Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Proc Natl Acad Sci U S A ; 114(8): E1500-E1508, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28174265

ABSTRACT

Many estrogen receptor alpha (ERα)-positive breast cancers initially respond to aromatase inhibitors (AIs), but eventually acquire resistance. Here, we report that serum- and glucocorticoid-inducible kinase 3 (SGK3), a kinase transcriptionally regulated by ERα in breast cancer, sustains ERα signaling and drives acquired AI resistance. SGK3 is up-regulated and essential for endoplasmic reticulum (EnR) homeostasis through preserving sarcoplasmic/EnR calcium ATPase 2b (SERCA2b) function in AI-resistant cells. We have further found that EnR stress response down-regulates ERα expression through the protein kinase RNA-like EnR kinase (PERK) arm, and SGK3 retains ERα expression and signaling by preventing excessive EnR stress. Our study reveals regulation of ERα expression mediated by the EnR stress response and the feed-forward regulation between SGK3 and ERα in breast cancer. Given SGK3 inhibition reduces AI-resistant cell survival by eliciting excessive EnR stress and also depletes ERα expression/function, we propose SGK3 inhibition as a potential effective treatment of acquired AI-resistant breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Aromatase Inhibitors/pharmacology , Breast Neoplasms/genetics , Drug Resistance, Neoplasm , Endoplasmic Reticulum Stress/drug effects , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis/genetics , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Down-Regulation , Endoplasmic Reticulum/physiology , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Xenograft Model Antitumor Assays
2.
Arthritis Rheumatol ; 66(5): 1153-64, 2014 May.
Article in English | MEDLINE | ID: mdl-24782179

ABSTRACT

OBJECTIVE: Engagement of Fcγ receptor IIb (FcγRIIb) suppresses B cell activation and represents a promising target for therapy in autoimmunity. The aim of this study was to characterize B cell immunosuppression mediated by the Fc-engineered antibody, XmAb5871, which coengages FcγRIIb with the B cell antigen receptor (BCR) complex and that is currently in clinical development for the treatment of rheumatoid arthritis (RA). Because rheumatoid factor (RF) might interfere with the binding of XmAb5871 to FcγRIIb, we correlated RF titers with the potency of XmAb5871. METHODS: We analyzed the expression of CD19, FcγRIIb, and CD86 on naive and memory B cells from 50 patients with RA and 66 healthy donors, quantified XmAb5871-induced promotion of FcγRIIb phosphorylation and suppression of calcium flux in activated B cells, measured CD86 inhibition in whole blood, and correlated RF and anti-citrullinated protein antibody (ACPA) levels with drug potency. We engrafted RA peripheral blood mononuclear cells (PBMCs) into SCID mice, treated them with XmAb5871, and quantified human total IgG, total IgM, and anti-tetanus IgG antibody levels in vivo. RESULTS: B cells from all donors expressed CD19 and FcγRIIb, and the expression of FcγRIIb was higher on naive, but not memory, B cells from donors with RA compared with healthy donors. BCR-mediated calcium flux was suppressed by XmAb5871 and was associated with FcγRIIb phosphorylation. XmAb5871 inhibited CD86 induction, and the levels of RF and ACPAs did not affect efficacy. XmAb5871 suppressed B cell activation regardless of disease severity. In SCID mice engrafted with PBMCs from a patient with RA, XmAb5871 suppressed humoral responses. CONCLUSION: Coengagement of the BCR complex and FcγRIIb by XmAb5871 inhibits B cell activation and function. The similar potency in patients with RA and healthy donors and the absence of autoantibody interference suggest that XmAb5871 may represent a new therapeutic strategy to suppress autoreactive B cells in RA.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Antigens, CD19/immunology , Arthritis, Rheumatoid/pathology , B-Lymphocytes/drug effects , Receptors, Antigen, B-Cell/drug effects , Receptors, IgG/drug effects , Animals , Antibodies, Anti-Idiotypic/metabolism , Antigens, CD19/metabolism , Arthritis, Rheumatoid/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B7-2 Antigen/metabolism , C-Reactive Protein/metabolism , Case-Control Studies , Female , Heterografts , Humans , Leukocytes, Mononuclear/pathology , Mice , Mice, SCID , Peptides, Cyclic/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, IgG/metabolism
3.
MAbs ; 5(3): 384-96, 2013.
Article in English | MEDLINE | ID: mdl-23549103

