Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cancer Res ; 61(3): 916-20, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11221882

ABSTRACT

The role of the Jun family of proteins (c-Jun, JunB, and JunD) in oncogenesis has been extensively studied, but the distinct biological roles of each Jun protein is not known. For example, whereas c-Jun can transform primary cells in cooperation with an activated ras oncogene, JunD antagonizes ras-mediated transformation. We have discovered that two isoforms of the JunD transcription factor are ubiquitously expressed, resulting from use of an alternative translation start codon within the JunD mRNA. Here we report the first characterized functional difference between these JunD isoforms; only the full-length isoform of JunD binds to the Menin tumor suppressor protein. Furthermore, Menin suppresses transcriptional activity of the full-length but not the truncated isoform of JunD, which identifies the full-length JunD isoform as a functional target of Menin.


Subject(s)
Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins , Animals , CHO Cells/metabolism , Cricetinae , Genes, Tumor Suppressor , Mice , Neoplasm Proteins/genetics , Phosphorylation , Point Mutation , Protein Binding , Protein Isoforms , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcriptional Activation , Transfection
2.
EMBO J ; 17(19): 5615-26, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9755162

ABSTRACT

Stimulation by UV irradiation, TNFalpha, as well as PDGF or EGF activates the JNK/SAPK signalling pathway in mouse fibroblasts. This results in the phosphorylation of the N-terminal domain of c-Jun, increasing its transactivation potency. Using an antibody that specifically recognizes c-Jun phosphorylated at Ser63, we show that culture confluency drastically inhibited c-Jun N-terminal phosphorylation due to the inhibition of the JNK/SAPK pathway. Transfection experiments demonstrate that the inhibition occurs at the same level as, or upstream of, the small G-proteins cdc42 and Rac1. In contrast, the classical MAPK pathway was insensitive to confluency. The inhibition of JNK/SAPK activation depended on the integrity of the actin microfilament network. These results were confirmed and extended in monolayer wounding experiments. After PDGF, EGF or UV stimulation, c-Jun was predominantly phosphorylated in cells bordering the wound, which are the cells that move to occupy the wounded area. Thus, modulation of the stress-dependent signal cascade by confluency will restrict c-Jun N-terminal phosphorylation in response to mitogenic or chemotactic agents to cells that border a wounded area.


Subject(s)
Fibroblasts/cytology , Mitogen-Activated Protein Kinases , Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Actins/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Count , Cell Cycle Proteins/metabolism , Cell Movement , Cytoskeleton/metabolism , Fibroblasts/drug effects , Fibroblasts/radiation effects , GTP-Binding Proteins/metabolism , Growth Substances/pharmacology , JNK Mitogen-Activated Protein Kinases , Mice , Phosphorylation , Signal Transduction , Ultraviolet Rays , cdc42 GTP-Binding Protein , p38 Mitogen-Activated Protein Kinases , rac GTP-Binding Proteins
3.
Oncogene ; 15(8): 981-9, 1997 Aug 18.
Article in English | MEDLINE | ID: mdl-9285693

ABSTRACT

Activation of the AP-1 transcription factor and TGF-beta1 growth factor by ionizing radiation was studied both in vivo in pig skin, and in vitro in human fibroblasts and keratinocytes. Three and 6 h after irradiation, the Fos and Jun proteins and their binding activity to an AP-1 consensus sequence were strongly induced by high doses of gamma-rays. c-Fos, c-Jun and JunB proteins were found to be present in gel-shift complexes by probing with specific antibodies. Both keratinocytes and fibroblasts exhibited heightened AP-1 activity following irradiation. As we previously found that TGF-beta1 is involved in the development of skin lesions induced by radiation, TGF-beta1 gene expression was also examined. Two and 6 h after irradiation, the levels of TGF-beta1 transcripts were increased in skin. By immunostaining, TGF-beta1 protein levels were found to be increased in fibroblasts, keratinocytes and endothelial cells. As the TGF-beta1 promoter contains AP-1 binding sites, the relation between AP-1 activity and TGF-beta1 induction was addressed. The -365 TGF-beta1 promoter fragment, which contains a high affinity AP-1 site, exhibited increased binding to Jun and Fos proteins following irradiation. These results suggest that stress-inducible TGF-beta1 expression is mediated by the activation of AP-1 transcription factor.


Subject(s)
Skin/radiation effects , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta/biosynthesis , Animals , Cells, Cultured , Consensus Sequence , Dose-Response Relationship, Radiation , Gene Expression/radiation effects , Humans , Oxidative Stress , Promoter Regions, Genetic , Skin/metabolism , Swine , Transforming Growth Factor beta/genetics
4.
Oncogene ; 14(7): 819-30, 1997 Feb 20.
Article in English | MEDLINE | ID: mdl-9047389

ABSTRACT

We have analysed the different Jun and Fos proteins as NIH3T3 fibroblasts pass from exponential growth to quiescence and during the first 24 h after their re-entry into the cell cycle following serum stimulation. We show that these proteins can be divided into 3 subgroups based on their pattern of expression. The first contains c-Jun, Jun-D and Fra-2 which are expressed at high level in cycling cells and are only mildly induced by serum. The second contains Jun-B, c-Fos, Fos-B and deltaFos-B whose levels are low in cycling cells but increase strongly and rapidly after stimulation by serum. The third group contains only Fra-1, which is absent from cycling cells and behaves as a delayed early response protein after serum stimulation. AP-1 binding activity is low both in cycling and quiescent fibroblasts but increases after stimulation by serum with kinetics matching the induction of the various Jun and Fos proteins. Antibody supershift analyses demonstrate that the composition of AP-1 binding activity reflects the relative abundance of each Jun and Fos protein. Furthermore, the state of post-translational modification varies continuously for all of the AP-1 proteins as growth conditions change. These data indicate that AP-1 activity during the G0-G1 transition is finely regulated and complex, involving changes both in protein expression and in posttranslational modification.


Subject(s)
Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-jun/analysis , Transcription Factor AP-1/metabolism , 3T3 Cells , Animals , Antibody Specificity , Cell Cycle , DNA/metabolism , Mice , Molecular Weight , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism
5.
Oncogene ; 14(7): 837-47, 1997 Feb 20.
Article in English | MEDLINE | ID: mdl-9047391

ABSTRACT

The Ras proteins play a central role in regulating cell growth and their mutation can lead to abnormal proliferation. To analyse the potential link betwen AP1 activity, encoded by members of the jun and fos gene families, and Ras-mediated cellular transformation, we have studied several NIH3T3 clones which overexpress the Ha-Ras or Ki-Ras oncogenes. These transformed fibroblasts accumulated higher levels of cJun, JunB, Fra1 and Fra2 proteins relative to their normal counterparts. They also displayed increased AP1 DNA binding activity which was predominantly composed of cJun and Fra1 containing dimers. Following serum stimulation of Ras clones, the elevated levels of cJun and Fral remained steady, while the induction of JunB and Fra2 was partially attenuated. Moreover, deregulated Ras signaling resulted in a complete loss of the serum inducibility of cFos and FosB. Ectopic co-expression of cJun and Fra1 in NIH3T3 fibroblasts led to a transformed phenotype, attenuation of cFos serum inducibility, increased AP1 activity and Cyclin D1 accumulation, all characteristics of oncogenic Ras expressing cells. These results demonstrate that cJun and Fra1 are crucial mediators of the Ras-transformation process.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Transcription Factor AP-1/analysis , ras Proteins/physiology , 3T3 Cells , Animals , Cyclin D1 , Cyclins/analysis , DNA-Binding Proteins/analysis , Fos-Related Antigen-2 , Mice , Oncogene Proteins/analysis , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-jun/analysis , Transcription Factor AP-1/metabolism , Transcription Factors/analysis
6.
J Cell Sci ; 110 ( Pt 22): 2771-9, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9427286

ABSTRACT

The muscle regulatory factor, myf5, is involved in the establishment of skeletal muscle precursor cells. Little is known, however, about the control of the expression of the gene encoding this basic helix-loop-helix (bHLH) factor. We have addressed this question in the mouse myogenic cell line, C2, and in a derivative of this cell line where the myf5 gene is the only muscle-specific bHLH factor to be expressed at the myoblast stage. We present evidence that the synthetic glucocorticoid dexamethasone, and the pharmacological agent anisomycin, act synergistically to rapidly up-regulate the levels of myf5 transcript and protein. The glucocorticoid antagonist RU 486 abolishes this synergy, demonstrating the involvement of the glucocorticoid receptor. The expression of a dominant negative mutant of c-jun which interferes with the transactivating properties of all AP-1 family members also blocks the induction of myf5 by anisomycin and dexamethasone. An activator of protein kinase C (PKCs), 12-O-tetradecanoyl phorbol 13-acetate (TPA), abolishes the up-regulation of myf5 gene expression by dexamethasone and anisomycin, and its effect is counteracted by an inhibitor of PKCs, GF 109203X. These results point to the possible involvement of PKCs in the negative control of myf5. Evidence that both positive and negative regulation of myf5 transcripts, described here, does not require the fresh synthesis of transcription factors suggests that myf5 may behave like an immediate early gene.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Receptors, Glucocorticoid/genetics , Trans-Activators , Transcription Factor AP-1/genetics , Animals , Cell Line , Helix-Loop-Helix Motifs , Mice , Muscle Proteins/metabolism , Myogenic Regulatory Factor 5 , Receptors, Glucocorticoid/metabolism , Transcription Factor AP-1/metabolism , Transfection
7.
J Biol Chem ; 271(38): 23176-84, 1996 Sep 20.
Article in English | MEDLINE | ID: mdl-8798512

ABSTRACT

The urokinase-type plasminogen activator receptor (u-PAR) facilitates extracellular matrix proteolysis by accelerating plasmin formation at the cell surface. The present study was undertaken to identify elements in the u-PAR promoter required for the elevated expression of this binding site. Toward this end, we used two cultured colon cancer cell lines; one (RKO) has a transcriptionally activated u-PAR gene, and the other (GEO) overexpresses the receptor only after phorbol ester treatment. A chloramphenicol acetyltransferase (CAT) reporter driven by 398 nucleotides of 5' regulatory sequence of the u-PAR gene was strongly activated in the RKO cells, which displays approximately 3 x 10(5) receptors/cell. A region of this promoter between -197 and -8 was required for optimal expression, as indicated using a CAT reporter driven by 5' deleted fragments. DNase I footprinting revealed three protected regions (I, -190 to -171; II, -148 to -124; and III, -99 to -70) in this part of the promoter. Mutation of an AP-1 binding site at -184 within region I reduced activation of the promoter by 85%. Deletion of either region II or III also reduced promoter activity by over 60%. An oligonucleotide spanning the AP-1 motif at -184 bound, specifically, nuclear factors from RKO cells, and antibodies specific for Jun-D, c-Jun, or Fra-1 proteins supershifted the complex indicating the presence of these proteins. The amount of these factors was reduced in GEO cells in which the u-PAR gene is only weakly transcriptionally activated. Expression of a vector encoding a wild-type Jun-D cDNA increased u-PAR promoter activity in GEO cells. Conversely, transfection of RKO cells with a transactivation domain-lacking Jun-D expression construct resulted in a dose-dependent decrease in u-PAR promoter activity. Treatment of GEO cells with phorbol ester increased u-PAR mRNA and the activity of a CAT reporter driven by the wild-type but not the AP-1 (-184)-mutated u-PAR promoter, and this was associated with a strong induction in the amount of Jun-D, c-Jun, and c-Fos. Methylation interference studies using a fragment of the u-PAR promoter (spanning -201 to -150) bound with nuclear extracted proteins from RKO cells, and phorbol 12-myristate 13-acetate-treated and -untreated GEO cells showed that the contact points corresponded to the AP-1 binding site at -184. Thus, the elevated expression of u-PAR in RKO cells, which constitutively produces this binding site, as well as in phorbol 12-myristate 13-acetate-stimulated GEO cells requires an AP-1 motif located 184 bp upstream of the transcriptional start site.


Subject(s)
Receptors, Cell Surface/genetics , Regulatory Sequences, Nucleic Acid , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/metabolism , Transcriptional Activation/drug effects , Base Sequence , Binding Sites , Genes, Reporter , Molecular Sequence Data , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured
8.
Mol Cell Biol ; 16(5): 1881-8, 1996 May.
Article in English | MEDLINE | ID: mdl-8628254

ABSTRACT

Among the Jun family of transcription factors, only c-Jun displays full transforming potential in cooperation with activated c-Ha-Ras in primary rat embryo fibroblasts. c-Jun in combination with Ras can both induce foci of transformed cells from rat embryo fibroblast monolayers and promote the establishment of these foci as tumoral cell lines. JunB can also cooperate with Ras to induce foci but is unable to promote immortalization. We report here that JunD, in cooperation with Ras, induces foci with an efficiency similar to that of JunB. Artificial Jun/eb1 derivatives from each of the three Jun proteins were also analyzed. These constructs carry a heterologous homodimerization domain from the viral EB1 transcription factor and are thought to form only homodimers in the cell. We show here that these Jun/eb1 chimeras are potent transactivators of AP1 sites and that they can cooperate with c-Ha-Ras to induce foci. However, among all the Ras-Jun and Ras-Jun/eb1 combinations tested, only foci from Ras-c-Jun can be efficiently expanded and maintained as long-term growing cultures. Therefore, we suggest that a heterodimer containing c-Jun might be required for in vitro establishment of these primary mammalian cells.


Subject(s)
Cell Transformation, Neoplastic , Genes, jun , Genes, myc , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Blotting, Western , Cell Line, Transformed , Cells, Cultured , Embryo, Mammalian , Fibroblasts , Kinetics , Luciferases/biosynthesis , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins p21(ras)/biosynthesis , Rats , Rats, Wistar , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Transfection
9.
Oncogene ; 11(6): 1069-78, 1995 Sep 21.
Article in English | MEDLINE | ID: mdl-7566966

ABSTRACT

Interleukin-6 (IL-6) is a proinflammatory cytokine which also acts as a growth factor for some murine hybridomas (7TD1) or human myelomas (U266). We demonstrate that elevation of cAMP cellular content inhibits IL-6-stimulated cell growth, by blocking cells mainly in G1 phase. This inhibition is associated with increased expression of the Fos family protein Fra-2. Treatment of cells with 8Br-cAMP results in increased DNA-binding activity of two distinct AP-1 complexes; JunD/Fra-2 and JunB/Fra-2, and also in elevated AP-1 transactivation. When 8Br-cAMP is withdrawn from the medium, cells enter S phase and Fra-2 protein levels and AP-1 DNA-binding activity decrease to their basal value indicating that a temporally correlation exists between the 8Br-cAMP-mediated induction of JunD/Fra-2 AP-1 complex and the 7TD1 and U266 cell growth inhibition.


Subject(s)
Cyclic AMP/physiology , DNA-Binding Proteins/metabolism , Interleukin-6/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Base Sequence , Cell Division , Cell Line , Colforsin/pharmacology , DNA/metabolism , Fos-Related Antigen-2 , Humans , Mice , Molecular Sequence Data , Proto-Oncogene Proteins c-fos/biosynthesis , Transcriptional Activation
10.
Neuron ; 14(5): 927-39, 1995 May.
Article in English | MEDLINE | ID: mdl-7748560

ABSTRACT

Sympathetic neurons depend on nerve growth factor (NGF) for survival and die by apoptosis in its absence. We have investigated the pattern of expression of the Jun and Fos family of transcription factors in dying sympathetic neurons using antibodies specific for each family member. When sympathetic neurons are deprived of NGF, the level of c-Jun protein significantly increases, whereas the levels of the other members of the Jun and Fos family remain relatively constant. c-Jun also becomes more phosphorylated, probably on its amino terminal transactivation domain. When microinjected into sympathetic neurons, an expression vector for a c-Jun dominant negative mutant protects them against NGF withdrawal-induced death, indicating that AP-1 activity is essential for neuronal cell death. Furthermore, overexpression of the full-length c-Jun protein is, in itself, sufficient to induce apoptosis in sympathetic neurons.


Subject(s)
Apoptosis/genetics , Ganglia, Sympathetic/cytology , Genes, jun , Mutation , Neurons/physiology , Animals , Base Sequence , Cells, Cultured , Gene Expression , Molecular Sequence Data , Nerve Growth Factors/pharmacology , Nerve Growth Factors/physiology , Proto-Oncogene Proteins c-jun/physiology , Rats , Rats, Sprague-Dawley
11.
Oncogene ; 10(3): 495-507, 1995 Feb 02.
Article in English | MEDLINE | ID: mdl-7845674

ABSTRACT

The closely-related proteins c-Jun, JunB and JunD form a family of transcription factors which require dimerization for DNA-binding and transcriptional activity. Dimerization is mediated by a conserved amphipathic alpha-helix located adjacent to a highly charged DNA-binding domain. The Jun proteins can form both homo- and heterodimers within the Jun family and can also cross-dimerize with the Fos proteins. When expressed at high levels in primary chicken cells, each mouse Jun displays distinct transforming capacities: c-Jun transforms efficiently, JunB transforms poorly, and JunD does not transform at all. The composition of the transforming dimers, however, is unknown. To study the activity of Jun-Jun homodimers we constructed artificial derivatives, denoted Juneb1, in which the naturally occurring dimerization domain has been replaced by an heterologous homodimerization domain from the Epstein-Barr virus transcription factor EB1. These derivatives were introduced into chicken cells and assayed for their ability to affect growth. Unexpectedly, all three Juneb1 proteins conferred a transformed phenotype to primary cultures, promoting sustained growth in low-serum medium and colony formation from single cells in agar. These data demonstrate that when forced to accumulate as homodimers, both JunB and JunD can transform cells. They also suggest that the poor transforming activity of JunB and the absence of transforming activity of JunD may be due to their inability to accumulate to high levels as homodimers.


Subject(s)
Cell Transformation, Neoplastic , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/physiology , Amino Acid Sequence , Animals , Biopolymers/physiology , Cell Division , Cells, Cultured , Chickens , Mice , Molecular Sequence Data , Rabbits , Recombinant Fusion Proteins , Structure-Activity Relationship , Transcriptional Activation/physiology
12.
Oncogene ; 9(7): 1957-64, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8208542

ABSTRACT

We have studied the expression of the c-jun gene during dimethyl-sulfoxide (DMSO) induced differentiation of Friend erythroleukemia (F-MEL) cells. No expression of c-jun was detected in a differentiation-competent F-MEL cell line (745A) either before or after treatment with DMSO. By contrast, c-jun expression was constitutive in a F-MEL cell line (TFP10) resistant to DMSO-induced differentiation and increased with DMSO. We have investigated the possible role of c-jun in conferring this resistance by stably transfecting either sense or antisense c-jun constructs into both differentiation-sensitive 745A and defective TFP10 cell lines. Inhibition of c-jun expression by antisense transcripts in the TFP10 cells restored their ability to undergo erythroid differentiation when exposed to DMSO while expression of junB or junD antisense vectors failed to do so. In addition, c-jun overexpression in the 745A cells resulted in decreased DMSO-induced differentiation. These results indicate a correlation between the level of c-jun expression and the ability of F-MEL cells to undergo DMSO-induced differentiation and suggest that c-Jun may be an important negative regulator in this process.


Subject(s)
Erythropoiesis/genetics , Genes, jun , RNA, Antisense/pharmacology , Animals , Dimethyl Sulfoxide/pharmacology , Drug Resistance , Erythropoiesis/drug effects , Leukemia, Erythroblastic, Acute , Mice , RNA, Antisense/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection , Tumor Cells, Cultured
13.
Cell ; 76(4): 747-60, 1994 Feb 25.
Article in English | MEDLINE | ID: mdl-8124713

ABSTRACT

As NIH 3T3 fibroblasts become quiescent, the level of c-Jun protein decreases while JunD accumulates. When resting cells are stimulated with fresh serum, nuclear-localized JunD is rapidly degraded, followed by resynthesis of both c-Jun and JunD later in G1. Overexpression of JunD results in slower growth and an increase in the percentage of cells in G0/G1 while c-Jun overexpression produces larger S/G2 and M phase populations. In addition, JunD partially suppresses transformation by an activated ras gene whereas c-Jun cooperates with ras to transform cells. These data indicate that two closely related transcription factors can function in an opposing manner.


Subject(s)
Cell Cycle , Cell Transformation, Neoplastic/genetics , Genes, ras , Proto-Oncogene Proteins c-jun/genetics , Amino Acid Sequence , Animals , Fibroblasts/cytology , Mice , Molecular Sequence Data , Peptides/chemistry , Proto-Oncogene Proteins c-jun/metabolism
14.
Oncogene ; 8(8): 2311-5, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8336952

ABSTRACT

Among the murine Jun family of transcription factors, c-Jun and JunD are closely-related proteins with similar dimerization, DNA binding and transactivating properties. However, when expressed from a self-replicating retroviral RCAS vector, c-jun, but not junD, transforms chick embryo fibroblasts. We attempted to map the regions of c-jun which are important for transformation by constructing hybrids between c-jun and junD. Using common restriction sites, we prepared six different chimeric molecules. All of these c-jun:junD hybrids code for transactivators of AP1-containing promoters. An N-terminal segment of 79 amino acids of c-Jun converts JunD into a strong transforming protein, while other segments of c-Jun contribute to a lesser extent. Contrary to what has been reported with rat embryo fibroblasts, a c-Jun derivative with serines substituted by alanines in positions 63 and 73 still transforms CEFs efficiently.


Subject(s)
Cell Transformation, Neoplastic , Genes, jun , Recombinant Fusion Proteins/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Chick Embryo , Mice , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-jun/chemistry , Recombinant Fusion Proteins/pharmacology , Structure-Activity Relationship , Trans-Activators/genetics , Trans-Activators/pharmacology
15.
Cell Growth Differ ; 4(8): 689-97, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8398910

ABSTRACT

The effects that three different growth inhibitory cytokines exert on expression and function of members of the Jun family were studied in this work. M1 myeloblastic cells were chosen for this purpose because of their high growth sensitivity to interleukin 6 (IL-6), transforming growth factor beta 1 and alpha- and beta-interferons. It is reported here that IL-6 elevated the junB and c-jun mRNA levels and induced the formation of a novel DNA-protein complex with high sequence specificity to 12-O-tetradecanoylphorbol-13-acetate response element (TRE) oligonucleotides. This IL-6 induced TRE binding complex was abolished by anti-Jun specific antibodies and was efficiently competed by an oligonucleotide that comprises the mouse homologue of a previously described human c-myc negative DNA element. It persisted in cells for at least 48 h after IL-6 treatment and failed to be induced by alpha- and beta-interferons or by transforming growth factor beta 1, which affected differently the pattern of jun mRNA expression. To further explore regulatory and functional aspects of this induced TRE binding activity, an IL-6 resistant M1 clone was isolated and further analyzed. This clone carried a postreceptor deficiency that abrogated completely the growth inhibitory responses to IL-6 but did not interfere with the induction of two differentiation related cell surface markers. Interestingly, the IL-6 resistant clone had lost two molecular responses to IL-6, induction of TRE binding activity and suppression of the c-myc gene. The data correlate the IL-6 induced AP1 activity with the suppression of c-myc and growth inhibition.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-6/pharmacology , Leukemia, Myeloid, Acute/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Division/drug effects , Clone Cells/drug effects , Genes, jun , Interferon Type I/pharmacology , Mice , Molecular Sequence Data , RNA, Messenger/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured
17.
Nature ; 345(6272): 263-5, 1990 May 17.
Article in English | MEDLINE | ID: mdl-2139717

ABSTRACT

Recent evidence suggests that the force for poleward movement of chromosomes during mitosis is generated at or close to the kinetochores. Chromosome movement depends on motion relative to microtubules, but the identities of the motors remain uncertain. One candidate for a mitotic motor is dynein, a large multimeric enzyme which can move along microtubules toward their slow growing end. Dyneins were originally found in axonemes of cilia and flagella where they power microtubule sliding. Recently, cytoplasmic dyneins have also been found, and specific antibodies have been raised against them. The cellular localization of dynein has previously been studied with several antibodies raised against flagellar dynein, but the relevance of these data to the distribution of cytoplasmic dynein is not known. Antibodies raised against cytoplasmic dyneins have shown localization of dynein antigens to the mitotic spindles in Caenorhabditis elegans embryos (Lye et al., personal communication) and punctate cytoplasmic structures in Dictyostelium amoebae. Using antibodies that recognize subunits of cytoplasmic dyneins, we show here that during mitosis, cytoplasmic dynein antigens concentrate near the kinetochores, centrosomes and spindle fibres of HeLa and PtK1 cells, whereas at interphase they are distributed throughout the cytoplasm. This is consistent with the hypothesis that cytoplasmic dynein is a mitotic motor.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromosomes/ultrastructure , Dyneins/metabolism , Microtubules/ultrastructure , Mitosis , Animals , Cell Line , Chromosomes/metabolism , Cytoplasm/enzymology , Dyneins/analysis , Dyneins/isolation & purification , Fluorescent Antibody Technique , HeLa Cells/enzymology , Humans , Immunoblotting , Microtubules/metabolism , Molecular Weight
SELECTION OF CITATIONS
SEARCH DETAIL
...