Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Womens Health (Larchmt) ; 30(2): 187-193, 2021 02.
Article in English | MEDLINE | ID: mdl-33147099

ABSTRACT

Maternal dysglycemia-including diabetes, impaired glucose tolerance, and impaired fasting glucose-affects one in six pregnancies worldwide and represents a significant health risk to the mother and the fetus. Maternal dysglycemia is an independent risk factor for perinatal mortality, major congenital anomalies, and miscarriages. Furthermore, it increases the longer-term risk of type 2 diabetes mellitus, metabolic syndrome, cardiovascular morbidity, malignancies, and ophthalmic, psychiatric, and renal diseases in the mother. The most commonly encountered form of maternal dysglycemia is gestational diabetes. Currently, international consensus does not exist for diagnostic criteria defining gestational diabetes at 24-28 weeks gestation, and potential diagnostic glucose thresholds earlier in gestation require further investigation. Likewise, recommendations regarding the timing and modality (e.g., lifestyle or pharmacological) of treatment vary greatly. Because a precise diagnosis determines the appropriate treatment and outcome of the pregnancy, it is imperative that a better definition of maternal dysglycemia and its treatment be achieved. This article will address some of the controversies related to diagnosing and managing maternal dysglycemia. In addition, the article will discuss the impact of maternal dysglycemia on complications experienced by the mother and infant, both at birth and in later life.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetes, Gestational , Prediabetic State , Diabetes Mellitus, Type 2/epidemiology , Diabetes, Gestational/diagnosis , Diabetes, Gestational/epidemiology , Female , Glucose , Glucose Tolerance Test , Humans , Infant, Newborn , Pregnancy , Pregnancy Outcome/epidemiology
3.
Oncology ; 83(6): 305-20, 2012.
Article in English | MEDLINE | ID: mdl-22964943

ABSTRACT

OBJECTIVE: Estrogen treatment limits the cytotoxic effects of chemotherapy in estrogen receptor-positive (ER+) breast cancer cell lines, suggesting that estrogen pathway signaling may confer chemotherapeutic resistance. This study investigates the molecular responses of ER+ breast cancer cell lines to the chemotherapeutic agent, doxorubicin, in the presence or absence of estrogen. METHODS: ER+ MCF-7 and T47-D cells were cultured in hormone-starved or estrogen-containing media with or without doxorubicin at concentrations mimicking the low concentrations seen in plasma and tumor microenvironments in humans following typical bolus administration. Protein levels, phosphorylations, and interactions of estrogen-signaling molecules were assessed following these treatments, as well the effects of ER signaling inhibitors on cell proliferation. RESULTS: Surprisingly, estrogen and doxorubicin co-treatment markedly induced pro-growth alterations compared to doxorubicin alone and modestly enhanced estrogen alone-induced changes. Several inhibitors suppressed cell proliferation in the presence of doxorubicin and estrogen. CONCLUSIONS: These findings demonstrate that molecular changes caused by doxorubicin in ER+ breast cancer cells can be reversed by estrogen, providing molecular evidence for the poorer responses of ER+ tumors to doxorubicin in the presence of physiologic estrogen levels. Our results also suggest that the addition of drugs targeting the ER, EGFR, the SFKs, MEK, PI3K, and/or the MMP proteins to a conventional chemotherapy regimen may improve chemosensitivity.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Estradiol/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Interactions , Drug Resistance, Neoplasm , Female , Humans , MCF-7 Cells , Phosphorylation/drug effects , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
4.
BMC Cancer ; 11: 184, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21595984

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is a highly aggressive malignant primary brain tumor, characterized by rapid growth, diffuse infiltration of cells into both adjacent and remote brain regions, and a generalized resistance to currently available treatment modalities. Recent reports in the literature suggest that Signal Transducers and Activators of Transcription (STATs) play important roles in the regulation of GBM pathophysiology. METHODS: STAT6 protein expression was analyzed by Western blotting in GBM cell lines and by immunohistochemistry in a tissue microarray (TMA) of glioma patient tissues. We utilized shRNA against STAT6 to investigate the effects of prolonged STAT6 depletion on the growth and invasion of two STAT6-positive GBM cell lines. Cell proliferation was assessed by measuring (3)H-Thymidine uptake over time. Invasion was measured using an in vitro transwell assay in which cells invade through a type IV collagen matrix toward a chemoattractant (Fetal Bovine Serum). Cells were then stained and counted. Kaplan-Meyer survival curves were generated to show the correlation between STAT6 gene expression and patient survival in 343 glioma patients and in a subset of patients with only GBM. Gene expression microarray and clinical data were acquired from the Rembrandt 1 public data depository (https://caintegrator.nci.nih.gov/rembrandt/). Lastly, a genome-wide expression microarray analysis was performed to compare gene expression in wild-type GBM cells to expression in stable STAT6 knockdown clones. RESULTS: STAT6 was expressed in 2 GBM cell lines, U-1242MG and U-87MG, and in normal astrocytes (NHA) but not in the U-251MG GBM cell line. In our TMA study, STAT6 immunostaining was visible in the majority of astrocytomas of all grades (I-IV) but not in normal brain tissue. In positive cells, STAT6 was localized exclusively in the nuclei over 95% of the time. STAT6-deficient GBM cells showed a reduction in (3)H-Thymidine uptake compared to the wild-type. There was some variation among the different shRNA- silenced clones, but all had a reduction in (3)H-Thymidine uptake ranging from 35%- 70% in U-1242MG and 40- 50% in U-87MG cells. Additionally, STAT6- depleted cells were less invasive than controls in our in vitro transmembrane invasion assay. Invasiveness was decreased by 25-40% and 30-75% in U-1242MG and U-87MG cells, respectively. The microarray analysis identified matrix metalloproteinase 1 (MMP-1) and urokinase Plasminogen activator (uPA) as potential STA6 target genes involved in the promotion of GBM cell invasion. In a Kaplan-Meier survival curve based on Rembrandt 1 gene expression microarray and clinical data, there was a significant difference in survival (P < 0.05) between glioma patients with up- and down-regulated STAT6. Decreased STAT6 expression correlated with longer survival times. In two subsets of patients with either grade IV tumors (GBM) or Grade II/III astrocytomas, there was a similar trend that however did not reach statistical significance. CONCLUSIONS: Taken together, these findings suggest a role for STAT6 in enhancing cell proliferation and invasion in GBM, which may explain why up-regulation of STAT6 correlates with shorter survival times in glioma patients. This report thus identifies STAT6 as a new and potentially promising therapeutic target.


Subject(s)
Brain Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic , Glioblastoma/physiopathology , STAT6 Transcription Factor/metabolism , Astrocytoma/physiopathology , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , Databases, Factual , Epidermal Growth Factor/pharmacology , Gene Expression Profiling , Gene Silencing , Glioblastoma/mortality , Glioma/physiopathology , Humans , Neoplasm Invasiveness/physiopathology , Phosphorylation/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , STAT6 Transcription Factor/genetics , Survival Analysis , Tyrosine/metabolism
5.
Genes Dev ; 24(14): 1456-64, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20634312

ABSTRACT

The objective of the workshop was to gain a better understanding of the link between circadian rhythms and human health and disease. The impacts of circadian rhythms on metabolic gene regulation, as well as the effect of nutrient uptake and balance on the molecular components of the clock, were discussed. Topics included the neural circuitry underlying the central clock; the effect of the environment and diet on the central clock as well as peripheral, tissue-specific clocks; and the transcriptional, post-transcriptional, and post-translational (e.g., epigenomic) mechanisms through which these signals are transduced. Evidence presented during the meeting demonstrated that circadian rhythms and metabolism are intricately linked, and that disruption in these rhythms have profound consequences-many times leading to metabolic disease. The mechanisms by which circadian rhythms are maintained and the cross-talk with metabolic signaling are just beginning to be elucidated. However, the interactions between these fields and the knowledge learned will clearly have a profound impact on our understanding of metabolic disease and lead to novel therapeutic approaches in the future.


Subject(s)
Circadian Rhythm , Metabolic Diseases/physiopathology , Animals , Biological Clocks , Humans
6.
Breast Cancer Res ; 11(4): R52, 2009.
Article in English | MEDLINE | ID: mdl-19630967

ABSTRACT

INTRODUCTION: Signal transducer and activator of transcription (STAT) 5b is a transcription factor involved in pro-proliferative and pro-survival signaling in a number of solid tumors, including breast cancer. The contribution of STAT5b to breast cancer cell motility has not been explored. This work aims to elucidate the role of STAT5b in breast cancer cell migration. METHODS: STAT5b was knocked down by using siRNA in two aggressive, highly migratory breast cancer cell lines (BT-549 and MDA-MB-231), and transwell migration assays were performed to determine the importance of STAT5b for their migration. Knockdown-rescue experiments were used to validate the specificity of STAT5b knockdown and to determine which regions/functions of STAT5b are necessary for its role in migration. Live-cell imaging of wound healing and spreading was carried out to examine cell morphology and motility after STAT5b knockdown. RESULTS: Knockdown of STAT5b, but not STAT5a, inhibited migration of BT-549 and MDA-MB-231 breast cancer cells to serum by 60% to 80%, and inhibited migration equally over a range of serum concentrations (0.1% to 10% serum). Migratory inhibition upon STAT5b knockdown could be rescued by reintroduction of wild-type STAT5b, as well as Y699F- and dominant-negative STAT5b mutants, but not an SH2 domain defective R618K-STAT5b mutant. beta1- integrin-mediated migration of breast cancer cells to fibronectin was inhibited with STAT5b knockdown, and loss of STAT5b correlated with loss of directional migration and formation of multiple, highly contractile protrusions upon attachment to fibronectin. CONCLUSIONS: The data presented here demonstrate that STAT5b is integral to breast cancer cell migration and identify a novel, SH2-dependent function of STAT5b in regulating beta1-integrin-mediated migration of highly aggressive breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Integrin beta1/physiology , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/physiology , STAT5 Transcription Factor/physiology , Cell Line, Tumor/drug effects , Cell Line, Tumor/pathology , Cell Movement/physiology , Female , Fibronectins/pharmacology , Gene Knockdown Techniques , Humans , Neoplasm Proteins/chemistry , RNA Interference , RNA, Small Interfering/pharmacology , STAT5 Transcription Factor/chemistry , STAT5 Transcription Factor/genetics , Wound Healing , src Homology Domains
7.
Mol Endocrinol ; 22(8): 1781-96, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18550772

ABSTRACT

17beta-Estradiol (E2) acts through the estrogen receptor alpha (ERalpha) to stimulate breast cancer proliferation. Here, we investigated the functional relationship between ERalpha and signal transducer and activator of transcription (STAT)5b activity in ER+ MCF-7 and T47D human breast cancer cells after specific knockdown of STAT5b. STAT5b small interfering RNA (siRNA) inhibited E2-induced bromodeoxyuridine (BrdU) incorporation in both cell lines, as well as the E2-induced increase in MCF-7 cell number, cyclin D1 and c-myc mRNA, and cyclin D1 protein expression, indicating that STAT5b is required for E2-stimulated breast cancer proliferation. E2 treatment stimulated STAT5b tyrosine phosphorylation at the activating tyrosine Y699, resulting in increased STAT5-mediated transcriptional activity, which was inhibited by a Y669F STAT5b mutant. E2-induced STAT5-mediated transcriptional activity was inhibited by overexpressing a kinase-defective epidermal growth factor receptor (EGFR), or the EGFR tyrosine kinase inhibitor tyrphostin AG1478, indicating a requirement for EGFR kinase activity. Both E2-induced STAT5b tyrosine phosphorylation and STAT5-mediated transcription were also inhibited by the ER antagonist ICI 182,780 and the c-Src inhibitor PP2, indicating additional requirements for the ER and c-Src kinase activity. EGFR and c-Src kinase activities were also required for E2-induced cyclin D1 and c-myc mRNA. Together, these studies demonstrate positive cross talk between ER, c-Src, EGFR, and STAT5b in ER+ breast cancer cells. Increased EGFR and c-Src signaling is associated with tamoxifen resistance in ER+ breast cancer cells. Here we show that constitutively active STAT5b not only increased basal DNA synthesis, but also conferred tamoxifen resistance. Because STAT5b plays an integral role in E2-stimulated proliferation and tamoxifen resistance, it may be an effective therapeutic target in ER+ breast tumors.


Subject(s)
Breast Neoplasms/enzymology , ErbB Receptors/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm , Humans , Phosphorylation/drug effects , Phosphotyrosine/metabolism , STAT5 Transcription Factor/genetics , Tamoxifen/pharmacology , Transcription, Genetic/drug effects
8.
Breast Cancer Res ; 9(6): R79, 2007.
Article in English | MEDLINE | ID: mdl-17997837

ABSTRACT

INTRODUCTION: Signal transducers and activators of transcription (STATs) are mediators of cytokine and growth factor signaling. In recent years, STAT5b has emerged as a key regulator of tumorigenesis. STAT5b phosphorylation and activation is mediated by several kinases known to be overexpressed in breast cancer, such as epidermal growth factor receptor, HER2, and c-Src. Breast tumor kinase (Brk), also known as protein tyrosine kinase 6, is a nonreceptor tyrosine kinase expressed in more than 60% of breast cancers. Only a few substrates of the Brk tyrosine kinase have been identified, the most recent being STAT3. In the present article we investigate the potential role of Brk in the phosphorylation and activation STAT5b. METHODS: To determine whether Brk can phosphorylate STAT5b, transient transfection and in vitro kinase assays were performed. Luciferase reporter assays were used to measure Brk-induced STAT5b transcriptional activity. siRNA technology was utilized to investigate the biological significance of Brk-induced activation of STAT5b in breast cancer cell models. RESULTS: Phosphospecific antibodies, mutational analysis, and in vitro kinase assays demonstrated that Brk specifically mediated STAT5b phosphorylation at the activating tyrosine, Y699. Transient transfection of Brk into the Brk-negative BT-549 breast cancer cell line enhanced STAT5b transcriptional activity, as measured by a STAT5-specific luciferase reporter. Furthermore, overexpression of kinase active c-Src enhanced Brk-induced STAT5b transcriptional activity. In Brk-positive breast cancer cell lines BT-20 and SKBr3, knockdown of Brk protein or of STAT5b protein using siRNA methodology resulted in a decrease in DNA synthesis. Knockdown of Brk and STAT5b together did not further decrease DNA synthesis compared with each alone, suggesting that Brk and STAT5b converge on the same pathway, ultimately leading to cellular proliferation. CONCLUSION: Our studies demonstrate that Brk phosphorylates STAT5b on Y699, leading to increased STAT5b transcriptional activity. Furthermore, analysis of DNA synthesis suggests that STAT5b and Brk are converging upon the same proproliferative signaling pathway in breast cancer cells. We propose that Brk, like other tyrosine kinases, signals downstream to STAT5b to mediate proliferation of breast cancer cells. These results further establish STAT5b as well as Brk as potential targets for breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , STAT5 Transcription Factor/metabolism , CSK Tyrosine-Protein Kinase , Cell Proliferation , Female , Humans , Luciferases , Luminescent Agents , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , Transfection , src-Family Kinases
9.
Biochem Biophys Res Commun ; 362(4): 1026-30, 2007 Nov 03.
Article in English | MEDLINE | ID: mdl-17822672

ABSTRACT

As mediators of cytokine and growth factor signaling, signal transducers and activators of transcription (STATs) transmit signals from the membrane and cytoplasm to the nucleus. While Y699 phosphorylation is required for STAT5b transcriptional activity, our previous studies show that mutation of two tyrosines in the transactivation domain of STAT5b (Y740/743F) increases Y699 phosphorylation leading to increased transcriptional activity and DNA synthesis in breast cancer cells [A.M. Weaver, C.M. Silva, Modulation of signal transducer and activator of transcription 5b activity in breast cancer cells by mutation of tyrosines within the transactivation domain, Molecular Endocrinology 20 (2006) 2392-2405]. In many instances, phosphorylation of serines in the transactivation domain also modulates STAT5b activity. Here, we demonstrate for the first time that EGF stimulation enhances S731 phosphorylation. Furthermore, we show that the increased activity of the Y740/743F STAT5b mutant requires S731. As STAT5b is implicated in several cancers, understanding how its activity is regulated through tyrosine and serine phosphorylation is vital for the development of potential novel cancer therapeutics.


Subject(s)
Breast Neoplasms/metabolism , Cysteine/metabolism , Gene Expression Regulation , STAT5 Transcription Factor/metabolism , Transcriptional Activation , Tyrosine/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Humans , Protein Structure, Tertiary , STAT5 Transcription Factor/genetics
10.
Mol Endocrinol ; 21(7): 1499-512, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17456797

ABSTRACT

The signaling pathways that are critical to the development and growth of breast cancer include those activated downstream of the estrogen receptor (ER) and the human epidermal growth factor receptor family. Many of these pathways, including the signal transducer and activator of transcription pathway, are common to both. The well-described genomic actions of ER involve its role as a transcription factor, either by binding directly to DNA through estrogen response elements, or by tethering to DNA through interaction with other proteins. Nongenomic signaling by the ER involves interaction with membrane-associated signaling proteins such as the c-Src tyrosine kinase and adapter proteins p130Cas and moderator of nongenomic activity of ER. Interactions with the signal transducer and activator of transcription pathway are important in both ER signaling pathways and are critical for estrogen-induced proliferation and tumorigenesis. These mechanisms of signaling cross talk and their role in resistance to antiestrogen therapies are discussed.


Subject(s)
Breast Neoplasms/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Estrogen Receptor Modulators/therapeutic use , Female , Growth Substances/genetics , Growth Substances/metabolism , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Models, Biological , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Steroids/metabolism , Transcription, Genetic
11.
Cancer Res ; 66(14): 7007-15, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16849545

ABSTRACT

High expression of the adaptor molecule Cas has been linked to resistance to the antiestrogen tamoxifen, both in tissue culture and in human tumors. The aim of this study was to elucidate the mechanism(s) by which overexpression of Cas confers resistance to tamoxifen. Cas overexpression in MCF-7 breast cancer cells was shown to alleviate both tamoxifen-mediated growth inhibition and induction of apoptosis. This enhancement of cell proliferation/survival occurred in the absence of detectable effects on estrogen receptor (ER) transcriptional activity under conditions where tamoxifen was present, indicating that Cas-dependent tamoxifen resistance is not the result of a switch to an ER-negative phenotype or enhanced responses to the partial agonist activity of tamoxifen. Instead, we present evidence, suggesting that Cas promotes tamoxifen resistance by deregulation of alternative cell proliferation pathways, particularly those mediated through enhanced c-Src protein tyrosine kinase activity arising from Cas/c-Src interactions. Overexpression of Cas was found to drive endogenous c-Src into complex with Cas, a process that has been shown previously to cause up-regulation of c-Src tyrosine kinase activity. MCF-7 cells overexpressing Cas exhibited increased phosphorylation of two c-Src substrates, Tyr845 in the kinase domain of the epidermal growth factor receptor (EGFR) and signal transducer and activator of transcription (STAT) 5b. Importantly, Cas-dependent protection from the antiproliferative effects of tamoxifen was reversed by the expression of dominant inhibitory variants of these substrates (Y845F EGFR and COOH-terminally truncated STAT5b). Based on these findings, we suggest that the Cas/c-Src/EGFR/STAT5 signaling axis is a major regulator of tamoxifen-resistant breast cancer cell growth and survival.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Crk-Associated Substrate Protein/metabolism , Protein-Tyrosine Kinases/metabolism , STAT5 Transcription Factor/metabolism , Tamoxifen/pharmacology , Animals , COS Cells , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Chlorocebus aethiops , Drug Resistance, Neoplasm , Humans , Signal Transduction , src-Family Kinases
12.
Mol Endocrinol ; 20(10): 2392-405, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16772534

ABSTRACT

The signal transducer and activator of transcription (STAT) proteins are latent transcription factors activated by a variety of cytokines and growth factors. Activation leads to phosphorylation on a conserved tyrosine residue. Although phosphorylation of STAT5b on Y699 is required for activation, it was previously shown that in epidermal growth factor receptor (EGFR)-overexpressing cell lines, three tyrosines (Y725, Y740, and Y743) in the STAT5b transactivation domain are also phosphorylated upon epidermal growth factor stimulation. The significance of these additional tyrosine phosphorylation sites was analyzed in the context of the human breast cancer cell line SKBr3, which overexpresses the EGFR and c-Src. When compared with wild-type STAT5b, mutation of Y725 decreased basal and epidermal growth factor-induced DNA synthesis. In contrast, mutation of Y740 and/or Y743 enhanced basal STAT5b Y699 phosphorylation, basal transcriptional activity, and basal DNA synthesis compared with wtSTAT5b. This indicates that Y699 and Y725 are positive regulators and Y740 and Y743 are negative regulators for STAT5b activity. Anti-phospho-Y740/743-specific antibodies demonstrated that the c-Src tyrosine kinase inhibits the phosphorylation of these two sites. Furthermore, Y740 and Y743 were not detectably phosphorylated in breast cancer cells overexpressing c-Src, but the Y740/743F mutant increased basal activity suggesting that the conformation of the transactivation domain is important in regulating STAT5b activity. Mechanistic insight into the inhibitory action of Y740 and Y743 may lead to the development of therapeutics that specifically modulate the activity of STAT5b in breast cancer and potentially other EGFR/c-Src-overexpressing cancers.


Subject(s)
Breast Neoplasms/genetics , DNA Replication/genetics , Gene Expression Regulation, Neoplastic/genetics , Mutation/genetics , STAT5 Transcription Factor/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Immunoblotting , Immunoprecipitation , Phosphorylation , Tyrosine/genetics
13.
Mol Carcinog ; 44(4): 262-73, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16167350

ABSTRACT

Signaling networks play important roles in cancer progression. For example, overexpression of the epidermal growth factor receptor (EGFR) is a poor prognostic indicator in multiple tumor types. Recent studies have postulated that the EGFR functions as a central conduit for signaling by different classes of cell surface receptors. In this study, we demonstrated that c-Src-dependent phosphorylation of tyrosine 845 (Tyr 845) on EGFR was required for DNA synthesis induced by the G protein-coupled agonists, endothelin (ET) and lysophosphatidic acid (LPA), and the cytokine, growth hormone (GH), in murine fibroblast and breast cancer model systems. In addition, we showed that a dominant interfering form of signal transducer and activator of transcription (STAT)5b (a downstream effector of phospho-Tyr 845 [pY845] in fibroblasts) abrogates DNA synthesis induced by all agonists in the breast cancer model. To further characterize the role of Tyr 845, a pY845-containing peptide was microinjected into SKBr3 breast cancer cells and murine fibroblasts, and was found to ablate EGF-stimulated S-phase entry in both cell systems. Taken together, these findings suggested that pY845 is critical for DNA synthesis induced by a variety of mitogens and that its signaling effectors may include but are not limited to STAT5b.


Subject(s)
DNA/biosynthesis , ErbB Receptors/agonists , Trans-Activators/physiology , Tyrosine/metabolism , Animals , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Bromodeoxyuridine , Cells, Cultured , Chickens , Endothelins/pharmacology , ErbB Receptors/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Growth Hormone/pharmacology , Humans , Immunoprecipitation , Lysophospholipids/pharmacology , Mice , Mice, Inbred C3H , Peptide Fragments/administration & dosage , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/physiology , S Phase , STAT5 Transcription Factor/metabolism
14.
Mol Endocrinol ; 19(11): 2660-70, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15976008

ABSTRACT

Breast cancer cell growth may be stimulated by 17beta-estradiol (E2) or growth factors like epidermal growth factor (EGF). However, tumors typically depend on only one of these pathways and may overexpress either estrogen receptor (ER) or EGF receptor (EGFR) and related family members. Tumors overexpressing EGFR are more aggressive than those expressing ER. Intracellular mediators of these growth-stimulatory pathways are not completely defined, but one potential common mediator of EGF and E2 signaling is the transcription factor signal transducer and activator of transcription 5 (STAT5). To investigate the role of STAT5 in potential crosstalk between E2 and EGF, MDA-MB231 and SKBr3 breast cancer cells, which are ER-negative and overexpress human EGF family receptors, were used. Introduction of ERalpha and treatment with E2 decreased EGF-induced tyrosine phosphorylation of STAT5b, basal and EGF-induced STAT5-mediated transcription, and EGF-stimulated DNA synthesis in these cells. Suppressive effects of E2-EpsilonRalpha were specific for STAT5, as EGF stimulation of MAPK was unaffected. Deletion/mutation analysis of ERalpha demonstrated that the DNA-binding domain was insufficient, and that the ligand-binding domain was required for these responses. ERalpha transcriptional activity was not necessary for suppression of STAT5 activity. Overexpression of c-Src did not prevent suppression of STAT5 activity by E2 and ERalpha. However, ERalpha did prevent basal increases in STAT5 activity with overexpressed c-Src. In the context of human EGF receptor family overexpression, E2-ER opposes EGF signaling by regulating STAT5 activity. STAT5 may be a crucial point of signaling for both E2 and growth factors in breast cancer cells, allowing targeted therapy for many types of breast tumors.


Subject(s)
Breast Neoplasms/metabolism , Epidermal Growth Factor/antagonists & inhibitors , ErbB Receptors/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , STAT5 Transcription Factor/antagonists & inhibitors , Breast Neoplasms/genetics , Bromodeoxyuridine/metabolism , Enzyme Activation , Estrogen Receptor alpha/genetics , Genes, Reporter , Humans , Luciferases/analysis , Luciferases/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Transcription, Genetic , Transfection , Tumor Cells, Cultured , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
15.
J Biol Chem ; 280(17): 17449-57, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15728176

ABSTRACT

The urokinase-type plasminogen activator (uPA) receptor (uPAR) functions in concert with co-receptors, including integrins, FPR-like receptor-1/lipoxin A4 receptor, and the epidermal growth factor receptor (EGFR), to initiate cell signaling. uPAR co-receptors may be dynamically organized into a multiprotein signaling receptor complex. In Chinese hamster ovary-K1 (CHO-K1) cells, uPA-binding to uPAR activates ERK/MAP kinase, even though these cells do not express the EGFR; however, when CHO-K1 cells are transfected to express the EGFR, ERK activation becomes EGFR-dependent. In this study, we demonstrate that ERK activation in response to uPA follows equivalent biphasic kinetics in EGFR-expressing and -deficient CHO-K1 cells. In both cell types, the response is pertussis toxin-sensitive; however, uPA promotes cell proliferation exclusively in the EGFR-expressing cells. uPA-induced mitogenic activity requires activation of both STAT5b and ERK. STAT5b was tyrosine-phosphorylated, in response to uPA, only in EGFR-expressing cells. uPA-induced cell proliferation was blocked by dominant-negative MEK1, dominant-negative STAT5b, and by expression of an EGFR that is mutated at Tyr-845, which is essential for STAT5b activation. In two cell culture models of uPA-stimulated breast cancer growth, MDA-MB 468 cells treated with uPA and MCF-7 cells treated with uPA-plasminogen activator inhibitor-1 complex, proliferation was completely inhibited when EGFR expression or activity was blocked. We conclude that expression and assembly of uPAR co-receptors in a specific cell type determines the response to uPA. The EGFR selectively cooperates with uPAR to mediate mitogenesis.


Subject(s)
Signal Transduction , Urokinase-Type Plasminogen Activator/metabolism , Animals , Bromodeoxyuridine/pharmacology , CHO Cells , Cell Line, Tumor , Cell Proliferation , Coloring Agents/pharmacology , Cricetinae , DNA/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , Kinetics , Milk Proteins/metabolism , Mutation , Pertussis Toxin/pharmacology , Phosphorylation , Prognosis , Protein Binding , RNA, Small Interfering/metabolism , STAT5 Transcription Factor , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Trans-Activators/metabolism , Transfection , Tyrosine/metabolism , src-Family Kinases/metabolism
16.
Oncogene ; 23(48): 8017-23, 2004 Oct 18.
Article in English | MEDLINE | ID: mdl-15489919

ABSTRACT

The signal transducers and activators of transcription (STATs) were originally identified in the signaling pathway activated by the nontyrosine kinase containing cytokine receptors. The role of these STATs in hematopoietic cell signaling has been well described. In the case of cytokine receptors, activation of STAT tyrosine phosphorylation occurs through ligand-induced recruitment, and activation of the intracellular JAK kinases. However, STATs can also be activated by growth factor receptors, particularly the EGFR; as well as by members of the Src Family of Kinases (SFKs), particularly c-Src. In many cases, there is a differential activation of the STATs by these tyrosine kinases as compared to activation by the cytokine receptors. This difference provides for the potential of unique actions of STATs in response to growth factor receptor and SFK activation. Since there are many cancers in which SFKs and c-Src in particular, are co-overexpressed with growth factor receptors, it is not surprising that STATs play an important role in the tumorigenesis process induced by c-Src. The activation paradigm and role of STATs in these cancers, with particular emphasis on breast cancer models, is discussed.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/metabolism , Milk Proteins/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , src-Family Kinases/metabolism , ErbB Receptors/metabolism , Female , Humans , Neoplasms/enzymology , Neoplasms/etiology , Neoplasms/metabolism , Receptors, Growth Factor/metabolism , STAT3 Transcription Factor , STAT5 Transcription Factor
17.
J Biol Chem ; 278(3): 1671-9, 2003 Jan 17.
Article in English | MEDLINE | ID: mdl-12429742

ABSTRACT

Overexpression of the epidermal growth factor receptor (EGFR) and its association with the tyrosine kinase, c-Src, is correlated with increased cellular proliferation and tumorigenesis. Previous studies have shown that EGFR and c-Src co-overexpression and association leads to the c-Src-mediated phosphorylation of tyrosine 845 of the EGFR and that mutation of Tyr(845) ablates epidermal growth factor (EGF)-induced DNA synthesis. Here, we investigate the contribution of the signal transducers and activators of transcription (STAT5b) in the signaling pathways regulated by EGFR and c-Src overexpression in human breast tumor cell lines as well as in a mouse fibroblast model (C3H10T1/2). We demonstrate that 1) activation of STAT5b by EGF requires overexpression of the EGFR, 2) co-overexpression of c-Src alone does not result in EGF-induced activation of STAT5b but enhances that seen in EGFR-overexpressing cells, and 3) EGF-induced tyrosine phosphorylation of STAT5b requires Tyr(845) of the EGFR. Furthermore, the stable overexpression of a kinase-defective c-Src in the context of EGFR overexpression results in a decrease in the tyrosine phosphorylation of STAT5b in response to EGF and a more dramatic decrease in EGF-induced transcriptional activation of STAT5b, suggesting an integral role for c-Src in the physiological actions of STAT5b. Using a dominant negative STAT5b, we provide evidence that one such physiological action is to mediate EGF-induced DNA-synthesis. Finally, the use of site-specific tyrosine mutants demonstrates that EGF-induced phosphorylation of STAT5b involves not only tyrosine 699 of STAT5b, which is required for its transcriptional activation, but also three previously identified tyrosines in the C terminus of STAT5b (Tyr(725)/Tyr(740)/Tyr(743)).


Subject(s)
DNA-Binding Proteins/physiology , ErbB Receptors/physiology , Milk Proteins , Proto-Oncogene Proteins pp60(c-src)/physiology , Trans-Activators/physiology , Animals , DNA Replication/physiology , ErbB Receptors/chemistry , Mice , Mice, Inbred C3H , Phosphorylation , STAT5 Transcription Factor , Tumor Cells, Cultured , Tyrosine/metabolism
18.
Growth Horm IGF Res ; 12(5): 374-80, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12213191

ABSTRACT

Placental growth hormone, or growth hormone variant (GH-V), is a member of the growth hormone gene family. Its physiologic role during pregnancy remains undefined. Although extensive work has been done characterizing the signaling pathways activated by hGH, the signaling pathways which are activated by GH-V have not been characterized. We investigated cellular signaling by GH-V in model systems in which signaling by hGH has been well characterized. GH-V activates the STAT5b transcription factor in the IM-9 human lymphocyte and 3T3-F442A preadipocyte cell lines, and in primary porcine smooth muscle cells, which all homologously express GH receptors. In contrast, hCS does not activate signaling in these GH receptor-expressing cells. Like hGH, signaling by GH-V is inhibited by the GH antagonist (G120K). These results provide preliminary evidence that GH-V plays a major role in affecting target cells expressing the GH receptor, thus potentially exerting significant GH-like effects on maternal physiology during pregnancy.


Subject(s)
Growth Hormone/metabolism , Milk Proteins , Placental Hormones/metabolism , 3T3 Cells , Animals , Cell Line , DNA-Binding Proteins/metabolism , Female , Growth Hormone/genetics , Humans , Mice , Placenta/metabolism , Placental Hormones/genetics , Placental Lactogen/metabolism , Pregnancy , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , STAT5 Transcription Factor , Signal Transduction , Swine , Trans-Activators/metabolism
19.
Endocrinology ; 143(7): 2610-7, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12072393

ABSTRACT

We have investigated and compared GH and epidermal growth factor (EGF) signaling in primary human skin fibroblasts from normal subjects and subjects with GH-binding protein-positive Laron syndrome (LS). In normal human fibroblasts, GH and EGF activate the tyrosine phosphorylation of signal transducer and activator of transcription (STAT)1 and STAT5b; in LS fibroblasts, EGF does, but GH does not. GH also activates the tyrosine phosphorylation of Janus kinase (JAK)2 in normal, but not LS, fibroblasts. Similarly, both GH and EGF activate MAPK in normal fibroblasts, but only EGF does in the LS fibroblasts. As in the 3T3-F442A mouse preadipocyte cell line, GH signaling to mitogen-activated protein kinase is partially inhibited by wortmannin treatment, indicating a role for phosphatidylinositol 3-kinase (PI3K) in this signaling pathway. The exogenous expression of the GH receptor in one family of LS fibroblasts (H1) but not the other (M) restores signaling to a STAT5 reporter element. Together, these results indicate that the mechanism of defective GH signaling in two families of LS fibroblasts are different but that both occur at a level close to, and specific for, the GH receptor.


Subject(s)
Abnormalities, Multiple/metabolism , Epidermal Growth Factor/pharmacology , Growth Hormone/pharmacology , Proto-Oncogene Proteins , Signal Transduction/drug effects , Androstadienes/pharmacology , Blotting, Western , Child , Female , Fibroblasts/drug effects , Humans , Indicators and Reagents , Janus Kinase 2 , Luciferases/physiology , Mitogen-Activated Protein Kinases/physiology , Phosphorylation , Precipitin Tests , Protein-Tyrosine Kinases/physiology , Receptors, Somatotropin/genetics , Receptors, Somatotropin/physiology , Syndrome , Transfection , Tyrosine/metabolism , Wortmannin
20.
J Biol Chem ; 277(10): 8693-701, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11751923

ABSTRACT

Of the seven signal transducers and activators of transcription that have been identified, STATs 1, 3, and 5a/5b can be activated not only by a multitude of cytokines but also by some growth factors. The data presented here demonstrate that, in contrast to activation by the cytokine, growth hormone (GH), the activation of STAT5b by the growth factor, epidermal growth factor (EGF), requires overexpression of the EGF receptor (EGFR). We have shown that EGF activates STAT5b not only in a HEK293 cell model in which the EGFR is stably overexpressed but also in the MDA-MB468 breast cancer cell line. Furthermore, EGF (but not GH) is able to activate tyrosine phosphorylation of a Tyr-699 mutant of STAT5b. Using metabolic labeling studies as well as site-directed mutagenesis, we have identified three novel EGF-induced tyrosine phosphorylation sites, Tyr-725, Tyr-740, and Tyr-743. Luciferase assays using a STAT5-specific DNA sequence demonstrate that, although Tyr-699 is absolutely required for transcriptional activation, tyrosines 725, 740, and 743 may be involved in a negative regulation of transcription. Because overexpression of the EGFR is common in many cancers, including advanced breast cancer, characterization of EGF-induced STAT5b may have direct implications in therapeutic applications.


Subject(s)
DNA-Binding Proteins/metabolism , Epidermal Growth Factor/metabolism , Milk Proteins , Trans-Activators/metabolism , Amino Acid Sequence , Blotting, Western , Cell Line , Epitopes , ErbB Receptors/metabolism , Genetic Vectors , Humans , Immunoblotting , Luciferases/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Phosphorylation , Precipitin Tests , STAT5 Transcription Factor , Sequence Homology, Amino Acid , Signal Transduction , Transcription, Genetic , Transcriptional Activation , Transfection , Trypsin/chemistry , Trypsin/metabolism , Tumor Cells, Cultured , Tyrosine/chemistry , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...