Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Metab ; 79: 101838, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37995884

ABSTRACT

OBJECTIVE: Carbohydrate Response Element Binding Protein (ChREBP) is a glucose 6-phosphate (G6P)-sensitive transcription factor that acts as a metabolic switch to maintain intracellular glucose and phosphate homeostasis. Hepatic ChREBP is well-known for its regulatory role in glycolysis, the pentose phosphate pathway, and de novo lipogenesis. The physiological role of ChREBP in hepatic glycogen metabolism and blood glucose regulation has not been assessed in detail, and ChREBP's contribution to carbohydrate flux adaptations in hepatic Glycogen Storage Disease type 1 (GSD I) requires further investigation. METHODS: The current study aimed to investigate the role of ChREBP as a regulator of glycogen metabolism in response to hepatic G6P accumulation, using a model for acute hepatic GSD type Ib. The immediate biochemical and regulatory responses to hepatic G6P accumulation were evaluated upon G6P transporter inhibition by the chlorogenic acid S4048 in mice that were either treated with a short hairpin RNA (shRNA) directed against ChREBP (shChREBP) or a scrambled shRNA (shSCR). Complementary stable isotope experiments were performed to quantify hepatic carbohydrate fluxes in vivo. RESULTS: ShChREBP treatment normalized the S4048-mediated induction of hepatic ChREBP target genes to levels observed in vehicle- and shSCR-treated controls. In parallel, hepatic shChREBP treatment in S4048-infused mice resulted in a more pronounced accumulation of hepatic glycogen and further reduction of blood glucose levels compared to shSCR treatment. Hepatic ChREBP knockdown modestly increased glucokinase (GCK) flux in S4048-treated mice while it enhanced UDP-glucose turnover as well as glycogen synthase and phosphorylase fluxes. Hepatic GCK mRNA and protein levels were induced by shChREBP treatment in both vehicle- and S4048-treated mice, while glycogen synthase 2 (GYS2) and glycogen phosphorylase (PYGL) mRNA and protein levels were reduced. Finally, knockdown of hepatic ChREBP expression reduced starch domain binding protein 1 (STBD1) mRNA and protein levels while it inhibited acid alpha-glucosidase (GAA) activity, suggesting reduced capacity for lysosomal glycogen breakdown. CONCLUSIONS: Our data show that ChREBP activation controls hepatic glycogen and blood glucose levels in acute hepatic GSD Ib through concomitant regulation of glucose phosphorylation, glycogenesis, and glycogenolysis. ChREBP-mediated control of GCK enzyme levels aligns with corresponding adaptations in GCK flux. In contrast, ChREBP activation in response to acute hepatic GSD Ib exerts opposite effects on GYS2/PYGL enzyme levels and their corresponding fluxes, indicating that GYS2/PYGL expression levels are not limiting to their respective fluxes under these conditions.


Subject(s)
Blood Glucose , Glycogen Storage Disease Type I , Animals , Mice , Carbohydrate Metabolism , Disease Models, Animal , Glucose/metabolism , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Glycogen Synthase/metabolism , Liver Glycogen/metabolism , Phosphates , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
J Exp Med ; 171(3): 861-73, 1990 Mar 01.
Article in English | MEDLINE | ID: mdl-2307934

ABSTRACT

IL-4, a pleiotropic cytokine produced by T lymphocytes, plays an important role in immune responsiveness by regulating proliferation and differentiation of a variety of lymphoid and myeloid cells via binding to high affinity receptors. In this report we describe the isolation and functional expression of a human IL-4-R cDNA. When transfected into COS-7 cells, the cDNA encodes a 140-kD cell-surface protein. After transfection into a murine T cell line, the cDNA encodes a protein that binds human IL-4 with high affinity and can confer responsiveness to human IL-4. The predicted extracellular domain of the IL-4-R exhibits significant amino acid sequence homology with the beta subunit of the IL-2-R (p75), and the receptors for IL-6, erythropoietin, and prolactin. These receptors comprise a novel superfamily with extracellular domains characterized by four conserved cysteine residues and a double tryptophan-serine (WSXWS) motif located proximal to the transmembrane region.


Subject(s)
Interleukin-4/metabolism , Receptors, Mitogen/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA/analysis , Humans , Interleukin-4/pharmacology , Mice , Molecular Sequence Data , RNA, Messenger/analysis , Receptors, Interleukin-4 , Receptors, Mitogen/analysis , Signal Transduction
3.
Science ; 238(4832): 1386-92, 1987 Dec 04.
Article in English | MEDLINE | ID: mdl-2825349

ABSTRACT

Nuclear oncogene products have the potential to induce alterations in gene regulation leading to the genesis of cancer. The biochemical mechanisms by which nuclear oncoproteins act remain unknown. Recently, an oncogene, v-jun, was found to share homology with the DNA binding domain of a yeast transcription factor, GCN4. Furthermore, GCN4 and the phorbol ester-inducible enhancer binding protein, AP-1, recognize very similar DNA sequences. The human proto-oncogene c-jun has now been isolated, and the deduced amino acid sequence indicates more than 80 percent identity with v-jun. Expression of cloned c-jun in bacteria produced a protein with sequence-specific DNA binding properties identical to AP-1. Antibodies raised against two distinct peptides derived from v-jun reacted specifically with human AP-1. In addition, partial amino acid sequence of purified AP-1 revealed tryptic peptides in common with the c-jun protein. The structural and functional similarities between the c-jun product and the enhancer binding protein suggest that AP-1 may be encoded by c-jun. These findings demonstrate that the proto-oncogene product of c-jun interacts directly with specific target DNA sequences to regulate gene expression, and therefore it may now be possible to identify genes under the control of c-jun that affect cell growth and neoplasia.


Subject(s)
DNA-Binding Proteins/physiology , Protein Kinases , Proto-Oncogene Proteins/physiology , Proto-Oncogenes , Saccharomyces cerevisiae Proteins , Transcription Factors/physiology , Amino Acid Sequence , Antibodies/immunology , Avian Sarcoma Viruses/genetics , Base Sequence , Cross Reactions , DNA/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Enhancer Elements, Genetic , Fungal Proteins/genetics , Gene Expression Regulation , Genes, Viral , Humans , Molecular Sequence Data , Oncogene Protein p65(gag-jun) , Oncogenes , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-jun , Recombinant Proteins/genetics , Retroviridae Proteins/genetics , Saccharomyces cerevisiae/genetics , Sequence Homology, Nucleic Acid , Transcription Factors/genetics , Transcription Factors/immunology , Transcription, Genetic
4.
Science ; 240(4855): 1010-6, 1988 May 20.
Article in English | MEDLINE | ID: mdl-3130660

ABSTRACT

The Fos protein complex and several Fos-related antigens (FRA) bind specifically to a sequence element referred to as the HeLa cell activator protein 1 (AP-1) binding site. A combination of structural and immunological comparisons has identified the Fos-associated protein (p39) as the protein product of the jun proto-oncogene (c-Jun). The p39/Jun protein is one of the major polypeptides identified in AP-1 oligonucleotide affinity chromatography extracts of cellular proteins. These preparations of AP-1 also contain Fos and several FRA's. Some of these proteins bind to the AP-1 site directly whereas others, like Fos, appear to bind indirectly via protein-protein interactions. Cell-surface stimulation results in an increase in c-fos and c-jun products. Thus, the products of two protooncogenes (and several related proteins), induced by extracellular stimuli, form a complex that associates with transcriptional control elements containing AP-1 sites, thereby potentially mediating the long-term responses to signals that regulate growth control and development.


Subject(s)
Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Cell Transformation, Neoplastic , HeLa Cells/analysis , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins/isolation & purification , Proto-Oncogene Proteins c-jun
5.
Trends Biochem Sci ; 14(5): 172-5, 1989 May.
Article in English | MEDLINE | ID: mdl-2505415

ABSTRACT

Jun is a transcription factor that can also induce oncogenic transformation. Its DNA-binding domain is conserved from yeast to man and shows homology to several other transcriptional regulators. Jun dimerizes with the fos protein through an alpha-helical domain termed the leucine zipper, and the jun-fos heterodimers bind to DNA and regulate transcription of numerous specific unlinked genes.


Subject(s)
DNA-Binding Proteins/metabolism , Retroviridae Proteins/metabolism , Animals , Humans , Oncogene Protein p65(gag-jun)
6.
Bone ; 43(3): 459-68, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18583211

ABSTRACT

The mammalian skeleton consists of bones that are formed in two different ways: long bones via endochondral ossification and flat bones via intramembranous ossification. These different formation modes may result in differences in the composition of the two bone types. Using the 2D-difference in gel electrophoresis technique and mass spectrometry, we analyzed the composition of murine mineral-associated proteins of calvaria and long bone. Considerable differences in protein composition were observed. Flat bones (calvariae) contained more soluble collagen (8x), pigment epithelium derived factor (3x) and osteoglycin (4x); whereas long bones expressed more chondrocalcin (3x), thrombospondin- 1 (4x), fetuin (4x), secreted phosphoprotein 24 (3x), and thrombin (7x). Although cystatin motifs containing proteins, such as secreted phosphoprotein 24 and fetuin are highly expressed in long bone, they did not inhibit the activity of the cysteine proteinases cathepsin B and K. The solubility of collagen differed which coincided with differences in collagen crosslinking, long bone containing 3x more (hydroxylysine)-pyridinoline. The degradation of long bone collagen by MMP2 (but not by cathepsin K) was impaired. These differences in collagen crosslinking may explain the differences in the proteolytic pathways osteoclasts use to degrade bone. Our data demonstrate considerable differences in protein composition of flat and long bones and strongly suggest functional differences in formation, resorption, and mechanical properties of these bone types.


Subject(s)
Biomechanical Phenomena/methods , Bone Resorption , Bone and Bones/metabolism , Collagen/chemistry , Skull/metabolism , Animals , Bone and Bones/physiology , Calcium-Binding Proteins/metabolism , Collagen Type II/metabolism , Electrophoresis, Gel, Two-Dimensional , Eye Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mass Spectrometry/methods , Mice , Models, Biological , Nerve Growth Factors/metabolism , Serpins/metabolism , Skull/physiology , Stress, Mechanical , Thrombospondins/metabolism
7.
J Inherit Metab Dis ; 31(1): 88-96, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18188679

ABSTRACT

The outcome was determined of population-wide neonatal screening for medium-chain acyl-CoA dehydrogenase (MCAD) deficiency using tandem mass spectrometry (MS/MS) in The Netherlands, between October 2003 and September 2005. Prospective population-wide neonatal screening for MCAD deficiency was performed in the northern part of The Netherlands. In newborns with blood octanoylcarnitine (C(8:0)) concentrations > or =0.3 micromol/L, clinical and laboratory follow-up was initiated, including MCAD enzymatic measurements which played a decisive role. In a 2-year period, 66 216 newborns were investigated for MCAD deficiency and follow-up was initiated in 28 newborns. True-positives (n = 14) were identified based upon MCAD enzyme activity <50%, measured with hexanoyl-CoA as substrate. The observed prevalence of MCAD deficiency was 1/6600 (95% CI: 1/4100-1/17 400). In addition to an elevated C(8:0) concentration, a C(8:0)/C(10:0) molar ratio >5.0 turned out to differentiate between false-positives and true-positives. Measurement of MCAD activity using phenylpropionyl-CoA as a substrate further discriminated between newborns with MCAD deficiency and so-called mild MCAD deficiency. To summarize, neonatal screening for MCAD deficiency in the northern part of The Netherlands resulted in the predicted number of affected newborns. Measurement of MCAD activity in leukocytes or lymphocytes using phenylpropionyl-CoA as a substrate can be regarded as the gold standard to diagnose MCAD deficiency upon initial positive screening test results.


Subject(s)
Acyl-CoA Dehydrogenase/deficiency , Lipid Metabolism, Inborn Errors/diagnosis , Neonatal Screening , Acyl Coenzyme A/metabolism , Acyl-CoA Dehydrogenase/analysis , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase/metabolism , Cells, Cultured , DNA Mutational Analysis , False Positive Reactions , Follow-Up Studies , Genotype , Humans , Infant, Newborn , Leukocytes/enzymology , Lipid Metabolism, Inborn Errors/epidemiology , Lipid Metabolism, Inborn Errors/genetics , Lymphocytes/enzymology , Molecular Diagnostic Techniques/standards , Netherlands , Pilot Projects , Prevalence
8.
J Clin Invest ; 89(6): 1974-80, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1602003

ABSTRACT

To determine whether alkaline phosphatase (ALP) can cause the mineralization of collagenous matrices in vivo, bovine intestinal ALP was covalently bound to slices of guanidine-extracted demineralized bovine dentin (DDS). The preparations were implanted subcutaneously over the right half of the rat skull. Control slices not treated with the enzyme were implanted over the left half of the skull of the same animals. Specimens were harvested after periods varying from 1 to 4 wk. It was shown that ALP-coupled DDS rapidly accumulated hydroxyapatite crystals. 4 wk after implantation, the content of calcium and phosphate per microgram of hydroxyproline amounted up to 80 and 60%, respectively, of that found in normal bovine dentin. Our observations present direct evidence that ALP may play a crucial role in the induction of hydroxyapatite deposition in collagenous matrices in vivo.


Subject(s)
Alkaline Phosphatase/metabolism , Collagen/metabolism , Intestines/enzymology , Minerals/metabolism , Animals , Calcium/metabolism , Cattle , Collagen/ultrastructure , Dentin/metabolism , Dentin/ultrastructure , Female , Humans , Microscopy, Electron , Phosphates/metabolism , Prostheses and Implants , Rats , Substrate Specificity
9.
Mol Cell Biol ; 5(9): 2181-9, 1985 Sep.
Article in English | MEDLINE | ID: mdl-3016520

ABSTRACT

We have investigated the site of surface expression of the neuraminidase (NA) glycoprotein of influenza A virus, which, in contrast to the hemagglutinin, is bound to membranes by hydrophobic residues near the NH2-terminus. Madin-Darby canine kidney or primary African green monkey kidney cells infected with influenza A/WSN/33 virus and subsequently labeled with monoclonal antibody to the NA and then with a colloidal gold- or ferritin-conjugated second antibody exhibited specific labeling of apical surfaces. Using simian virus 40 late expression vectors, we also studied the surface expression of the complete NA gene (SNC) and a truncated NA gene (SN10) in either primary or a polarized continuous line (MA104) of African green monkey kidney cells. The polypeptides encoded by the cloned NA cDNAs were expressed on the surface of both cell types. Analysis of [3H]mannose-labeled polypeptides from recombinant virus-infected MA104 cells showed that the products of cloned NA cDNA comigrated with glycosylated NA from influenza virus-infected cells. Both the complete and the truncated glycoproteins were found to be preferentially expressed on apical plasma membranes, as detected by immunogold labeling. These results indicate that the NA polypeptide contains structural features capable of directing the transport of the protein to apical cell surfaces and the first 10 amino-terminal residues of the NA polypeptide are not involved in this process.


Subject(s)
Epithelium/ultrastructure , Glycoproteins/analysis , Influenza A virus/analysis , Membrane Proteins/analysis , Neuraminidase/analysis , Viral Envelope Proteins/analysis , Animals , Cattle , Cell Line , Chlorocebus aethiops , Cricetinae , DNA , Dogs , Epithelium/analysis , Glycoproteins/genetics , Hemagglutinin Glycoproteins, Influenza Virus , Hemagglutinins, Viral/analysis , Influenza A virus/genetics , Membrane Proteins/genetics , Neuraminidase/genetics , Protein Processing, Post-Translational , Recombinant Proteins/analysis , Simian virus 40/genetics , Vesicular stomatitis Indiana virus/analysis , Viral Envelope Proteins/genetics
10.
Mol Cell Biol ; 9(3): 1255-62, 1989 Mar.
Article in English | MEDLINE | ID: mdl-2725496

ABSTRACT

Transforming growth factor beta (TGF beta) is a multifunctional polypeptide that regulates proliferation, differentiation, and other functions of many cell types. The pathway of TGF beta signal transduction in cells is unknown. We report here that an early effect of TGF beta is an enhancement of the expression of two genes encoding serum- and phorbol ester tumor promoter-regulated transcription factors: the junB gene and the c-jun proto-oncogene, respectively. This stimulation was observed in human lung adenocarcinoma A549 cells which were growth inhibited by TGF beta, AKR-2B mouse embryo fibroblasts which were growth stimulated by TGF beta, and K562 human erythroleukemia cells, which were not appreciably affected in their growth by TGF beta. The increase in jun mRNA occurred with picomolar TGF beta concentrations within 1 h of TGF beta stimulation, reached a peak between 1 and 5 h in different cells, and declined gradually to base-line levels. This mRNA response was followed by a large increase in the biosynthesis of the c-jun protein (AP-1), as shown by metabolic labeling and immunoprecipitation analysis. However, differential and cell type-specific regulation appeared to determine the timing and magnitude of the response of each jun gene in a given cell. In AKR-2B and NIH 3T3 cells, only junB was induced by TGF beta, evidently in a protein synthesis-independent fashion. The junB response to TGF beta was maintained in c-Ha-ras and neu oncogene-transformed cells. Thus, one of the earliest genomic responses to TGF beta may involve nuclear signal transduction and amplification by the junB and c-jun transcription factors in concert with c-fos, which is also induced. The differential activation of the jun genes may explain some of the pleiotropic effects of TGF beta.


Subject(s)
Transcription Factors/genetics , Transforming Growth Factors/pharmacology , Animals , Cell Line , Cell Transformation, Neoplastic , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Proto-Oncogene Mas , Proto-Oncogenes/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/biosynthesis , Transfection , Transforming Growth Factors/administration & dosage
11.
Oncogene ; 8(7): 1895-903, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8510933

ABSTRACT

A direct comparison of the relative DNA-binding capabilities of in vivo Jun-containing complexes derived from overexpression of the highly transforming viral Jun (VJ-1 CEF), the weakly transforming chicken cellular Jun (CJ-3 CEF) or background endogenous Jun (RCAS CEF) was assessed by gel mobility-shift assays using a synthetic oligonucleotide containing the consensus sequence TGACTCA (consensus AP-1). Chicken embryo fibroblasts (CEFs) expressing background c-Jun levels (RCAS CEF) contain almost undetectable levels of c-Jun but retain significant DNA-binding activity with two distinct complexes capable of binding specifically to the consensus AP-1 site. CEFs overexpressing either v-Jun or c-Jun contain these same two complexes and, while showing marked increases in Jun protein levels, do not exhibit any increase in DNA binding or transcriptional activation activity, suggesting that much of the overexpressed protein is inactive. Gel-shift assays performed in the presence of a Jun-specific antibody revealed a reduction in binding by both complexes, suggesting that each contains Jun or a Jun cross-reactive protein. Antibodies specific for Jun B, c-Fos, Fos B and CREB failed to interact with either complex. However, antibody specific for Fra-2 caused a slight supershift, suggesting that one or both complexes may contain Fra-2. Gel-shift competition assays with 16 'AP-1- and CREB-like' target sequences revealed that, within each cell type, the two protein complexes varied in their ability to recognize the mutant target sequences. These results clearly indicate differences in potential target recognition by each specific in vivo complex, and suggest that each may preferentially bind its own subset of target DNAs. In addition, a comparison of binding by individual complexes derived from CEFs overexpressing v-Jun and c-Jun also revealed differences in target recognition. Thus, in vivo complexes formed by overexpression of v-Jun and c-Jun vary in their ability to recognize and bind to a number of 'AP-1- and CREB-like' target sequences. This has important implications with regard to the mechanisms involved in cell transformation by v-Jun.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , DNA/metabolism , Oncogene Protein p65(gag-jun)/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Animals , Base Sequence , Cells, Cultured , Chick Embryo , Gene Expression , Genes, jun , Molecular Sequence Data , Oncogene Protein p65(gag-jun)/genetics , Proto-Oncogene Proteins c-jun/genetics , Trans-Activators
12.
Oncogene ; 19(42): 4876-85, 2000 Oct 05.
Article in English | MEDLINE | ID: mdl-11039905

ABSTRACT

Overexpression of v-Jun in chicken embryo fibroblasts (CEF) leads to oncogenic transformation phenotypically characterized by anchorage independent growth and release from contact inhibition (focus formation). The mechanisms involved in this oncogenic conversion however, are not yet clear. Because Jun is a transcription factor, it has been assumed that oncogenic transformation results directly from deregulated AP-1 target gene expression. However, a number of experimental observations in avian cell culture models fail to correlate oncogenesis with AP-1 activity suggesting that transformation induced by v-Jun may occur through an indirect mechanism. To test this possibility, we introduced point mutations into the basic DNA binding domain of v-Jun and created mutants that exhibit altered binding specificity. When expressed in CEF, these mutants fail to deregulate three known v-Jun target genes (JTAP-1, apolipoprotein A1, c-Jun) thus demonstrating in vivo specificity changes. Each of the binding specificity mutants was also tested for its ability to induce oncogenic transformation. Interestingly, expression of these mutants in CEF results in a phenotype indistinguishable from the vector control with respect to growth rate, focus formation and the ability to form colonies in soft agar. These results are consistent with a model requiring direct AP-1 target deregulation as a prerequisite of v-Jun induced cell transformation. With this in mind, we generated a series of additional mutants that retain the ability to bind AP-1 sequence elements, but vary in their oncogenic potential. We demonstrate the use of these mutants to screen v-Jun induced gene targets for a functional role in cell transformation.


Subject(s)
Amino Acid Substitution , Cell Transformation, Viral/genetics , DNA/metabolism , Genes, jun , Oncogene Protein p65(gag-jun)/genetics , Animals , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , Avian Sarcoma Viruses/genetics , Avian Sarcoma Viruses/physiology , Cells, Cultured , Chick Embryo , Fibroblasts , Gene Expression Regulation, Viral , Models, Biological , Mutagenesis, Site-Directed , Oncogene Protein p65(gag-jun)/chemistry , Point Mutation , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-jun/biosynthesis , Recombinant Fusion Proteins/metabolism , Transcription Factor AP-1/metabolism , Transfection
13.
Oncogene ; 6(10): 1759-66, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1656361

ABSTRACT

Myoblasts from skeletal muscle of chicken or Japanese quail embryos were infected with avian sarcoma virus 17 (ASV-17), a retrovirus carrying the jun oncogene. At high multiplicities of infection ASV-17-induced morphologic transformation inhibited fusion of myoblasts into myotubes and stimulated extended replication. The expression of the muscle-specific proteins desmin, myosin and creatine phosphokinase was inhibited in ASV-17-infected cultures. Immunofluorescent staining detected strong expression of the ASV-17 Gag-Jun fusion protein in the nuclei of infected mononuclear myoblasts, but Gag-Jun was not detectable in multinucleated myotubes that occurred in clonal populations of ASV-17-infected quail myoblasts. This result suggests that the nuclear expression of viral jun and myogenic differentiation are mutually exclusive events. A mutant of ASV-17, ts jun-1, is partly temperature-sensitive in its ability to transform chicken embryo fibroblasts. At the non-permissive temperature of 41.5 degrees C, multinucleated myotubes readily formed in ts jun-1-infected myoblast cultures and expressed muscle-specific proteins detectable by immunofluorescent staining. These myotubes also showed strong immunofluorescent staining for Gag-Jun in the cell nuclei. The nuclear expression of a Jun protein that is defective in its transforming function appears therefore to be compatible with myogenesis. Several retroviral constructs carrying various viral and cellular jun inserts, as well as jun deletion mutants and recombinants between c-jun and v-jun, were tested for their effect on myogenic differentiation. There was an approximate correlation between the ability of a construct to transform chicken embryo fibroblasts and its effectiveness in interfering with myogenic differentiation. We conclude that the expression of an oncogenic jun gene in myoblasts strongly inhibits myogenic differentiation, and that a highly transforming Jun protein cannot be expressed in the nuclei of differentiating myotubes, while the presence of transformation-defective variants of Jun is compatible with differentiation.


Subject(s)
Cell Transformation, Viral/genetics , Genes, jun/physiology , Muscles/cytology , Animals , Avian Sarcoma Viruses/genetics , Cell Differentiation/genetics , Cells, Cultured , Chick Embryo , Coturnix , Muscles/ultrastructure , Mutation/genetics , Myofibrils/ultrastructure , Temperature
14.
Oncogene ; 12(1): 135-42, 1996 Jan 04.
Article in English | MEDLINE | ID: mdl-8552384

ABSTRACT

The oncogenic potential of Jun in chicken embryo fibroblasts (CEF) varies depending on its structure. V-Jun, which has a number of structural differences from c-Jun is highly transforming and tumorigenic. C-Jun however, is only weakly transforming and is not tumorigenic. We have used this difference in oncogenic potential between v-Jun and c-Jun to screen for downstream target genes associated with the v-Jun induced transformed phenotype. We describe here the identification, cloning and characterization of one of these genes, JTAP-1. JTAP-1 is consistently overexpressed 7 to 10-fold in CEF transformed by v-Jun compared with c-Jun overexpressing or normal CEF. This pattern of expression suggests that JTAP-1 is associated with the transformed phenotype. DNA and amino acid homology search analysis revealed that JTAP-1 shares a high degree of similarity with over 100 cysteine proteases from a variety of species and is likely the chicken homolog of cathepsin O. Analysis of expression of JTAP-1 in CEF overexpressing other oncogenes including v-Ha-ras, v-Src, c-Fos, and Myc revealed that it's overexpression is unique to v-Jun transformed cells. Thus, JTAP-1 is likely a specific target of v-Jun overexpression and not simply a consequence of cell transformation.


Subject(s)
Cathepsins/genetics , Cell Transformation, Neoplastic , Cysteine Endopeptidases/genetics , Endopeptidases , Gene Expression Regulation , Genes, jun , Amino Acid Sequence , Animals , Base Sequence , Cathepsin K , Cathepsin L , Chickens , Cloning, Molecular , Humans , Molecular Sequence Data , Rabbits , Up-Regulation
15.
Oncogene ; 16(5): 655-60, 1998 Feb 05.
Article in English | MEDLINE | ID: mdl-9482111

ABSTRACT

The product of the Jun oncogene influences a variety of processes including cell proliferation and differentiation. Jun exerts its influence by binding to the promoter and enhancer regions of a number of different target genes resulting in their activation or repression. We describe here the isolation and characterization of a gene differentially downregulated upon overexpression of v-Jun but not c-Jun. DNA and amino acid homology search analysis revealed this gene to be identical to chicken apolipoprotein A-1, the major component of high density lipoprotein (HDL). The half life of apolipoprotein A-1 RNA remains constant in the presence or absence of v-Jun overexpression suggesting downregulation by v-Jun is at the level of promoter activity. Consistent with this hypothesis, apolipoprotein A-1 upstream promoter fragments active in normal and c-Jun expressing CEF are inactive in v-Jun transformed CEF. Analysis of expression of apolipoprotein A-1 in CEF overexpressing other oncogenes revealed a similar downregulation by Myc and v-Src but not c-Fos, v-Ha-Ras, c-Src or c-Ski. Our findings point to a potential regulatory affect on cholesterol metabolism by v-Jun, as a result of altered levels of apolipoprotein A-1 message expression.


Subject(s)
Apolipoprotein A-I/physiology , Genes, jun , Proto-Oncogene Proteins c-jun/physiology , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Base Sequence , Chick Embryo , DNA/genetics , DNA/isolation & purification , Down-Regulation , Gene Expression , Gene Expression Regulation, Developmental/physiology , Molecular Sequence Data , Proto-Oncogene Proteins c-jun/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid
16.
Oncogene ; 4(2): 123-6, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2494630

ABSTRACT

The products of the proto-oncogenes c-jun and c-fos are known to form a complex in vivo. Complex formation appears to stabilize protein-DNA interactions and is thought to play an important functional role in transcriptional regulation. Here we show that the viral Jun oncoprotein, which differs structurally from cellular Jun, is also capable of complex formation with Fos. Thus the oncogenic potency of viral Jun is unlikely to be due to an altered affinity for Fos. We have also defined, by deletion analysis, the domain of v-Jun responsible for complex formation to reside in the carboxy terminus encompassing the leucine zipper motif. We find that complex formation with c-Fos does not occur with v-Jun deletions affecting one or more leucine residues in the zipper domain. Our results are consistent with the hypothesis that the leucine zipper mediates Jun-Fos interaction.


Subject(s)
Proto-Oncogene Proteins/metabolism , Retroviridae Proteins/metabolism , DNA/metabolism , DNA-Binding Proteins/metabolism , Mutation , Oncogene Protein p65(gag-jun) , Protein Biosynthesis , Protein Conformation , Proto-Oncogene Proteins c-fos , Proto-Oncogene Proteins c-jun , Transcription Factors/metabolism
17.
Oncogene ; 19(24): 2836-45, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10851087

ABSTRACT

The 5' untranslated region (UTR) of the chicken c-jun message is exceptionally GC rich and has the potential to form a complex and extremely stable secondary structure. Because stable RNA secondary structures can serve as obstacles to scanning ribosomes, their presence suggests inefficient translation or initiation through alternate mechanisms. We have examined the role of the c-jun 5' UTR with respect to its ability to influence translation both in vitro and in vivo. We find, using rabbit reticulocyte lysates, that the presence of the c-jun 5' UTR severely inhibits translation of both homologous and heterologous genes in vitro. Furthermore, translational inhibition correlates with the degree of secondary structure exhibited by the 5' UTR. Thus, in the rabbit reticulocyte lysate system, the c-jun 5' UTR likely impedes ribosome scanning resulting in inefficient translation. In contrast to our results in vitro, the c-jun 5' UTR does not inhibit translation in a variety of different cell lines suggesting that it may direct an alternate mechanism of translational initiation in vivo. To distinguish among the alternate mechanisms, we generated a series of bicistronic expression plasmids. Our results demonstrate that the downstream cistron, in the bicistronic gene, is expressed to a much higher level when directly preceded by the c-jun 5' UTR. In addition, inhibition of ribosome scanning on the bicistronic message, through insertion of a synthetic stable hairpin, inhibits translation of the first cistron but does not inhibit translation of the cistron downstream of the c-jun 5' UTR. These results are consistent with a model by which the c-jun message is translated through cap independent internal initiation. Oncogene (2000) 19, 2836 - 2845


Subject(s)
5' Untranslated Regions/physiology , Protein Biosynthesis , Proto-Oncogene Proteins c-jun/genetics , 3T3 Cells , 5' Untranslated Regions/chemistry , Animals , Base Sequence , Chick Embryo , Mice , Molecular Sequence Data , RNA, Messenger/chemistry , Rabbits
18.
Oncogene ; 7(6): 1119-25, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1594243

ABSTRACT

Mutants in the leucine zipper and basic regions of mouse c-jun were tested for transformation in chicken embryo fibroblast cultures. Reduction or elimination of the ability of Jun to dimerize or to bind to DNA severely decreased transformation. A chicken v-jun gene from which the major transactivation domain was deleted also failed to transform. We conclude that an intact leucine zipper, basic region and transactivation domain are required for Jun-induced oncogenic transformation. Coexpression of chicken c-Fos increased formation of transformed foci by Jun proteins of moderate to low oncogenic potency but had no effect on highly transforming Jun. Chicken c-Fos could also transform chicken embryo fibroblasts on its own, albeit after prolonged culture and at a low efficiency.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, fos , Genes, jun , Leucine Zippers/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Amino Acid Sequence , Animals , Blotting, Western , Cells, Cultured , Chick Embryo , Chickens , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Biosynthesis , Protein Conformation , Transcription, Genetic , Transcriptional Activation , Transfection
19.
Oncogene ; 9(11): 3241-7, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7936648

ABSTRACT

We have isolated and characterized cDNA clones that encode the rat homologue of a binding protein, LERK-2, for the receptor tyrosine kinase, elk. The cDNAs contain an open reading frame of 1527 nucleotides capable of encoding a protein 345 amino acid residues in length. The nucleotide sequence of the present clones is > 90% identical to the previously identified human LERK-2 cDNA, and the predicted proteins encoded by the rat and human clones are identical at 95% of amino acid residues. Recombinant proteins expressed from the rat cDNAs bind to elk with high affinity, similar to recombinant human LERK-2 and an endogenously-expressed rat elk-binding protein. Expression of the rat LERK-2 mRNA was detected in embryonic brain, kidney, lung, skeletal muscle, thymus, liver, and heart, and diminished in the early post-natal period. Significant LERK-2 mRNA expression in the young adult rat was restricted to the lung, kidney, heart and testes.


Subject(s)
Conserved Sequence , DNA-Binding Proteins , Gene Expression Regulation , Proteins/genetics , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Transcription Factors , Amino Acid Sequence , Animals , Base Sequence , Biological Evolution , Cloning, Molecular , DNA, Complementary , Ephrin-B1 , Humans , Male , Molecular Sequence Data , Open Reading Frames , Protein Binding , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sequence Homology, Amino Acid , Tissue Distribution , Tumor Cells, Cultured , ets-Domain Protein Elk-1
20.
Oncogene ; 18(44): 6063-70, 1999 Oct 28.
Article in English | MEDLINE | ID: mdl-10557095

ABSTRACT

We have previously demonstrated decreased Jun/AP-1 activity in the breast cancer cell line MCF-7 when compared to normal or immortalized mammary epithelial cells. In this paper, we overexpress Jun in MCF-7 cells (MCF7Jun) and demonstrate that it results in diverse biologic and biochemical changes, which mimic those seen clinically in breast cancer. Overexpression of Jun causes significant alterations in the composition of AP-1, decreased junB and increased fra-1 expression and results in an increased biologic aggressiveness. MCF7Jun cells exhibit increased cellular motility, increased expression of a matrix degrading enzyme MMP-9, increased in vitro chemoinvasion and tumor formation in nude mice in the absence of exogenous estrogens. Furthermore, MCF7Jun cells are unresponsive to the growth stimulating effects of estrogen and growth inhibitory effects of tamoxifen. Analysis of the estrogen receptor (ER) expression and activity showed that the MCF7Jun cells have no detectable ER. MCF-7 cells overexpressing mutant forms of cJun were responsive to the growth stimulatory effects of estrogen indicating that full-length cJun is required to acquire the estrogen-independent phenotype in breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Proto-Oncogene Proteins c-jun/genetics , Animals , Breast Neoplasms/drug therapy , Cell Movement/genetics , Estrogen Antagonists/pharmacology , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, fos , Humans , Mice , Mice, Nude , Mutation , Neoplasm Invasiveness , Phenotype , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Transcription Factor AP-1/metabolism , Transfection , Tumor Cells, Cultured , Vimentin/genetics , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL