Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
Add more filters

Publication year range
1.
Nat Med ; 3(6): 632-8, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9176489

ABSTRACT

We demonstrate here that synthetic 22-mer peptide 46, corresponding to the carboxy-terminal amino acid residues 361-382 of p53, can activate specific DNA binding of wild-type p53 in vitro and can restore the transcriptional transactivating function of at least some mutant p53 proteins in living cells. Introduction of peptide 46 in Saos-2 cells carrying a Tet-regulatable His-273 mutant p53 construct caused growth inhibition and apoptosis in the presence of mutant p53 but not in its absence, confirming that the effect of the peptide is mediated by reactivation of mutant p53. Moreover, peptide 46 caused apoptosis in mutant as well as wild-type p53-carrying human tumor cell lines of different origin, whereas p53 null tumor cells were not affected. These findings raise possibilities for developing drugs that restore the tumor suppressor function of mutant p53 proteins, thus selectively eliminating tumor cells.


Subject(s)
Apoptosis , Recombinant Fusion Proteins/administration & dosage , Tumor Suppressor Protein p53/chemistry , Cell Division/drug effects , Doxycycline/pharmacology , Fluorescent Antibody Technique , HeLa Cells , Humans , Osteosarcoma/genetics , Osteosarcoma/pathology , Peptides/pharmacology , Transcriptional Activation , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
J Cell Biol ; 122(1): 199-208, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8100231

ABSTRACT

The pathogenicity of the human c-erbB-2 oncogene was evaluated in transgenic mice. A DNA sequence comprising the promoter-enhancer region of the MMTV LTR and a constitutively activated allele of the human c-erbB-2 growth factor receptor gene was introduced into the germ line of mice. Expression of the transgene was observed in kidney, lung, mammary gland, salivary gland, Harderian gland, and in epithelial cells of the male reproductive tract. All transgenic mice expressing the c-erbB-2 receptor died within four months of birth. Histopathological analysis suggests that preneoplastic lesions in kidney and lung most likely caused organ failure and the early death of the transgenic mice. Focal dilatation and atypical proliferation of the tubular epithelial cells was found in the kidney. These hyperplastic lesions were found adjacent to normal tubules. Immunohistochemistry showed that normal renal structures were completely negative for c-erbB-2 protein expression. Atypical pseudopapillary proliferation of bronchial and bronchiolar epithelial cells narrowed the bronchial lumen in lung. Alveoli appeared normal. The expression of c-erbB-2 protein was strictly limited to the proliferating epithelial cells and not detected in normal tissue. The mammary glands of two parous mice were underdeveloped, lacking lobular-alveolar structures and were lactation deficient. Only a few ducts were interspersed in the fat pad. A virgin mouse developed a focal adenocarcinoma infiltrating the mammary fat pad. Expression of the c-erbB-2 protein was enhanced in the proliferating epithelial cells. Transgenic males were sterile. Epithelial hyperplasia and hypertrophy in the epididymis, vas deferens and seminal vesicles was found. The transgene is not uniformly expressed in the tissues where the MMTV LTR is transcriptionally active. The scattered transgene expression invariably coincides with epithelial hyperplasia.


Subject(s)
Gene Expression Regulation , Kidney/pathology , Lung/pathology , Precancerous Conditions/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Receptors, Cell Surface/genetics , Alleles , Animals , Death , Epididymis/pathology , Female , Genetic Vectors , Humans , Longevity , Male , Mammary Glands, Animal/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Organ Specificity , Precancerous Conditions/genetics , Proto-Oncogene Proteins/biosynthesis , Receptor, ErbB-2 , Receptors, Cell Surface/biosynthesis , Repetitive Sequences, Nucleic Acid
3.
J Cell Biol ; 108(3): 1127-38, 1989 Mar.
Article in English | MEDLINE | ID: mdl-2466037

