Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Clin Exp Immunol ; 159(3): 327-37, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20015271

ABSTRACT

Wegener's granulomatosis (WG) is a life-threatening autoimmune vasculitis that affects lungs, kidneys and other organs. A hallmark of WG is the presence of classic anti-neutrophil cytoplasmic antibodies (c-ANCA) against self-proteinase 3 (PR3). Little is known about the role of these antibodies and PR3-specific immune responses in disease development. In this study, we demonstrate that PR3-specific autoimmune responses are pathogenic in non-obese diabetic (NOD) mice with an impaired regulatory arm of the immune response. Immunization of autoimmunity prone NOD mice with rmPR3 (recombinant mouse PR3) in complete Freund's adjuvant (CFA) resulted in high levels of c-ANCA, without detectable disease development. However, when splenocytes from these immunized mice were transferred into immunodeficient NOD-severe combined immunodeficiency (SCID) mice, the recipient mice developed vasculitis and severe segmental and necrotizing glomerulonephritis. No disease developed in NOD-SCID mice that received splenocytes from the CFA-alone-immunized donors (controls), indicating that disease development depends upon PR3-specific immune responses. In contrast to the pathology observed in NOD-SCID mice, no disease was observed when splenocytes from rmPR3-immunized C57BL/6 mice were transferred into immunodeficient C57BL/6-RAG-1(-/-) mice, suggesting that complex and probably multi-genetic factors play a role in the regulation of disease development.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/immunology , Antibody Specificity/immunology , Autoimmune Diseases/immunology , Glomerulosclerosis, Focal Segmental/immunology , Granulomatosis with Polyangiitis/immunology , Myeloblastin/immunology , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/pathology , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/pathology , Granulomatosis with Polyangiitis/chemically induced , Granulomatosis with Polyangiitis/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Species Specificity
2.
Am J Pathol ; 157(1): 221-36, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10880392

ABSTRACT

Tumor necrosis factor (TNF) is a mediator of the acute phase response in the liver and can initiate proliferation and cause cell death in hepatocytes. We investigated the mechanisms by which TNF causes apoptosis in hepatocytes focusing on the role of oxidative stress, antioxidant defenses, and mitochondrial damage. The studies were conducted in cultured AML12 cells, a line of differentiated murine hepatocytes. As is the case for hepatocytes in vivo, AML12 cells were not sensitive to cell death by TNF alone, but died by apoptosis when exposed to TNF and a small dose of actinomycin D (Act D). Morphological signs of apoptosis were not detected until 6 hours after the treatment and by 18 hours approximately 50% of the cells had died. Exposure of the cells to TNF+Act D did not block NFkappaB nuclear translocation, DNA binding, or its overall transactivation capacity. Induction of apoptosis was characterized by oxidative stress indicated by the loss of NAD(P)H and glutathione followed by mitochondrial damage that included loss of mitochondrial membrane potential, inner membrane structural damage, and mitochondrial condensation. These changes coincided with cytochrome C release and the activation of caspases-8, -9, and -3. TNF-induced apoptosis was dependent on glutathione levels. In cells with decreased levels of glutathione, TNF by itself in the absence of transcriptional blocking acted as an apoptotic agent. Conversely, the antioxidant alpha-lipoic acid, that protected against the loss of glutathione in cells exposed to TNF+Act D completely prevented mitochondrial damage, caspase activation, cytochrome C release, and apoptosis. The results demonstrate that apoptosis induced by TNF+Act D in AML12 cells involves oxidative injury and mitochondrial damage. As injury was regulated to a larger extent by the glutathione content of the cells, we suggest that the combination of TNF+Act D causes apoptosis because Act D blocks the transcription of genes required for antioxidant defenses.


Subject(s)
Apoptosis/drug effects , Homeostasis , I-kappa B Proteins , Liver/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/physiology , Caspases/drug effects , Caspases/metabolism , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , DNA-Binding Proteins/genetics , Dactinomycin/pharmacology , Enzyme Activation/drug effects , Glutathione/drug effects , Glutathione/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Liver/cytology , Liver/ultrastructure , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/physiology , NADP/drug effects , NADP/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Oxidation-Reduction , Protein Binding/drug effects , Reactive Oxygen Species/metabolism , Thioctic Acid/pharmacology , Transcriptional Activation/drug effects
3.
Mol Cell Biochem ; 199(1-2): 169-78, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10544965

