Cargando…

MIIP remodels Rac1-mediated cytoskeleton structure in suppression of endometrial cancer metastasis

BACKGROUND: Endometrial carcinoma (EC) is one of the most common malignancies of the female reproductive system. Migration and invasion inhibitory protein (MIIP) gene was recently discovered candidate tumor suppress gene which located at chromosome 1p36.22. 1p36 deletion was found in many types of t...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Yingmei, Hu, Limei, Ji, Ping, Teng, Fei, Tian, Wenyan, Liu, Yuexin, Cogdell, David, Liu, Jinsong, Sood, Anil K., Broaddus, Russell, Xue, Fengxia, Zhang, Wei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5069779/
https://www.ncbi.nlm.nih.gov/pubmed/27760566
http://dx.doi.org/10.1186/s13045-016-0342-6
_version_ 1782461001250111488
author Wang, Yingmei
Hu, Limei
Ji, Ping
Teng, Fei
Tian, Wenyan
Liu, Yuexin
Cogdell, David
Liu, Jinsong
Sood, Anil K.
Broaddus, Russell
Xue, Fengxia
Zhang, Wei
author_facet Wang, Yingmei
Hu, Limei
Ji, Ping
Teng, Fei
Tian, Wenyan
Liu, Yuexin
Cogdell, David
Liu, Jinsong
Sood, Anil K.
Broaddus, Russell
Xue, Fengxia
Zhang, Wei
author_sort Wang, Yingmei
collection PubMed
description BACKGROUND: Endometrial carcinoma (EC) is one of the most common malignancies of the female reproductive system. Migration and invasion inhibitory protein (MIIP) gene was recently discovered candidate tumor suppress gene which located at chromosome 1p36.22. 1p36 deletion was found in many types of tumor including EC. In the present study, we will determine the role and mechanism of MIIP in EC metastasis. METHODS: Immunohistochemistry was used to measure MIIP expression in normal and EC tissue. Both gain-of-function (infection) and loss-of-function (siRNA) assays were used to alter MIIP expression levels. The effect of MIIP on cell migration and invasion was measured by transwell assay. F-actin immunofluorescence staining was used to observe the cell morphology. The activation of GTP-loaded Rac1 was evaluated by Rac activity assay kit. Immunoprecipitation/WB was used to measure the interaction between MIIP and PAK1. RESULTS: We demonstrate that MIIP expression was significantly decreased in EC patients comparing to the normal ones, and decreased MIIP expression in EC tissues is associated with deep myometrial invasion, advanced stage, and the presence of lymph node metastasis. Using both gain-of-function (infection) and loss-of-function (siRNA) assays, we show that MIIP markedly blocked EC cell migration, whereas loss of MIIP led to increase in EC cell migration. We demonstrate that elevated expression of MIIP resulted in cytoskeleton reorganization with decreased formation of lamellipodia. We also provide evidence that MIIP is a key molecule in directing Rac1 signaling cascades in EC. Ectopically expressed MIIP consistently competed with Rac1-GTP for binding with the PAK1 p21-binding domain. Our data show that MIIP and PAK1 bind each other and that a C-terminal polyproline domain of MIIP is required for PAK1 binding. Deletion of the PAK1-binding domain of MIIP reduced cell migration-inhibiting activity. CONCLUSIONS: MIIP may function as a tumor suppressor gene for endometrial carcinoma. MIIP attenuates Rac1 signaling through a protein interaction network, and loss of this regulator may contribute to EC metastasis. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13045-016-0342-6) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-5069779
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-50697792016-10-24 MIIP remodels Rac1-mediated cytoskeleton structure in suppression of endometrial cancer metastasis Wang, Yingmei Hu, Limei Ji, Ping Teng, Fei Tian, Wenyan Liu, Yuexin Cogdell, David Liu, Jinsong Sood, Anil K. Broaddus, Russell Xue, Fengxia Zhang, Wei J Hematol Oncol Research BACKGROUND: Endometrial carcinoma (EC) is one of the most common malignancies of the female reproductive system. Migration and invasion inhibitory protein (MIIP) gene was recently discovered candidate tumor suppress gene which located at chromosome 1p36.22. 1p36 deletion was found in many types of tumor including EC. In the present study, we will determine the role and mechanism of MIIP in EC metastasis. METHODS: Immunohistochemistry was used to measure MIIP expression in normal and EC tissue. Both gain-of-function (infection) and loss-of-function (siRNA) assays were used to alter MIIP expression levels. The effect of MIIP on cell migration and invasion was measured by transwell assay. F-actin immunofluorescence staining was used to observe the cell morphology. The activation of GTP-loaded Rac1 was evaluated by Rac activity assay kit. Immunoprecipitation/WB was used to measure the interaction between MIIP and PAK1. RESULTS: We demonstrate that MIIP expression was significantly decreased in EC patients comparing to the normal ones, and decreased MIIP expression in EC tissues is associated with deep myometrial invasion, advanced stage, and the presence of lymph node metastasis. Using both gain-of-function (infection) and loss-of-function (siRNA) assays, we show that MIIP markedly blocked EC cell migration, whereas loss of MIIP led to increase in EC cell migration. We demonstrate that elevated expression of MIIP resulted in cytoskeleton reorganization with decreased formation of lamellipodia. We also provide evidence that MIIP is a key molecule in directing Rac1 signaling cascades in EC. Ectopically expressed MIIP consistently competed with Rac1-GTP for binding with the PAK1 p21-binding domain. Our data show that MIIP and PAK1 bind each other and that a C-terminal polyproline domain of MIIP is required for PAK1 binding. Deletion of the PAK1-binding domain of MIIP reduced cell migration-inhibiting activity. CONCLUSIONS: MIIP may function as a tumor suppressor gene for endometrial carcinoma. MIIP attenuates Rac1 signaling through a protein interaction network, and loss of this regulator may contribute to EC metastasis. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13045-016-0342-6) contains supplementary material, which is available to authorized users. BioMed Central 2016-10-19 /pmc/articles/PMC5069779/ /pubmed/27760566 http://dx.doi.org/10.1186/s13045-016-0342-6 Text en © The Author(s). 2016 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
Wang, Yingmei
Hu, Limei
Ji, Ping
Teng, Fei
Tian, Wenyan
Liu, Yuexin
Cogdell, David
Liu, Jinsong
Sood, Anil K.
Broaddus, Russell
Xue, Fengxia
Zhang, Wei
MIIP remodels Rac1-mediated cytoskeleton structure in suppression of endometrial cancer metastasis
title MIIP remodels Rac1-mediated cytoskeleton structure in suppression of endometrial cancer metastasis
title_full MIIP remodels Rac1-mediated cytoskeleton structure in suppression of endometrial cancer metastasis
title_fullStr MIIP remodels Rac1-mediated cytoskeleton structure in suppression of endometrial cancer metastasis
title_full_unstemmed MIIP remodels Rac1-mediated cytoskeleton structure in suppression of endometrial cancer metastasis
title_short MIIP remodels Rac1-mediated cytoskeleton structure in suppression of endometrial cancer metastasis
title_sort miip remodels rac1-mediated cytoskeleton structure in suppression of endometrial cancer metastasis
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5069779/
https://www.ncbi.nlm.nih.gov/pubmed/27760566
http://dx.doi.org/10.1186/s13045-016-0342-6
work_keys_str_mv AT wangyingmei miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT hulimei miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT jiping miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT tengfei miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT tianwenyan miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT liuyuexin miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT cogdelldavid miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT liujinsong miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT soodanilk miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT broaddusrussell miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT xuefengxia miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis
AT zhangwei miipremodelsrac1mediatedcytoskeletonstructureinsuppressionofendometrialcancermetastasis