Deletion of mdig enhances H3K36me3 and metastatic potential of the triple negative breast cancer cells

Summary: In this report, we provide evidence showing diminished expression of the mineral dust-induced gene (mdig), a previously identified oncogenic gene, in human triple negative breast cancer (TNBC). Using a mouse model of orthotopic xenograft of the TNBC MDA-MB-231 cells, we demonstrate that mdi...

Full description

Bibliographic Details
Main Authors: Chitra Thakur, Yiran Qiu, Qian Zhang, Nicholas J. Carruthers, Miaomiao Yu, Zhuoyue Bi, Yao Fu, Priya Wadgaonkar, Bandar Almutairy, Akimasa Seno, Paul M. Stemmer, Fei Chen
Format: Article
Language:English
Published: Elsevier 2022-10-01
Series:iScience
Subjects:
Online Access:http://www.sciencedirect.com/science/article/pii/S2589004222013293
_version_ 1811261159221755904
author Chitra Thakur
Yiran Qiu
Qian Zhang
Nicholas J. Carruthers
Miaomiao Yu
Zhuoyue Bi
Yao Fu
Priya Wadgaonkar
Bandar Almutairy
Akimasa Seno
Paul M. Stemmer
Fei Chen
author_facet Chitra Thakur
Yiran Qiu
Qian Zhang
Nicholas J. Carruthers
Miaomiao Yu
Zhuoyue Bi
Yao Fu
Priya Wadgaonkar
Bandar Almutairy
Akimasa Seno
Paul M. Stemmer
Fei Chen
author_sort Chitra Thakur
collection DOAJ
description Summary: In this report, we provide evidence showing diminished expression of the mineral dust-induced gene (mdig), a previously identified oncogenic gene, in human triple negative breast cancer (TNBC). Using a mouse model of orthotopic xenograft of the TNBC MDA-MB-231 cells, we demonstrate that mdig promotes the growth of primary tumors but inhibits metastasis of these cells in vivo. Knockout of mdig resulted in an enhancement of H3K36me3 in the genome and upregulation of some X chromosome-linked genes for cell motility, invasion, and metastasis. Silencing MAGED2, one of the most upregulated and H3K36me3-enriched genes resulted from mdig depletion, can partially reverse the invasive migration of the mdig knockout cells. The anti-metastatic and inhibitory role of mdig on H3K36me3 was cross-validated in another cell line, A549 lung cancer cells. Together, our data suggest that mdig is antagonist against H3K36me3 that enforces expression of genes, such as MAGED2, for cell invasion and metastasis.
first_indexed 2024-04-12T18:58:18Z
format Article
id doaj.art-1c2cbcbf074541f6a44b5dbfde9a3421
institution Directory Open Access Journal
issn 2589-0042
language English
last_indexed 2024-04-12T18:58:18Z
publishDate 2022-10-01
publisher Elsevier
record_format Article
series iScience
spelling doaj.art-1c2cbcbf074541f6a44b5dbfde9a34212022-12-22T03:20:14ZengElsevieriScience2589-00422022-10-012510105057Deletion of mdig enhances H3K36me3 and metastatic potential of the triple negative breast cancer cellsChitra Thakur0Yiran Qiu1Qian Zhang2Nicholas J. Carruthers3Miaomiao Yu4Zhuoyue Bi5Yao Fu6Priya Wadgaonkar7Bandar Almutairy8Akimasa Seno9Paul M. Stemmer10Fei Chen11Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USAStony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USADepartment of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USAInstitute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USADepartment of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA; Cancer Hospital of China Medical University, 44 Xiaoheyan Road, Dadong District, Shenyang, 110042 Liaoning Province, ChinaStony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USAStony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USADepartment of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USADepartment of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA; College of Pharmacy, Al-Dawadmi Campus, Shaqra University, P.O. Box 11961, Riyadh, Saudi ArabiaDepartment of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA; Faculty of Engineering, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, JapanInstitute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USAStony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USA; Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA; Corresponding authorSummary: In this report, we provide evidence showing diminished expression of the mineral dust-induced gene (mdig), a previously identified oncogenic gene, in human triple negative breast cancer (TNBC). Using a mouse model of orthotopic xenograft of the TNBC MDA-MB-231 cells, we demonstrate that mdig promotes the growth of primary tumors but inhibits metastasis of these cells in vivo. Knockout of mdig resulted in an enhancement of H3K36me3 in the genome and upregulation of some X chromosome-linked genes for cell motility, invasion, and metastasis. Silencing MAGED2, one of the most upregulated and H3K36me3-enriched genes resulted from mdig depletion, can partially reverse the invasive migration of the mdig knockout cells. The anti-metastatic and inhibitory role of mdig on H3K36me3 was cross-validated in another cell line, A549 lung cancer cells. Together, our data suggest that mdig is antagonist against H3K36me3 that enforces expression of genes, such as MAGED2, for cell invasion and metastasis.http://www.sciencedirect.com/science/article/pii/S2589004222013293Molecular biologyMolecular mechanism of gene regulationCancer
spellingShingle Chitra Thakur
Yiran Qiu
Qian Zhang
Nicholas J. Carruthers
Miaomiao Yu
Zhuoyue Bi
Yao Fu
Priya Wadgaonkar
Bandar Almutairy
Akimasa Seno
Paul M. Stemmer
Fei Chen
Deletion of mdig enhances H3K36me3 and metastatic potential of the triple negative breast cancer cells
iScience
Molecular biology
Molecular mechanism of gene regulation
Cancer
title Deletion of mdig enhances H3K36me3 and metastatic potential of the triple negative breast cancer cells
title_full Deletion of mdig enhances H3K36me3 and metastatic potential of the triple negative breast cancer cells
title_fullStr Deletion of mdig enhances H3K36me3 and metastatic potential of the triple negative breast cancer cells
title_full_unstemmed Deletion of mdig enhances H3K36me3 and metastatic potential of the triple negative breast cancer cells
title_short Deletion of mdig enhances H3K36me3 and metastatic potential of the triple negative breast cancer cells
title_sort deletion of mdig enhances h3k36me3 and metastatic potential of the triple negative breast cancer cells
topic Molecular biology
Molecular mechanism of gene regulation
Cancer
url http://www.sciencedirect.com/science/article/pii/S2589004222013293
work_keys_str_mv AT chitrathakur deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT yiranqiu deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT qianzhang deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT nicholasjcarruthers deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT miaomiaoyu deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT zhuoyuebi deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT yaofu deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT priyawadgaonkar deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT bandaralmutairy deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT akimasaseno deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT paulmstemmer deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells
AT feichen deletionofmdigenhancesh3k36me3andmetastaticpotentialofthetriplenegativebreastcancercells