Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 34(10): 108822, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691110

ABSTRACT

MED1 (mediator subunit 1) co-amplifies with HER2, but its role in HER2-driven mammary tumorigenesis is still unknown. Here, we generate MED1 mammary-specific overexpression mice and cross them with mouse mammary tumor virus (MMTV)-HER2 mice. We observe significantly promoted onset, growth, metastasis, and multiplicity of HER2 tumors by MED1 overexpression. Further studies reveal critical roles for MED1 in epithelial-mesenchymal transition, cancer stem cell formation, and response to anti-HER2 therapy. Mechanistically, RNA sequencing (RNA-seq) transcriptome analyses and clinical sample correlation studies identify Jab1, a component of the COP9 signalosome complex, as the key direct target gene of MED1 contributing to these processes. Further studies reveal that Jab1 can also reciprocally regulate the stability and transcriptional activity of MED1. Together, our findings support a functional cooperation between these co-amplified genes in HER2+ mammary tumorigenesis and their potential usage as therapeutic targets for the treatment of HER2+ breast cancers.


Subject(s)
Mammary Neoplasms, Experimental/pathology , Mediator Complex Subunit 1/metabolism , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/therapeutic use , COP9 Signalosome Complex/antagonists & inhibitors , COP9 Signalosome Complex/genetics , COP9 Signalosome Complex/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Lapatinib/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse/genetics , Mediator Complex Subunit 1/genetics , Mice , Mice, Nude , Neoplasm Metastasis , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Transcriptional Activation
2.
J Zhejiang Univ Sci B ; 20(5): 381-390, 2019 May.
Article in English | MEDLINE | ID: mdl-31090264

ABSTRACT

Breast cancer, one of the most frequent cancer types, is a leading cause of death in women worldwide. Estrogen receptor (ER) α is a nuclear hormone receptor that plays key roles in mammary gland development and breast cancer. About 75% of breast cancer cases are diagnosed as ER-positive; however, nearly half of these cancers are either intrinsically or inherently resistant to the current anti-estrogen therapies. Recent studies have identified an ER coactivator, Mediator Subunit 1 (MED1), as a unique, tissue-specific cofactor that mediates breast cancer metastasis and treatment resistance. MED1 is overexpressed in over 50% of human breast cancer cases and co-amplifies with another important breast cancer gene, receptor tyrosine kinase HER2. Clinically, MED1 expression highly correlates with poor disease-free survival of breast cancer patients, and recent studies have reported an increased frequency of MED1 mutations in the circulating tumor cells of patients after treatment. In this review, we discuss the biochemical characterization of MED1 and its associated MED1/Mediator complex, its crosstalk with HER2 in anti-estrogen resistance, breast cancer stem cell formation, and metastasis both in vitro and in vivo. Furthermore, we elaborate on the current advancements in targeting MED1 using state-of-the-art RNA nanotechnology and discuss the future perspectives as well.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mediator Complex Subunit 1/genetics , Amino Acid Motifs , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Survival , Disease-Free Survival , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Humans , Mutation , Nanotechnology , Neoplasm Metastasis , Neoplastic Stem Cells/cytology , RNA/analysis , RNA Polymerase II/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction
3.
Cancer Res ; 78(2): 422-435, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29187405

ABSTRACT

Studies of the estrogen receptor (ER) coactivator protein Mediator subunit 1 (MED1) have revealed its specific roles in pubertal mammary gland development and potential contributions to breast tumorigenesis, based on coamplification of MED1 and HER2 in certain breast cancers. In this study, we generated a mouse model of mammary tumorigenesis harboring the MMTV-HER2 oncogene and mutation of MED1 to evaluate its role in HER2-driven tumorigenesis. MED1 mutation in its ER-interacting LxxLL motifs was sufficient to delay tumor onset and to impair tumor growth, metastasis, and cancer stem-like cell formation in this model. Mechanistic investigations revealed that MED1 acted directly to regulate ER signaling through the downstream IGF1 pathway but not the AREG pathway. Our findings show that MED1 is critical for HER2-driven breast tumorigenesis, suggesting its candidacy as a disease-selective therapeutic target.Significance: These findings identify an estrogen receptor-binding protein as a critical mediator of HER2-driven breast tumorigenesis, suggesting its candidacy as a disease-selective therapeutic target. Cancer Res; 78(2); 422-35. ©2017 AACR.


Subject(s)
Breast Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/secondary , Mediator Complex Subunit 1/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mediator Complex Subunit 1/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
ACS Nano ; 11(1): 335-346, 2017 01 24.
Article in English | MEDLINE | ID: mdl-27966906

ABSTRACT

Most breast cancers express estrogen receptor (ER) α, and the antiestrogen drug tamoxifen has been widely used for their treatment. Unfortunately, up to half of all ERα-positive tumors have intrinsic or acquired endocrine therapy resistance. Our recent studies revealed that the ER coactivator Mediator Subunit 1 (MED1) plays a critical role in tamoxifen resistance through cross-talk with HER2. Herein, we assembled a three-way junction (3-WJ) pRNA-HER2apt-siMED1 nanoparticle to target HER2-overexpressing human breast cancer via an HER2 RNA aptamer to silence MED1 expression. We found that these ultracompact RNA nanoparticles are very stable under RNase A, serum, and 8 M urea conditions. These nanoparticles specifically bound to HER2-overexpressing breast cancer cells, efficiently depleted MED1 expression, and significantly decreased ERα-mediated gene transcription, whereas point mutations of the HER2 RNA aptamer on these nanoparticles abolished such functions. The RNA nanoparticles not only reduced the growth, metastasis, and mammosphere formation of the HER2-overexpressing breast cancer cells but also sensitized them to tamoxifen treatment. These biosafe nanoparticles efficiently targeted and penetrated into HER2-overexpressing tumors after systemic administration in orthotopic xenograft mouse models. In addition to their ability to greatly inhibit tumor growth and metastasis, these nanoparticles also led to a dramatic reduction in the stem cell content of breast tumors when combined with tamoxifen treatment in vivo. Overall, we have generated multifunctional RNA nanoparticles that specifically targeted HER2-overexpressing human breast cancer, silenced MED1, and overcame tamoxifen resistance.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Silencing/drug effects , Nanoparticles/chemistry , RNA, Viral/chemistry , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/chemistry , Bacteriophage lambda/chemistry , Humans , Particle Size , Surface Properties , Tamoxifen/chemistry , Tumor Cells, Cultured
5.
Methods Mol Biol ; 1296: 225-33, 2015.
Article in English | MEDLINE | ID: mdl-25791605

ABSTRACT

Small non-coding RNAs (sncRNAs) such as small interfering RNAs (siRNAs), microRNAs (miRNAs) and RNA aptamers have recently emerged as highly versatile and valuable tools in disease diagnostics and therapeutics, largely due to their key regulatory functions in many human diseases including cancer, viral infections, genetic disorders, etc. Recent technological advancements as described in the previous chapters have greatly aided the discovery of sncRNAs and their applications for disease detection and therapy. Here, we describe the advantages of using sncRNAs as diagnostic and therapeutic tools, followed by some of the most recent examples of their use and a vision for the future perspectives.


Subject(s)
Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/therapeutic use , Molecular Diagnostic Techniques/methods , RNA, Small Untranslated/genetics , RNA, Small Untranslated/therapeutic use , Humans , Nanotechnology/methods
6.
PLoS One ; 8(7): e70641, 2013.
Article in English | MEDLINE | ID: mdl-23936234

ABSTRACT

Pure anti-estrogen fulvestrant has been shown to be a promising ER antagonist for locally advanced and metastatic breast cancer. Unfortunately, a significant proportion of patients developed resistance to this type of endocrine therapy but the molecular mechanisms governing cellular responsiveness to this agent remain poorly understood. Here, we've reported that knockdown of estrogen receptor coactivator MED1 sensitized fulvestrant resistance breast cancer cells to fulvestrant treatment. We found that MED1 knockdown further promoted cell cycle arrest induced by fulvestrant. Using an orthotopic xenograft mouse model, we found that knockdown of MED1 significantly reduced tumor growth in mice. Importantly, knockdown of MED1 further potentiated tumor growth inhibition by fulvestrant. Mechanistic studies indicated that combination of fulvestrant treatment and MED1 knockdown is able to cooperatively inhibit the expression of ER target genes. Chromatin immunoprecipitation experiments further supported a role for MED1 in regulating the recruitment of RNA polymerase II and transcriptional corepressor HDAC1 on endogenous ER target gene promoter in the presence of fulvestrant. These results demonstrate a role for MED1 in mediating resistance to the pure anti-estrogen fulvestrant both in vitro and in vivo.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Estradiol/analogs & derivatives , Estrogen Receptor Modulators/pharmacology , Gene Silencing , Mediator Complex Subunit 1/genetics , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Estradiol/pharmacology , Female , Fulvestrant , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone Deacetylase 1/metabolism , Humans , Mice , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Receptors, Estrogen/metabolism , Trefoil Factor-1 , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
7.
Int J Biochem Mol Biol ; 4(1): 27-40, 2013.
Article in English | MEDLINE | ID: mdl-23638319

ABSTRACT

RNA Aptamers refer to RNA oligonulceotides that are capable of binding to specific targets with high affinity and specificity. Through a process called Systematic Evolution of Ligands by EXponential enrichment (SELEX), a number of RNA aptamers have been identified against various targets including organic compounds, nucleotides, proteins and even whole cells and organisms. RNA aptamers have proven to be of high therapeutic and diagnostic value with recent FDA approval of the first aptamer drug and additional ones in the clinical pipelines. It has also been found to be a particularly useful tool for cell-type specific delivery of other RNA therapeutics like siRNA. All these establish RNA aptamers as one of the pivotal tools of the emerging RNA nanotechnology field in the fight against human diseases including cancer, viral infections and other diseases. This article summarizes the current advancement in the identification of RNA aptamers and also provides some examples of their therapeutic and diagnostic applications.

SELECTION OF CITATIONS
SEARCH DETAIL
...