ABSTRACT

The CTLA4-Ig fusion proteins abatacept and belatacept are clinically proven immunosuppressants used for rheumatoid arthritis and renal transplant, respectively. Given that both biologics are typically administered chronically by infusion, a need exists for a next-generation CTLA4-Ig with more convenient dosing. We used structure-based protein engineering to optimize the affinity of existing CTLA4-Ig therapeutics for the ligands CD80 and CD86, and for the neonatal Fc receptor, FcRn. From a rationally designed library, we identified four substitutions that enhanced binding to human CD80 and CD86. Coupled with two IgG1 Fc substitutions that enhanced binding to human FcRn, these changes comprise the novel CTLA4-Ig fusion protein, XPro9523. Compared with abatacept, XPro9523 demonstrated 5.9-fold, 23-fold, and 12-fold increased binding to CD80, CD86, and FcRn, respectively; compared with belatacept, CD80, CD86, and FcRn binding increased 1.5-fold, 7.7-fold, and 11-fold, respectively. XPro9523 and belatacept suppressed human T cell proliferation and IL-2 production more potently than abatacept. XPro9523 also suppressed inflammation in the mouse collagen-induced arthritis model. In cynomolgus monkeys, XPro9523 saturated CD80 and CD86 more effectively than abatacept and belatacept, potently inhibited IgM and IgG immunization responses, and demonstrated longer half-life. Pharmacokinetic modeling of its increased potency and persistence suggests that, in humans, XPro9523 may demonstrate superior efficacy and dosing convenience compared with abatacept and belatacept.


Subject(s)
Arthritis, Experimental/therapy , Arthritis, Rheumatoid/therapy , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Graft Rejection/therapy , Histocompatibility Antigens Class I/metabolism , Immunoconjugates/metabolism , Protein Binding/drug effects , Receptors, Fc/metabolism , Recombinant Fusion Proteins/metabolism , Abatacept , Animals , Antibody Affinity , Antibody Formation/drug effects , B7-1 Antigen/immunology , B7-2 Antigen/immunology , Cells, Cultured , Female , Histocompatibility Antigens Class I/immunology , Humans , Immunoconjugates/genetics , Immunoconjugates/pharmacology , Immunosuppression Therapy , Kidney Transplantation , Lymphocyte Activation/drug effects , Macaca fascicularis , Male , Mice , Mice, Inbred DBA , Mutation/genetics , Protein Binding/immunology , Protein Engineering , Receptors, Fc/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Structure-Activity Relationship
4.
J Nutr ; 141(10): 1805-12, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21880954

ABSTRACT

Previous studies in our laboratory demonstrated that blueberry (BB) extract exhibited antitumor activity against MDA-MB-231 triple negative breast cancer (TNBC) cells and decreased metastatic potential in vitro. The current study tested 2 doses of whole BB powder, 5 and 10% (wt:wt) in the diet, against MDA-MB-231 tumor growth in female nude mice. In this study, tumor volume was 75% lower in mice fed the 5% BB diet and 60% lower in mice fed the 10% BB diet than in control mice (P ≤ 0.05). Tumor cell proliferation (Ki-67) was lower in the 5 and 10% BB-fed mice and cell death (Caspase 3) was greater in the 10% BB-fed mice compared to control mice (P ≤ 0.05). Gene analysis of tumor tissues from the 5% BB-fed mice revealed significantly altered expression of genes important to inflammation, cancer, and metastasis, specifically, Wnt signaling, thrombospondin-2, IL-13, and IFNγ. To confirm effects on Wnt signaling, analysis of tumor tissues from 5% BB-fed mice revealed lower ß-catenin expression and glycogen synthase kinase-3ß phosphorylation with greater expression of the ß-catenin inhibitory protein adenomatous polyposis coli compared to controls. A second study tested the ability of the 5% BB diet to inhibit MDA-MB-231-luc-D3H2LN metastasis in vivo. In this study, 5% BB-fed mice developed 70% fewer liver metastases (P = 0.04) and 25% fewer lymph node metastases (P = 0.09) compared to control mice. This study demonstrates the oral antitumor and metastasis activity of whole BB powder against TNBC in mice.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Blueberry Plants/chemistry , Breast Neoplasms/diet therapy , Dietary Supplements , Fruit/chemistry , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Apoptosis , Blueberry Plants/adverse effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Dietary Supplements/adverse effects , Female , Fruit/adverse effects , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Lymphatic Metastasis/prevention & control , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Random Allocation , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays
5.
J Biochem ; 150(4): 439-50, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21693545

