Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Adv Exp Med Biol ; 1212: 1-29, 2020.
Article in English | MEDLINE | ID: mdl-30850960

ABSTRACT

Induced pluripotent stem cells (iPSCs) offer great promise in the areas of disease modeling, basic research, drug development, and regenerative medicine. Much of their value comes from the fact that they can be used to create otherwise inaccessible cell types, such as cardiomyocytes, which are genetically matched to a patient or any other individual of interest. A consistent issue plaguing the iPSC platform, however, involves excessive variability exhibited in the differentiated products. This includes discrepancies in genetic, epigenetic, and transcriptional features, cell signalling, the cell types produced from cardiac differentiation, and cardiomyocyte functionality. These properties can result from both the somatic source cells and environmental conditions related to the derivation and handling of these cells. Understanding the potential sources of variability, along with determining which factors are most relevant to a given application, are essential in advancing iPSC-based technologies.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Regenerative Medicine , Cell Differentiation , Humans
2.
Stem Cells Dev ; 29(2): 75-89, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31744402

ABSTRACT

The functional maturation status of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) has a notable impact upon their use in pharmacological studies, disease modeling, and therapeutic applications. Non-cardiomyocytes (non-CMs) produced in the differentiation process have previously been identified as having an extrinsic influence upon hiPSC-CM development, yet the underlying mechanisms are not fully understood. Herein, we aimed to modulate electrophysiological properties of hiPSC-CMs within co-cultures containing varied proportions of non-CMs and investigate the nature of interactions between these different cell types. Therefore, we sorted cardiac differentiations on day 10 and subsequently replated the cells at ratios of 7:3, 1:1, 3:7, and 1:9 non-CMs to CMs. After a month of co-culture, we evaluated electrophysiological properties through the genetically encoded voltage indicator ArcLight. We ultimately identified that co-cultures with approximately 70%-90% CM purity demonstrated the highest action potential (AP) amplitude and maximum upstroke velocity by day 40 of differentiation, indicative of enhanced electrophysiological maturation, as well as more ventricular-like AP morphologies. Notably, these findings were distinct from those observed for co-cultures of hiPSC-CMs and dermal fibroblasts. We determined that the co-culture phenotypes could not be attributed to paracrine effects of non-CMs due to the inability of conditioned media to recapitulate the observed effects. This led to the further observation of a distinctive expression pattern of connexin 43 (Cx43) at cell-cell interfaces between both CMs and non-CMs. Depletion of Cx43 by short hairpin RNA (shRNA) specifically in the non-CM population within a co-culture environment was able to recapitulate electrophysiological phenotypes of a purer hiPSC-CM population. Collectively, our data demonstrate that abundant non-CM content exerts a significant negative influence upon the electrophysiological maturation of hiPSC-CMs through Cx43-mediated cell-cell-contacts, and thus should be considered regarding the future production of purpose-built hiPSC-CM systems.


Subject(s)
Action Potentials/physiology , Cell Communication/physiology , Cell Differentiation/physiology , Connexin 43/metabolism , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Cells, Cultured , Connexin 43/genetics , Culture Media, Conditioned/pharmacology , Electrophysiological Phenomena/drug effects , Female , Fluorescent Antibody Technique , Gene Expression , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Patch-Clamp Techniques
3.
Stem Cells Transl Med ; 6(10): 1829-1839, 2017 10.
Article in English | MEDLINE | ID: mdl-28924979

ABSTRACT

Human induced pluripotent stem cells (hiPSC) hold great promise in diagnostic and therapeutic applications. However, translation of hiPSC technology depends upon a means of assessing hiPSC quality that is quantitative, high-throughput, and can decipher malignant teratocarcinoma clones from normal cell lines. These attributes are lacking in current approaches such as detection of cell surface makers, RNA profiling, and/or teratoma formation assays. The latter remains the gold standard for assessing clone quality in hiPSCs, but is expensive, time-consuming, and incompatible with high-throughput platforms. Herein, we describe a novel method for determining hiPSC quality that exploits pluripotent cells' documented hypersensitivity to the topoisomerase inhibitor etoposide (CAS No. 33419-42-0). Based on a study of 115 unique hiPSC clones, we established that a half maximal effective concentration (EC50) value of <300 nM following 24 hours of exposure to etoposide demonstrated a positive correlation with RNA profiles and colony morphology metrics associated with high quality hiPSC clones. Moreover, our etoposide sensitivity assay (ESA) detected differences associated with culture maintenance, and successfully distinguished malignant from normal pluripotent clones independent of cellular morphology. Overall, the ESA provides a simple, straightforward method to establish hiPSC quality in a quantitative and functional assay capable of being incorporated into a generalized method for establishing a quality control standard for all types of pluripotent stem cells. Stem Cells Translational Medicine 2017;6:1829-1839.