ABSTRACT

Recent studies have suggested that MAP kinase phosphatase 1 (MKP-1) is overexpressed in prostate cancer. To evaluate the role of MKP-1 in regulating cell death and tumor growth in prostate cancer, MKP-1 was conditionally overexpressed in the human prostate cancer cell line DU145. Overexpression of MKP-1 in DU145 cells blocked activation of stress-activated protein kinase (SAPK/JNK). MKP-1 overexpression in DU-145 cells was also found to inhibit Fas ligand (FasL)-induced apoptosis, as well as block the activation of caspases by Fas engagement. In addition, MKP-1 blocked the activation of apoptosis by transfected MEKK-1 and ASK-1, presumably through its inhibition of the SAPK/JNK family of enzymes. MKP-1 blocked the ability of FasL to induce loss of mitochondrial transmembrane potential (delta Psi(m)), suggesting that MKP-1 acts upstream of mitochondrial pro-apoptotic events induced by FasL and that the SAPK/JNK pathway may form the signaling link between Fas receptor and mitochondrial dysfunction. Thus, MKP-1 overexpression in prostate cancer may play a role in promoting prostate carcinogenesis by inhibiting FasL-induced cell death.


Subject(s)
Apoptosis , Cell Cycle Proteins , Immediate-Early Proteins/metabolism , MAP Kinase Kinase Kinase 1 , Membrane Glycoproteins/metabolism , Mitochondria/metabolism , Phosphoprotein Phosphatases , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases , Protein Tyrosine Phosphatases/metabolism , Caspases/metabolism , Dual Specificity Phosphatase 1 , Enzyme Activation , Fas Ligand Protein , Humans , Immediate-Early Proteins/genetics , MAP Kinase Kinase Kinase 5 , MAP Kinase Kinase Kinases/metabolism , Male , Membrane Potentials , Metallothionein/genetics , Mitogen-Activated Protein Kinase 10 , Mitogen-Activated Protein Kinases/metabolism , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/genetics , Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Cells, Cultured
4.
Ann Oncol ; 10(11): 1361-7, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10631466

ABSTRACT

BACKGROUND: Etoposide mediates its cytotoxicity by inducing apoptosis. Thus, mechanisms which regulate apoptosis should also affect drug resistance. Oxidants and antioxidants have been shown to participate in the regulation of apoptosis. We were interested in studying whether responsiveness of acute myeloblastic leukemia (AML) cells to etoposide is mediated by oxidative stress and glutathione levels. PATIENTS AND METHODS: Two subclones of the OCI/AML-2 cell line which are etoposide-sensitive (ES), and etoposide-resistant (ER), were established by the authors at the University of Oulu, and used as models. Assays for apoptosis included externalization of phosphatidylserine (as evidenced by annexin V binding), and caspase activation as indicated by cleavage of poly(ADP-ribose)polymerase (Western blotting). Peroxide formation was analyzed by flow cytometry. Glutathione and gamma-glutamylcysteine synthetase (gamma-GCS) levels were determined spectrophotometrically and by Western blotting, respectively. RESULTS: Etoposide-induced apoptosis was evident 12 hours after treatment in the ES subclone, but was apparent in the ER subclone only after 24 hours. The basal glutathione and gamma-GCS levels were higher in the ER than the ES subclone. Etoposide increased peroxide formation in both subclones after 12-hour exposure. Significant depletion of glutathione was observed in the ES subclone during etoposide exposure, while glutathione levels were maintained in the ER subclone. In neither of the subclones was induction of gamma-GCS observed during 24-hour exposure to etoposide. Furthermore, the catalytic subunit of gamma-GCS was cleaved during apoptosis, concurrent with depletion of intracellular glutathione. When glutathione was depleted by treatment with buthionine sulfoximine, a direct inhibitor of gamma-GCS, the sensitivity to etoposide was increased, particularly in the ER subclone. CONCLUSIONS: The results underline the significance of glutathione biosynthesis in the responsiveness of AML cells to etoposide. The molecular mechanisms mediating glutathione depletion during etoposide exposure might include the cleavage of the catalytic subunit of gamma-GCS.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/physiology , Etoposide/pharmacology , Glutamate-Cysteine Ligase/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Blotting, Western , Drug Resistance, Neoplasm , Flow Cytometry , Glutamate-Cysteine Ligase/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Sensitivity and Specificity , Tumor Cells, Cultured
5.
Endocrinol Metab Clin North Am ; 27(3): 641-54, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9785058

ABSTRACT

The major goal of the evaluation and management of DLP in children is to provide safe and effective therapy with lifestyle modification. There is a strong rationale for the initiation of DLP treatment in childhood to limit the earliest stages of atherosclerosis, to establish lifelong lifestyle changes in diet and activity, and to limit the acquisition of additional CVD risk factors such as smoking and obesity. The NCEP has recommended screening for children with a parent with total cholesterol of 240 mg/dL or greater or a parent or grandparent with onset of CVD before age 55 years. Clinical evaluation and management are based on an LDL-C level of 130 mg/dL or greater. This approach to screening has a low sensitivity to identify children with DLP. Initial therapy is with a step 1 diet followed by the step 2 diet if necessary. Medications are reserved for older children with LDL-C of 190 mg/dL or greater after diet therapy or 160 mg/dL or greater with other CVD risk factors.