ABSTRACT

We previously isolated phage antibodies from a phage library displaying human single chain antibodies (scFvs) by screening with a mannotriose (Man3)-bearing lipid. Of four independent scFv genes originally characterized, 5A3 gene products were purified as fusion proteins such as a scFv-human IgG1 Fc form, but stable clones secreting 1A4 and 1G4 scFv-Fc proteins had never been established. Thus, bacterial expression systems were used to purify 1A4 and 1G4 scFv gene products as soluble forms. Purification of 1A4 and 1G4 scFv proteins from inclusion bodies was also carried out together with purification of 5A3 scFv protein in order to compare their Man3-binding abilities. The present studies demonstrated that 1A4 and 1G4 scFv proteins have a higher affinity for Man3 than 5A3 scFv protein, which may determine whether scFv-Fc proteins expressed in mammalian cells are retained in the ER or secreted. Furthermore, the inhibitory effects of anti-Man3 1G4 scFv and anti-Tn antigen scFv proteins on MCF-7 cell growth were evaluated. Despite the fact that no obvious difference was detected in cell growth, microscopic observations revealed inhibition of foci formation in cells grown in the presence of the anti-carbohydrate scFv proteins. This finding provides a basis for the development of cancer therapeutics.


Subject(s)
Bacteriophage T7/genetics , Escherichia coli/genetics , Mannose/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Antineoplastic Agents/analysis , Antineoplastic Agents/pharmacology , Humans , Mannose/chemistry , Single-Chain Antibodies/analysis , Single-Chain Antibodies/pharmacology , Tumor Cells, Cultured
6.
Mol Endocrinol ; 25(1): 72-82, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21084382

ABSTRACT

Serum- and glucocorticoid-inducible kinase 3 (SGK3) is a protein kinase of the AGC family of protein kinase A, protein kinase G, and protein kinase C and functions downstream of phosphatidylinositol 3-kinase (PI3K). Recent study revealed that SGK3 plays a pivotal role in Akt/protein kinase B independent signaling downstream of oncogenic PI3KCA mutations in breast cancer. Here we report that SGK3 is an estrogen receptor (ER) transcriptional target and promotes estrogen-mediated cell survival of ER-positive breast cancer cells. Through a meta-analysis on 22 microarray studies of breast cancer in the Oncomine database, we found that the expression of SGK3 is significantly higher (5.7-fold, P < 0.001) in ER-positive tumors than in ER-negative tumors. In ER-positive breast cancer cells, SGK3 expression was found to be induced by 17ß-estradiol (E(2)) in a dose- and time-dependent manner, and the induction of SGK3 mRNA by E(2) is independent of newly synthesized proteins. We identified two ERα-binding regions at the sgk3 locus through chromatin immunoprecipitation with massively parallel DNA sequencing. Promoter analysis revealed that ERα stimulates the activity of sgk3 promoters by interaction with these two ERα-binding regions on E(2) treatment. Loss-of-function analysis indicated that SGK3 is required for E(2)-mediated cell survival of MCF-7 breast carcinoma cells. Moreover, overexpression of SGK3 could partially protect MCF-7 cells against apoptosis caused by antiestrogen ICI 182,780. Together, our study defines the molecular mechanism of regulation of SGK3 by estrogen/ER and provides a new link between the PI3K pathway and ER signaling as well as a new estrogen-mediated cell survival mechanism mediated by SGK3 in breast cancer cells.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Estrogens/pharmacology , Protein Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Cytoprotection/drug effects , Enzyme Induction/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fulvestrant , Genetic Loci/genetics , Humans , Models, Biological , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects
7.
Cancer Res ; 70(9): 3594-605, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20388778

ABSTRACT

Dietary phytochemicals are known to exhibit a variety of anticarcinogenic properties. This study investigated the chemopreventive activity of blueberry extract in triple-negative breast cancer cell lines in vitro and in vivo. Blueberry decreased cell proliferation in HCC38, HCC1937, and MDA-MB-231 cells with no effect on the nontumorigenic MCF-10A cell line. Decreased metastatic potential of MDA-MB-231 cells by blueberry was shown through inhibition of cell motility using wound-healing assays and migration through a polyethylene terephthalate membrane. Blueberry treatment decreased the activity of matrix metalloproteinase-9 and the secretion of urokinase-type plasminogen activator while increasing tissue inhibitor of metalloproteinase-1 and plasminogen activator inhibitor-1 secretion in MDA-MB-231 conditioned medium as shown by Western blotting. Cell signaling pathways that control the expression/activation of these processes were investigated via Western blotting and reporter gene assay. Treatment with blueberry decreased phosphatidylinositol 3-kinase (PI3K)/AKT and NFkappaB activation in MDA-MB-231 cells, where protein kinase C and extracellular signal-regulated kinase (ERK) were not affected. In vivo, the efficacy of blueberry to inhibit triple-negative breast tumor growth was evaluated using the MDA-MB-231 xenograft model. Tumor weight and proliferation (Ki-67 expression) were decreased in blueberry-treated mice, where apoptosis (caspase-3 expression) was increased compared with controls. Immunohistochemical analysis of tumors from blueberry-fed mice showed decreased activation of AKT and p65 NFkappaB signaling proteins with no effect on the phosphorylation of ERK. These data illustrate the inhibitory effect of blueberry phytochemicals on the growth and metastatic potential of MDA-MB-231 cells through modulation of the PI3K/AKT/NFkappaB pathway.


