Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-29138646

ABSTRACT

OBJECTIVE: The aim of this review is to characterize current status of global TCM clinical trials registered in ClinicalTrials.gov. METHODS: We examined all the trials registered within ClinicalTrials.gov up to 25 September 2015, focusing on study interventions to identify TCM-related trials, and extracted 1,270 TCM trials from the data set. RESULTS: Overall, 691 (54.4%) trials were acupuncture, and 454 (35.8%) trials were herbal medicines. Differences in TCM trial intervention types were also evident among the specific therapeutic areas. Among all trials, 55.7% that were small studies enrolled <100 subjects, and only 8.7% of completed studies had reported results of trials. As for the location, the United States was second to China in conducting the most TCM trials. CONCLUSION: This review is the first snapshot of the landscape of TCM clinical trials registered in ClinicalTrials.gov, providing the basis for treatment and prevention of diseases within TCM and offering useful information that will guide future research on TCM.

2.
Mol Endocrinol ; 28(6): 846-59, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24678732

ABSTRACT

Transcription of the HER2 oncogene can be repressed by estrogen (E2). We now show that, a splice isoform of the nuclear receptor coactivator AIB1, AIB1-Δ4, is able to reverse E2 repression of HER2 gene expression in breast cancer cells. The first 224 amino acids of AIB1 that are absent in AIB1-Δ4, bind a co-repressor, ANCO1. Using chromatin immunoprecipitation assay approaches in MCF7 and BT474 cell lines, we demonstrate that AIB1 and AIB1-Δ4 can bind to the E2 regulatory site in the first intron of the HER2 gene, after E2 treatment, but only full-length AIB1 recruits ANCO1. Consistent with E2-induced chromatin repression, the AIB1-ANCO1 complex recruits HDAC3 and HDAC4 to the intronic estrogen response element and the proximal promoter acquires the repressive chromatin mark H3K9me3 and loses H3K4me1. In contrast, AIB1-Δ4 does not recruit ANCO 1, HDAC3, or HDAC4 and the proximal promoter retains activation marks of H3K4me1. In cell lines with low levels of ANCO1 (T47D), E2 does not repress HER2 gene transcription but the repressive response can be restored by overexpression of ANCO1. ANCO1 can also repress other E2-responsive genes, indicating that AIB1, AIB1-Δ4 and ANCO1 are important determinants of endocrine and growth factor responsiveness in breast cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Receptor Coactivator 3/physiology , Receptor, ErbB-2/genetics , Repressor Proteins/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chromatin Assembly and Disassembly , Estradiol/physiology , Female , HEK293 Cells , Histone Deacetylases/metabolism , Humans , MCF-7 Cells , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Oncogenes , Protein Binding , Receptor, ErbB-2/metabolism , Response Elements , Signal Transduction , Transcription, Genetic
3.
Mol Endocrinol ; 27(7): 1113-27, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23660594

ABSTRACT

The oncogene nuclear receptor coactivator amplified in breast cancer 1 (AIB1) is a transcriptional coactivator that is overexpressed in various types of human cancers. However, the molecular mechanisms controlling AIB1 expression in the majority of cancers remain unclear. In this study, we identified a novel interacting protein of AIB1, forkhead-box protein G1 (FoxG1), which is an evolutionarily conserved forkhead-box transcriptional corepressor. We show that FoxG1 expression is low in breast cancer cell lines and that low levels of FoxG1 are correlated with a worse prognosis in breast cancer. We also demonstrate that transient overexpression of FoxG1 can suppress endogenous levels of AIB1 mRNA and protein in MCF-7 breast cancer cells. Exogenously expressed FoxG1 in MCF-7 cells also leads to apoptosis that can be rescued in part by AIB1 overexpression. Using chromatin immunoprecipitation, we determined that FoxG1 is recruited to a region of the AIB1 gene promoter previously characterized to be responsible for AIB1-induced, positive autoregulation of transcription through the recruitment of an activating, multiprotein complex, involving AIB1, E2F transcription factor 1, and specificity protein 1. Increased FoxG1 expression significantly reduces the recruitment of AIB1, E2F transcription factor 1 and E1A-binding protein p300 to this region of the endogenous AIB1 gene promoter. Our data imply that FoxG1 can function as a pro-apoptotic factor in part through suppression of AIB1 coactivator transcription complex formation, thereby reducing the expression of the AIB1 oncogene.


