Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Oncogene ; 33(18): 2385-94, 2014 May 01.
Article in English | MEDLINE | ID: mdl-23708659

ABSTRACT

Activation of the mitogen-activated protein kinase (MAPK) cascade in mammalian cell lines positively regulates the G2/M transition. The molecular mechanism underlying this biological phenomenon remains poorly understood. Ribosomal S6 kinase (RSK) is a key downstream element of the MAPK cascade. Our previous studies established roles of RSK2 in Cdc25C activation during progesterone-induced meiotic maturation of Xenopus oocytes. In this study we demonstrate that both recombinant RSK and endogenous RSK in Xenopus egg extracts phosphorylate all three isoforms of human Cdc25 at a conserved motif near the catalytic domain. In human HEK293 and PC-3mm2 cell lines, RSK preferentially phosphorylates Cdc25A and Cdc25B in mitotic cells. Phosphorylation of the RSK sites in these Cdc25 isoforms increases their M-phase-inducing activities. Inhibition of RSK-mediated phosphorylation of Cdc25 inhibits G2/M transition. Moreover, RSK is likely to be more active in mitotic cells than in interphase cells, as evidenced by the phosphorylation status of T359/S363 in RSK. Together, these findings indicate that RSK promotes G2/M transition in mammalian cells through activating phosphorylation of Cdc25A and Cdc25B.


Subject(s)
G2 Phase Cell Cycle Checkpoints , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , cdc25 Phosphatases/metabolism , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Molecular Sequence Data , Oocytes/enzymology , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Xenopus , cdc25 Phosphatases/genetics
2.
Oncogene ; 32(24): 2973-83, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-22890325

ABSTRACT

Castration-resistant prostate cancer (PCa) is refractory to hormone therapy and new strategies for treatment are urgently needed. We found that androgen-insensitive (AI) PCa cells, LNCaP-AI, are reprogrammed to upregulate the mitotic kinase Plk1 (Polo-like kinase 1) and other M-phase cell-cycle proteins, which may underlie AI PCa growth. In androgen-depleted media, LNCaP-AI cells showed exquisite sensitivity to growth inhibition by subnanomolar concentrations of a small molecule inhibitor of Plk1, BI2536, suggesting that these cells are dependent on Plk1 for growth. In contrast, the androgen-responsive parental LNCaP cells showed negligible responses to BI2536 treatment under the same condition. BI2536 treatment of LNCaP-AI cells resulted in an increase in cell death marker PARP-1 (polymerase-1) but did not activate caspase-3, an apoptosis marker, suggesting that the observed cell death was caspase-independent. BI2536-treated LNCaP-AI cells formed multinucleated giant cells that contain clusters of nuclear vesicles indicative of mitotic catastrophe. Live-cell time-lapse imaging revealed that BI2536-treated giant LNCaP-AI cells underwent necroptosis, as evidenced by 'explosive' cell death and partial reversal of cell death by a necroptosis inhibitor. Our studies suggest that LNCaP-AI cells underwent reprogramming in both their cell growth and cell death pathways, rendering them highly sensitive to Plk1 inhibition that induces necroptosis. Harnessing necroptosis through Plk1 inhibition may be explored for therapeutic intervention of castration-resistant PCa.


Subject(s)
Androgens/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Up-Regulation/drug effects , Aneuploidy , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mitosis/drug effects , Necrosis/chemically induced , Necrosis/metabolism , Polo-Like Kinase 1
3.
Oncogene ; 29(22): 3196-207, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20228840

ABSTRACT

Dasatinib, a dual Src family kinase and Abl inhibitor, is being tested clinically for the treatment of prostate cancer bone metastasis. Bidirectional interactions between osteoblasts and prostate cancer cells are critical in the progression of prostate cancer in bone, but the effect of dasatinib on osteoblasts is unknown. We found that dasatinib inhibited proliferation of primary mouse osteoblasts isolated from mouse calvaria and the immortalized MC3T3-E1 cell line. In calvarial osteoblasts from Col-luc transgenic mice carrying osteoblast-specific Col1alpha1 promoter reporter, luciferase activity was inhibited. Dasatinib also inhibited fibroblast growth factor-2-induced osteoblast proliferation, but strongly promoted osteoblast differentiation, as reflected by stimulation of alkaline phosphatase activity, osteocalcin secretion and osteoblast mineralization. To determine how dasatinib blocks proliferative signaling in osteoblasts, we analyzed the expression of a panel of tyrosine kinases, including Src, Lyn, Fyn, Yes and Abl, in osteoblasts. In the Src family kinases, only Src was activated at a high level. Abl was expressed at a low level in osteoblasts. Phosphorylation of Src-Y419 or Abl-Y245 was inhibited by dasatinib treatment. Knockdown of either Src or Abl by lenti-shRNA in osteoblasts enhances osteoblast differentiation, suggesting that dasatinib enhances osteoblast differentiation through inhibition of both Src and Abl.