Subject(s)
Blueberry Plants , Breast Neoplasms/drug therapy , Fruit/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/pharmacology , Animals , Apoptosis/drug effects , Beverages , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Female , Flavonoids/analysis , Flavonoids/pharmacology , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred BALB C , Mice, Nude , Phenols/analysis , Phenols/pharmacology , Plant Extracts/analysis , Plasminogen Activator Inhibitor 1/metabolism , Polyphenols , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Xenograft Model Antitumor Assays
8.
Breast Cancer Res Treat ; 124(1): 89-99, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20054641

ABSTRACT

Resistance to endocrine therapy agents has presented a clinical obstacle in the treatment of hormone-dependent breast cancer. Our laboratory has initiated a study of microRNA regulation of signaling pathways that may result in breast cancer progression on aromatase inhibitors (AI). Microarray analysis of hormone refractory cell lines identified 115 differentially regulated microRNAs, of which 49 microRNAs were believed to be hormone-responsive. A group of microRNAs were inversely expressed in the AI-resistant lines versus LTEDaro and tamoxifen-resistant. We focused our work on hsa-miR-128a which was hormone-responsive and selectively up-regulated in the letrozole-resistant cell lines. Human miR-128a was predicted to target the TGFß signaling pathway and indeed sensitivity to TGFß was compromised in the letrozole-resistant cells, as compared to parental MCF-7aro. Human miR-128a was shown to negatively target TGFßRI protein expression by binding to the 3'UTR region of the gene. Inhibition of endogenous miR-128a resulted in resensitization of the letrozole-resistant lines to TGFß growth inhibitory effects. These data suggest that the hormone-responsive miR-128a can modulate TGFß signaling and survival of the letrozole-resistant cell lines. To our knowledge, this is the first study to address the role of microRNA regulation as well as TGFß signaling in AI-resistant breast cancer cell lines. We believe that in addition to estrogen-modulation of gene expression, hormone-regulated microRNAs may provide an additional level of post-transcriptional regulation of signaling pathways critically involved in breast cancer progression and AI-resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Aromatase Inhibitors/pharmacology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , MicroRNAs/metabolism , Nitriles/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Triazoles/pharmacology , 3' Untranslated Regions , Aromatase/genetics , Aromatase/metabolism , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Estradiol/metabolism , Female , Gene Expression Profiling/methods , Humans , Letrozole , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Testosterone/metabolism , Transfection
9.
J Steroid Biochem Mol Biol ; 118(4-5): 277-82, 2010 Feb 28.
Article in English | MEDLINE | ID: mdl-19897035

ABSTRACT

To determine potential genes involved in mediating resistance to aromatase inhibitors (AIs), a microarray study was performed using MCF-7aro (aromatase overexpressing) cells that are resistant to letrozole (T+LET R), anastrozole (T+ANA R) and exemestane (T+EXE R), as well as LTEDaro and tamoxifen-resistant (T+TAM R) lines for comparison. Based on hierarchical clustering, estrogen-responsive genes were found to be differentially expressed in AI-resistant lines versus LTEDaro and T+TAM R. Additional genome-wide analysis showed that gene expression profiles of the non-steroidal AI-resistant lines were most closely correlated and that T+EXE R lines exhibit differing profiles. Also, LTEDaro and T+TAM R lines are inherently different from expression profiles of AI-resistant lines. Further characterization of these resistant lines revealed that T+LET R, T+ANA R and LTEDaro cells contain a constitutively active estrogen receptor alpha (ERalpha) that does not require the ligand estrogen for activation. Ligand-independent activation of ERalpha does not activate identical estrogen-responsive gene profiles in AI-resistant lines as in LTEDaro lines, thereby establishing differing mechanisms of resistance. This ligand-independent activation of ER was not observed in the parental cell lines MCF-7aro, T+EXE R or T+TAM R cells. Based on the steroidal structure of EXE, our laboratory has shown that this AI has weak estrogen-like properties, and that EXE resistance involves an ER-dependent crosstalk with EGFR growth factor signaling. Recent studies in our laboratory pertaining to pre-clinical models of AI treatment revealed that intermittent use of EXE delays the onset of acquired resistance in comparison to continuous treatment. Specific molecular mechanisms involved in intermittent use of EXE are currently being explored, based on microarray gene expression profiling. Lastly, our laboratory has initiated a study of microRNAs and their potential role in regulating target genes involved in AI-resistance. Overall, we propose a model of acquired resistance that progresses from hormone-dependence (T+TAM R and T+EXE R) to hormone-independence (T+LET R and T+ANA R), eventually resulting in hormone-independence that does not rely on conventional ER signaling (LTEDaro).