Subject(s)
Colony-Forming Units Assay/methods , Etoposide/pharmacology , Induced Pluripotent Stem Cells/drug effects , Topoisomerase Inhibitors/pharmacology , Cells, Cultured , Clinical Trials as Topic , Humans , Induced Pluripotent Stem Cells/metabolism , Transcriptome
4.
Nucleic Acids Res ; 45(9): 5170-5182, 2017 May 19.
Article in English | MEDLINE | ID: mdl-28201653

ABSTRACT

We have previously demonstrated that TGFß Inducible Early Gene-1 (TIEG1), also known as KLF10, plays important roles in mediating skeletal development and homeostasis in mice. TIEG1 has also been identified in clinical studies as one of a handful of genes whose altered expression levels or allelic variations are associated with decreased bone mass and osteoporosis in humans. Here, we provide evidence for the first time that TIEG1 is involved in regulating the canonical Wnt signaling pathway in bone through multiple mechanisms of action. Decreased Wnt signaling in the absence of TIEG1 expression is shown to be in part due to impaired ß-catenin nuclear localization resulting from alterations in the activity of AKT and GSK-3ß. We also provide evidence that TIEG1 interacts with, and serves as a transcriptional co-activator for, Lef1 and ß-catenin. Changes in Wnt signaling in the setting of altered TIEG1 expression and/or activity may in part explain the observed osteopenic phenotype of TIEG1 KO mice as well as the known links between TIEG1 expression levels/allelic variations and patients with osteoporosis.


Subject(s)
Bone and Bones/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Bone and Bones/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Female , Gene Expression Regulation/drug effects , Ligands , Lithium Chloride/pharmacology , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Osteoblasts/drug effects , Osteoblasts/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Skull/cytology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
5.
Blood ; 123(7): 1055-8, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24335105

ABSTRACT

Activation of the Janus kinase family/signal transducer and activator of transcription (JAK/STAT) signaling pathway has been associated with the pathogenesis and progression of both solid and hematologic malignancies. We have detected constitutive activation of STAT5 in malignant B cells derived from patients with Waldenström's macroglobulinemia (WM). Although short hairpin RNA-mediated knockdown of the STAT5A and STAT5B isoforms did not affect cellular proliferation, loss of STAT5 significantly decreased immunoglobulin M (IgM) secretion. A similar dose-dependent inhibition of IgM secretion was observed when WM cell lines were treated with a small molecule inhibitor of STAT5. These data suggest that STAT5 is involved in regulating IgM production in WM and that inhibition of STAT5 may represent a novel therapeutic strategy for lowering IgM levels in WM patients.


Subject(s)
Immunoglobulin M/metabolism , STAT5 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics , Waldenstrom Macroglobulinemia/genetics , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , Humans , RNA, Small Interfering/pharmacology , STAT5 Transcription Factor/metabolism , Secretory Pathway/drug effects , Secretory Pathway/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Tumor Suppressor Proteins/metabolism , Waldenstrom Macroglobulinemia/immunology , Waldenstrom Macroglobulinemia/metabolism
6.
PLoS One ; 8(3): e59456, 2013.
Article in English | MEDLINE | ID: mdl-23555036

ABSTRACT

While the effect of TGF-ß on malignant B cells in non-Hodgkin lymphoma (NHL) has been previously evaluated, studies to specifically define the role of TGF-ß in tumor immunity in B-cell NHL are limited. We found that soluble TGF-ß, secreted by both lymphoma cells and intratumoral T cells, is present in the serum of patients with B-cell NHL. Soluble TGF-ß promoted regulatory T (T(reg)) cells by enhancing expression of Foxp3 in CD4(+) T cells and suppressed effector helper T (T(H)) cells by inhibiting expression of IFN-γ and IL-17. Blockade of the IL-2 signaling pathway diminished the effect of soluble TGF-ß on T cell differentiation. Furthermore, we found that membrane-bound TGF-ß is expressed specifically on the surface of malignant B cells in B-cell NHL. TGF-ß was able to bind to the surface of lymphoma B cells through an interaction with heparan sulfate (HS) but not through the TGF-ß receptor. We showed that pretreatment of lymphoma B cells with TGF-ß significantly inhibits the proliferation and cytokine production of intratumoral T cells. Taken together, these results suggest that tumor-associated soluble and membrane-bound TGF-ß are involved in the regulation of intratumoral T cell differentiation and function in B-cell NHL.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Cell Membrane/metabolism , Lymphoma, Non-Hodgkin/pathology , T-Lymphocytes/cytology , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/metabolism , B-Lymphocytes/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Interleukin-2/biosynthesis , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Solubility , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
7.
Blood ; 120(18): 3774-82, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22976953

ABSTRACT

Cytokines within the tumor microenvironment play an important role in supporting the growth and survival of B-cell malignancies. One such cytokine, IL-21, promotes the growth of myeloma and Hodgkin lymphoma cells while inducing apoptosis in chronic lymphocytic leukemia. However, the biologic significance of IL-21 has not been examined in Waldenstrom macroglobulinemia (WM), a B-cell lymphoma characterized by elevated serum IgM and a lymphoplasmacytic bone marrow infiltrate. We report here on the presence of IL-21 in the bone marrow of patients with WM and have identified activated T cells as the source of this cytokine. We readily detected the IL-21 receptor on malignant WM B cells and show that IL-21 significantly increases both IgM secretion and cellular proliferation of these cells with no effect on viability. IL-21 rapidly induces phosphorylation of STAT3 in WM cells, and treatment of the WM cell line MWCL-1 with a STAT3 inhibitor abolished the IL-21-mediated increases in cellular proliferation and IgM secretion. IL-21 also increased the expression of known STAT3 targets involved in B-cell differentiation, including BLIMP-1, XBP-1, IL-6, and IL-10. Overall, our data indicate that IL-21 in the bone marrow microenvironment significantly affects the biology of WM tumor cells through a STAT3-dependent mechanism.


Subject(s)
Cell Proliferation , Immunoglobulin M/biosynthesis , Interleukins/metabolism , Tumor Microenvironment/immunology , Waldenstrom Macroglobulinemia/metabolism , Waldenstrom Macroglobulinemia/pathology , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoblotting , Immunoglobulin M/immunology , Immunohistochemistry , Interleukins/immunology , Lymphocyte Activation/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Waldenstrom Macroglobulinemia/immunology
8.
J Biol Chem ; 286(13): 10950-9, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21270130

ABSTRACT

Lymphoid enhancer-binding factor (Lef) 1 is a high mobility group protein best known as a Wnt-responsive transcription factor that associates with ß-catenin. Lef1ΔN is a short isoform of Lef1 that lacks the first 113 amino acids and a well characterized high affinity ß-catenin binding domain present in the full-length protein. Both Lef1 isoforms bind DNA and regulate gene expression. We previously reported that Lef1 is expressed in proliferating osteoblasts and blocks osteocalcin expression. In contrast, Lef1ΔN is only detectable in the later stages of osteoblast differentiation and promotes osteogenesis in vitro. Here, we show that Lef1ΔN retains the ability to interact physically and functionally with ß-catenin. Unlike what has been reported in T cells and colon cancer cell lines, Lef1ΔN activated gene transcription in the absence of exogenous ß-catenin and cooperated with constitutively active ß-catenin to stimulate gene transcription in mesenchymal and osteoblastic cells. Residues at the N terminus of Lef1ΔN were required for ß-catenin binding and the expression of osteoblast differentiation genes. To determine the role of Lef1ΔN on bone formation in vivo, a Lef1ΔN transgene was expressed in committed osteoblasts using the 2.3-kb fragment of the type 1 collagen promoter. The Lef1ΔN transgenic mice had higher trabecular bone volume in the proximal tibias and L5 vertebrae. Histological analyses of tibial sections revealed no differences in osteoblast, osteoid, or osteoclast surface areas. However, bone formation and mineral apposition rates as well as osteocalcin levels were increased in Lef1ΔN transgenic mice. Together, our data indicate that Lef1ΔN binds ß-catenin, stimulates Lef/Tcf reporter activity, and promotes terminal osteoblast differentiation.


Subject(s)
Cell Differentiation/physiology , Lymphoid Enhancer-Binding Factor 1/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , beta Catenin/metabolism , Animals , COS Cells , Chlorocebus aethiops , Collagen Type I/biosynthesis , Collagen Type I/genetics , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Mice , Mice, Transgenic , Organ Size/physiology , Osteoblasts/cytology , Osteocalcin/biosynthesis , Osteocalcin/genetics , Promoter Regions, Genetic/physiology , Protein Binding/physiology , Protein Isoforms , Protein Structure, Tertiary , Spine/cytology , Spine/metabolism , Tibia/cytology , Tibia/metabolism , beta Catenin/genetics
9.
Bone ; 48(5): 1117-26, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21255693

ABSTRACT

Histone deacetylase (Hdac) inhibitors are used clinically to treat cancer and epilepsy. Although Hdac inhibition accelerates osteoblast maturation and suppresses osteoclast maturation in vitro, the effects of Hdac inhibitors on the skeleton are not understood. The purpose of this study was to determine how the pan-Hdac inhibitor, suberoylanilide hydroxamic acid (SAHA; a.k.a. vorinostat or Zolinza(TM)) affects bone mass and remodeling in vivo. Male C57BL/6J mice received daily SAHA (100mg/kg) or vehicle injections for 3 to 4weeks. SAHA decreased trabecular bone volume fraction and trabecular number in the distal femur. Cortical bone at the femoral midshaft was not affected. SAHA reduced serum levels of P1NP, a bone formation marker, and also suppressed tibial mRNA levels of type I collagen, osteocalcin and osteopontin, but did not alter Runx2 or osterix transcripts. SAHA decreased histological measures of osteoblast number but interestingly increased indices of osteoblast activity including mineral apposition rate and bone formation rate. Neither serum (TRAcP 5b) nor histological markers of bone resorption were affected by SAHA. P1NP levels returned to baseline in animals which were allowed to recover for 4weeks after 4weeks of daily SAHA injections, but bone density remained low. In vitro, SAHA suppressed osteogenic colony formation, decreased osteoblastic gene expression, induced cell cycle arrest, and caused DNA damage in bone marrow-derived adherent cells. Collectively, these data demonstrate that bone loss following treatment with SAHA is primarily due to a reduction in osteoblast number. Moreover, these decreases in osteoblast number can be attributed to the deleterious effects of SAHA on immature osteoblasts, even while mature osteoblasts are resistant to the harmful effects and demonstrate increased activity in vivo, indicating that the response of osteoblasts to SAHA is dependent upon their differentiation state. These studies suggest that clinical use of SAHA and other Hdac inhibitors to treat cancer, epilepsy or other conditions may potentially compromise skeletal structure and function.


Subject(s)
Bone Resorption/chemically induced , Cell Differentiation/drug effects , Hydroxamic Acids/adverse effects , Hydroxamic Acids/pharmacology , Osteoblasts/drug effects , Osteoblasts/pathology , Acetylation/drug effects , Animals , Biomarkers/blood , Body Weight/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Remodeling/drug effects , Bone Resorption/blood , Cell Count , Cell Cycle/drug effects , Colony-Forming Units Assay , DNA Damage , Femur/drug effects , Femur/metabolism , Femur/pathology , Gene Expression Regulation/drug effects , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteogenesis/drug effects , Peptide Fragments/metabolism , Procollagen/metabolism , Time Factors , Vorinostat
10.
PLoS One ; 5(7): e11492, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20628553

ABSTRACT

Histone deacetylase (Hdac)3 is a nuclear enzyme that contributes to epigenetic programming and is required for embryonic development. To determine the role of Hdac3 in bone formation, we crossed mice harboring loxP sites around exon 7 of Hdac3 with mice expressing Cre recombinase under the control of the osterix promoter. The resulting Hdac3 conditional knockout (CKO) mice were runted and had severe deficits in intramembranous and endochondral bone formation. Calvarial bones were significantly thinner and trabecular bone volume in the distal femur was decreased 75% in the Hdac3 CKO mice due to a substantial reduction in trabecular number. Hdac3-CKO mice had fewer osteoblasts and more bone marrow adipocytes as a proportion of tissue area than their wildtype or heterozygous littermates. Bone formation rates were depressed in both the cortical and trabecular regions of Hdac3 CKO femurs. Microarray analyses revealed that numerous developmental signaling pathways were affected by Hdac3-deficiency. Thus, Hdac3 depletion in osterix-expressing progenitor cells interferes with bone formation and promotes bone marrow adipocyte differentiation. These results demonstrate that Hdac3 inhibition is detrimental to skeletal health.


Subject(s)
Adipogenesis/physiology , Bone Density/physiology , Bone Marrow Cells/cytology , Histone Deacetylases/metabolism , Osteogenesis/physiology , Stem Cells/cytology , Stem Cells/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Blotting, Western , Bone Density/genetics , Bone Marrow Cells/metabolism , Genotype , Growth Plate/cytology , Growth Plate/metabolism , Histone Deacetylases/genetics , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Osteogenesis/genetics , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sp7 Transcription Factor , Transcription Factors/genetics , X-Ray Microtomography
11.
Curr Osteoporos Rep ; 7(2): 64-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19631031

ABSTRACT

Bone is one of the few tissues in the body with the capacity to regenerate and repair itself. Fractures usually are completely repaired in a relatively short time, but in a small percentage of cases, healing never occurs and nonunion is the result. Fracture repair and bone regeneration require the localized reactivation of signaling cascades that are crucial for skeletal development. The Wnt/beta-catenin signaling pathway is one such developmental pathway whose role in bone formation and regeneration recently has been appreciated. During the past decade, much has been learned about how Wnt pathways regulate bone mass. Small molecules and biologics aimed at this pathway are now being tested as potential new anabolic agents. This article reviews recent data demonstrating that Wnt pathways are active during fracture repair and that increasing the activities of Wnt pathway components accelerates bone regeneration.


Subject(s)
Fracture Healing/physiology , Signal Transduction/physiology , Wnt Proteins/physiology , Bone Regeneration/physiology , Fractures, Bone/physiopathology , Humans , beta Catenin/physiology
12.
Expert Opin Ther Targets ; 13(4): 485-96, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19335070

ABSTRACT

BACKGROUND: There is a need to develop new bone anabolic agents because current bone regeneration regimens have limitations. The Wingless-type MMTV integration site (Wnt) pathway has emerged as a regulator of bone formation and regeneration. OBJECTIVE: To review the molecular basis for Wnt pathway modulation and discuss strategies that target it and improve bone mass. METHODS: Data in peer-reviewed reports and meeting abstracts are discussed. RESULTS/CONCLUSIONS: Neutralizing inhibitors of Wnt signaling have emerged as promising strategies. Small-molecule inhibitors of glycogen synthase kinase 3beta increase bone mass, lower adiposity and reduce fracture risk. Neutralizing antibodies to Dickkopf 1, secreted Frizzled-related protein 1 and sclerostin produce similar outcomes in animal models. These drugs are exciting breakthroughs but are not without risks. The challenges include tissue-specific targeting and consequently, long-term safety.


Subject(s)
Bone Diseases/drug therapy , Osteoblasts/drug effects , Signal Transduction/drug effects , Wnt Proteins/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Bone Diseases/physiopathology , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/physiology , Bone Resorption/drug therapy , Drug Evaluation, Preclinical , Fractures, Bone/drug therapy , Gene Expression Regulation/drug effects , Genetic Markers/physiology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , LDL-Receptor Related Proteins/deficiency , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/physiology , Low Density Lipoprotein Receptor-Related Protein-5 , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Knockout , Organ Specificity , Osteoblasts/physiology , Osteogenesis/drug effects , Osteoporosis/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Regeneration/physiology , Wnt Proteins/physiology , beta Catenin/biosynthesis , beta Catenin/genetics
13.
J Cell Biochem ; 101(5): 1125-47, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17520659

ABSTRACT

The actions of 17beta-estradiol (E2) and selective estrogen receptor modulators (SERMs) have been extensively investigated regarding their ability to act through estrogen receptor-alpha (ERalpha) to perturb estrogen receptor positive (ER+) breast cancer (BC) growth. However, many BCs also express ERbeta, along with multiple estrogen receptor (ER) splice variants such as ERbetacx, an ERbeta splice variant incapable of binding ligand. To gain a more comprehensive understanding of ER action in BC cells, we stably expressed ERalpha, ERbeta, or ERbetacx under doxycycline (Dox) control in Hs578T cells. Microarrays performed on E2 or 4OH-tamoxifen (4HT) treated Hs578T ERalpha and ERbeta cells revealed distinct ligand and receptor-dependent patterns of gene regulation, while the induction of ERbetacx did not alter gene expression patterns. E2 stimulation of Hs578T ERbeta cells resulted in a 27% decrease in cellular proliferation, however, no significant change in proliferation was observed following the exposure of Hs578T ERalpha or ERbeta cells to 4HT. Expression of ERbetacx in Hs578T cells did not effect cellular proliferation. Flow cytometry assays revealed a 50% decrease in E2-stimulated Hs578T ERbeta cells entering S-phase, along with a 17% increase in G0/G1 cell-cycle arrest. We demonstrate here that ERalpha and ERbeta regulate unique gene expression patterns in Hs578T cells, and such regulation likely is responsible for the observed isoform-specific changes in cell proliferation. Hs578T ER expressing cell-lines provide a unique BC model system, permitting the comparison of ERalpha, ERbeta, and ERbetacx actions in the same cell-line.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Doxycycline/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm , Humans , Oligonucleotide Array Sequence Analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
14.
J Biol Chem ; 281(39): 28596-604, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16873370

ABSTRACT

Estrogen (E2) is involved in mediating many important functions relevant to osteoblast biology through the actions of the estrogen receptors (ER) alpha and beta. To further understand the mechanisms of ER-specific regulation, we used microarray and reverse transcription-PCR analyses of E2-treated U2OS-ERalpha or -ERbeta cells and identified retinoblastoma-binding protein 1 (RBBP1) as a major E2-regulated gene. RBBP1 is a retinoblastoma cofactor involved in the control of osteoblastic proliferation. Although RBBP1 mRNA levels rapidly increased after 2 h of E2 treatment in both U2OS-ER-expressing lines, a sustained induction was only observed in U2OS-ERalpha cells. Examination of the RBBP1 genomic sequence revealed an ER response element and a Sp1 site located within the first intron. Chromatin immunoprecipitation analyses demonstrated that E2-dependent ERalpha binding to the intron 1 enhancer region was constitutive, whereas ERbeta binding was transient, consistent with the mRNA time course. Interestingly, transient transfection and receptor mutational studies revealed that RBBP1 induction by ERalpha only requires the Sp1 site, whereas ERbeta utilizes both the Sp1 and estrogen response elements binding sites for maximal E2-dependent activation. Stable U2OS transfectants containing a deletion of the ERalpha activation function 1 (AF1) resulted in a temporal mRNA induction profile similar to that of wild type ERbeta. Further, overexpression and chromatin immunoprecipitation analyses also demonstrated that E2-dependent RBBP1 induction is SRC2-dependent for both ER isoforms. These results describe an E2-dependent, ER isoform-specific transcriptional activation of the RBBP1 gene, which in part, is explained by the differential activity of ER AF1 and enhancer element binding.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Enhancer Elements, Genetic , Gene Expression Regulation , Receptors, Estrogen/chemistry , Base Sequence , Binding Sites , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Humans , Molecular Sequence Data , Osteoblasts/metabolism , Protein Isoforms , Protein Structure, Tertiary , Receptors, Estrogen/metabolism , Retinoblastoma-Binding Protein 1
15.
Mol Endocrinol ; 19(6): 1555-68, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15802376

ABSTRACT

The 17beta-estradiol (E2) receptor isoforms [estrogen receptor (ER) alpha and ERbeta] bind E2 and selective ER modulators (SERMs) as homodimers (alpha/alpha or beta/beta) or heterodimers (alpha/beta) to regulate gene expression. Although recent studies have shown that ER homodimers regulate unique sets of E2-responsive genes, little information exists regarding the transcriptional actions of the ERalpha/beta heterodimer. This paper describes the development of a U2OS human osteosarcoma (osteoblast) cell line stably expressing both ERalpha and ERbeta isoforms at a ratio of 1:4, a ratio reported to exist in normal, mature osteoblast cells derived from cancellous bone. The regulation of endogenous genes by E2 and 4-hydroxy-tamoxifen were measured in these cells using gene microarrays and real-time RT-PCR. Both E2 and 4-hydroxy-tamoxifen were shown to regulate unique sets of endogenous genes in the U2OS-ERalpha/beta heterodimer cell line (20% and 27% of total, respectively), compared with all the genes regulated in U2OS-ER homodimer cell lines. Furthermore, two novel E2-regulated genes, retinoblastoma binding protein 1 and 7-dehydrocholesterol reductase, were found to contain estrogen response element-like sequences that directly bind the ERalpha/beta heterodimer. These results suggest that the expression of both ER isoforms, forming functional ERalpha/beta heterodimers, result in unique patterns of gene regulation, many of which are distinct from the genes regulated by the ER homodimers.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Gene Expression Regulation , Tamoxifen/pharmacology , Blotting, Western , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Chromatin Immunoprecipitation , Dimerization , Down-Regulation , Estradiol/metabolism , Humans , Immunoprecipitation , Models, Genetic , Oligonucleotide Array Sequence Analysis , Osteoblasts/metabolism , Osteosarcoma/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Isoforms , Response Elements , Retinoblastoma-Binding Protein 1 , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...