ABSTRACT

We have established and characterized a spontaneously immortalized, nontumorigenic mouse mammary cell line, designated IM-2. IM-2 cells synthesize large amounts of the milk protein beta-casein upon addition of lactogenic hormones. The induction of beta-casein occurs rapidly and does not require any exogenous extracellular matrix components. The IM-2 cell line is morphologically heterogeneous and could be separated into cell clones with epithelial and fibroblastic characteristics. In monoculture, none of the epithelial clones could be induced to synthesize caseins. Coculture of epithelial and fibroblastic clones, however, rendered the epithelial cells competent to differentiate functionally; the addition of lactogenic hormones to these cocultures resulted in the synthesis of beta-casein in amounts comparable to that seen with the original IM-2 line. Using this unique cell system, we have investigated the interrelationships between different steps in differentiation leading to hormone-induced casein production. Independent of hormones, epithelial-fibroblastic cell contacts led to the formation of characteristic structures showing the deposition of laminin. We found that the epithelial cells located in these structures also exhibited significantly increased levels of cytokeratin intermediate filament polypeptides. Double immunofluorescence revealed that the cells inducible by hormones to synthesize casein, colocalized exactly with the areas of laminin deposition and with the cells showing greatly intensified cytokeratin expression. These results suggest that hormone-independent differentiation events take place in response to intercellular epithelial-mesenchymal contacts. These events in turn bring about a state of competence for functional differentiation after lactogenic hormonal stimulation.


Subject(s)
Mammary Glands, Animal/cytology , Animals , Caseins/biosynthesis , Caseins/genetics , Cell Communication , Cell Differentiation , Cell Line , Clone Cells , Electrophoresis, Polyacrylamide Gel , Epithelial Cells , Fibroblasts/cytology , Fluorescent Antibody Technique , Hydrocortisone/pharmacology , Insulin/pharmacology , Keratins/analysis , Keratins/metabolism , Laminin/metabolism , Mammary Glands, Animal/analysis , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred BALB C , Prolactin/pharmacology , RNA, Messenger/genetics , Vimentin/analysis
4.
Science ; 222(4630): 1341-3, 1983 Dec 23.
Article in English | MEDLINE | ID: mdl-6318311

ABSTRACT

Steroid hormones, when complexed to their receptors, recognize and bind specific DNA sequences and subsequently induce increased levels of transcription. The mechanisms of steroid hormone action were analyzed by constructing chimeric DNA molecules from portions of mouse mammary tumor virus envelope and long terminal repeat (LTR) regions ligated to the thymidine kinase (tk) gene of herpes simplex virus. This construction allowed the tk gene to be expressed in a hormone-responsive fashion upon transfection into Ltk- cells. Comparison of transcription data with in vitro binding data showed that hormone-responsive transcription can be directly correlated to the presence of steroid hormone receptor binding sites on the DNA. There are at least two such receptor binding sites in the LTR region, one between -202 and -137 and another between -137 and -50 base pairs from the RNA cap site, as well as a site near the 5' end of the envelope region. These results strengthen the hypothesis that steroid-receptor complexes regulate genes primarily by binding to DNA sites near the promoter region and thereby modulate transcription.


Subject(s)
DNA, Viral/metabolism , Glucocorticoids/pharmacology , Mammary Tumor Virus, Mouse/analysis , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/metabolism , Transcription, Genetic/drug effects , Animals , Binding Sites , Cell Line , Chimera , Glucocorticoids/metabolism , Mice , Repetitive Sequences, Nucleic Acid , Transfection , Triamcinolone Acetonide/metabolism
5.
Curr Opin Genet Dev ; 5(5): 587-94, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8664546

ABSTRACT

Mammary epithelial cells grow and develop with the onset of sexual maturity. In addition, lobular alveolar structures are formed during pregnancy, and quiescent differentiated cells secrete high levels of milk proteins after parturition. These events are governed by multiple hormones and growth factors and involve the sequential and synergistic action of functionally distinct signal transduction pathways. Milk protein genes have been analyzed and composite response elements have been identified in the promoter sequences. Transcription factors, which relay the hormonal signals, bind to these sequences. The factor that confers prolactin simulation to milk protein gene transcription has recently been identified. MGF/Stat5 is a latent transcription factor that becomes activated by a tyrosine-specific protein kinase, Jak2, associated with the prolactin receptor. Tyrosine phosphorylation converts the latent factor into one with DNA-binding and transcriptional activation potential. The regulation of MGF/Stat5 in vitro and in vivo indicates that it is a central component of the lactogenic hormone signaling pathway. Involvement of MGF/Stat5 in the signaling by other cytokines indicates that the same factor might be involved in regulation of growth-promoting genes, primarily in hematopoietic cells.