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors/therapeutic use , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Drug Resistance, Neoplasm , Gene Expression Profiling , Anastrozole , Androstadienes/therapeutic use , Aromatase/genetics , Aromatase/metabolism , Breast Neoplasms/drug therapy , Female , Gene Expression Regulation, Neoplastic , Humans , Letrozole , Nitriles/therapeutic use , Tamoxifen/therapeutic use , Triazoles/therapeutic use
10.
Breast Cancer Res Treat ; 116(3): 461-70, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18677558

ABSTRACT

Third generation aromatase inhibitors (AI) have shown good clinical efficacy in comparison to the anti-estrogen tamoxifen. The steroidal AI, exemestane (EXE) has previously been shown to act as an androgen, but this report demonstrates the estrogen-like activity of EXE. Based on genome-wide microarray analysis, high correlation was seen between EXE-Only (EXE O, hormone-free) and hormone-containing AI-resistant lines. In addition, the top regulated genes in the EXE O lines were mostly estrogen-responsive genes. This estrogen-like activity of EXE was further validated using estrogen receptor (ER) activity assays, where in comparison to 17beta-estradiol (E2), EXE was able to induce ER activity, though at a higher concentration. Also, this EXE-mediated ER activity was blocked by the ER antagonist ICI as well as the ERalpha-specific antagonist methyl-piperidino-pyrazole (MPP). Similarly, EXE was able to induce proliferation of breast cancer cell lines, MCF-7 and MCF-7aro, as well as activate transcription of known estrogen-responsive genes, i.e., PGR, pS2 and AREG. These results suggest that EXE does have weak estrogen-like activity.


Subject(s)
Androstadienes/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Anastrozole , Aromatase Inhibitors/pharmacology , Breast Neoplasms/pathology , Cell Proliferation , Estrogen Receptor alpha/genetics , Female , Gene Expression Profiling , Humans , Microarray Analysis , Nitriles/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Triazoles/pharmacology , Tumor Cells, Cultured
11.
Nutr Cancer ; 60(6): 744-56, 2008.
Article in English | MEDLINE | ID: mdl-19005974

ABSTRACT

White button mushrooms are a widely consumed food containing phytochemicals beneficial to cancer prevention. The purpose of this research was to evaluate the effects of white button mushroom extract and its major component, conjugated linoleic acid (CLA) on prostate cancer cell lines in vitro and mushroom extract in vivo. In all cell lines tested, mushroom inhibited cell proliferation in a dose-dependent manner and induced apoptosis within 72 h of treatment. CLA inhibited proliferation in the prostate cancer cell lines in vitro. DU145 and PC3 prostate tumor size and tumor cell proliferation were decreased in nude mice treated with mushroom extract, whereas tumor cell apoptosis was increased compared to pair-fed controls. Microarray analysis of tumors identified significant changes in gene expression in the mushroom-fed mice as compared to controls. Gene network analysis identified alterations in networks involved in cell death, growth and proliferation, lipid metabolism, the TCA cycle and immune response. The data provided by this study illustrate the anticancer potential of phytochemicals in mushroom extract both in vitro and in vivo and supports the recommendation of white button mushroom as a dietary component that may aid in the prevention of prostate cancer in men.


Subject(s)
Agaricus , Apoptosis , Linoleic Acids, Conjugated/pharmacology , Prostatic Neoplasms/prevention & control , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Citric Acid Cycle , Dinoprostone/antagonists & inhibitors , Humans , Linoleic Acids, Conjugated/therapeutic use , Male , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Phosphatidylserines/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
12.
Cancer Res ; 68(12): 4910-8, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559539

ABSTRACT

Acquired resistance to either tamoxifen or aromatase inhibitors (AI) develops after prolonged treatment in a majority of hormone-responsive breast cancers. In an attempt to further elucidate mechanisms of acquired resistance to AIs, MCF-7aro cells resistant to letrozole (T+LET R), anastrozole (T+ANA R), and exemestane (T+EXE R), as well as long-term estrogen deprived (LTEDaro) and tamoxifen-resistant (T+TAM R) lines were generated. This is the first complete panel of endocrine therapy-resistant cell lines, which were generated as multiple independent biological replicates for unbiased genome-wide analysis using affymetrix microarrays. Although similarities are apparent, microarray results clearly show gene signatures unique to AI-resistance were inherently different from LTEDaro and T+TAM R gene expression profiles. Based on hierarchical clustering, unique estrogen-responsive gene signatures vary depending on cell line, with some genes up-regulated in all lines versus other genes up-regulated only in the AI-resistant lines. Characterization of these resistant lines showed that LTEDaro, T+LET R, and T+ANA R cells contained a constitutively active estrogen receptor (ER)alpha that does not require estrogen for activation. This ligand-independent activation of ER was not observed in the parental cells, as well as T+EXE R and T+TAM R cells. Further characterization of these resistant lines was performed using cell cycle analysis, immunofluorescence experiments to visualize ER subcellular localization, as well as cross-resistance studies to determine second-line inhibitor response. Using this well-defined model system, our studies provide important information regarding differences in resistance mechanisms to AIs, TAM, and LTEDaro, which are critical in overcoming resistance when treating hormone-responsive breast cancers.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Estrogens/deficiency , Genome, Human , Tamoxifen/pharmacology , Aromatase/metabolism , Blotting, Western , Breast Neoplasms/pathology , Cell Cycle/drug effects , Chromatin Immunoprecipitation , Estrogen Receptor alpha/genetics , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Microarray Analysis , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
13.
Cancer Res ; 68(7): 2259-65, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381432

ABSTRACT

Exemestane-resistant breast cancer cell lines (i.e., ExeR), derived from MCF-7 cells expressing a high level of aromatase (MCF-7aro), were generated in our laboratory. The epidermal growth factor (EGF)-like protein amphiregulin (AREG) was highly expressed in ExeR cells based on cDNA microarray analysis. The high levels of AREG mRNA in ExeR cell lines were confirmed by real-time reverse transcription-PCR. The high levels of AREG protein in ExeR cell lysates and culture media were confirmed by Western blot analysis and ELISA, respectively. Furthermore, our Western blot analysis showed that whereas no AREG was detected in the DMSO control, overnight treatment of parental MCF-7aro cells with 1 micromol/L exemestane strongly induced the expression of AREG. This induction was totally blocked by 100 nmol/L of pure antiestrogen ICI 182,780, implying estrogen receptor (ER) dependence of exemestane-induced AREG expression. MCF-7aro cells were not able to proliferate in hormone-free medium, but were able to proliferate in conditioned medium from ExeR cells, similar to the treatment of recombinant human AREG. Small interference RNA targeting AREG inhibited ExeR proliferation, confirming that AREG is truly functioning as a growth factor of ExeR cells. The specific inhibitors to ER (ICI 182,780), EGF receptor (EGFR; AG1478), and mitogen-activated protein kinase (MAPK; U0126) all showed dose-dependent suppression of the proliferation of ExeR cells, indicating the involvement of the ER, EGFR, and MAPK pathways. Based on these findings, we propose a possible mechanism that underlies exemestane resistance: exemestane induces AREG in an ER-dependent manner. AREG then activates the EGFR pathway and leads to the activation of the MAPK pathway that drives cell proliferation.


Subject(s)
Androstadienes/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Glycoproteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Amphiregulin , Androstadienes/antagonists & inhibitors , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , EGF Family of Proteins , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Glycoproteins/antagonists & inhibitors , Glycoproteins/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , RNA, Small Interfering/genetics , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Recombinant Proteins/pharmacology
14.
J Steroid Biochem Mol Biol ; 106(1-5): 8-15, 2007.
Article in English | MEDLINE | ID: mdl-17611102

ABSTRACT

Clinical trials have demonstrated the importance of aromatase inhibitor (AI) therapy in the effective treatment of hormone-dependent breast cancers. In contrast to tamoxifen, an antagonist of the estrogen receptor (ER), AIs have shown to be better tolerated along with decreased recurrence rates of the disease. Currently, three third-generation AIs are being used: exemestane, letrozole, and anastrozole. Our laboratory is attempting to understand several aspects of AI functionality. In this paper, we first review recent findings from our structure-function studies of aromatase as well as the molecular characterization of the interaction between AIs and aromatase. Based on these studies, we propose new evidence for the interaction of letrozole and exemestane with aromatase. In addition, we will discuss recent results generated from our AI-resistant cell lines. Our laboratory has generated MCF-7aro cells that are resistant to letrozole, anastrozole, exemestane, and tamoxifen. Basic functional characterization of aromatase and ERalpha in these resistant cell lines has been done and microarray analysis has been employed in order to better understand the mechanism responsible for AI resistance on a genome-wide scale. The results generated so far suggest the presence of at least four types of resistant cell lines. Overall, the information presented in this paper supplements our understanding of AI function, and such information can be valuable for the development of treatment strategies against AI resistant breast cancers.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/metabolism , Drug Resistance/drug effects , Models, Biological , Aromatase/chemistry , Aromatase Inhibitors/chemistry , Humans , Tamoxifen/pharmacology
15.
Cancer Res ; 66(24): 12026-34, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17178902

ABSTRACT

White button mushrooms (Agaricus bisporous) are a potential breast cancer chemopreventive agent, as they suppress aromatase activity and estrogen biosynthesis. Therefore, we evaluated the activity of mushroom extracts in the estrogen receptor-positive/aromatase-positive MCF-7aro cell line in vitro and in vivo. Mushroom extract decreased testosterone-induced cell proliferation in MCF-7aro cells but had no effect on MCF-10A, a nontumorigenic cell line. Most potent mushroom chemicals are soluble in ethyl acetate. The major active compounds found in the ethyl acetate fraction are unsaturated fatty acids such as linoleic acid, linolenic acid, and conjugated linoleic acid. The interaction of linoleic acid and conjugated linoleic acid with aromatase mutants expressed in Chinese hamster ovary cells showed that these fatty acids inhibit aromatase with similar potency and that mutations at the active site regions affect its interaction with these two fatty acids. Whereas these results suggest that these two compounds bind to the active site of aromatase, the inhibition kinetic analysis indicates that they are noncompetitive inhibitors with respect to androstenedione. Because only conjugated linoleic acid was found to inhibit the testosterone-dependent proliferation of MCF-7aro cells, the physiologically relevant aromatase inhibitors in mushrooms are most likely conjugated linoleic acid and its derivatives. The in vivo action of mushroom chemicals was shown using nude mice injected with MCF-7aro cells. The studies showed that mushroom extract decreased both tumor cell proliferation and tumor weight with no effect on rate of apoptosis. Therefore, our studies illustrate the anticancer activity in vitro and in vivo of mushroom extract and its major fatty acid constituents.


Subject(s)
Aromatase Inhibitors/isolation & purification , Aromatase Inhibitors/pharmacology , Breast Neoplasms/prevention & control , Phytotherapy , Plant Extracts/pharmacology , Agaricus , Animals , Anticarcinogenic Agents/isolation & purification , Anticarcinogenic Agents/pharmacology , Aromatase/metabolism , CHO Cells , Cell Line, Tumor , Cricetinae , Female , Humans , Kinetics , Mice , Mice, Nude , Placenta/drug effects , Placenta/physiology , Pregnancy , Transplantation, Heterologous
16.
J Steroid Biochem Mol Biol ; 102(1-5): 232-40, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17055257

ABSTRACT

Clinical trials have demonstrated the importance of aromatase inhibitor (AI) therapy in the effective treatment of hormone-dependent breast cancers. Yet, as with all prolonged drug therapy, resistance to aromatase inhibitors does develop. To date, the precise mechanism responsible for resistance to aromatase inhibitors is not completely understood. In this paper, several mechanisms of de novo/intrinsic resistance and acquired resistance to AIs are discussed. These mechanisms are hypothesized based on important findings from a number of laboratories. To better understand this question, our lab has generated, in vitro, breast cancer cell lines that are resistant to aromatase inhibitors. Resistant cell lines were generated over a prolonged period of time using the MCF-7aro (aromatase overexpressed) breast cancer line. These cell lines are resistant to the aromatase inhibitors letrozole, anastrozole and exemestane and the anti-estrogen tamoxifen, for comparison. Two types of resistant cell lines have been generated, those that grow in the presence of testosterone (T) which is needed for cell growth, and resistant lines that are cultured in the presence of inhibitor only (no T). In addition to functional characterization of aromatase and ERalpha in these resistant cell lines, microarray analysis has been employed in order to determine differential gene expression within the aromatase inhibitor resistant cell lines versus tamoxifen, in order to better understand the mechanism responsible for AI resistance on a genome-wide scale. We anticipate that our studies will generate important information on the mechanisms of AI resistance. Such information can be valuable for the development of treatment strategies against AI-resistant breast cancers.


Subject(s)
Aromatase Inhibitors/therapeutic use , Aromatase/chemistry , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Animals , Aromatase/metabolism , Breast Neoplasms/enzymology , Humans
17.
Cancer Res ; 66(11): 5960-7, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740737

ABSTRACT

Aromatase is the enzyme that converts androgen to estrogen. It is expressed at higher levels in breast cancer tissues than normal breast tissues. Grape seed extract (GSE) contains high levels of procyanidin dimers that have been shown in our laboratory to be potent inhibitors of aromatase. In this study, GSE was found to inhibit aromatase activity in a dose-dependent manner and reduce androgen-dependent tumor growth in an aromatase-transfected MCF-7 (MCF-7aro) breast cancer xenograft model, agreeing with our previous findings. We have also examined the effect of GSE on aromatase expression. Reverse transcription-PCR experiments showed that treatment with 60 mug/mL of GSE suppressed the levels of exon I.3-, exon PII-, and exon I.6-containing aromatase mRNAs in MCF-7 and SK-BR-3 cells. The levels of exon I.1-containing mRNA, however, did not change with GSE treatment. Transient transfection experiments with luciferase-aromatase promoter I.3/II or I.4 reporter vectors showed the suppression of the promoter activity in a dose-dependent manner. The GSE treatment also led to the down-regulation of two transcription factors, cyclic AMP-responsive element binding protein-1 (CREB-1) and glucocorticoid receptor (GR). CREB-1 and GR are known to up-regulate aromatase gene expression through promoters I.3/II and I.4, respectively. We believe that these results are exciting in that they show GSE to be potentially useful in the prevention/treatment of hormone-dependent breast cancer through the inhibition of aromatase activity as well as its expression.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/biosynthesis , Breast Neoplasms/enzymology , Plant Extracts/pharmacology , Vitis , Animals , Aromatase/genetics , Aromatase/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Promoter Regions, Genetic , Seeds , Xenograft Model Antitumor Assays
18.
Cancer Res ; 63(23): 8516-22, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14679019

ABSTRACT

In breast cancer, in situ estrogen production has been demonstrated to play a major role in promoting tumor growth. Aromatase is the enzyme responsible for the conversion of androgen substrates into estrogens. This enzyme is highly expressed in breast cancer tissue compared with normal breast tissue. A wine extract fraction was recently isolated from red wine that exhibited a potent inhibitory action on aromatase activity. Using UV absorbance analysis, high-performance liquid chromatography profiling, accurate mass-mass spectrometry, and nanospray tandem mass spectrometry, most of the compounds in our red wine fraction were identified as procyanidin B dimers that were shown to be aromatase inhibitors. These chemicals have been found in high levels in grape seeds. Inhibition kinetic analysis on the most potent procyanidin B dimer has revealed that it competes with the binding of the androgen substrate with a K(i) value of 6 micro M. Because mutations at Asp-309, Ser-378, and His-480 of aromatase significantly affected the binding of the procyanidin B dimer, these active site residues are thought to be important residues that interact with this phytochemical. The in vivo efficacy of procyanidin B dimers was evaluated in an aromatase-transfected MCF-7 breast cancer xenograft model. The procyanidin B dimers were able to reduce androgen-dependent tumor growth, indicating that these chemicals suppress in situ estrogen formation. These in vitro and in vivo studies demonstrated that procyanidin B dimers in red wine and grape seeds could be used as chemopreventive agents against breast cancer by suppressing in situ estrogen biosynthesis.


Subject(s)
Biflavonoids , Catechin/pharmacology , Estrogen Antagonists/pharmacology , Estrogens/biosynthesis , Proanthocyanidins , Vitis/chemistry , Wine , Animals , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/pharmacology , Aromatase/genetics , Aromatase Inhibitors , Catechin/chemistry , Chromatography, High Pressure Liquid , Dimerization , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Estrogen Antagonists/chemistry , Female , Kinetics , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Nude , Mutagenesis, Site-Directed , Seeds
SELECTION OF CITATIONS
SEARCH DETAIL
...