Subject(s)
Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Receptor Coactivator 3/genetics , Repressor Proteins/metabolism , Transcription, Genetic , Apoptosis/genetics , Down-Regulation/genetics , E2F1 Transcription Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , MCF-7 Cells , Models, Biological , Nuclear Receptor Coactivator 3/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Stability , Sp1 Transcription Factor/metabolism
4.
PLoS One ; 6(6): e20410, 2011.
Article in English | MEDLINE | ID: mdl-21738574

ABSTRACT

BACKGROUND: Estrogen is a known growth promoter for estrogen receptor (ER)-positive breast cancer cells. Paradoxically, in breast cancer cells that have been chronically deprived of estrogen stimulation, re-introduction of the hormone can induce apoptosis. METHODOLOGY/PRINCIPAL FINDINGS: Here, we sought to identify signaling networks that are triggered by estradiol (E2) in isogenic MCF-7 breast cancer cells that undergo apoptosis (MCF-7:5C) versus cells that proliferate upon exposure to E2 (MCF-7). The nuclear receptor co-activator AIB1 (Amplified in Breast Cancer-1) is known to be rate-limiting for E2-induced cell survival responses in MCF-7 cells and was found here to also be required for the induction of apoptosis by E2 in the MCF-7:5C cells. Proteins that interact with AIB1 as well as complexes that contain tyrosine phosphorylated proteins were isolated by immunoprecipitation and identified by mass spectrometry (MS) at baseline and after a brief exposure to E2 for two hours. Bioinformatic network analyses of the identified protein interactions were then used to analyze E2 signaling pathways that trigger apoptosis versus survival. Comparison of MS data with a computationally-predicted AIB1 interaction network showed that 26 proteins identified in this study are within this network, and are involved in signal transduction, transcription, cell cycle regulation and protein degradation. CONCLUSIONS: G-protein-coupled receptors, PI3 kinase, Wnt and Notch signaling pathways were most strongly associated with E2-induced proliferation or apoptosis and are integrated here into a global AIB1 signaling network that controls qualitatively distinct responses to estrogen.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Estradiol/pharmacology , Proteomics/methods , Apoptosis/genetics , Female , Humans , Immunoprecipitation , Mass Spectrometry , Nuclear Receptor Coactivator 3/genetics , Nuclear Receptor Coactivator 3/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Tumor Cells, Cultured , Wnt Proteins/genetics , Wnt Proteins/metabolism
5.
J Biol Chem ; 286(30): 26813-27, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21636853

ABSTRACT

The oncogene amplified in breast cancer 1 (AIB1) is a nuclear receptor coactivator that plays a major role in the progression of various cancers. We previously identified a splice variant of AIB1 called AIB1-Δ4 that is overexpressed in breast cancer. Using mass spectrometry, we define the translation initiation of AIB1-Δ4 at Met(224) of the full-length AIB1 sequence and have raised an antibody to a peptide representing the acetylated N terminus. We show that AIB1-Δ4 is predominantly localized in the cytoplasm, although leptomycin B nuclear export inhibition demonstrates that AIB1-Δ4 can enter and traffic through the nucleus. Our data indicate an import mechanism enhanced by other coactivators such as p300/CBP. We report that the endogenously and exogenously expressed AIB1-Δ4 is recruited as efficiently as full-length AIB1 to estrogen-response elements of genes, and it enhances estrogen-dependent transcription more effectively than AIB1. Expression of an N-terminal AIB1 protein fragment, which is lost in the AIB1-Δ4 isoform, potentiates AIB1 as a coactivator. This suggests a model whereby the transcriptional activity of AIB1 is squelched by a repressive mechanism utilizing the N-terminal domain and that the increased coactivator function of AIB1-Δ4 is due to the loss of this inhibitory domain. Finally, we show, using Scorpion primer technology, that AIB1-Δ4 expression is correlated with metastatic capability of human cancer cell lines.


Subject(s)
Cell Nucleus/metabolism , Nuclear Receptor Coactivator 3/metabolism , Transcription, Genetic , Active Transport, Cell Nucleus/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , CHO Cells , COS Cells , Cell Nucleus/genetics , Chlorocebus aethiops , Cricetinae , Cricetulus , Cytoplasm/genetics , Cytoplasm/metabolism , Dogs , Fatty Acids, Unsaturated/pharmacology , HEK293 Cells , Humans , Mice , Nuclear Receptor Coactivator 3/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Response Elements/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...