Subject(s)
Gene Expression Regulation , Mammary Glands, Animal/metabolism , Milk Proteins/biosynthesis , Prolactin/physiology , Animals , Base Sequence , Caseins/biosynthesis , Caseins/genetics , Conserved Sequence , DNA-Binding Proteins/metabolism , Epithelium/drug effects , Epithelium/metabolism , Female , Gene Expression Regulation/drug effects , Mammary Glands, Animal/drug effects , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , Prolactin/drug effects , Regulatory Sequences, Nucleic Acid , STAT5 Transcription Factor , Sexual Maturation , Trans-Activators/metabolism , Transcription, Genetic , Transcriptional Activation
6.
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
7.
Mol Cell Biol ; 14(9): 6004-12, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8065333

ABSTRACT

beta-Casein gene expression in mammary epithelial cells is under the control of the lactogenic hormones, glucocorticoids, insulin, and prolactin. The hormonal control affects gene transcription, and several regulatory elements in the beta-casein gene promoter between positions -80 and -221 have previously been identified. A region located in the promoter between positions -170 and -221 contains overlapping sequences for negative and positive regulatory elements. A sequence-specific single-stranded DNA-binding factor (STR), composed of two proteins with molecular masses of 35 and 54 kDa, recognizes the upper strand of this region and has a repressing role in transcription. High-level STR binding activity was observed in nuclear extracts from mammary glands of pregnant and postlactating mice and from noninduced HC11 mammary epithelial cells, cells with a low level of transcriptional activity of the beta-casein gene. STR activity is downregulated in mammary epithelial cells during lactation of the animals and after lactogenic hormone induction of HC11 cells in culture. These cells strongly transcribe the beta-casein gene. We investigated the mechanism of downregulation and found that a lactogenic-hormone-induced molecule (I-STR) inhibits STR from binding to its DNA target. I-STR is composed of RNA. STR is sequestered into the cytoplasm by I-STR after lactogenic hormone induction of mammary epithelial cells and remains present in an RNA-bound form. A high-affinity STR binding site was found in the 5' untranslated region of beta-casein mRNA. We propose that beta-casein mRNA can function as I-STR. beta-Casein mRNA may positively regulate its own transcription by translocating STR from the nucleus to the cytoplasm. The beta-casein STR binding sequence increases expression of a transfected beta-galactosidase gene when it is placed into the 5' untranslated region sequence of the mRNA. STR may have a positive role in posttranscriptional regulation.


Subject(s)
Caseins/metabolism , Gene Expression Regulation , Mammary Glands, Animal/physiology , Promoter Regions, Genetic , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Animals , Base Sequence , Lactation , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Rats , Transcription, Genetic , Vitellogenins/genetics
8.
Mol Cell Biol ; 13(12): 7303-10, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8246951

ABSTRACT

Transcription of the beta-casein gene in mammary epithelial cells is regulated by the lactogenic hormones insulin, glucocorticoids, and prolactin. The DNA sequence elements in the promoter which confer the action of the hormones on the transcriptional machinery and the nuclear proteins binding to this region have been investigated. We found that 221 nucleotides of promoter sequence 5' of the RNA start site are sufficient to mediate the induction of a chloramphenicol acetyltransferase reporter gene in transfected HC11 mammary epithelial cells. Deletion of 5' sequences to position -183 results in a construct with enhanced basal activity which still retains inducibility. A -170 beta-casein promoter-chloramphenicol acetyltransferase construct has very low transcriptional activity, which indicates the presence of a negative regulatory in the region between -221 and -183 and a positive regulatory element between -183 and -170. Band shift analysis showed that the promoter region between -194 and -163 specifically binds two nuclear proteins. The proteins are sequence-specific, single-stranded DNA-binding proteins which exclusively recognize the upper DNA strand and most likely play a repressing role in transcription. DNA binding activity of these nuclear proteins was observed only in nuclear extracts from mammary glands of mice in late pregnancy and postlactation, not during lactation. Hormonal control of the DNA binding activity of these proteins was also observed in the mammary epithelial cell line HC11. Mixing experiments showed that extracts from mammary tissue of lactating mice and from lactogenic hormone-treated HC11 cells contain an activity which can suppress the DNA binding of the single-stranded DNA-binding proteins.2+ identical specificity to the single-stranded DNA.


Subject(s)
Caseins/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Promoter Regions, Genetic , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA, Single-Stranded/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Insulin/pharmacology , Lactation/genetics , Lactation/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mice , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Phosphorylation , Pregnancy , Prolactin/pharmacology , Rats , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation , Transfection
9.
Mol Cell Biol ; 14(1): 128-37, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8264581

ABSTRACT

Expression of the beta-casein milk protein gene in the mammary epithelial cell line HC11 is primarily regulated at the transcriptional level. A 338-bp segment of promoter sequence 5' of the transcription start site is sufficient to confer inducibility by the lactogenic hormones insulin, glucocorticoid hormone, and prolactin. Positively and negatively acting promoter elements and specific DNA binding proteins have been identified. The binding of the mammary gland factor MGF to a site between -80 and -100 is indispensable for hormonal induction of transcription. Binding of MGF activity to DNA is greatly enhanced by the action of the lactogenic hormones. Repression of transcription in the uninduced state is mediated by a promoter element located adjacent to the MGF binding site at positions -110 to -150. This repressor element consists of two interacting protein binding sites. A nuclear factor that binds specifically to the proximal site between positions -110 and -120 has been characterized and found to be identical with the nuclear factor YY1 (delta, NF-E1). YY1 does not bind to the distal site. The simultaneous mutation in the proximal and the distal sites results in high, hormone-independent transcription. This finding suggests that YY1 plays a functional role in the repression and acts in conjunction with a second DNA binding protein. Comparison of YY1 DNA binding activity in uninduced and hormone-induced cells showed that relief of repression is not mediated by changes in the concentration or binding affinity of YY1. Infection of HC11 cells with a YY1-expressing recombinant retrovirus resulted in overexpression of YY1 but did not suppress hormonal induction. The addition of purified MGF decreased YY1 binding to its DNA recognition site in vitro. This finding indicates that MGF regulates the DNA binding activity of YY1 and thereby may cause the relief of transcriptional repression.


Subject(s)
Caseins/genetics , DNA-Binding Proteins/metabolism , Mammary Glands, Animal/metabolism , Milk Proteins , Promoter Regions, Genetic , Trans-Activators , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites/genetics , Caseins/biosynthesis , Cell Line , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/pharmacology , Dexamethasone/pharmacology , Epithelium/metabolism , Erythroid-Specific DNA-Binding Factors , Female , Gene Expression Regulation/drug effects , Insulin/pharmacology , Mammary Glands, Animal/drug effects , Mice , Models, Biological , Molecular Sequence Data , Prolactin/pharmacology , Promoter Regions, Genetic/drug effects , Rats , STAT5 Transcription Factor , YY1 Transcription Factor
10.
Mol Cell Biol ; 18(4): 1783-92, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9528750

ABSTRACT

The lactogenic hormones, i.e., prolactin and glucocorticoids, act in concert to stimulate transcription factors responsible for hormone-dependent milk protein gene expression. In the mammary gland, prolactin activates Stat5a and Stat5b and glucocorticoids activate the glucocorticoid receptor (GR). Immunoprecipitation experiments revealed that in mammary cells, Stat5a, Stat5b, and the GR are physically associated in vivo. The association is not dependent on lactogenic hormone treatment and is evident at all stages of mammary gland development. Immunodepletion experiments indicated that a fraction of GR and Stat5 proteins are not associated, suggesting that there are different intracellular pools of these proteins. Lactogenic hormone treatment of HC11 mammary cells resulted in tyrosine phosphorylation of Stat5a and Stat5b, dimerization, and rapid nuclear translocation of both Stat5 proteins. Following hormone treatment, Stat5a-Stat5b heterodimers were detected by their coimmunoprecipitation. In addition, immunodepletion experiments followed by gel shift analyses revealed the presence of active Stat5a and Stat5b homodimers. In mammary cells, Stat5b homodimers are less abundant than Stat5a homodimers. Although the GR does not bind the Stat5 DNA binding site directly, it could be detected with the Stat5-DNA complex. These results suggest that glucocorticoids affect milk protein gene expression via association of the GR with Stat5. Thus, there is a functional coupling between Stat-dependent and nuclear hormone receptor-dependent gene transcription.


Subject(s)
DNA-Binding Proteins/metabolism , Mammary Glands, Animal/metabolism , Milk Proteins , Receptors, Glucocorticoid/metabolism , Trans-Activators/metabolism , 3T3 Cells , Animals , Cell Differentiation , Cell Nucleus/metabolism , DNA/metabolism , DNA-Binding Proteins/chemistry , Dimerization , Female , Kinetics , Macromolecular Substances , Mice , Phosphorylation , STAT5 Transcription Factor , Trans-Activators/chemistry , Transcription, Genetic , Tyrosine/metabolism
11.
Mol Cell Biol ; 17(11): 6708-16, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9343435

ABSTRACT

Prolactin and glucocorticoid hormone are signals which regulate the transcription of milk protein genes in mammary epithelial cells. We have investigated the molecular mechanisms by which these hormones cooperate in the induction of transcription. Both hormones activate latent transcription factors in the cytoplasm of mammary epithelial cells. Prolactin exerts its effect through binding to the extracellular domain of the prolactin receptor and through receptor dimerization. This leads to the activation of a protein tyrosine kinase (Jak2), which is noncovalently associated with the cytoplasmic domain of the prolactin receptor. Jak2 phosphorylates the signal transducer and transcription activator (Stat5) which causes its dimerization and nuclear translocation where Stat5 specifically binds to sequence elements in the promoter regions of milk protein genes. In comparison, the glucocorticoid receptor is activated by a lipophilic steroid ligand in the cytoplasm which causes allosteric changes in the molecule, dimerization, and nuclear localization. It has been demonstrated that Stat5 and the glucocorticoid receptor form a molecular complex which cooperates in the induction of transcription of the beta-casein gene. We have defined the DNA sequence requirements for this cooperative mechanism and have delimited the functional domains in Stat5 and the glucocorticoid receptor that are necessary for the functional interaction. We find that the Stat5 response element (Stat5RE) within the beta-casein gene promoter is sufficient to elicit the cooperative action of Stat5 and the glucocorticoid receptor on transcription. Activation of Stat5 through phosphorylation of tyrosine 694 is an absolute prerequisite for transcription. Deletion of the transactivation domain of Stat5 results in a molecule which cannot mediate transactivation by itself but can still cooperate with the glucocorticoid receptor. Mutated variants of the glucocorticoid receptor with a nonfunctional DNA binding domain or a DNA binding domain contributed by the estrogen receptor are still able to cooperate with Stat5 in transcriptional induction. Deletion of the ligand binding domain of the glucocorticoid receptor does not impede cooperation with Stat5, whereas deletion of the AF-1 transactivation domain does prevent cooperation. Our results indicate that the glucocorticoid receptor acts as a ligand-dependent coactivator of Stat5 independently of its DNA binding function.


Subject(s)
Caseins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Milk Proteins , Receptors, Glucocorticoid/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Animals , Binding Sites , COS Cells , Caseins/biosynthesis , DNA-Binding Proteins/genetics , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Ligands , Models, Genetic , Phosphorylation , Prolactin/pharmacology , Promoter Regions, Genetic , Protein Binding , Receptors, Glucocorticoid/genetics , Recombinant Fusion Proteins/biosynthesis , STAT5 Transcription Factor , Sequence Deletion , Signal Transduction , Trans-Activators/genetics , Tyrosine/metabolism
12.
Mol Cell Biol ; 11(7): 3745-55, 1991 Jul.
Article in English | MEDLINE | ID: mdl-2046676

ABSTRACT

Transcription from the beta-casein milk protein gene promoter is induced by the synergistic action of glucocorticoid and prolactin hormones in the murine mammary epithelial cell line, HC11. We analyzed the binding of nuclear proteins to the promoter and determined their binding sites. Site-directed mutagenesis was used to determine the function of nuclear factor binding. During lactogenic hormone induction of HC11 cells, the binding of two nuclear factors increased. The binding of two other nuclear factors, present in uninduced cells, decreased. The basal activity of the promoter could be increased to and above the level of the induced wild-type promoter when the recognition sequences of the negatively regulated factors were mutated. This suggests that the beta-casein promoter is regulated by the relief of the repression of transcription. An essential tissue-specific factor was also found in nuclear extracts from the mammary glands of mice. Mutation of its recognition sequence in the beta-casein promoter led to the abolition of the induction of transcription by lactogenic hormones. The DNA sequences recognized by all five of these nuclear factors are conserved in the promoters of different casein genes from several species, confirming their importance in the regulation of milk protein gene transcription.


Subject(s)
Caseins/genetics , Cell Nucleus/physiology , Dexamethasone/pharmacology , Gene Expression Regulation , Mammary Glands, Animal/physiology , Nuclear Proteins/metabolism , Prolactin/pharmacology , Promoter Regions, Genetic , Transcription, Genetic , Animals , Base Sequence , Epithelium/drug effects , Epithelium/physiology , Female , Gene Expression Regulation/drug effects , Insulin/pharmacology , Mammary Glands, Animal/drug effects , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligonucleotide Probes , Pregnancy , Promoter Regions, Genetic/drug effects , Transcription, Genetic/drug effects
13.
Mol Cell Biol ; 10(8): 4027-34, 1990 Aug.
Article in English | MEDLINE | ID: mdl-2196443

ABSTRACT

The HC11 cell line was isolated from mammary gland cells of pregnant mice. The cells displayed a normal phenotype and retained some characteristics of mammary epithelial cell differentiation. After treatment with the lactogenic hormones prolactin and glucocorticoids, the HC11 cells expressed the milk protein beta-casein. Various oncogenes were transfected and expressed in HC11 cells. The oncogenes were tested for their transformation ability and for their effects upon the differentiation of the HC11 cells. All of the oncogenes tested, including activated human Ha-ras, human transforming growth factor-alpha, activated rat neuT, and human c-erbB-2 activated by a point mutation in the transmembrane domain, caused transformation of the HC11 cells, as shown by tumor formation in nude mice. HC11 cells expressing the neuT and activated c-erbB-2 genes synthesized beta-casein in response to lactogenic hormones, whereas those expressing the Ha-ras or transforming growth factor-alpha oncogenes were no longer able to respond to the lactogenic hormones. This inhibition of beta-casein production occurs at the transcriptional level and in the transforming growth factor-alpha-transformed cells is due to an autocrine mechanism involving the activation of the epidermal growth factor receptor. This suggests that, although the c-erbB-2 and epidermal growth factor receptors are structurally quite similar, their activation has different effects upon mammary epithelial cell differentiation.


Subject(s)
Caseins/genetics , Dexamethasone/pharmacology , Epidermal Growth Factor/pharmacology , ErbB Receptors/physiology , Gene Expression Regulation/drug effects , Genes , Prolactin/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Transfection , Animals , Antibodies, Monoclonal , Cell Line , Cell Transformation, Neoplastic , Clone Cells , Epithelium , ErbB Receptors/drug effects , Female , Insulin/pharmacology , Mammary Glands, Animal , Mice , Plasmids , Pregnancy , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/biosynthesis , Rats , Transforming Growth Factors/biosynthesis , Transforming Growth Factors/genetics
14.
Mol Cell Biol ; 17(7): 3663-78, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9199300

ABSTRACT

Stat (signal transducers and activators of transcription) and Jak (Janus kinases) proteins are central components in the signal transduction events in hematopoietic and epithelial cells. They are rapidly activated by various cytokines, hormones, and growth factors. Upon ligand binding and cytokine receptor dimerization, Stat proteins are phosphorylated on tyrosine residues by Jak kinases. Activated Stat proteins form homo- or heterodimers, translocate to the nucleus, and induce transcription from responsive genes. Stat5 and Stat6 are transcription factors active in mammary epithelial cells and immune cells. Prolactin activates Stat5, and interleukin-4 (IL-4) activates Stat6. Both cytokines are able to stimulate cell proliferation, differentiation, and survival. We investigated the transactivation potential of Stat6 and found that it is not restricted to lymphocytes. IL-4-dependent activation of Stat6 was also observed in HC11 mammary epithelial cells. In these cells, Stat6 activation led to the induction of the beta-casein gene promoter. The induction of this promoter was confirmed in COS7 cells. The glucocorticoid receptor was able to further enhance IL-4-induced gene transcription through the action of Stat6. Deletion analysis of the carboxyl-terminal region of Stat6 and recombination of this region with a heterologous DNA binding domain allowed the delimitation and characterization of the transactivation domain of Stat6. The potencies of the transactivation domains of Stat5, Stat6, and viral protein VP16 were compared. Stat6 had a transactivation domain which was about 10-fold stronger than that of Stat5. In pre-B cells (Ba/F3), the transactivation domain of Stat6 was IL-4 regulated, independently from its DNA binding function.


Subject(s)
DNA-Binding Proteins/genetics , Lymphocytes/physiology , Mammary Glands, Animal/physiology , Milk Proteins , Trans-Activators/genetics , Amino Acid Sequence , Animals , Caseins/genetics , Cells, Cultured , DNA-Binding Proteins/physiology , Epithelium/physiology , Gene Expression Regulation , Humans , Immediate-Early Proteins/genetics , Interleukin-4/physiology , Mice , Molecular Sequence Data , Oncostatin M , Peptides/genetics , Prolactin/physiology , Receptors, Glucocorticoid/physiology , STAT5 Transcription Factor , STAT6 Transcription Factor , Sequence Alignment , Sequence Homology, Amino Acid , Suppressor of Cytokine Signaling Proteins , Transcriptional Activation
15.
Mol Cell Biol ; 5(1): 268-72, 1985 Jan.
Article in English | MEDLINE | ID: mdl-3982419

ABSTRACT

A line of mouse mammary epithelial cells (NMuMG) has been characterized for its ability to be stably transfected with exogenous DNA. A transfection frequency of at least 1 cell per 1,000 was obtained with the pSV2neo plasmid. Several thousand G418-resistant NMuMG cell clones can easily be generated in cotransfection of genomic DNA and pSV2neo. The NMuMG cells were isolated from normal mammary glands and do not form malignant lesions when injected into nude mice. We have cotransfected NMuMG cells with pSV2neo and genomic DNA from the human EJ bladder carcinoma line, a cell line which contains an activated c-rasH oncogene. When a pool of 4,700 G418-resistant colonies was injected into nude mice, tumors were obtained. These tumors contain a transfected human rasH gene. Genomic DNA transfection into a line of mouse epithelial cells, in combination with the selection of stable transfectants and tumor induction in nude mice, can be used to screen human tumor DNA for the presence of activated oncogenes.


Subject(s)
Cell Transformation, Neoplastic , Mammary Neoplasms, Experimental/genetics , Oncogenes , Animals , Cell Line , Female , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Transfection
16.
Mol Cell Biol ; 16(10): 5691-700, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8816482

ABSTRACT

The Stat (signal transducer and activator of transcription) factors transmit cytokine, growth factor, and hormone responses. Seven members of the Stat gene family are known. MGF-Stat5a has been discovered as a mediator of the prolactin response in mammary epithelial cells. Two closely related variants of Stat5, Stat5a and Stat5b, are encoded by distinct genes. We examined the functional properties of the carboxyl termini of these molecules. Wild-type Stat5a (794 amino acids) and the carboxyl-terminal deletion mutant Stat5a delta 772 supported prolactin-induced transcription of a beta-casein promoter-reporter construct in COS7 cells; Stat5a delta 750 did not. Upon prolactin activation, tyrosine phosphorylation and the specificity of DNA binding were indistinguishable among the three Stat5a variants. Tyrosine dephosphorylation and the downregulation of the DNA-binding activity were delayed in the Stat5a delta 750 mutant. The carboxyl-terminal transactivation domain of Stat5a, amino acids 722 to 794, can be conferred to the DNA-binding domain of the yeast transcription factor GAL4. Coexpression of Stat5a or Stat5b and of the carboxyl-terminal deletion mutants resulted in the suppression of transcriptional induction in COS or Ba/F3 cells. We propose that Stat5a delta 750 and Stat5b delta 754 are lacking functional transactivation domains and exert their dominant negative effects by blocking the DNA-binding site in Stat5-responsive gene promoters.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Milk Proteins , Saccharomyces cerevisiae Proteins , Sequence Deletion , Trans-Activators/metabolism , Transcription Factors , Transcription, Genetic , Transcriptional Activation , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Caseins/genetics , Cattle , Cell Nucleus/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/chemistry , Fungal Proteins/biosynthesis , Fungal Proteins/metabolism , Genes, Reporter , Humans , Kinetics , Luciferases/biosynthesis , Molecular Sequence Data , Mutagenesis , Oligodeoxyribonucleotides , Phenotype , Phosphotyrosine/analysis , Promoter Regions, Genetic , Protein Structure, Secondary , STAT5 Transcription Factor , Sequence Homology, Amino Acid , Sheep , Trans-Activators/biosynthesis , Trans-Activators/chemistry , Transfection , Tumor Suppressor Proteins
17.
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.

18.
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
20.
Cancer Res ; 52(22): 6310-7, 1992 Nov 15.
Article in English | MEDLINE | ID: mdl-1358432

ABSTRACT

A high percentage of human breast and ovarian tumors display amplified c-erbB-2 gene copies, leading to overexpression of the growth factor receptor. Its membrane location and elevated expression make the erbB-2 protein an appropriate target for a directed tumor therapy. We have used recombinant DNA technology to produce a single-chain antibody-exotoxin A (scFv-ETA) fusion protein which specifically binds the human erbB-2 receptor. The scFv portion is composed of the heavy- and light-chain variable domains of a monoclonal antibody which recognizes the extracellular domain of the human erbB-2 receptor. The bacterially produced scFv-ETA protein was shown to bind specifically to cells expressing the human erbB-2 protein. The scFv-ETA inhibits protein synthesis in erbB-2-expressing tumor cells at doses ranging from 2 to 200 ng/ml and is cytotoxic for these cells at equivalent doses. In athymic nude mice, administration of the scFv-ETA inhibited the growth of erbB-2-overexpressing human ovarian carcinoma cells.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Carrier Proteins/pharmacology , Exotoxins/pharmacology , Immunotoxins/pharmacology , Mammary Neoplasms, Experimental/therapy , Ovarian Neoplasms/therapy , Proto-Oncogene Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Virulence Factors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Base Sequence , Carrier Proteins/genetics , Cell Division/drug effects , Chimera/genetics , DNA, Recombinant/genetics , Exotoxins/genetics , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Proteins/biosynthesis , Neoplasm Transplantation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Receptor, ErbB-2 , Recombinant Fusion Proteins/genetics , Recombinant Proteins , Transplantation, Heterologous , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL