Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
1.
Leukemia ; 31(10): 2132-2142, 2017 10.
Article in English | MEDLINE | ID: mdl-28074064

ABSTRACT

The signal transducer and activator of transcription 5 (STAT5) regulates differentiation, survival, proliferation and transformation of hematopoietic cells. Upon cytokine stimulation, STAT5 tyrosine phosphorylation (pYSTAT5) is transient, while in diverse neoplastic cells persistent overexpression and enhanced pYSTAT5 are frequently found. Post-translational modifications might contribute to enhanced STAT5 activation in the context of transformation, but the strength and duration of pYSTAT5 are incompletely understood. We found that O-GlcNAcylation and tyrosine phosphorylation act together to trigger pYSTAT5 levels and oncogenic transcription in neoplastic cells. The expression of a mutated hyperactive gain-of-function (GOF) STAT5 without O-GlcNAcylation resulted in decreased tyrosine phosphorylation, oligomerization and transactivation potential and complete loss of oncogenic transformation capacity. The lack of O-GlcNAcylation diminished phospho-ERK and phospho-AKT levels. Our data show that O-GlcNAcylation of STAT5 is an important process that contributes to oncogenic transcription through enhanced STAT5 tyrosine phosphorylation and oligomerization driving myeloid transformation. O-GlcNAcylation of STAT5 could be required for nutrient sensing and metabolism of cancer cells.


Subject(s)
Acetylglucosamine/metabolism , Cell Transformation, Neoplastic , Myeloproliferative Disorders/etiology , Protein Processing, Post-Translational , STAT5 Transcription Factor/metabolism , Transcriptional Activation , Tumor Suppressor Proteins/metabolism , Animals , Cell Line , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Glycosylation , Humans , Interleukin-3/pharmacology , Lymphoid Tissue/cytology , Male , Mice , Mutagenesis, Site-Directed , Myeloproliferative Disorders/genetics , Phosphorylation , Phosphotyrosine/metabolism , Radiation Chimera , Recombinant Fusion Proteins/metabolism , STAT5 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Threonine/metabolism , Tumor Suppressor Proteins/genetics
2.
Medchemcomm ; 8(1): 96-102, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-30108694

ABSTRACT

Tumor cell resistance to drug treatment severely limits the therapeutic success of treatment. Tumor cells, exposed to chemotherapeutic drugs, have developed intricate strategies to escape the cytotoxic effects and adapt to adverse conditions. The molecular mechanisms causing drug resistance can be based upon modifications of drug transport or metabolism, structural alterations of drug targets or adaptation of cellular signaling. An important component in the transformation of cells and the emergence of drug resistance is the activation of the transcription factor Stat3. The persistent, inappropriate activation of Stat3 causes the expression of target genes which promote tumor cell proliferation, survival, invasion and immune suppression, and it is instrumental in the process of the emergence of resistance to both conventional chemotherapeutic agents and novel targeted compounds. For these reasons, Stat3 inhibition is being pursued as a promising therapeutic strategy. We have investigated the effects of the tyrosine kinase inhibitor canertinib on the glioma cell line Tu-2449. In these cells Stat3 is persistently phosphorylated and activated downstream of the oncogenic driver v-Src and its effector, the cytoplasmic tyrosine kinase Bmx. Canertinib exposure of Tu-2449 cells rapidly caused the inhibition of the Bmx kinase and the deactivation of Stat3. Prolonged exposure of the cells to canertinib caused the death of the large majority of the cells. Only a few cells became resistant to canertinib and survived in tight clusters. These cells have become drug resistant. When the canertinib resistant cells were expanded and cultured at lower cell densities, they regained their sensitivity towards canertinib. We measured the extent of Stat3 activation as a function of cell density and found that higher cell densities are accompanied by increased Stat3 activation and a higher expression of Stat3 target genes. We suggest that Stat3 induction through tight cell-cell interactions, most likely through the engagement of cadherins, can counteract the inhibitory effects exerted by canertinib on Bmx. Cell-cell interactions induced Stat3 and compensated for the suppression of Stat3 by canertinib, thus transiently protecting the cells from the cytotoxic effects of the inhibitor.

3.
Oncogene ; 31(7): 869-83, 2012 Feb 16.
Article in English | MEDLINE | ID: mdl-21765473

ABSTRACT

BRCA1 mutation-associated breast cancer originates in oestrogen receptor-alpha-negative (ER(-)) progenitors in the mammary luminal epithelium. These cells also express high levels of the Kit gene and a recent study demonstrated a correlation between Brca1 loss and Kit over-expression in the mammary epithelium. However, the functional significance of c-Kit expression in the mammary gland is unknown. To address this, c-Kit(-) and c-Kit(+) mammary epithelial subsets were isolated by flow cytometry, characterised for expression of lineage-specific cell markers and functionally analysed by in vitro colony forming and in vivo transplantation assays. The results confirm that the majority of luminal ER(-) progenitors are c-Kit(+), but also that most stem cells and the differentiated cell populations are c-Kit(-). A subset of c-Kit(+) cells with high proliferative potential was found in the luminal ER(+) population, however, suggesting the existence of a distinct luminal ER(+) progenitor cell type. Analysis of mouse Brca1 mammary tumours demonstrated that they expressed Kit and its downstream effector Lyn at levels comparable to the most strongly c-Kit(+) luminal ER(-) progenitors. Consistent with c-Kit being a progenitor cell marker, in vitro three-dimensional differentiation of c-Kit(+) cells resulted in a loss of c-Kit expression, whereas c-Kit over-expression prevented normal differentiation in vivo. Furthermore, c-Kit was a functional marker of proliferative potential, as c-Kit inhibition by short hairpin knockdown prevented normal epithelial growth and caused cells to undergo apoptosis. Therefore, c-Kit defines distinct progenitor populations in the mammary epithelium and is critical for mammary progenitor survival and proliferation. Importantly, c-Kit is only the second mammary epithelial stem/progenitor marker to be shown to have a functional role in the mammary epithelium and the first marker to be shown to be required for progenitor cell function. The c-Kit signalling network has potential as a target for therapy and/or prevention in BRCA1-associated breast cancer.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Cell Proliferation , Mutation , Proto-Oncogene Proteins c-kit/genetics , Animals , Apoptosis/genetics , BRCA1 Protein/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation/genetics , Cell Survival/genetics , Cells, Cultured , Epithelial Cells/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , NIH 3T3 Cells , Proto-Oncogene Proteins c-kit/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Stem Cells/metabolism
4.
Lab Anim ; 42(1): 104-10, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18348772

ABSTRACT

Transplantation of epithelial cells into cleared fat pads is a widely used technique in the study of mammary gland biology. It was first described in 1959 and has remained a valuable technique, most recently in conjunction with the analysis of mammary anlagen from knockout mice with an embryonic lethal phenotype or reproductive defect, and for mammary epithelial stem-cell assays or analysis of precancerous cells. Mammary glands, unlike most other organs, mainly develop postnatally. When the small amount of endogenous epithelium present in the fat pad of a prepubertal mouse is removed, this clearance leaves a natural microenvironment that can be repopulated with exogenously supplied epithelial cells. Cells with the appropriate developmental potential (stem cells or progenitor cells) can regenerate the epithelial portion of the mammary gland after puberty and pregnancy. The conventional clearance of the fat pad is an involved surgical procedure. We have improved the technique and minimized surgery and recovery time, while maintaining an efficient removal of endogenous epithelium from the mammary fat pad.


Subject(s)
Adipose Tissue/surgery , Cell Transplantation/methods , Epithelial Cells/transplantation , Epithelium/surgery , Mammary Glands, Animal/surgery , Adipose Tissue/cytology , Animals , Epithelial Cells/cytology , Female , Mammary Glands, Animal/anatomy & histology , Mammary Glands, Animal/cytology , Mice , Mice, Inbred C57BL
6.
Mol Cell Endocrinol ; 183(1-2): 101-12, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11604230

ABSTRACT

The signal transducers and activators of transcriptions (Stats) are central mediators of cytokine responses especially in hematopoietic cells. The detailed molecular mechanisms of Stat activation, particularly the role of post-translational modifications and co-operation with cellular transcription factors are subject to intense investigation. The phosphorylation of a tyrosine residue in the carboxyl terminal domain is a common characteristic for the biologically active state of all known Stats. We studied the biological potential of purified recombinant murine Stat5a and Stat5b. These proteins were expressed in Sf9 insect cells upon infection with Stat5 encoding baculoviruses. We also obtained the tyrosine phosphorylated, activated forms of the Stat5 proteins by expressing the tyrosine kinase Janus kinase2 (Jak) in the same cells through co-infection with a kinase encoding virus. After purification, only the tyrosine phosphorylated form was able to bind specifically in vitro to the Stat5 DNA response element. This activated form of Stat5 is also able to support specific cell free in vitro transcription of a gene with a Stat5 response element in its promoter region. The recombinant purified Stat5 proteins were treated with the tyrosine specific protein phosphatase or with potato acidic phosphatase, which removes phosphate groups from serine and tyrosine residues. Phosphatase treatment resulted in the loss of specific DNA binding ability. This property could be restored by an in vitro reaction with recombinant, purified EGF or PDGF receptor kinases. Tyrosine rephosphorylation in vitro also restored the transactivation potential of Stat5. This modification is, therefore, a sufficient prerequisite for transcriptional induction by Stat5.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Milk Proteins , Trans-Activators/metabolism , Transcriptional Activation , Animals , COS Cells , Chromatography , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , ErbB Receptors/metabolism , Genes, Reporter , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microinjections , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , STAT5 Transcription Factor , Trans-Activators/genetics , Trans-Activators/isolation & purification , Tyrosine/metabolism
7.
J Hematother Stem Cell Res ; 10(4): 523-34, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11522235

ABSTRACT

T lymphocytes recognize specific antigens through interaction of the T cell receptor (TCR) with short peptides presented by major histocompatibility complex (MHC) class I or II molecules. For initial activation and clonal expansion, naïve T cells are dependent on professional antigen-presenting cells (APCs) that provide additional co-stimulatory signals. TCR activation in the absence of co-stimulation can result in unresponsiveness and clonal anergy. To bypass immunization, different approaches for the derivation of cytotoxic effector cells with grafted recognition specificity have been developed. Chimeric antigen receptors have been constructed that consist of binding domains derived from natural ligands or antibodies specific for cell-surface antigens, genetically fused to effector molecules such as the TCR alpha and beta chains, or components of the TCR-associated CD3 complex. Upon antigen binding, such chimeric receptors link to endogenous signaling pathways in the effector cell and generate activating signals similar to those initiated by the TCR complex. Since the first reports on chimeric antigen receptors, this concept has steadily been refined and the molecular design of chimeric receptors has been optimized. Aided by advances in recombinant antibody technology, chimeric antigen receptors targeted to a wide variety of antigens on the surface of cancer cells and of cells infected by human immunodeficiency virus (HIV) have been generated. In initial clinical studies, infusion of such cells into patients proved to be safe and transient therapeutic effects have been observed.


Subject(s)
Receptors, Antigen/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Genetic Engineering , Humans , Immunotherapy, Adoptive/methods , Receptors, Antigen/genetics , Recombinant Fusion Proteins/genetics
8.
Gynecol Oncol ; 80(2): 145-55, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11161852

ABSTRACT

OBJECTIVES: The HER-2 proto-oncogene is overexpressed in 15-30% of ovarian cancers, providing a target for therapy in a fraction of patients. We previously described the ability of a recombinant single-chain anti-p185HER-2-Pseudomonas exotoxin A fusion protein, scFv(FRP5)-ETA, to inhibit growth of cancer cells in culture and tumor xenografts in vivo. We have also described synergistic interaction between an anti-p185HER-2-ricin A chain immunotoxin and ionizing radiation for inhibiting growth of ovarian and breast cancer cells that overexpress HER-2. Our objective in this report was to evaluate the in vitro and in vivo effects of scFv(FRP5)-ETA against SKOv3 ovarian cancer cells that have been transfected to express >10(6) p185HER-2 receptors per cell (clone-9002-18). METHODS: Inhibition of clonogenic growth was measured in vitro by limiting dilution analysis. Inhibition of tumor growth was measured in vivo using heterografts established with the same ovarian cancer cell lines. RESULTS: Clone-9002-18 cells were substantially more sensitive than parental SKOv3 cells to the cytotoxic effects of scFv(FRP5)-ETA. Exotoxin fusion protein induced apoptosis in clone-9002-18 cells, but did not affect parental SKOv3 cells. Treatment of clone-9002-18 cells with 200- to 2000-cGy external beam irradiation in combination with scFv(FRP5)-ETA produced synergistic elimination of clonogenic tumor cells in culture. In vivo, subcutaneous or intraperitoneal growth of tumor xenografts in nu/nu mice was significantly inhibited (P = 0.004) by treatment for 10 days with scFv(FRP5)-ETA or with 131I-labeled-520C9 anti-p185HER-2 radionuclide conjugate, but additive effects were not observed with combined treatment. CONCLUSION: ScFv(FRP5)-ETA deserves further evaluation for intraperitoneal therapy of ovarian cancers that overexpress p185HER-2.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Exotoxins/toxicity , Immunotoxins/toxicity , Iodine Radioisotopes/toxicity , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Receptor, ErbB-2/immunology , Recombinant Fusion Proteins/toxicity , Virulence Factors , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/toxicity , Cell Division/drug effects , Cell Division/radiation effects , Combined Modality Therapy , Drug Synergism , Epitopes/immunology , Epitopes/metabolism , Female , Growth Inhibitors/toxicity , Humans , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/toxicity , Immunotoxins/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/radiotherapy , Proto-Oncogene Mas , Receptor, ErbB-2/biosynthesis , Recombinant Fusion Proteins/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
9.
J Biol Chem ; 276(13): 10485-91, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11133982

ABSTRACT

The growth hormone family of cytokines transduces intracellular signals through the Jak2-Stat5 pathway to activate the transcription of target genes. Amino acids within the C termini of Stats constitute the transactivation domain but also regulate the time course of tyrosine phosphorylation and extent of DNA binding. We mutated Thr(757) in the C-terminal of Stat5a (Thr-Stat5) to Val (Val-Stat5) and Asp (Asp-Stat5) and examined the effect on nuclear translocation, DNA binding, and prolactin-induced transcriptional activation of a Stat5-responsive luciferase reporter gene. Val-Stat5 produced a 5-fold higher increase in transcriptional activity relative to Thr-Stat5; Asp-Stat5 produced a similar response to Thr-Stat5. The increased transactivation was ligand induced and was not due to differences in basal expression of Val-Stat5 or to a constitutively activated Stat5 protein. Similar rates of loss of DNA binding ability and phosphorylation of Val- and Thr-Stat5 were observed following a single pulse of prolactin, indicating that the dephosphorylation pathways were unaltered. The serine-threonine kinase inhibitor H7 inhibited the transactivation potential of Thr-, Val-, and Asp-Stat5 to a similar extent, eliminating phosphorylation of Thr(757) as a regulatory mechanism. The results suggest that Thr(757) modulates the transactivation potential of Stat5 by a mechanism(s) that is dependent on the formation of Stat5 dimers and/or their nuclear translocation.


Subject(s)
DNA-Binding Proteins/physiology , Milk Proteins , Threonine/chemistry , Trans-Activators/physiology , Valine/chemistry , Aspartic Acid/chemistry , Blotting, Western , Cell Line , Cell Nucleus/metabolism , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Ligands , Luciferases/metabolism , Phosphorylation , Plasmids/metabolism , Prolactin/pharmacology , Protein Structure, Tertiary , STAT5 Transcription Factor , Signal Transduction , Time Factors , Trans-Activators/chemistry , Trans-Activators/metabolism , Transcriptional Activation , Transfection , Tumor Suppressor Proteins , beta-Galactosidase/metabolism
10.
Methods Mol Med ; 39: 749-56, 2001.
Article in English | MEDLINE | ID: mdl-21340836

ABSTRACT

Cellular transformation does not necessarily require the expression of proteins with neoantigenic properties, and for this reason, immunosurveillance does not register all tumor cells. They frequently express potentially immunogenic components, but are able to escape elimination by immune mechanisms. One explanation for this escape is poor antigen presentation by the tumor cells, resulting in little or no measurable antitumor immunity in immunocompetent hosts. T cells remain naive or even become anergic to the tumor cells. Reasons for the deficient antigen presentation by the tumor cells include the reduced or absent expression of major histocompatibility complex (MHC) molecules and the absence of tumor antigens in the groove of class I or class II MHC molecules as a consequence of defective protein processing. Other reasons are the absence or inadequate levels of expression of adhesion molecules, the absence or inadequate levels of costimulatory molecules or the expression of lymphocyte suppressive cytokines like transforming growth factor (TGF-ß) or interleukin 10 (IL-10) by tumor cells (1-5).

11.
Growth Horm IGF Res ; 10 Suppl B: S15-20, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10984248

ABSTRACT

Extracellular hormones, growth factors and cytokines relay their effects on the transcription of genes through the recognition of specific receptors and intracellular signalling molecules. Signal transducers and activators of transcription (STATs) have been recognized as crucial intracellular signalling molecules. The cytokine receptor-associated Janus kinases (JAKs) convert the latent monomeric form of the STAT molecules to the activated dimeric form through tyrosine phosphorylation. The dimers bind to specific DNA response elements and are able to induce transcription. This induction requires the full-length form of the STAT molecules. Negative regulatory potential is exerted by the short form of the molecule, which lacks the trans-activation domain. This short form is activated and dimerized, but dephosphorylation is impaired. The short form of STAT occupies the DNA-binding sites in a stable fashion and acts as a strong suppressor of wild-type action. Positive enhancement of STAT5 trans-activation potential is provided by the glucocorticoid receptor. Ligand activation of this receptor causes the formation of a complex with STAT5 and deviation to the STAT5 DNA-binding site. An additional regulatory loop is provided by the reactivation of the short form of STAT5 through glucocorticoid receptor association. Conversely, classical glucocorticoid-responsive genes are negatively affected by STAT5 activation.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Gene Expression Regulation , Milk Proteins , Proto-Oncogene Proteins , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Animals , Binding Sites , Caseins/genetics , DNA-Binding Proteins/genetics , Dimerization , Genes, Dominant , Janus Kinase 2 , Ligands , Models, Biological , Phosphorylation , Promoter Regions, Genetic , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Receptors, Glucocorticoid/metabolism , Response Elements , STAT5 Transcription Factor , Signal Transduction , Trans-Activators/genetics , Transcription, Genetic , Tyrosine/metabolism
12.
Cancer Res ; 60(14): 3971-7, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919676

ABSTRACT

Interleukin-9 (IL-9) is a growth factor for T cells and various hematopoietic and lymphoid tumor cells. IL-9 signaling involves activation of Janus kinase (JAK)1 and JAK3 kinases, and signal transducer and activator of transcription (STAT)1, STAT3 and STAT5. Using a dominant negative form of STAT5 (STAT5delta), we demonstrated that this factor is an important mediator of IL-9-dependent Ba/F3 cell growth. Mutation of the STAT binding site of the IL-9 receptor (tyr116phe) results in an important decrease in STAT activation and inhibition of proliferation in the presence of IL-9. A small number of cells escape this inhibition, and IL-9-dependent cell lines could be derived. The selected cells required activation of STAT5 for growth, which was blocked by STAT5delta expression and enhanced by overexpression of wild-type STAT5. In contrast to parental cells, Ba/F3-Phe116 cells growing in the presence of IL-9 further progress to cytokine-independent tumorigenic clones. These tumorigenic clones exhibited a strong cytokine-independent activation of JAK1 and STAT5, which most likely supports their proliferation. Transfection of a constitutively activated variant of STAT5 promoted the growth of wild-type Ba/F3 cells in the absence of cytokine. Finally, the expression of the proto-oncogene pim-1 was correlated with STAT5 activation and cell growth. Our data suggest that STAT5 is an important mediator of IL-9-driven proliferation and that dysregulation of STAT5 activation favors tumorigenesis of lymphoid cells.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-9/metabolism , Lymphocytes/metabolism , Milk Proteins , Protein Serine-Threonine Kinases , Trans-Activators/metabolism , Animals , Binding Sites , CHO Cells , Cell Division/drug effects , Cell Division/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cricetinae , DNA, Complementary/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Enzyme Activation , Female , Humans , Janus Kinase 1 , Janus Kinase 2 , Lymphocytes/pathology , Mice , Mice, SCID , Mutation , Neoplasm Transplantation , Plasmids , Precipitin Tests , Protein Synthesis Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-pim-1 , STAT5 Transcription Factor , Tetracycline/pharmacology , Time Factors , Trans-Activators/chemistry , Trans-Activators/genetics , Transfection , Tumor Cells, Cultured
13.
J Immunol ; 164(11): 5689-97, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10820245

ABSTRACT

IFN-gamma and glucocorticoids regulate inflammatory and immune responses through Stat1 and glucocorticoid receptor (GR) transcription factors, respectively. The biological responses to these polypeptides are determined by integration of various signaling pathways in a cell-type and promoter-dependent manner. In this study we have characterized the molecular basis for the functional cooperation between IFN-gamma and dexamethasone (Dex) in the induction of the high-affinity Fc gamma receptor I (Fc gamma RI) in monocytes. Dex did not affect IFN-gamma-induced Stat1 DNA binding activity or induce novel DNA-binding complexes to the Fc gamma RI promoter. By using cell systems lacking functional GR or Stat1, we showed that GR stimulated Stat1-dependent transcription in a ligand-dependent manner, while Stat1 did not influence GR-dependent transcription. The cooperation required phosphorylation of Tyr701, DNA binding, and the trans-activation domain of Stat1, but did not involve Ser727 phosphorylation of Stat1 or physical interaction between GR and Stat1. The costimulatory effect of Dex was not dependent on a consensus glucocorticoid response element in the Stat1-responsive promoters, but required the DNA-binding and trans-activation functions of GR, and Dex-induced protein synthesis. GR activated the natural Fc gamma RI promoter construct, and this response required both Stat1 and the Ets family transcription factor PU.1. Previously, physical association between GR and Stat5 has been shown to enhance Stat5-dependent and suppress GR-dependent transcription. The results shown here demonstrate a distinct, indirect mechanism of cross-modulation between cytokine and steroid receptor signaling that integrates Stat1 and GR pathways with cell type-specific PU.1 transcription factor in the regulation of Fc gamma RI gene transcription.


Subject(s)
DNA-Binding Proteins/physiology , Milk Proteins , Monocytes/metabolism , Proto-Oncogene Proteins/physiology , Receptors, Glucocorticoid/physiology , Receptors, IgG/metabolism , Trans-Activators/physiology , Transcriptional Activation/immunology , DNA/metabolism , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Drug Synergism , Humans , Interferon-gamma/physiology , Monocytes/drug effects , Promoter Regions, Genetic/immunology , Protein Binding/drug effects , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Glucocorticoid/metabolism , Receptors, IgG/biosynthesis , Receptors, IgG/genetics , STAT1 Transcription Factor , STAT5 Transcription Factor , Signal Transduction/immunology , Trans-Activators/metabolism , Tumor Cells, Cultured
14.
J Biol Chem ; 275(14): 10247-55, 2000 Apr 07.
Article in English | MEDLINE | ID: mdl-10744710

ABSTRACT

The activity of transcription factors of the Stat family is controlled by phosphorylation of a conserved, carboxyl-terminal tyrosine residue. Tyrosine phosphorylation is essential for Stat dimerization, nuclear translocation, DNA binding, and transcriptional activation. Phosphorylation of Stats on specific serine residues has also been described. We have previously shown that in HC11 mammary epithelial cells Stat5a is phosphorylated on Tyr(694) in a prolactin-sensitive manner, whereas serine phosphorylation is constitutive (Wartmann, M., Cella, N., Hofer, P., Groner, B., Xiuwen, L., Hennighausen, L., and Hynes, N. E. (1996) J. Biol. Chem. 271, 31863-31868). By using mass spectrometry and site-directed mutagenesis, we have now identified Ser(779), located in a unique Stat5a SP motif, as the site of serine phosphorylation. By using phospho-Ser(779)-specific antiserum, we have determined that Ser(779) is constitutively phosphorylated in mammary glands taken from different developmental stages. Stat5a isolated from spleen, heart, brain, and lung was also found to be phosphorylated on Ser(779). Ser(725) in Stat5a has also been identified as a phosphorylation site (Yamashita, H., Xu, J., Erwin, R. A., Farrar, W. L., Kirken, R. A., and Rui, H. (1998) J. Biol. Chem. 273, 30218-30224). Here we show that mutagenesis of Ser(725), Ser(779), or a combination of Ser(725/779) to an Ala had no effect on prolactin-induced transcriptional activation of a beta-casein reporter construct. However, following prolactin induction the Ser(725) mutant displayed sustained DNA binding activity compared with that of wild type Stat5a. The results suggest that Ser(725) phosphorylation has an impact on signal duration.


Subject(s)
DNA-Binding Proteins/metabolism , Mammary Glands, Animal/metabolism , Milk Proteins , Prolactin/pharmacology , Serine , Trans-Activators/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Brain/metabolism , COS Cells , DNA-Binding Proteins/chemistry , Female , Lung/metabolism , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Myocardium/metabolism , Peptide Fragments/chemistry , Peptide Mapping , Phosphates/metabolism , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Phosphorylation , Phosphoserine , Receptors, Prolactin/genetics , Receptors, Prolactin/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , STAT5 Transcription Factor , Spleen/metabolism , Trans-Activators/chemistry , Transfection
15.
Int J Cancer ; 86(2): 269-75, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10738256

ABSTRACT

Overexpression of the ErbB2 and epidermal growth factor receptor (EGFR) tyrosine kinases is frequently observed in squamous cell carcinomas of the head and neck, and has been correlated with shorter overall survival. By immunoblot analysis, we have found EGFR and ErbB2 expression in 6 out of 6 established head and neck cancer cell lines. Elevated EGFR protein levels were noted in 3 and elevated ErbB2 levels in 5 of them. Significant expression of EGFR and ErbB2 was also detected in 17 of 47 and 26 of 45 primary tumor samples. Due to their enhanced expression on the tumor cell surface, these receptors can be regarded as suitable targets for directed cancer therapy. We have analyzed the antitumoral activity of recombinant single-chain antibody toxins specific for ErbB2 and EGFR against head and neck cancer cells in vitro and in vivo. The recombinant toxins consist of the variable domains of the heavy and light chains of monoclonal antibodies (MAbs) genetically fused to a truncated Pseudomonas exotoxin A (ETA). At low concentrations, the ErbB2-specific single-chain antibody (scFv) toxin scFv(FRP5)-ETA and the EGFR-specific toxins scFv(225)-ETA and scFv(14E1)-ETA inhibited the in vitro growth of established head and neck cancer cell lines and primary tumor cells. In a nude mouse tumor model, intratumoral injection of the antibody toxins resulted in the rapid regression of subcutaneously growing CAL 27 tumor xenografts, with scFv(FRP5)-ETA and scFv(14E1)-ETA treatment being most effective and leading to the cure of up to 50% of the animals. Our results suggest that EGFR and ErbB2-specific antibody toxins may become valuable therapeutic reagents for the treatment of squamous cell carcinomas of the head and neck.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Carcinoma, Squamous Cell/pathology , ErbB Receptors/immunology , Exotoxins/pharmacology , Head and Neck Neoplasms/pathology , Immunotoxins/pharmacology , Receptor, ErbB-2/immunology , Virulence Factors , Animals , Antibodies, Monoclonal , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/drug therapy , Cell Division , ErbB Receptors/analysis , Exotoxins/administration & dosage , Exotoxins/therapeutic use , Head and Neck Neoplasms/chemistry , Head and Neck Neoplasms/drug therapy , Humans , Immunotoxins/immunology , Immunotoxins/therapeutic use , Mice , Mice, Nude , Neoplasm Transplantation , Receptor, ErbB-2/analysis , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Tumor Cells, Cultured , Pseudomonas aeruginosa Exotoxin A
16.
Breast Cancer Res ; 2(3): 149-53, 2000.
Article in English | MEDLINE | ID: mdl-11250703

ABSTRACT

The mammary epithelium undergoes cyclical periods of cellular proliferation, differentiation and regression. These processes are under the control of the hormones secreted during pregnancy, lactation and involution. Signaling pathways have been identified that connect the hormonal stimuli with the transcription of genes responsible for the determination of the cellular fate. The kinetics of induction and deinduction have suggested that cytokine-activated Stat proteins play a crucial role. Stat5 is strongly activated towards the end of pregnancy, persists in an activated state during pregnancy and is rapidly inactivated after cessation of suckling. Stat3 activation is hardly detectable during lactation, but is strongly induced at the onset of involution. The phenotypes of mice in which these genes have been inactivated through homologous recombination corroborate some of the functional assignments deducted from the activation pattern. Stat3 activation seems to be a driving force in the induction of apoptosis early in the involution period.


Subject(s)
Breast/physiology , Cell Survival/genetics , Signal Transduction , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Survival/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Drug Design , Epithelium/physiology , Female , Hematopoiesis/genetics , Hematopoiesis/physiology , Hormones/metabolism , Hormones/physiology , Mice , Pregnancy , Receptors, Cell Surface/metabolism , STAT3 Transcription Factor , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors , Transcriptional Activation
17.
Anal Biochem ; 276(1): 97-104, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10585749

ABSTRACT

A highly sensitive method for determination of insulin receptor (IR) kinase activity in whole cells, which is based on a STAT5 (signal transducer and activator of transcription 5)-dependent reporter gene assay, has been developed. We show in Rat1 fibroblasts stably overexpressing the human IR (Rat1-HIR-cl5) an insulin-dependent direct association and phosphorylation of STAT5b by IR kinase. Rat1-HIR cells transfected with a luciferase gene reporter construct under control of a STAT5-inducible promoter showed insulin-mediated induction of STAT5-dependent luciferase activity, with peak activities around 8 h of insulin treatment over a wide dose range. Transient STAT5b but not STAT5a cotransfection significantly enhanced reporter gene activity, yielding up to a fivefold induction. Addition of the IR kinase inhibitor tyrphostin AG1024 down-regulated luciferase induction in a dose-dependent manner. This is the first assay allowing determination of IR kinase activity in intact cells in a 24-well culture and a microtiter format. Kinetics of this cellular response, sensitivity range, and signal amplitude make it well suited for automation and offer the potential for establishing high-throughput screening systems for both insulin mimetic substances and IR kinase antagonists in a simple nonradioactive assay.


Subject(s)
DNA-Binding Proteins/metabolism , Genes, Reporter , Luciferases/genetics , Milk Proteins , Receptor, Insulin/metabolism , Trans-Activators/metabolism , Animals , Cell Line , Gene Expression/drug effects , Humans , Insulin/pharmacology , Kinetics , Rats , Receptor, Insulin/analysis , STAT5 Transcription Factor , Sensitivity and Specificity , Signal Transduction , Transfection , Tumor Suppressor Proteins , beta-Galactosidase/genetics
18.
Eur J Oral Sci ; 107(5): 338-43, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10515198

ABSTRACT

The amplification and overexpression of the erbB-2 oncogene and its involvement in tumorigenesis makes this receptor an appropriate target for specific agents directed towards tumor cells. The purpose of this study was to evaluate the in vitro effect of the bacterially produced recombinant immunotoxin scFv(FRP5)-ETA on the protein synthesis and adenosine triphosphate (ATP) reduction in oral squamous cell carcinoma (OSCC) cells. This agent recognizes the erbB-2 receptor and inhibits protein synthesis in receptor-overexpressing cells. OSCC cells were selected for this study, and amplification and expression levels of the erbB-2 receptor were determined. Cell suspensions were cultured for 6 d with various concentrations of scFv(FRP5)-ETA (1-1000 ng/ml). A431 and MDA-MB468 cell lines were used as controls. Chemosensibility of tumor cells was measured by [3H]leucine incorporation assay and by an ATP luminescence assay. In OSCC cells with amplification and overexpression of erbB-2 inhibition, up to 92% of protein synthesis and 90% of ATP reduction was observed when cells were exposed to 1,000 ng/ml immunotoxin. In OSCC cells showing a deletion of erbB-2 and in erbB-2-negative MDA-MB468 cells, protein synthesis was inhibited by 22% and 8%, respectively. These results indicate that the effectiveness of a recombinant immunotoxin targeting erbB-2 receptors in OSCC cells depends on the level of erbB-2 amplification and expression, that it is highly specific for tumor cells expressing these receptors, and that a dose-dependency can be observed.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Exotoxins/therapeutic use , Immunotoxins/therapeutic use , Mouth Neoplasms/drug therapy , Poly(ADP-ribose) Polymerases/therapeutic use , Pseudomonas aeruginosa , Receptor, ErbB-2/antagonists & inhibitors , Virulence Factors , Adenosine Triphosphate/antagonists & inhibitors , Antibodies, Monoclonal/therapeutic use , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Gene Amplification , Gene Deletion , Gene Expression Regulation, Neoplastic , Genes, erbB-2/genetics , Humans , Leucine/metabolism , Luminescent Measurements , Neoplasm Proteins/antagonists & inhibitors , Radiopharmaceuticals , Recombinant Proteins , Tritium , Tumor Cells, Cultured , Pseudomonas aeruginosa Exotoxin A
19.
J Virol Methods ; 81(1-2): 71-5, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10488763

ABSTRACT

Retroviral vectors are used widely as gene transfer vehicles. Vector particles are generated by packaging cell lines, which supply the structural proteins gag, pol and env needed to package the retroviral vector RNA. The most efficient way to introduce the vector genome into the packaging cell line is cross-infection with a retroviral vector. Since the infection of a packaging cell line by the produced virus is blocked due to the down regulation of the retrovirus receptor by the envelope glycoprotein, the vector genome should be introduced by a virus with a host tropism different from the one of the packaging cell line. The murine ecotropic retrovirus receptor was expressed in the human amphotropic packaging cell line FLYA13 to generate a cell line which can be infected by murine ecotropic retroviruses. Vector transfer can now be facilitated by cross-infection with the appropriate ecotropic retroviral vectors and provides a simple and efficient method for the generation of amphotropic packaging lines.


Subject(s)
DNA, Recombinant/genetics , Genetic Vectors/genetics , Leukemia Virus, Murine/genetics , Membrane Glycoproteins/genetics , Receptors, Virus/genetics , Retroviridae/genetics , Virion/genetics , Virus Assembly/genetics , 3T3 Cells , Animals , Gene Transfer Techniques , Humans , Membrane Glycoproteins/biosynthesis , Mice , Receptors, Virus/biosynthesis , Retroviridae/physiology , Tumor Cells, Cultured
20.
Virology ; 261(1): 70-8, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10441556

ABSTRACT

The incorporation of human immunodeficiency virus-type-2 (HIV-2) envelope glycoprotein into murine leukemia virus (MuLV) particles was studied in a transient transfection packaging cell system. We observed that wild-type HIV-2 envelope protein or a frameshift mutant with 187 unrelated carboxyl-terminal residues did not allow the formation of infectious retroviral particles. In view of recent findings that an HIV-1 envelope protein variant with a shortened cytoplasmic domain was incorporated into MuLV particles, we constructed carboxyl-terminal truncations of the HIV-2 envelope protein. An envelope variant with 18 cytoplasmic amino acids formed only very few viral pseudotypes. The further removal of an additional 11 amino acids allowed the efficient pseudotyping of MuLV particles. As with the HIV-1 envelope protein, an HIV-2 envelope variant with 7 cytoplasmic amino acids was incorporated into functional MuLV particles. The pseudotyped vectors obtained are able to infect human CD4/CXCR4-expressing cells. Cell lines expressing human CD4 and other coreceptors could not be infected. This retroviral vector will prove useful for the study of HIV infection events mediated by the HIV-2 envelope glycoproteins, as well as for the targeting of CD4+ cells in the context of gene therapy of AIDS.


Subject(s)
Gene Products, env/genetics , Genetic Vectors , HIV-2/chemistry , HIV-2/genetics , Leukemia Virus, Murine/genetics , Animals , CD4 Antigens/metabolism , Gene Products, env/chemistry , Gene Products, env/metabolism , Gene Transfer Techniques , Genes, env , HIV-1/chemistry , HIV-1/genetics , HIV-1/metabolism , HIV-2/metabolism , HeLa Cells , Humans , Leukemia Virus, Murine/physiology , Mice , Receptors, CXCR4/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...