Subject(s)
Hyperlipoproteinemias/diagnosis , Hyperlipoproteinemias/therapy , Adolescent , Cardiovascular Diseases/prevention & control , Child , Cholesterol, LDL/blood , Diet, Fat-Restricted , Exercise , Humans , Hyperlipoproteinemias/complications , Obesity/complications , Obesity/prevention & control
6.
Proc Natl Acad Sci U S A ; 95(6): 3014-9, 1998 Mar 17.
Article in English | MEDLINE | ID: mdl-9501207

ABSTRACT

UV irradiation induces apoptosis in U937 human leukemic cells that is accompanied by the activation of both the stress-activated protein kinase (SAPK) and p38 mitogen-activated protein kinase (MAPK) signal transduction pathways. The MAPK phosphatase, MKP-1, is capable of inactivating both SAPK and p38 MAPK in vivo. To determine whether MKP-1-mediated inhibition of SAPK and/or p38 MAPK activity provided cytoprotection against UV-induced apoptosis, a U937 cell line conditionally expressing MKP-1 from the human metallothionein IIa promoter was established. Conditional expression of MKP-1 was found to abolish UV-induced SAPK and p38 MAPK activity, and inhibit UV-induced apoptosis as judged by both morphological criteria and DNA fragmentation. MKP-1 was also found to inhibit other biochemical events associated with apoptosis, including activation of caspase-3 and the proteolytic cleavage of the caspase-3 substrate, poly(ADP ribose) polymerase. These findings demonstrate that MKP-1 acts at a site upstream of caspase activation within the apoptotic program. The cytoprotective properties of MKP-1 do not appear to be mediated by its ability to inhibit p38 MAPK because the p38 MAPK specific inhibitor SB203580 had no effect on UV-induced apoptosis in U937 cells. Furthermore, by titrating the level of MKP-1 expression it was found that MKP-1 inhibited UV-induced SAPK activity, DNA fragmentation, and caspase-3 activation in a similar dose-dependent manner. The dual-specificity phosphatase, PAC1, which does not inhibit UV-induced activation of SAPK, did not provide a similar cytoprotection against UV-induced apoptosis. These results are consistent with a model whereby MKP-1 provides cytoprotection against UV-induced apoptosis by inhibiting UV-induced SAPK activity.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins , Immediate-Early Proteins/metabolism , Mitogen-Activated Protein Kinases , Phosphoprotein Phosphatases , Protein Tyrosine Phosphatases/metabolism , Radiation Tolerance/physiology , Ultraviolet Rays , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Dual Specificity Phosphatase 1 , Dual Specificity Phosphatase 2 , Humans , Immediate-Early Proteins/genetics , Protein Kinases/metabolism , Protein Phosphatase 1 , Protein Phosphatase 2 , Protein Tyrosine Phosphatases/genetics , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
7.
J Biol Chem ; 272(27): 16917-23, 1997 Jul 04.
Article in English | MEDLINE | ID: mdl-9202001