Subject(s)
Osteoblasts/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrimidines/pharmacology , Thiazoles/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Cell Differentiation/drug effects , Cell Growth Processes/drug effects , Dasatinib , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Transgenic , Neoplasm Invasiveness , Osteoblasts/cytology , Osteoblasts/enzymology , Osteonectin/biosynthesis , Phosphorylation , Proto-Oncogene Proteins c-abl/metabolism , src-Family Kinases/metabolism
4.
Oncogene ; 27(39): 5195-203, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18490922

ABSTRACT

ErbB3 is a transmembrane growth factor receptor that has been implicated in the pathogenesis of human cancer. After finding that a truncated form of ErbB3 was present and upregulated in metastatic prostate cancer cells in lymph nodes and bone, we explored the pathophysiological functions of this unusual form of ErbB3 in the context of mouse calvaria as well as osteoblasts in vitro and the femur microenvironment in vivo. Here we demonstrate that prostate cancer cells expressed an alternatively spliced transcript that encodes a 45-kDa glycosylated protein (p45-sErbB3). The recombinant p45-sErbB3 purified from conditioned medium stimulated calvarial bone formation and induced osteoblast differentiation. Overexpression of p45-sErbB3 in the osteolytic prostate cancer cell line PC-3 converted its phenotype from bone lysing to bone forming upon injection into the femurs of immunodeficient mice. Further, we detected sErbB3 in plasma samples from patients with castration-resistant prostate cancer with bone metastasis. These observations establish that p45-sErbB3 is a structurally and functionally unique gene product of ErbB3 and suggest that p45-sErbB3 is likely one of the factors involved in the osteoblastic bone metastases of prostate cancer.


Subject(s)
Bone Development/physiology , Prostatic Neoplasms/metabolism , Receptor, ErbB-3/physiology , Alternative Splicing , Animals , Base Sequence , Bone Neoplasms/secondary , Cell Line, Tumor , Culture Media, Conditioned , DNA Primers , Humans , Male , Mice , Osteoblasts/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Am J Physiol Lung Cell Mol Physiol ; 284(1): L108-18, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12388333

ABSTRACT

Interferon gamma (IFN-gamma), a potent cytokine inducing a wide range of immunologic activities, is increased in the airway secondary to viral infection or during an inflammatory response. This increase in IFN-gamma concentration may alter the expression of specific airway epithelial cell genes that regulate adaptation of airway inflammatory responses. One protein induced by IFN-gamma is Clara cell secretory protein (CCSP), which may contribute to the attenuation of airway inflammation. This study was done to investigate the molecular mechanism by which IFN-gamma stimulates the expression of the CCSP gene in mouse transformed Clara cells and transgenic mice. Deletion mapping and linker-scanning mutations demonstrated that IFN-gamma-induced expression of CCSP was regulated, in part, at the level of transcription. In vitro and in vivo studies verified that the minimal IFN-gamma-responsive segment was localized to the proximal 166 bp of the 5'-flanking region. Additionally, IFN-gamma-induced expression of CCSP was mediated indirectly through an interferon regulatory factor-1-mediated increase in hepatocyte nuclear factor-3beta.


Subject(s)
Gene Expression Regulation/physiology , Interferon-gamma/physiology , Proteins/genetics , Transcription Factors , Transcription, Genetic/physiology , Uteroglobin , Animals , Binding Sites/physiology , Cell Line, Transformed , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Gene Expression/physiology , Hepatocyte Nuclear Factor 3-beta , Interferon Regulatory Factor-1 , Mice , Nuclear Proteins/metabolism , Phosphoproteins/physiology , Promoter Regions, Genetic , Proteins/chemistry , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Stereoisomerism , Trans-Activators/metabolism
6.
Int J Dev Biol ; 45(7): 839-43, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11732844

ABSTRACT

To identify gene products important for gastrulation in the amphibian Pleurodeles waltl, a screen for regional differences in new protein expression at the early gastrula stage was performed. A 45 kDa protein whose synthesis was specific for progenitor endodermal cells was identified. Microsequencing and cDNA cloning showed that P45 is highly homologous to rat NUDC, a protein suggested to play a role in nuclear migration. Although PNUDC can be detected in all regions of the embryo, its de novo synthesis is tightly regulated spatially and temporally throughout oogenesis and embryonic development. New PNUDC synthesis in the progenitor endodermal cells depends on induction by the mesodermal cells in the gastrula. During development, PNUDC is localized in the egg cortical cytoplasm, at the cleavage furrow during the first embryonic division, around the nuclei and cortical regions of bottle cells in the gastrula, and at the basal region of polarized tissues in the developing embryo. These results show for the first time the expression and compartmentalization of PNUDC at distinct stages during amphibian development.


Subject(s)
Embryo, Nonmammalian/cytology , Embryonic Development , Fungal Proteins/metabolism , Pleurodeles/embryology , Amino Acid Sequence , Animals , Cloning, Molecular , Electrophoresis, Gel, Two-Dimensional , Embryo, Nonmammalian/metabolism , Fungal Proteins/genetics , Fungal Proteins/isolation & purification , Gastrula/cytology , Gastrula/metabolism , Immunoenzyme Techniques , Immunologic Tests , Molecular Sequence Data , Oogenesis/physiology , Pleurodeles/genetics , Pleurodeles/growth & development , Sequence Homology, Amino Acid
7.
Mol Endocrinol ; 15(12): 2172-81, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11731618

ABSTRACT

PRL has been shown to stimulate mRNA expression of both ERalpha and ERbeta in the rat corpus luteum and decidua of pregnancy. To investigate whether PRL may stimulate ER expression at the level of transcription and which transcription factors may mediate this stimulation, we have cloned the 5'-flanking regions of both rat ER genes. A constitutively active PRL receptor (PRL-R(CA)) stimulated both ERalpha and ERbeta promoter activity, indicating that PRL is acting to stimulate ER transcription. Putative signal transducer and activator of transcription (Stat)5 response elements were identified at -189 in the ERalpha promoter and at -330 in the ERbeta promoter. Mutation of these response elements or overexpression of dominant negative Stat5 prevented stimulation of ERalpha and ERbeta promoter activity, indicating that PRL regulation of ER expression requires both intact Stat5 binding sites as well as functional Stat5. Interestingly, either Stat5a or Stat5b could stimulate ERalpha transcription while stimulation of ERbeta occurred only in the presence of Stat5b. Through mutational analysis, a single nucleotide difference between the ERalpha and ERbeta Stat5 response elements was shown to be responsible for the lack of Stat5a-mediated stimulation of ERbeta. These findings indicate that PRL stimulation of ER expression occurs at the level of transcription and that PRL regulation of ERalpha can be mediated by either Stat5a or Stat5b, while regulation of ERbeta appears to be mediated only by Stat5b.


Subject(s)
DNA-Binding Proteins/physiology , Milk Proteins , Prolactin/physiology , Receptors, Estrogen/genetics , Trans-Activators/physiology , Animals , Base Sequence , Binding Sites , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Molecular Sequence Data , Mutagenesis, Site-Directed , Pregnancy , Promoter Regions, Genetic/physiology , Rats , Rats, Sprague-Dawley , Receptors, Estradiol/biosynthesis , Receptors, Estrogen/biosynthesis , STAT5 Transcription Factor , Signal Transduction/physiology , Transcriptional Activation/physiology
8.
J Neurosci ; 21(24): RC187, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11734602

ABSTRACT

NUDC is a highly conserved protein important for nuclear migration and viability in Aspergillus nidulans. Mammalian NudC interacts with Lis1, a neuronal migration protein important during neocorticogenesis, suggesting a conserved mechanism of nuclear movement in A. nidulans and neuronal migration in the developing mammalian brain (S. M. Morris et al., 1998). To further investigate this possibility, we show for the first time that NudC, Lis1, and cytoplasmic dynein intermediate chain (CDIC) colocalize at the microtubule organizing center (MTOC) around the nucleus in a polarized manner facing the leading pole of cerebellar granule cells with a migratory morphology. In neurons with stationary morphology, NudC is distributed throughout the soma and colocalizes with CDIC and tubulin in neurites as well as at the MTOC. At the subcellular level, NudC, CDIC, and p150 dynactin colocalize to the interphase microtubule array and the MTOC in fibroblasts. The observed colocalization is confirmed biochemically by coimmunoprecipitation of NudC with CDIC and cytoplasmic dynein heavy chain (CDHC) from mouse brain extracts. Consistent with its expression in individual neurons, a high level of NudC is detected in regions of the embryonic neocortex undergoing extensive neurogenesis as well as neuronal migration. These data suggest a biochemical and functional interaction of NudC with Lis1 and the dynein motor complex during neuronal migration in vivo.


Subject(s)
Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , Molecular Motor Proteins/metabolism , Neurons/metabolism , Proteins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Animals , Brain Chemistry , COS Cells , Cell Cycle Proteins , Cell Movement/physiology , Cell Polarity/physiology , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Choroid Plexus/cytology , Choroid Plexus/embryology , Choroid Plexus/metabolism , Ependyma/cytology , Ependyma/embryology , Ependyma/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/embryology , Lateral Ventricles/metabolism , Macromolecular Substances , Mice , Mice, Inbred Strains , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Neocortex/cytology , Neocortex/embryology , Neocortex/metabolism , Neurons/cytology , Nuclear Proteins , Precipitin Tests
9.
Mol Cell Endocrinol ; 184(1-2): 135-41, 2001 Nov 26.
Article in English | MEDLINE | ID: mdl-11694349

ABSTRACT

Transcription of the interferon regulatory factor-1 gene (IRF-1) is induced in a biphasic manner (G1 and G1/S phase) in Nb2 T cells in response to prolactin (PRL) stimulation. Signal transducer and activator of transcription 1 (Stat1) is required for PRL activation of the IRF-1 promoter. Mutation of a -200 bp Sp1 site in the IRF-1 promoter results in a loss of G1 but not G1/S IRF-1 transcriptional activity in response to PRL. These studies illustrate that the temporal transcription of the IRF-1 gene is mediated by not only Stat1 but also Sp1 in response to PRL stimulation.


Subject(s)
DNA-Binding Proteins/genetics , Phosphoproteins/genetics , Prolactin/pharmacology , Sp1 Transcription Factor/physiology , Transcriptional Activation/drug effects , Animals , Binding Sites , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , G1 Phase , Interferon Regulatory Factor-1 , Mutation , Phosphoproteins/drug effects , Prolactin/metabolism , Prolactin/physiology , Promoter Regions, Genetic , Rats , STAT1 Transcription Factor , Signal Transduction , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Trans-Activators/physiology , Transfection , Tumor Cells, Cultured
10.
Biol Reprod ; 64(5): 1392-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11319143

ABSTRACT

The enzymes which comprise the 2',5'-oligoadenylate synthetase (OAS) family are interferon (IFN) stimulated genes which regulate ribonuclease L antiviral responses and may play additional roles in control of cellular growth and differentiation. This study characterized OAS expression in the endometrium of cyclic and pregnant ewes as well as determined effects of IFNtau and progesterone on OAS expression in cyclic or ovariectomized ewes and in endometrial epithelial and stromal cell lines. In cyclic ewes, low levels of OAS protein were detected in the endometrial stroma (S) and glandular epithelium (GE). In early pregnant ewes, OAS expression increased in the S and GE on Day 15. OAS expression in the lumenal epithelium (LE) was not detected in uteri from either cyclic or pregnant ewes. Intrauterine administration of IFNtau stimulated OAS expression in the S and GE, and this effect of IFNtau was dependent on progesterone. Ovine endometrial LE, GE, and S cell lines responded to IFNtau with induction of OAS proteins. In all three cell lines, the 40/46-kDa OAS forms were induced by IFNtau, whereas the 100-kDa OAS form appeared to be constitutively expressed and not affected by IFNtau. The 69/71-kDa OAS forms were induced by IFNtau in the S and GE cell lines, but not in the LE. Collectively, these results indicate that OAS expression in the endometrial S and GE of the early pregnant ovine uterus is directly regulated by IFNtau from conceptus and requires the presence of progesterone.


Subject(s)
2',5'-Oligoadenylate Synthetase/genetics , Estrus/physiology , Gene Expression , Interferon Type I/pharmacology , Pregnancy Proteins/pharmacology , Sheep/metabolism , Uterus/metabolism , Animals , Cell Line , Endometrium/metabolism , Female , Interferon Type I/administration & dosage , Pregnancy , Pregnancy Proteins/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Uterus/drug effects
11.
Endocrinology ; 142(1): 98-107, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145571

ABSTRACT

Interferon-tau (IFNtau), a type I IFN produced by sheep conceptus trophectoderm, is the signal for maternal recognition of pregnancy. Although it is clear that IFNtau suppresses transcription of the estrogen receptor alpha and oxytocin receptor genes and induces expression of various IFN-stimulated genes within the endometrial epithelium, little is known of the signal transduction pathway activated by the hormone. This study determined the effects of IFNtau on signal transducer and activator of transcription (STAT) activation, expression, DNA binding, and transcriptional activation using an ovine endometrial epithelial cell line. IFNtau induced persistent tyrosine phosphorylation and nuclear translocation of STAT1 and -2 (10 min to 48 h), but transient phosphorylation and nuclear translocation of STAT3, -5a/b, and -6 (10 to <60 min). IFNtau increased expression of STAT1 and -2, but not STAT3, -5a/b, and -6. IFN-stimulated gene factor-3 and STAT1 homodimers formed and bound an IFN-stimulated response element (ISRE) and gamma-activated sequence (GAS) element, respectively. IFNtau increased transcription of GAS-driven promoters at 3 h, but suppressed their activity at 24 h. In contrast, the activity of an ISRE-driven promoter was increased at 3 and 24 h. These results indicate that IFNtau activates multiple STATs and has differential effects on ISRE- and GAS-driven gene transcription.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endometrium/physiology , Epithelial Cells/physiology , Interferon Type I/pharmacology , Pregnancy Proteins/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic/physiology , Animals , Cell Nucleus/metabolism , Cells, Cultured , Endometrium/cytology , Epithelial Cells/cytology , Female , Interferon Type I/physiology , Interferon-Stimulated Gene Factor 3 , Luciferases , Phosphorylation , Pregnancy Proteins/physiology , Promoter Regions, Genetic , Protein Transport , Recombinant Fusion Proteins/analysis , STAT1 Transcription Factor , STAT2 Transcription Factor , Sheep , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transfection
12.
Eur Cytokine Netw ; 11(3): 435-42, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11022129

ABSTRACT

It has been proposed that prolactin (PRL) is a lympho-hemopoietic growth and differentiation factor. We show here by Western blotting that PRL-receptors (PRL-R) are expressed in normal rat bone marrow and spleen cells. We also show that PRL stimulates the phosphorylation of the PRL-R-associated Janus tyrosine kinase (JAK)-2 in rat bone marrow and spleen cells. This leads to the activation and subsequent binding of signal transducer and activator of transcription (Stat) 5b to an interferon regulatory factor-1 (IRF-1) gamma activation sequence (GAS) as visualized by electromobility shift assay. As shown after reverse transcription of mRNA by polymerase chain reaction, PRL, at physiological concentrations (0.01 microg/ml), stimulates the expression of the IRF-1 gene in these normal cells. PRL could thus affect several aspects of the immune response.


Subject(s)
DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/immunology , Lymphocytes/immunology , Milk Proteins , Phosphoproteins/genetics , Prolactin/physiology , Receptors, Prolactin/genetics , Animals , Bone Marrow Cells/immunology , Cell Line , Cells, Cultured , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Interferon Regulatory Factor-1 , Interferon-gamma/physiology , Lymphocytes/cytology , Lymphocytes/drug effects , Prolactin/pharmacology , RNA, Messenger/genetics , Rats , STAT5 Transcription Factor , Spleen/immunology , Thymus Gland/immunology , Trans-Activators/metabolism , Transcription Factors/genetics
13.
J Clin Endocrinol Metab ; 84(11): 4260-5, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10566682

ABSTRACT

PRL expression in the human uterus is up-regulated during the mid to late secretory phase of the menstrual cycle. This coincides with up-regulation of the expression of the PRL receptor, which is localized primarily to the endometrial glandular epithelial cells. Recent data have demonstrated activation of the Jak (Janus kinase)/Stat (signal transducer and activator of transcription) signaling pathway in the secretory endometrium after stimulation with exogenous PRL. However, the target genes for the action of PRL on the endometrial epithelial cells have not been elucidated. In this study we have investigated the pattern/site of expression of the transcription factor interferon regulatory factor-1 (IRF-1) as well as the effect of exogenous PRL on the transcription of IRF-1 in the human endometrium during the mid to late secretory phase of the menstrual cycle. Expression of the IRF-1 gene was confirmed by RNase protection assays using a 260-bp homologous [alpha-32P]UTP-labeled IRF-1 complementary ribonucleic acid (RNA) probe and 10 microg total RNA extracted from human endometrium (n = 5) collected between days 19 and 26 of the menstrual cycle. Northern and Western blot analyses were conducted on secretory phase human endometrium (n = 3) using human [alpha-32P]dCTP-labeled IRF-1 complementary DNA and antihuman IRF-1 antibody. Expression of the IRF-1 gene in the secretory phase endometrium was encoded by a RNA transcript of approximately 2.1 kb and a protein of 48 kDa. Furthermore, expression of the IRF-1 gene in the secretory phase endometrium was localized by immunohistochemistry predominantly to the glandular epithelial cells as has been shown previously for the PRL receptor. To investigate the effect of PRL on expression of IRF-1, human endometrial biopsies (n = 3) collected between days 24-26 of the menstrual cycle were cultured in the presence of cycloheximide with or without 100 ng/mL human PRL for 2 and 4 h. Culture of endometrial tissue with PRL for 2 and 4 h resulted in 2.9 +/- 0.3-fold (P < 0.01) and 1.7 +/- 0.1-fold induction of expression of the IRF-1 gene, respectively. These data demonstrate the expression of the transcription factor IRF-1 in the glandular epithelium of the endometrium and its regulation by PRL during the secretory phase of the menstrual cycle. Previous observations of the temporal up-regulation of expression of both PRL and PRL receptors in the secretory human endometrium and their localization to the stromal and glandular compartments, respectively, suggest that endometrial PRL mediates transcription of the IRF-1 gene in a paracrine fashion.


Subject(s)
DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Endometrium/chemistry , Gene Expression Regulation/drug effects , Menstrual Cycle/physiology , Phosphoproteins/analysis , Phosphoproteins/genetics , Prolactin/pharmacology , Blotting, Northern , Blotting, Western , Culture Techniques , Endometrium/metabolism , Epithelium/chemistry , Epithelium/metabolism , Female , Humans , Immunohistochemistry , Interferon Regulatory Factor-1 , Prolactin/analysis , RNA Probes , RNA, Messenger/analysis , Receptors, Prolactin/analysis , Stromal Cells/chemistry , Stromal Cells/metabolism , Transcription Factors
14.
J Immunol ; 163(9): 4663-72, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10528163

ABSTRACT

The cytokines IL-4 and IFN-gamma exert biologically antagonistic effects that in part reflect opposing influences on gene transcription. While the molecular mechanisms for IL-4-mediated transcription activation have been extensively studied, little is known about molecular mechanisms required for IL-4 inhibition of IFN-gamma signaling. We have investigated IL-4 inhibition of the IFN-gamma-inducible promoter for IFN regulatory factor-1 (IRF-1). In a cell line with low endogenous Stat6, increasing levels of activated Stat6 at constant doses of IFN-gamma and IL-4 leads to inhibition of the IRF-1 promoter. The Stat1-dependent IFN-gamma activation sequence element of the IRF-1 promoter is a target for Stat6-mediated inhibition despite apparently normal Stat1 DNA binding. However, our data are inconsistent with competition between Stat1 and Stat6 for access to the IRF-1 IFN-gamma activation sequence or for an essential coactivator as a mechanism for this Stat6-mediated inhibition. Instead, the data demonstrate that a threshold of Stat6 transcription activation domains is required for IL-4-dependent inhibition. The findings provide evidence of a novel mechanism in which the Stat6 transcription activation domains play a critical role in the IL-4-mediated inhibition of an IFN-gamma-inducible promoter.


Subject(s)
Interferon-gamma/physiology , Peptide Fragments/genetics , Promoter Regions, Genetic/immunology , Signal Transduction/immunology , Trans-Activators/genetics , Trans-Activators/physiology , Transcriptional Activation/immunology , Binding, Competitive/genetics , Binding, Competitive/immunology , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Humans , Interferon Regulatory Factor-1 , Interferon-gamma/antagonists & inhibitors , Interleukin-4/pharmacology , Peptide Fragments/physiology , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Protein Binding/genetics , Protein Binding/immunology , Regulatory Sequences, Nucleic Acid/immunology , STAT6 Transcription Factor , Signal Transduction/genetics , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Tumor Cells, Cultured
15.
J Biol Chem ; 274(8): 5138-45, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-9988763

ABSTRACT

The mechanisms regulating the cellular distribution of STAT family transcription factors remain poorly understood. To identify regions of Stat5B required for ligand-induced nuclear accumulation, we constructed a cDNA encoding green fluorescent protein (GFP) fused to the N terminus of Stat5B and performed site-directed mutagenesis. When co-expressed with growth hormone (GH) receptor in COS-7 cells, GFP-Stat5B is tyrosyl-phosphorylated, forms dimers, and binds DNA in response to GH in a manner indistinguishable from untagged Stat5B. In multiple cell types, laser scanning confocal imaging of GFP-Stat5B co-expressed with GH receptor shows that GFP-Stat5B undergoes a rapid, dramatic accumulation in the nucleus upon GH stimulation. We introduced alanine substitutions in several regions of Stat5B and assayed for GH-dependent nuclear localization. Only the mutation that prevented binding to DNA (466VVVI469) abrogated GH-stimulated nuclear localization. This mutant fusion protein is tyrosyl-phosphorylated and dimerizes in response to GH. These results suggest that either high affinity binding to DNA contributes to nuclear accumulation of Stat5B or that this region is crucial for two functions, namely accumulation of Stat5B in the nucleus and DNA binding. Thus, we have identified a mutant Stat5 defective in nuclear localization despite its ability to be tyrosyl-phosphorylated and to dimerize.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Growth Hormone/metabolism , Milk Proteins , Trans-Activators/metabolism , Animals , Base Sequence , Binding Sites , COS Cells , DNA Primers , DNA-Binding Proteins/genetics , Dimerization , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , STAT5 Transcription Factor , Trans-Activators/genetics , Tumor Cells, Cultured
16.
Curr Biol ; 8(10): 603-6, 1998 May 07.
Article in English | MEDLINE | ID: mdl-9601647

ABSTRACT

Important clues to how the mammalian cerebral cortex develops are provided by the analysis of genetic diseases that cause cortical malformations [1-5]. People with Miller-Dieker syndrome (MDS) or isolated lissencephaly sequence (ILS) have a hemizygous deletion or mutation in the LIS1 gene [3,6]; both conditions are characterized by a smooth cerebral surface, a thickened cortex with four abnormal layers, and misplaced neurons [7,8]. LIS1 is highly expressed in the ventricular zone and the cortical plate [9,10], and its product, Lis1, has seven WD repeats [3]; several proteins with such repeats have been shown to interact with other polypeptides, giving rise to multiprotein complexes [11]. Lis1 copurifies with platelet-activating factor acetylhydrolase subunits alpha 1 and alpha 2 [12], and with tubulin; it also reduces microtubule catastrophe events in vitro [13]. We used a yeast two-hybrid screen to isolate new Lis1-interacting proteins and found a mammalian ortholog of NudC, a protein required for nuclear movement in Aspergillus nidulans [14]. The specificity of the mammalian NudC-Lis1 interaction was demonstrated by protein-protein interaction assays in vitro and by co-immunoprecipitation from mouse brain extracts. In addition, the murine mNudC and mLis1 genes are coexpressed in the ventricular zone of the forebrain and in the cortical plate. The interaction of Lis1 with NudC, in conjunction with the MDS and ILS phenotypes, raises the possibility that nuclear movement in the ventricular zone is tied to the specification of neuronal fates and thus to cortical architecture.


Subject(s)
Fungal Proteins/metabolism , Microtubule-Associated Proteins , Nerve Tissue Proteins/metabolism , Neurons/physiology , Proteins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Amino Acid Sequence , Animals , Cell Movement/physiology , Fungal Proteins/genetics , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Proteins/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid
17.
Biol Reprod ; 58(2): 295-301, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9475379

ABSTRACT

The peptide hormone prolactin (PRL) is known to regulate numerous target tissues. Among the less well-known targets are cells of the immune system, including T cells, B cells, and macrophages. Our laboratory has cloned a panel of PRL-inducible T-cell activation genes for use in studies investigating how PRL modulates the biology of cells of the immune system. This article focuses on two such PRL-inducible genes. One is a transcription factor called interferon regulatory factor-1, whose expression is regulated by signaling molecules along the PRL-inducible JAK/Stat signaling pathway. These signaling molecules include Stat1 and CBP as positive mediators and, unexpectedly, Stat5b as a negative mediator. A second PRL-inducible gene is c15/RNUDC, a novel nuclear movement protein, which may provide a link between PRL signaling and signaling via the lipid second messenger, platelet activating factor.


Subject(s)
Prolactin/physiology , Signal Transduction/physiology , Animals , Female , Humans , Pregnancy
18.
Exp Cell Res ; 238(1): 23-32, 1998 Jan 10.
Article in English | MEDLINE | ID: mdl-9457053

ABSTRACT

Prolactin and other cytokines regulate lymphocyte proliferation through the activation of a number of genes, one of which was identified as RnudC from a prolactin-dependent rat T cell line, Nb2. RnudC encodes a 45-kDa protein whose carboxy terminal 94 amino acids are similar to the carboxy terminus of the Aspergillus nidulans nuclear movement protein NUDC. In Nb2 T cells, RNUDC protein levels are induced two- to threefold by prolactin stimulation. This prolactin-inducible increase in RNUDC protein levels is due in part to an increase in RNUDC protein synthesis between 8 and 12 h, during the G1/S transition. Newly synthesized RNUDC protein is very stable, exhibiting a half-life of greater than 8 h. RNUDC has also been identified in many different cell types and species, ranging from fibroblasts to neuronal cells and from mouse to human, suggesting a highly conserved function. Immunocytochemical studies, using affinity-purified rabbit anti-rat RNUDC antibodies, have localized a significant fraction of the RNUDC protein to the region of the Golgi apparatus in interphase rat Nb2 T cells, monkey kidney fibroblast COS-1 cells, and human 2AG10 adenocarcinoma cells. Treatment of 2AG10 cells with the microtubule-depolymerizing drug nocodazole, which causes dispersion of the Golgi apparatus, led to a diffuse pattern of RNUDC staining. Removal of nocodazole, which allowed the reformation of the Golgi apparatus, led to the reconcentration of RNUDC staining to the Golgi region. Taken together, these studies suggest that a fraction of RNUDC is tightly associated with the Golgi apparatus in many different mammalian cell types.


Subject(s)
Golgi Apparatus/metabolism , Nuclear Proteins/biosynthesis , Prolactin/pharmacology , Proteins , Animals , Antibodies , Antibody Specificity , Aspergillus nidulans , COS Cells , Cell Line , Clone Cells , Fungal Proteins , Golgi Apparatus/ultrastructure , Humans , Mammals , Mice , Nuclear Proteins/analysis , RNA, Messenger/biosynthesis , Rabbits , Rats , T-Lymphocytes , Transcription, Genetic , Tumor Cells, Cultured
19.
Endocrinology ; 138(8): 3426-34, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9231797

ABSTRACT

GH is known to activate JAK2 tyrosine kinase and members of the Stat family of transcription factors, including Stats 1, 3, and 5. The recent observation that at least two Stat5 proteins (Stat5A and Stat5B) exist in mouse and human, raises the question of whether GH activates both Stat5A and Stat5B and, if so, whether the requirements for activation are the same. An initial report investigating this issue demonstrated GH-dependent activation of Stat5A but not Stat5B. In this paper, we demonstrate (in COS cells expressing rat GH receptor (rGHR) and either Stat5A or Stat5B, 3T3-F442A fibroblasts, and CHO cells expressing rGHR) that GH induces tyrosyl phosphorylation of both Stat5A and Stat5B. Similar time courses of phosphorylation were observed for the two proteins. Interestingly, the pattern of observed bands differs for the two forms of Stat5. Two closely migrating Stat5A bands can be detected in cells treated with or without GH. Both of these bands become tyrosyl phosphorylated in response to GH. Three species of Stat5B are observed in untreated cells. An additional, more slowly migrating Stat5B band, appears upon treatment with GH. The three more slower migrating Stat5B bands observed in response to GH contain phosphorylated tyrosyl residues. We further demonstrate that GH induces binding of Stat5A and Stat5B, as well as Stat1, to the GAS-like element in the beta-casein promoter. We and others have demonstrated previously that specific regions of GHR are required for GH-dependent activation of what is here identified as Stat5B. To gain insight into the mechanism by which GH promotes tyrosyl phosphorylation of Stat5A, GH-dependent tyrosyl phosphorylation of Stat5A was examined in CHO cells expressing truncated and mutated rGHR. The results indicate that Stat5A and Stat5B require the same regions of rGHR for maximal activation by GH: the C-terminal half of the cytoplasmic domain; tyrosines 333 and/or 338 in the N-terminal half of the cytoplasmic domain; and the regions required for JAK2 activation. To dissect further the mechanism by which GH activates Stat5A and B, the requirement for JAK2 in GH-dependent Stat5 tyrosyl phosphorylation was assessed using JAK2-deficient cells expressing GHR (gamma2A-GHR) and the wild-type parental cell line expressing GHR (2C4-GHR). GH-induced tyrosyl phosphorylation of Stat5B in 2C4-GHR cells but not in the JAK2 deficient, gamma2A-GHR cells, indicating that JAK2 is required for GH-dependent tyrosyl phosphorylation of Stat5B. Western blotting revealed that Stat5A is not expressed in this cell type. Taken together, these findings suggest that: 1) GH activates both Stat5A and Stat5B in several cell types; 2) the pattern of bands observed differs for Stat5A and Stat5B; 3) GH-dependent tyrosyl phosphorylation of Stat5A requires specific regions of GHR, and these requirements are the same as for Stat5B; and 4) JAK2 kinase is required for GH-dependent tyrosyl phosphorylation of Stat5B and, most likely, Stat5A.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Growth Hormone/pharmacology , Milk Proteins , Proto-Oncogene Proteins , Trans-Activators/metabolism , Tyrosine/metabolism , Animals , Antibodies/analysis , Antibodies/immunology , Antibody Specificity , Blotting, Western , CHO Cells/cytology , CHO Cells/metabolism , CHO Cells/physiology , COS Cells/cytology , COS Cells/metabolism , COS Cells/physiology , Cell Line , Cricetinae , DNA-Binding Proteins/immunology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/physiology , Janus Kinase 2 , Mice , Phosphorylation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Rats , Receptors, Somatotropin/genetics , STAT5 Transcription Factor , Trans-Activators/immunology , Transfection
20.
Proc Soc Exp Biol Med ; 215(1): 35-52, 1997 May.
Article in English | MEDLINE | ID: mdl-9142136

ABSTRACT

The immunoregulatory properties of prolactin, a pituitary peptide hormone, have received renewed attention. The prolactin receptor, a member of the hematopoietin/cytokine receptor superfamily, is ubiquitously expressed by cells in the immune system. Certain subpopulations of lymphocytes synthesize and secrete biologically active prolactin, which suggests that prolactin can act as an autocrine and/or paracrine factor to modulate the activities of cells of the immune system. This review focuses on the molecular actions of prolactin in the immune system. Emphasis is given to recent information about the molecular mechanisms of prolactin receptor signal transduction, and the signaling molecules and prolactin-inducible target genes that participate in these responses. In particular, the prolactin-inducible interferon regulatory factor-1 gene and its roles in mediating diverse immune responses are highlighted.


Subject(s)
Immune System/physiology , Prolactin/metabolism , Prolactin/pharmacology , Animals , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Humans , Immune System/drug effects , Interferon Regulatory Factor-1 , Lymphocytes/metabolism , Phosphoproteins/metabolism , Phosphoproteins/pharmacology , Receptors, Prolactin/chemistry , Receptors, Prolactin/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...