Cargando…

Rosiglitazone inhibits metastasis development of a murine mammary tumor cell line LMM3

BACKGROUND: Activation of peroxisome proliferator-activated receptors γ (PPARγ) induces diverse effects on cancer cells. The thiazolidinediones (TZDs), such as troglitazone and ciglitazone, are PPARγ agonists exhibiting antitumor activities; however, the underlying mechanism remains inconclusive. Ro...

Descripción completa

Detalles Bibliográficos
Autores principales: Magenta, Gabriela, Borenstein, Ximena, Rolando, Romina, Jasnis, María Adela
Formato: Texto
Lenguaje:English
Publicado: BioMed Central 2008
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2268944/
https://www.ncbi.nlm.nih.gov/pubmed/18261208
http://dx.doi.org/10.1186/1471-2407-8-47
_version_ 1782151711683510272
author Magenta, Gabriela
Borenstein, Ximena
Rolando, Romina
Jasnis, María Adela
author_facet Magenta, Gabriela
Borenstein, Ximena
Rolando, Romina
Jasnis, María Adela
author_sort Magenta, Gabriela
collection PubMed
description BACKGROUND: Activation of peroxisome proliferator-activated receptors γ (PPARγ) induces diverse effects on cancer cells. The thiazolidinediones (TZDs), such as troglitazone and ciglitazone, are PPARγ agonists exhibiting antitumor activities; however, the underlying mechanism remains inconclusive. Rosiglitazone (RGZ), a synthetic ligand of PPARγ used in the treatment of Type 2 diabetes, inhibits growth of some tumor cells and is involved in other processes related to cancer progression. Opposing results have also been reported with different ligands on tumor cells. The purpose of this study was to determine if RGZ and 15d-PGJ(2 )induce antitumor effects in vivo and in vitro on the murine mammary tumor cell line LMM3. METHODS: The effect on LMM3 cell viability and nitric oxide (NO) production of different doses of RGZ, 15-dPGJ(2), BADGE and GW9662 were determined using the MTS colorimetric assay and the Griess reaction respectively. In vivo effect of orally administration of RGZ on tumor progression was evaluated either on s.c. primary tumors as well as on experimental metastasis. Cell adhesion, migration (wound assay) and invasion in Transwells were performed. Metalloproteinase activity (MMP) was determined by zymography in conditioned media from RGZ treated tumor cells. PPARγ expression was detected by inmunohistochemistry in formalin fixed tumors and by western blot in tumor cell lysates. RESULTS: RGZ orally administered to tumor-bearing mice decreased the number of experimental lung metastases without affecting primary s.c. tumor growth. Tumor cell adhesion and migration, as well as metalloproteinase MMP-9 activity, decreased in the presence of 1 μM RGZ (non-cytotoxic dose). RGZ induced PPARγ protein expression in LMM3 tumors. Although metabolic activity -measured by MTS assay- diminished with 1–100 μM RGZ, 1 μM-treated cells recovered their proliferating capacity while 100 μM treated cells died. The PPARγ antagonist Biphenol A diglicydyl ether (BADGE) did not affect RGZ activity. On the contrary, the specific antagonist GW9662 completely abrogated RGZ-induced decrease in cell viability. A decrease in NO levels was detected in the presence of either 1 or 100 μM RGZ. The natural ligand 15d-PGJ(2 )did not affect metabolic activity although it induced a significant decrease in NO production. CONCLUSION: A significant decrease in the number of experimental LMM3 lung metastasis, but not on primary tumor growth, after oral RGZ administration was observed. In vitro, 100 μMRGZ also reduced cell viability and NO production, while no changes were observed in the presence of 15d-PGJ(2). BADGE did not reverse RGZ effect while the antagonist GW9662 completely abrogated it, suggesting a PPARγ- dependent mechanism. Inhibition of lung metastatic nodules by RGZ administered in vivo, might be associated with the observed decrease in MMP-9 expression, in cell adhesion, migration and invasion. RGZ augmented its expression. PPARγ was detected in cell lysates by western blot and by immunohistochemistry in tumors from RGZ-treated mice. In summary we can suggest that RGZ or any other TZDs might be possible future approaches in the treatment of metastasis of PPARγ-expressing cells.
format Text
id pubmed-2268944
institution National Center for Biotechnology Information
language English
publishDate 2008
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-22689442008-03-19 Rosiglitazone inhibits metastasis development of a murine mammary tumor cell line LMM3 Magenta, Gabriela Borenstein, Ximena Rolando, Romina Jasnis, María Adela BMC Cancer Research Article BACKGROUND: Activation of peroxisome proliferator-activated receptors γ (PPARγ) induces diverse effects on cancer cells. The thiazolidinediones (TZDs), such as troglitazone and ciglitazone, are PPARγ agonists exhibiting antitumor activities; however, the underlying mechanism remains inconclusive. Rosiglitazone (RGZ), a synthetic ligand of PPARγ used in the treatment of Type 2 diabetes, inhibits growth of some tumor cells and is involved in other processes related to cancer progression. Opposing results have also been reported with different ligands on tumor cells. The purpose of this study was to determine if RGZ and 15d-PGJ(2 )induce antitumor effects in vivo and in vitro on the murine mammary tumor cell line LMM3. METHODS: The effect on LMM3 cell viability and nitric oxide (NO) production of different doses of RGZ, 15-dPGJ(2), BADGE and GW9662 were determined using the MTS colorimetric assay and the Griess reaction respectively. In vivo effect of orally administration of RGZ on tumor progression was evaluated either on s.c. primary tumors as well as on experimental metastasis. Cell adhesion, migration (wound assay) and invasion in Transwells were performed. Metalloproteinase activity (MMP) was determined by zymography in conditioned media from RGZ treated tumor cells. PPARγ expression was detected by inmunohistochemistry in formalin fixed tumors and by western blot in tumor cell lysates. RESULTS: RGZ orally administered to tumor-bearing mice decreased the number of experimental lung metastases without affecting primary s.c. tumor growth. Tumor cell adhesion and migration, as well as metalloproteinase MMP-9 activity, decreased in the presence of 1 μM RGZ (non-cytotoxic dose). RGZ induced PPARγ protein expression in LMM3 tumors. Although metabolic activity -measured by MTS assay- diminished with 1–100 μM RGZ, 1 μM-treated cells recovered their proliferating capacity while 100 μM treated cells died. The PPARγ antagonist Biphenol A diglicydyl ether (BADGE) did not affect RGZ activity. On the contrary, the specific antagonist GW9662 completely abrogated RGZ-induced decrease in cell viability. A decrease in NO levels was detected in the presence of either 1 or 100 μM RGZ. The natural ligand 15d-PGJ(2 )did not affect metabolic activity although it induced a significant decrease in NO production. CONCLUSION: A significant decrease in the number of experimental LMM3 lung metastasis, but not on primary tumor growth, after oral RGZ administration was observed. In vitro, 100 μMRGZ also reduced cell viability and NO production, while no changes were observed in the presence of 15d-PGJ(2). BADGE did not reverse RGZ effect while the antagonist GW9662 completely abrogated it, suggesting a PPARγ- dependent mechanism. Inhibition of lung metastatic nodules by RGZ administered in vivo, might be associated with the observed decrease in MMP-9 expression, in cell adhesion, migration and invasion. RGZ augmented its expression. PPARγ was detected in cell lysates by western blot and by immunohistochemistry in tumors from RGZ-treated mice. In summary we can suggest that RGZ or any other TZDs might be possible future approaches in the treatment of metastasis of PPARγ-expressing cells. BioMed Central 2008-02-08 /pmc/articles/PMC2268944/ /pubmed/18261208 http://dx.doi.org/10.1186/1471-2407-8-47 Text en Copyright © 2008 Magenta et al; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( (http://creativecommons.org/licenses/by/2.0) ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Magenta, Gabriela
Borenstein, Ximena
Rolando, Romina
Jasnis, María Adela
Rosiglitazone inhibits metastasis development of a murine mammary tumor cell line LMM3
title Rosiglitazone inhibits metastasis development of a murine mammary tumor cell line LMM3
title_full Rosiglitazone inhibits metastasis development of a murine mammary tumor cell line LMM3
title_fullStr Rosiglitazone inhibits metastasis development of a murine mammary tumor cell line LMM3
title_full_unstemmed Rosiglitazone inhibits metastasis development of a murine mammary tumor cell line LMM3
title_short Rosiglitazone inhibits metastasis development of a murine mammary tumor cell line LMM3
title_sort rosiglitazone inhibits metastasis development of a murine mammary tumor cell line lmm3
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2268944/
https://www.ncbi.nlm.nih.gov/pubmed/18261208
http://dx.doi.org/10.1186/1471-2407-8-47
work_keys_str_mv AT magentagabriela rosiglitazoneinhibitsmetastasisdevelopmentofamurinemammarytumorcelllinelmm3
AT borensteinximena rosiglitazoneinhibitsmetastasisdevelopmentofamurinemammarytumorcelllinelmm3
AT rolandoromina rosiglitazoneinhibitsmetastasisdevelopmentofamurinemammarytumorcelllinelmm3
AT jasnismariaadela rosiglitazoneinhibitsmetastasisdevelopmentofamurinemammarytumorcelllinelmm3