ABSTRACT

Phorbol ester tumor promoters, such as phorbol 12-myristate 13-acetate (PMA), are potent activators of extracellular signal-regulated kinase 2 (ERK2), stress-activated protein kinase (SAPK), and p38 mitogen-activated protein kinase (MAPK) in U937 human leukemic cells. These kinases are regulated by the reversible dual phosphorylation of conserved threonine and tyrosine residues. The dual specificity protein phosphatase MAPK phosphatase-1 (MKP-1) has been shown to dephosphorylate and inactivate ERK2, SAPK, and p38 MAPK in transient transfection studies. Here we demonstrate that PMA treatment induces MKP-1 protein expression in U937 cells, which is detectable within 30 min with maximal levels attained after 4 h. This time course coincides with the rapid inactivation of PMA-induced SAPK activity, but not ERK2 phosphorylation, which remains elevated for up to 6 h. To examine directly the role of MKP-1 in the regulation of these protein kinases in vivo, we established a U937 cell line that conditionally expresses MKP-1 from the human metallothionein IIa promoter. Conditional expression of MKP-1 inhibited PMA-induced ERK2, SAPK, and p38 MAPK activity. By titrating the levels of MKP-1 expression from the human metallothionein IIa promoter, however, it was found that p38 MAPK and SAPK were much more sensitive to inhibition by MKP-1 than ERK2. This differential substrate specificity of MKP-1 can be functionally extended to nuclear transcriptional events in that PMA-induced c-Jun transcriptional activity was more sensitive to inhibition by MKP-1 than either Elk-1 or c-Myc. Conditional expression of MKP-1 also abolished the induction of endogenous MKP-1 protein expression in response to PMA treatment. This negative feedback regulatory mechanism is likely due to MKP-1-mediated inhibition of ERK2, as studies utilizing the MEK1/2 inhibitor PD98059 suggest that ERK2 activation is required for PMA-induced MKP-1 expression. These findings suggest that ERK2-mediated induction of MKP-1 may play an important role in preferentially attenuating signaling through the p38 MAPK and SAPK signal transduction pathways.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Cycle Proteins , Enzyme Inhibitors/pharmacology , Immediate-Early Proteins/pharmacology , Mitogen-Activated Protein Kinases , Phosphoprotein Phosphatases , Protein Tyrosine Phosphatases/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Dual Specificity Phosphatase 1 , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 1 , Protein Phosphatase 1 , Substrate Specificity , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
8.
Mol Immunol ; 34(5): 409-18, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9293774

ABSTRACT

Stress-activated protein kinases (SAPK; also known as JNK for c-Jun N-terminal kinase) phosphorylate Ser63 and Ser73 in the amino-terminus of the c-Jun protein and potentiate its transcriptional activity. We have analysed phosphorylation of GST fusion proteins containing the c-Jun N-terminal domain by lysates of Daudi human B lymphoblastoid cells stimulated with medium or anti-IgM. Crosslinking membrane IgM (mIgM) results in an increase in phosphorylation of GST-c-Jun (5-89) in an antibody dose-dependent manner. The kinase activity specifically phosphorylates the c-Jun N-terminal domain since it does not phosphorylate GST or GST-JunB. The activity preferentially phosphorylates the substrate that contains the sites for in vivo phosphorylation by SAPK/JNK and requires the delta domain of c-Jun, which is also required for SAPK/JNK activity. However, the c-Jun N-terminal kinase activity induced by mIgM ligation is not precipitatable with anti-SAPK/JNK antibodies. In addition, unlike SAPK/JNKs, the mIgM-dependent c-Jun N-terminal kinase activity is not detectable in assays for renaturable kinase activity (in-gel assay) or in assays that test activities that bind to c-Jun (solid-phase assay). The increased phosphorylation of c-Jun N-terminal domain in response to mIgM ligation is unlikely to be due to mIgM-activated ERKs as it was not suppressed by a selective MEK inhibitor. Thus, the mIgM-induced activity is distinct from the known SAPK/JNKs and may represent a novel mechanism for c-Jun phosphorylation in response to mIgM engagement in human B cells.


Subject(s)
B-Lymphocytes/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Immunoglobulin M/metabolism , Mitogen-Activated Protein Kinases , Receptors, Antigen, B-Cell/metabolism , B-Lymphocytes/immunology , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Enzyme Activation/immunology , Enzyme Induction/immunology , Humans , Immunoglobulin M/physiology , JNK Mitogen-Activated Protein Kinases , Lymphoma, B-Cell , Phosphorylation , Protein Structure, Tertiary , Receptors, Antigen, B-Cell/physiology , Tumor Cells, Cultured
9.
Oncogene ; 11(11): 2365-74, 1995 Dec 07.
Article in English | MEDLINE | ID: mdl-8570188

ABSTRACT

Treatment of U937 human leukemic cells with the phorbol ester PMA, activates both mitogen-activated protein kinase (MAPK) and stress-activated protein kinase (SAPK), stimulates c-Jun phosphorylation and transcriptional activity, and induces a macrophage-like differentiation of U937 cells. The involvement of the MAPK pathway in mediating both the early phosphorylation and transcriptional activation events and the chronic differentiation of U937 cells was examined utilizing constitutively active MAPK kinase (MEK1) mutants. Transient expression of an activated MEK1 construct in U937 cells was found to stimulate MAPK and SAPK activity, as well as enhancing AP1-, SRE- and c-Jun-mediated transcriptional activity. Transient transfection of MAPK phosphatase-1 (MKP-1), a protein phosphatase which preferentially dephosphorylates and inactivates MAPK, inhibited the functional effects of both PMA and the constitutively active MEK1 mutants. To determine whether specific activation of the MEK/MAPK pathway was sufficient to induce hematopoietic differentiation, U937 cell lines were established that conditionally expressed the activated MEK1 mutant under the control of the human IIa metallothionein promoter. The induction of constitutively active MEK1 protein expression resulted in an increase in MEK1 activity, c-Jun and AP-1 transcriptional activity and an inhibition of U937 cell growth. However, this growth inhibition was not accompanied by U937 cell differentiation. These results suggest that a cross-talk mechanism exists between the MAPK and SAPK signal transduction pathways in U937 cells and that PMA-mediated SAPK activation may involve the MAPK pathway. Furthermore, selective activation of the MEK/MAPK pathway utilizing a constitutively active MEK1 mutant, while growth inhibitory, was not sufficient to induce the macrophage-like differentiation of U937 cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins , Leukemia/enzymology , Mitogen-Activated Protein Kinases , Phosphoprotein Phosphatases , Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription, Genetic , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Cell Differentiation/genetics , Cell Division/genetics , Cell Line , Dual Specificity Phosphatase 1 , Enzyme Activation , Enzyme Induction , Haplorhini , Humans , Immediate-Early Proteins/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases , Mutation , Protein Kinase Inhibitors , Protein Kinases/genetics , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
10.
Oncogene ; 10(5): 849-55, 1995 Mar 02.
Article in English | MEDLINE | ID: mdl-7898927

ABSTRACT

The transactivating function of the c-Jun proto-oncogene component of the AP-1 transcription factor is acutely regulated by a wide variety of cellular signals via modulation of phosphorylation of two serines (63 and 73). The viral oncoprotein, v-Jun, while containing homologous serines, is not phosphorylated in cells. A novel family of stress-activated protein kinases (SAPKs), also termed Jun N-terminal domain kinases (JNKs), are responsible for mediating S63/73 phosphorylation in response to a variety of cellular stimuli including tumor necrosis factor-alpha, heat stress and u.v. light. The p54 alpha 1, alpha 2, p54 beta and p46 beta SAPKs are shown to bind directly to c-Jun but not to v-Jun, with an absolute requirement for c-Jun amino acids 31-47, a region deleted in v-Jun. Inactive SAPKs tightly bind c-Jun in resting cells and may be a manifestation of the 'delta' inhibitor, a previously described repressor of c-Jun function.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Mitogen-Activated Protein Kinases , Proto-Oncogene Proteins c-jun/physiology , Repressor Proteins/physiology , Cell Line , Humans , JNK Mitogen-Activated Protein Kinases , Phosphorylation , Proto-Oncogene Mas
11.
Biochem J ; 305 ( Pt 3): 967-74, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7848298

ABSTRACT

Transcriptional activator proteins interact with the general transcription factors TATA-binding protein (TBP), TFIIB and/or other TBP-associated factors (TAFs). Using affinity chromatography we demonstrate that members of the Jun family of transcriptional activators interact with both TBP and TFIIB in vitro. TBP binds to both the N-terminal activation domain and C-terminal bZIP regions of c-Jun, whereas TFIIB binds to only the c-Jun bZIP domain. This interaction requires the dimerization of the Jun protein. The ability of the N-terminal activation domains of c-Jun, JunB, JunD and v-Jun to interact with TBP in vitro correlates with their transcriptional activity in vivo. Domain mapping experiments indicate that c-Jun interacts with the conserved C-terminus of TBP. Studies using a set of TFIIB inframe deletion mutants demonstrate that C-terminal amino acids 178-201 and 238-316 play an important role in modulating the interaction between TFIIB and c-Jun. Although phosphorylation of the c-Jun N-terminal activation domain stimulates c-Jun transcriptional activity in vivo, it has no effect on the ability of c-Jun to interact with either TBP or TFIIB in vitro. These data suggest that the Jun family of activator proteins may activate transcription by interacting with the general transcription factors TBP and TFIIB.


Subject(s)
Proto-Oncogene Proteins c-jun/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Chickens , DNA/metabolism , Gene Deletion , Glutathione Transferase/genetics , Humans , Leucine Zippers , Macromolecular Substances , Osmolar Concentration , Peptide Fragments/chemistry , Phosphorylation , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/genetics , Recombinant Fusion Proteins , Structure-Activity Relationship , Transcription Factor TFIIB , Transcription Factor TFIID , Transcription Factors/chemistry , Transcription Factors/genetics
12.
Methods Enzymol ; 254: 550-64, 1995.
Article in English | MEDLINE | ID: mdl-8531714

ABSTRACT

Many transcription factors are regulated by post-translational phosphorylation mechanisms. This chapter described several approaches that have been utilized to examine the phosphorylation of the c-Jun transcription factor. A combination of the techniques described in this chapter can be used to determine whether a transcription factor is phosphorylated in vivo, to analyze the sites of phosphorylation in vitro, and to permit the identification of putative protein kinases that may mediate this phosphorylation in vivo. Proteins labeled with 32P by either in vivo or in vitro techniques can be further analyzed by tryptic phosphopeptide mapping or phosphoamino acid analysis. These procedures have been described in detail elsewhere.


Subject(s)
Protein Processing, Post-Translational , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Animals , Autoradiography/methods , Cells, Cultured , Cloning, Molecular/methods , Electrophoresis, Polyacrylamide Gel/methods , Glutathione Transferase/biosynthesis , Indicators and Reagents , Phosphates/metabolism , Phosphorus Radioisotopes , Phosphorylation , Protein Biosynthesis , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-jun/isolation & purification , Rabbits , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Reticulocytes/metabolism , Transcription Factors/biosynthesis , Transcription Factors/isolation & purification
13.
Cell Growth Differ ; 4(5): 377-85, 1993 May.
Article in English | MEDLINE | ID: mdl-8390855

ABSTRACT

A variety of protein kinases, including pp42 and pp54 mitogen-activated protein (MAP) kinases, p34cdc2, and a partially purified protein kinase from 4 beta-phorbol 12-myristate 13 alpha-acetate (PMA)-treated U937 cells have been shown to phosphorylate the NH2-terminal activation domain of c-Jun in vitro. To investigate the role of pp42 MAP kinase in mediating c-Jun phosphorylation in vivo, we have treated U937 monocytic leukemia cells with a variety of pharmacological agents, including PMA, cycloheximide, AIF4, and okadaic acid. Although all of these agents stimulated c-Jun phosphorylation, cycloheximide and okadaic acid had no effect on pp42 MAP kinase phosphorylation, suggesting that MAP kinase activation was not necessary for c-Jun phosphorylation in vivo. Because dominant-negative RasAsn17 has been shown to block the effects of PMA on pp42 MAP kinase phosphorylation, we assessed its effect on c-Jun phosphorylation by cotransfection with a truncated c-Jun construct (c-Jun234). We found that c-Jun234 was expressed only in the cytosol and was inducibly phosphorylated with kinetics similar to those of endogenous nuclear c-Jun. Furthermore, we found that RasAsn17 had no effect on PMA-induced phosphorylation of c-Jun234. Because Ha-Ras requires isoprenylation for membrane binding, we examined the effect of the isoprenylation inhibitors lovastatin and perillic acid on PMA-induced c-Jun phosphorylation. Pretreatment of U937 cells with these agents had no effect on PMA-induced c-Jun or pp42 MAP kinase phosphorylation.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cycloheximide/pharmacology , Ethers, Cyclic/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/physiology , Cell Line , Humans , Mitogen-Activated Protein Kinase 1 , Okadaic Acid , Phosphorylation , Protein Serine-Threonine Kinases/drug effects , Protein-Tyrosine Kinases/drug effects , Proto-Oncogene Proteins/biosynthesis , Recombinant Proteins/drug effects
14.
Oncogene ; 8(2): 407-15, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8426747

ABSTRACT

Phorbol esters, such as phorbol myristate acetate (PMA), cause differentiation of U937 human monomyelocytic cells along the macrophage pathway. Within 15 min of PMA treatment DNA binding of the c-jun transcription factor is increased and is accompanied by rapid changes in the phosphate content of the c-jun protein. Phorbol esters stimulate phosphorylation of serines 63 and 73 located within the A1 transactivation domain of c-Jun that have previously been shown to positively regulate activity. A protein kinase activity is detectable in extracts of phorbol ester-treated U937 cells that specifically targets these two serines. Using novel assays, the protein kinase activity has been purified over 1000-fold. The major portion of protein kinase activity co-chromatographs over three columns with pp42/44 mitogen-activated protein kinases as judged by immunological methods. The significance of these results with respect to mitogen-induced transcription of AP-1-responsive genes is discussed.


Subject(s)
Leukemia, Myeloid/enzymology , Protein Serine-Threonine Kinases/isolation & purification , Protein-Tyrosine Kinases/isolation & purification , Proto-Oncogene Proteins c-jun/isolation & purification , Tetradecanoylphorbol Acetate/pharmacology , Base Sequence , Humans , Mitogen-Activated Protein Kinase 1 , Molecular Sequence Data , Phosphorylation , Tumor Cells, Cultured
16.
Proc Natl Acad Sci U S A ; 89(15): 7247-51, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1496019

ABSTRACT

Phorbol ester tumor promoters activate gene transcription by regulating both the synthesis and posttranslational modification of the activator protein 1 (AP-1) transcription factor, c-Jun and JunB are components of the mammalian AP-1 complex. Here we demonstrate that in U-937 human leukemic cells, phorbol esters stimulate the phosphorylation of the amino terminus of human c-Jun (JUN) but not human JunB (JUNB). Mutational analysis indicates that serine-63 and -73, which reside within the putative regulatory domain of JUN, are required for both constitutive and phorbol 12-myristate 13-acetate-inducible N-terminal JUN phosphorylation. To determine the functional role of this N-terminal phosphorylation, we prepared several chimeric proteins containing the N-terminal 84 amino acids (positions 5-89) of human JUN or murine JUNB fused to the yeast GAL4 DNA-binding domain. This region was found to be sufficient for the phorbol ester-inducible transcriptional activity of JUN, but not JUNB. This induction was abolished by the mutation of serine-63 and -73 to leucine residues. Thus, we propose that phorbol esters enhance the trans-activation potential of JUN, but not JUNB, by the phosphorylation of the N-terminal regulatory domain of JUN.


Subject(s)
Proto-Oncogene Proteins c-jun/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic , Amino Acid Sequence , Animals , Cell Line , Genes, jun , Humans , Methionine/metabolism , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphates/metabolism , Phosphorus Radioisotopes , Phosphorylation , Proto-Oncogene Proteins c-jun/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Nucleic Acid , Sulfur Radioisotopes , Transcription, Genetic/drug effects , Transfection
17.
Proc Natl Acad Sci U S A ; 89(12): 5341-5, 1992 Jun 15.
Article in English | MEDLINE | ID: mdl-1608942

ABSTRACT

c-Jun and its oncogenic counterpart v-Jun are completely conserved within the region from Ser-63 to Ser-73; these serines are sites for phorbol ester-inducible c-Jun phosphorylation. Using a U937 human leukemic cell line stably expressing v-Jun, we have demonstrated that phorbol esters stimulate the in vivo phosphorylation of c-Jun but not v-Jun. We developed an in vitro protein kinase assay to characterize the c-Jun protein kinase and to examine the determinants underlying this differential phosphorylation. Fusion proteins between glutathione S-transferase and the N terminus of c-Jun, v-Jun, or several c-Jun mutants were used as substrates. A c-Jun kinase activity was affinity-purified 5000-fold by using glutathione S-transferase-c-Jun-glutathione-Sepharose beads and was found to phosphorylate the N terminus of c-Jun but not v-Jun or c-Jun containing a 27-amino acid N-terminal deletion found in v-Jun. These effects were also observed in vivo as phorbol 12-myristate 13-acetate did not induce the phosphorylation of v-Jun or the c-Jun deletion mutant in U937 cell lines stably expressing these proteins. These findings indicate that the delta domain of c-Jun (amino acids 34-60), which is deleted in v-Jun, plays a critical role in regulating N-terminal c-Jun phosphorylation.


Subject(s)
Genes, jun , Oncogene Protein p65(gag-jun)/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Amino Acid Sequence , Base Sequence , Cell Line , Humans , Kinetics , Methionine/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Oncogene Protein p65(gag-jun)/genetics , Phosphates/metabolism , Phosphorylation , Protein Kinases/isolation & purification , Proto-Oncogene Proteins c-jun/genetics , Sequence Homology, Nucleic Acid
18.
J Cell Physiol ; 151(2): 415-26, 1992 May.
Article in English | MEDLINE | ID: mdl-1315324

ABSTRACT

Okadaic acid, a protein phosphatase inhibitor, is a strong tumor promoter which activates protein phosphorylation. Because another activator of protein phosphorylation, phorbol esters, stimulates hematopoietic differentiation, we sought to determine whether okadaic acid could also induce the differentiation of the human leukemic cell line U937. Differentiation was assessed by measuring changes in the following: mRNA levels, cell growth, morphology, cell surface markers, and the ability to induce superoxide. We found that okadaic acid treatment of U937 cells induces immediate increases in total cellular levels of both c-jun and c-fos mRNAs. Nuclear run-on experiments demonstrate that initial increases are secondary to increases in transcription, whereas latter changes may be secondary to mRNA stabilization. Like phorbol esters, okadaic acid treatment also activates AP-1 enhancer activity and induces the phosphorylation of c-Jun protein. Approximately 6-12 hours after treatment with okadaic acid, mRNA levels of c-myc, p34cdc2, and p58GTA, two cell cycle regulated protein kinases, decrease. Okadaic acid inhibits the growth of U937 cells, induces changes in nuclear morphology, stimulates increases in Mac-1 and Leu 11 surface antigens, and induces these cells to produce superoxide. These changes, taken together, suggest that U937 cells have been induced by okadaic acid to differentiate towards a more mature cell type.


Subject(s)
Ethers, Cyclic/pharmacology , Leukemia/pathology , Phosphoprotein Phosphatases/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/metabolism , Base Sequence , Biomarkers, Tumor , Cell Differentiation/drug effects , Enhancer Elements, Genetic , Humans , Leukemia/metabolism , Molecular Sequence Data , Okadaic Acid , Oligonucleotide Probes/genetics , Phosphorylation , Tumor Cells, Cultured
19.
Biochim Biophys Acta ; 1134(2): 137-42, 1992 Mar 16.
Article in English | MEDLINE | ID: mdl-1554749

ABSTRACT

Phorbol esters, epidermal growth factor (EGF) and serum induce the transient expression of the c-jun and c-fos proto-oncogenes in quiescent fibroblasts. While phorbol esters such as phorbol 12-myristate 13-acetate (PMA) are thought to induce the transcription of these genes by activating protein kinase C (PKC), the signal transduction pathway(s) mediating the effects of EGF and serum are still unclear. We have investigated whether PKC and/or calcium play a role in mediating EGF-stimulated c-jun and c-fos RNA and protein expression in quiescent NIH 3T3 fibroblasts. PMA, EGF or serum stimulated a rapid, transient increase in c-jun and c-fos expression and cJun protein synthesis in quiescent NIH 3T3 cells. Depletion of whole cell PKC activity by pretreatment with PMA abolished any subsequent response to PMA, but had no effect on the ability of EGF or serum to induce c-jun and c-fos RNA and cJun protein expression. Nuclear run-on analysis indicated that EGF-induced gene expression was due to an increase in the rate of transcription of c-jun and c-fos in both naive and PKC-depleted cells. The role of calcium in the EGF-induced expression of c-jun and c-fos was also investigated using an NIH 3T3 cell line (HER-14) overexpressing the wild type human EGF receptor. Removal of extracellular calcium by chelation with excess EGTA or use of the non-specific calcium channel blocker lanthanide, both of which abolish the EGF-induced calcium transient in HER-14 cells, had no effect on the PMA or EGF induced c-jun or c-fos response. These findings suggest that EGF induces c-jun and c-fos transcription and cJun protein synthesis in a manner independent of an increase in intracellular calcium or activation of PKC in quiescent NIH 3T3 cells.


Subject(s)
Epidermal Growth Factor/pharmacology , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Transcription, Genetic/drug effects , 3T3 Cells , Animals , Calcium/metabolism , Cell Line , Mice , Precipitin Tests , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-jun/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology
20.
Nucleic Acids Res ; 20(4): 897-902, 1992 Feb 25.
Article in English | MEDLINE | ID: mdl-1542579

ABSTRACT

To understand the mechanism by which phorbol esters (PMA) stimulate c-jun transcription in human leukemic cell line U937, we have mutated specific enhancer sequences within the c-jun promoter. We find in the region of DNA from -132 to +170 containing Sp1, C-TF and AP-1 sequences that mutation of the AP-1 sequence alone is not sufficient to abrogate transcription, and mutation of the Sp1 sequence increases transcription 4-fold. Although mutation of the CTF site had no effect, CTF and AP-1 mutations together totally abrogate PMA-induced transcription. In comparison mutations of either of these sites alone or together in a construct containing -1639/+740 of the c-jun promoter had no effect on transcription. Because this data suggested the possibility of other upstream control regions, we sequenced the promoter from -142 to -1639. This sequence demonstrates a greater than 70% homology between human, and mouse c-jun promoters for the region from -142 to -441, and a second AP-1-like site in the -183 to -192 region. Mutation of this site did not influence transcription by PMA. By making constructs containing varying portions of the promoter, we have identified the region between -142 and -711 to be responsible for mediating PMA-induced c-jun transcription.


Subject(s)
Gene Expression Regulation/drug effects , Genes, jun/genetics , Promoter Regions, Genetic/genetics , Tetradecanoylphorbol Acetate/analogs & derivatives , Base Sequence , Binding Sites/genetics , Enhancer Elements, Genetic/genetics , Humans , Molecular Sequence Data , Proto-Oncogene Proteins c-jun/metabolism , Sequence Alignment , Sequence Homology, Nucleic Acid , Sp1 Transcription Factor/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...