin Rabold, Thomas Ulas, Dennis M. De Graaf, Bas Heinhuis, Kristian Händler, Joachim L. Schultze, Mihai G. Netea, Johannes W. A. Smit, Leo A. B. Joosten, Romana T. Netea-Maier

Published in: Cellular Oncology | Issue 5/2019

Login to get access

Abstract

Purpose

Interleukin 32 (IL-32) is a pro-inflammatory cytokine of which different isoforms have been identified. Recently, IL-32 has been shown to act as a potent inducer of cell migration in several types of cancer. Although previous research showed that IL-32 is expressed in differentiated thyroid cancer (TC) cells, the role of IL-32 in TC cell migration has not been investigated. Furthermore, tumour-associated macrophages (TAMs) may play a facilitating role in cancer cell migration. The aim of this study was to explore whether the interaction between TC cells and TAMs results in increased expression of IL-32 in TC cells and to investigate whether this affects TC cell migration.

Methods

TPC-1 cells were co-culture with TC-induced or naive macrophages. Next, transcriptome analysis on TPC-1 cells was performed and supernatants were used for stimulation of TPC-1 cells. IL-32β and IL-32γ were exogenously overexpressed in TPC-1 cells using transient transfection, after which an in vitro gap closure assay was performed to assess cell migration, and the expression of migratory factors was assessed using RT-qPCR.

Results

We found that TC-induced macrophages induced IL-32 expression in TC cells and that TAM-derived TNFα was the main inducer of IL-32β expression in TC cells. Overexpression of IL-32β and IL-32γ did not affect TC cell migration, but increased cell death. Finally, we found that IL-32β overexpression led to increased mRNA expression of the pro-survival cytokine IL-8, while the expression of other migratory factors was not affected.

Conclusions

From our data, we conclude that TAM-derived TNFα induces IL-32β in TC cells. Although IL-32β does not affect TC cell migration, alternative splicing of IL-32 towards the IL-32β isoform may be beneficial for TC cell survival through induction of the pro-survival cytokine IL-8.
Appendix
Available only for authorised users
Literature
13.
17.
go back to reference T.S. Plantinga, I. Costantini, B. Heinhuis, A. Huijbers, G. Semango, B. Kusters, M.G. Netea, A.R. Hermus, J.W. Smit, C.A. Dinarello, L.A. Joosten, R.T. Netea-Maier, A promoter polymorphism in human interleukin-32 modulates its expression and influences the risk and the outcome of epithelial cell-derived thyroid carcinoma. Carcinogenesis 34, 1529–1535 (2013). https://doi.org/10.1093/carcin/bgt092 CrossRefPubMed T.S. Plantinga, I. Costantini, B. Heinhuis, A. Huijbers, G. Semango, B. Kusters, M.G. Netea, A.R. Hermus, J.W. Smit, C.A. Dinarello, L.A. Joosten, R.T. Netea-Maier, A promoter polymorphism in human interleukin-32 modulates its expression and influences the risk and the outcome of epithelial cell-derived thyroid carcinoma. Carcinogenesis 34, 1529–1535 (2013). https://​doi.​org/​10.​1093/​carcin/​bgt092 CrossRefPubMed
18.
21.
go back to reference R.J.W. Arts, T.S. Plantinga, S. Tuit, T. Ulas, B. Heinhuis, M. Tesselaar, Y. Sloot, G.J. Adema, L.A.B. Joosten, J.W.A. Smit, M.G. Netea, J.L. Schultze, R.T. Netea-Maier, Transcriptional and metabolic reprogramming induce an inflammatory phenotype in non-medullary thyroid carcinoma-induced macrophages. Oncoimmunology 5, e1229725 (2016). https://doi.org/10.1080/2162402X.2016.1229725 R.J.W. Arts, T.S. Plantinga, S. Tuit, T. Ulas, B. Heinhuis, M. Tesselaar, Y. Sloot, G.J. Adema, L.A.B. Joosten, J.W.A. Smit, M.G. Netea, J.L. Schultze, R.T. Netea-Maier, Transcriptional and metabolic reprogramming induce an inflammatory phenotype in non-medullary thyroid carcinoma-induced macrophages. Oncoimmunology 5, e1229725 (2016). https://​doi.​org/​10.​1080/​2162402X.​2016.​1229725
22.
go back to reference R.E. Schweppe, J.P. Klopper, C. Korch, U. Pugazhenthi, M. Benezra, J.A. Knauf, J.A. Fagin, L.A. Marlow, J.A. Copland, R.C. Smallridge, B.R. Haugen, Deoxyribonucleic acid profiling analysis of 40 human thyroid cancer cell lines reveals cross-contamination resulting in cell line redundancy and misidentification. J Clin Endocrinol Metab 93, 4331–4341 (2008). https://doi.org/10.1210/jc.2008-1102 CrossRefPubMedPubMedCentral R.E. Schweppe, J.P. Klopper, C. Korch, U. Pugazhenthi, M. Benezra, J.A. Knauf, J.A. Fagin, L.A. Marlow, J.A. Copland, R.C. Smallridge, B.R. Haugen, Deoxyribonucleic acid profiling analysis of 40 human thyroid cancer cell lines reveals cross-contamination resulting in cell line redundancy and misidentification. J Clin Endocrinol Metab 93, 4331–4341 (2008). https://​doi.​org/​10.​1210/​jc.​2008-1102 CrossRefPubMedPubMedCentral
28.
go back to reference F. Vinals, J. Pouyssegur, Confluence of vascular endothelial cells induces cell cycle exit by inhibiting p42/p44 mitogen-activated protein kinase activity. Mol Cell Biol 19, 2763–2772 (1999)CrossRefPubMedPubMedCentral F. Vinals, J. Pouyssegur, Confluence of vascular endothelial cells induces cell cycle exit by inhibiting p42/p44 mitogen-activated protein kinase activity. Mol Cell Biol 19, 2763–2772 (1999)CrossRefPubMedPubMedCentral
34.
go back to reference Y.J.E. Sloot, K. Rabold, M.G. Netea, J.W.A. Smit, N. Hoogerbrugge, R.T. Netea-Maier, Effect of PTEN inactivating germline mutations on innate immune cell function and thyroid cancer-induced macrophages in patients with PTEN hamartoma tumor syndrome. Oncogene (2019). https://doi.org/10.1038/s41388-019-0685-x Y.J.E. Sloot, K. Rabold, M.G. Netea, J.W.A. Smit, N. Hoogerbrugge, R.T. Netea-Maier, Effect of PTEN inactivating germline mutations on innate immune cell function and thyroid cancer-induced macrophages in patients with PTEN hamartoma tumor syndrome. Oncogene (2019). https://​doi.​org/​10.​1038/​s41388-019-0685-x
Metadata
Title
Interplay between thyroid cancer cells and macrophages: effects on IL-32 mediated cell death and thyroid cancer cell migration
Authors
Yvette J. E. Sloot
Katrin Rabold
Thomas Ulas
Dennis M. De Graaf
Bas Heinhuis
Kristian Händler
Joachim L. Schultze
Mihai G. Netea
Johannes W. A. Smit
Leo A. B. Joosten
Romana T. Netea-Maier
Publication date
01-10-2019
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 5/2019
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-019-00457-9

Other articles of this Issue 5/2019

Cellular Oncology 5/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine