Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Lupus Sci Med ; 2(1): e000121, 2015.
Article in English | MEDLINE | ID: mdl-26719810

ABSTRACT

OBJECTIVE: To describe the pharmacodynamic monitoring of (immuno)proteasome inhibition following treatment with bortezomib in a therapy-refractory systemic lupus erythematosus (SLE) patient with life-threatening myocarditis and lupus nephritis. PATIENT AND METHODS: Inhibition of catalytic activities of the proteasome subunits ß5 (constitutive proteasome), ß5i and ß1i (immunoproteasome) were measured in peripheral blood mononuclear cells using subunit-specific fluorogenic peptide substrates in a patient who received three cycles of bortezomib (1.3 mg/m(2) subcutaneously, days 1, 4, 8 and 11; every three weeks) along with plasma exchange during the first two cycles. RESULTS: Proteasome ß5, ß5i and ß1i subunit activities were readily inhibited 1 h after bortezomib administration. Twenty-four hours post-bortezomib administration, ß5 and ß5i activities were largely restored, whereas inhibition of ß1i activity was sustained. Clinically, after three cycles, cardiac function had improved, with concurrent improvement of haemodynamic stability during haemodialysis. Anti-ds-DNA dropped from >400 to 12 IU/mL along with normalisation of complement C3 and C4. Bortezomib therapy was well tolerated, and patient now has a sustained remission for >16 months. CONCLUSIONS: This case illustrates the potential benefit of pharmacodynamic monitoring of (immune)proteasome subunit-specific activity after bortezomib dosing in patients with therapy refractory SLE. This tool may hold potential to guide personalised/precision dosing aiming to achieve maximal efficacy and minimal toxicity.

2.
Mol Cancer Ther ; 13(6): 1625-35, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672057

ABSTRACT

MLN4924 is an investigational small-molecule inhibitor of the Nedd8-activating enzyme currently in phase I clinical trials. MLN4924 induces DNA damage via rereplication in most cell lines. This distinct mechanism of DNA damage may affect its ability to combine with standard-of-care agents and may affect the clinical development of MLN4924. As such, we studied its interaction with other DNA-damaging agents. Mitomycin C, cisplatin, cytarabine, UV radiation, SN-38, and gemcitabine demonstrated synergy in combination with MLN4924 in vitro. The combination of mitomycin C and MLN4924 was shown to be synergistic in a mouse xenograft model. Importantly, depletion of genes within the ataxia telangiectasia and Rad3 related (ATR) and BRCA1/BRCA2 pathways, chromatin modification, and transcription-coupled repair reduced the synergy between mitomycin C and MLN4924. In addition, comet assay demonstrated increased DNA strand breaks with the combination of MLN4924 and mitomycin C. Our data suggest that mitomycin C causes stalled replication forks, which when combined with rereplication induced by MLN4924 results in frequent replication fork collisions, leading to cell death. This study provides a straightforward approach to understand the mechanism of synergy, which may provide useful information for the clinical development of these combinations.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Cyclopentanes/administration & dosage , Drug Synergism , Mitomycin/administration & dosage , Pyrimidines/administration & dosage , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Animals , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Cell Line, Tumor , Chromatin/drug effects , Chromatin/genetics , DNA Damage/drug effects , Humans , Mice , Ubiquitin-Activating Enzymes/genetics , Ultraviolet Rays , Xenograft Model Antitumor Assays
3.
J Hematol Oncol ; 7: 7, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24418325

ABSTRACT

BACKGROUND: Despite encouraging results with the proteasome inhibitor bortezomib in the treatment of hematologic malignancies, emergence of resistance can limit its efficacy, hence calling for novel strategies to overcome bortezomib-resistance. We previously showed that bortezomib-resistant human leukemia cell lines expressed significantly lower levels of immunoproteasome at the expense of constitutive proteasomes, which harbored point mutations in exon 2 of the PSMB5 gene encoding the ß5 subunit. Here we investigated whether up-regulation of immunoproteasomes by exposure to interferon-γ restores sensitivity to bortezomib in myeloma and leukemia cell lines with acquired resistance to bortezomib. METHODS: RPMI-8226 myeloma, THP1 monocytic/macrophage and CCRF-CEM (T) parental cells and sub lines with acquired resistance to bortezomib were exposed to Interferon-γ for 24-48 h where after the effects on proteasome subunit expression and activity were measured, next to sensitivity measurements to proteasome inhibitors bortezomib, carfilzomib, and the immunoproteasome selective inhibitor ONX 0914. At last, siRNA knockdown experiments of ß5i and ß1i were performed to identify the contribution of these subunits to sensitivity to proteasome inhibition. Statistical significance of the differences were determined using the Mann-Whitney U test. RESULTS: Interferon-γ exposure markedly increased immunoproteasome subunit mRNA to a significantly higher level in bortezomib-resistant cells (up to 30-fold, 10-fold, and 6-fold, in ß1i, ß5i, and ß2i, respectively) than in parental cells. These increases were paralleled by elevated immunoproteasome protein levels and catalytic activity, as well as HLA class-I. Moreover, interferon-γ exposure reinforced sensitization of bortezomib-resistant tumor cells to bortezomib and carfilzomib, but most prominently to ONX 0914, as confirmed by cell growth inhibition studies, proteasome inhibitor-induced apoptosis, activation of PARP cleavage and accumulation of polyubiquitinated proteins. This sensitization was abrogated by siRNA silencing of ß5i but not by ß1i silencing, prior to pulse exposure to interferon-γ. CONCLUSION: Downregulation of ß5i subunit expression is a major determinant in acquisition of bortezomib-resistance and enhancement of its proteasomal assembly after induction by interferon-γ facilitates restoration of sensitivity in bortezomib-resistant leukemia cells towards bortezomib and next generation (immuno) proteasome inhibitors.


Subject(s)
Boronic Acids/pharmacology , Drug Resistance, Neoplasm/drug effects , Interferon-gamma/pharmacology , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , Up-Regulation/drug effects , Apoptosis/drug effects , Blotting, Western , Bortezomib , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , HLA Antigens/genetics , HLA Antigens/metabolism , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/pharmacology , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
4.
Cancer Res ; 73(1): 225-34, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23100467

ABSTRACT

MLN4924 is an investigational small-molecule inhibitor of the NEDD8-activating enzyme (NAE) in phase I clinical trials. NAE inhibition prevents the ubiquitination and proteasomal degradation of substrates for cullin-RING ubiquitin E3 ligases that support cancer pathophysiology, but the genetic determinants conferring sensitivity to NAE inhibition are unknown. To address this gap in knowledge, we conducted a genome-wide siRNA screen to identify genes and pathways that affect the lethality of MLN4924 in melanoma cells. Of the 154 genes identified, approximately one-half interfered with components of the cell cycle, apoptotic machinery, ubiquitin system, and DNA damage response pathways. In particular, genes involved in DNA replication, p53, BRCA1/BRCA2, transcription-coupled repair, and base excision repair seemed to be important for MLN4924 lethality. In contrast, genes within the G(2)-M checkpoint affected sensitivity to MLN4924 in colon cancer cells. Cell-cycle analysis in melanoma cells by flow cytometry following RNAi-mediated silencing showed that MLN4924 prevented the transition of cells from S-G(2) phase after induction of rereplication stress. Our analysis suggested an important role for the p21-dependent intra-S-phase checkpoint and extensive rereplication, whereas the ATR-dependent intra-S-phase checkpoint seemed to play a less dominant role. Unexpectedly, induction of the p21-dependent intra-S-phase checkpoint seemed to be independent of both Cdt1 stabilization and ATR signaling. Collectively, these data enhance our understanding of the mechanisms by which inhibition of NEDD8-dependent ubiquitination causes cell death, informing clinical development of MLN4924.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclopentanes/pharmacology , DNA Damage/drug effects , Melanoma/metabolism , Pyrimidines/pharmacology , Ubiquitins/metabolism , Blotting, Western , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Flow Cytometry , Humans , NEDD8 Protein , Polymerase Chain Reaction
5.
Mol Cell Proteomics ; 10(11): M111.009183, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21873567

ABSTRACT

Cullin-RING ubiquitin ligases (CRLs) are responsible for the ubiquitination of many cellular proteins, thereby targeting them for proteasomal degradation. In most cases the substrates of the CRLs have not been identified, although many of those that are known have cancer relevance. MLN4924, an investigational small molecule that is a potent and selective inhibitor of the Nedd8-activating enzyme (NAE), is currently being explored in Phase I clinical trials. Inhibition of Nedd8-activating enzyme by MLN4924 prevents the conjugation of cullin proteins with NEDD8, resulting in inactivation of the entire family of CRLs. We have performed stable isotope labeling with amino acids in cell culture analysis of A375 melanoma cells treated with MLN4924 to identify new CRL substrates, confidently identifying and quantitating 5122-6012 proteins per time point. Proteins such as MLX, EID1, KLF5, ORC6L, MAGEA6, MORF4L2, MRFAP1, MORF4L1, and TAX1BP1 are rapidly stabilized by MLN4924, suggesting that they are novel CRL substrates. Proteins up-regulated at later times were also identified and siRNA against their corresponding genes were used to evaluate their influence on MLN4924-induced cell death. Thirty-eight proteins were identified as being particularly important for the cytotoxicity of MLN4924. Strikingly, these proteins had roles in cell cycle, DNA damage repair, and ubiquitin transfer. Therefore, the combination of RNAi with stable isotope labeling with amino acids in cell culture provides a paradigm for understanding the mechanism of action of novel agents affecting the ubiquitin proteasome system and a path to identifying mechanistic biomarkers.


Subject(s)
Cyclopentanes/pharmacology , Proteome/metabolism , Pyrimidines/pharmacology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Kinetics , Phenotype , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Proteome/genetics , Proteomics , RNA Interference , Ubiquitin-Activating Enzymes/metabolism , Ubiquitination
6.
Bioorg Med Chem Lett ; 20(22): 6581-6, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20875739

ABSTRACT

Starting from a tripeptide screening hit, a series of dipeptide inhibitors of the proteasome with Thr as the P3 residue has been optimized with the aid of crystal structures in complex with the ß-5/6 active site of y20S. Derivative 25, (ß5 IC(50)=7.4 nM) inhibits only the chymotryptic activity of the proteasome, shows cellular activity against targets in the UPS, and inhibits proliferation.


Subject(s)
Chymotrypsin/antagonists & inhibitors , Dipeptides/chemistry , Proteasome Endopeptidase Complex/metabolism , Threonine/chemistry , Humans , Models, Molecular
7.
Biochem J ; 430(3): 461-76, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20632995

ABSTRACT

The mammalian 26S proteasome is a 2500 kDa multi-catalytic complex involved in intracellular protein degradation. We describe the synthesis and properties of a novel series of non-covalent di-peptide inhibitors of the proteasome based [corrected] on a capped tri-peptide that was first identified by high-throughput screening of a library of approx. 350000 compounds for inhibitors of the ubiquitin-proteasome system in cells. We show that these compounds are entirely selective for the beta5 (chymotrypsin-like) site over the beta1 (caspase-like) and beta2 (trypsin-like) sites of the 20S core particle of the proteasome, and over a panel of less closely related proteases. Compound optimization, guided by X-ray crystallography of the liganded 20S core particle, confirmed their non-covalent binding mode and provided a structural basis for their enhanced in vitro and cellular potencies. We demonstrate that such compounds show low nanomolar IC50 values for the human 20S beta5 site in vitro, and that pharmacological inhibition of this site in cells is sufficient to potently inhibit the degradation of a tetra-ubiquitin-luciferase reporter, activation of NFkappaB (nuclear factor kappaB) in response to TNF-alpha (tumour necrosis factor-alpha) and the proliferation of cancer cells. Finally, we identified capped di-peptides that show differential selectivity for the beta5 site of the constitutively expressed proteasome and immunoproteasome in vitro and in B-cell lymphomas. Collectively, these studies describe the synthesis, activity and binding mode of a new series of non-covalent proteasome inhibitors with unprecedented potency and selectivity for the beta5 site, and which can discriminate between the constitutive proteasome and immunoproteasome in vitro and in cells.


Subject(s)
Oligopeptides/pharmacology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Amino Acid Sequence , Binding Sites , Boronic Acids/pharmacology , Bortezomib , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , HCT116 Cells , HT29 Cells , Humans , Kinetics , Luciferases/genetics , Luciferases/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , NF-kappa B/genetics , NF-kappa B/metabolism , Oligopeptides/chemistry , Oligopeptides/metabolism , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Pyrazines/pharmacology , RNA Interference , Sequence Homology, Amino Acid , Ubiquitin/genetics , Ubiquitin/metabolism
8.
Cancer Res ; 70(11): 4318-26, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20460535

ABSTRACT

Multiple pathways have been proposed to explain how proteasome inhibition induces cell death, but mechanisms remain unclear. To approach this issue, we performed a genome-wide siRNA screen to evaluate the genetic determinants that confer sensitivity to bortezomib (Velcade (R); PS-341). This screen identified 100 genes whose knockdown affected lethality to bortezomib and to a structurally diverse set of other proteasome inhibitors. A comparison of three cell lines revealed that 39 of 100 genes were commonly linked to cell death. We causally linked bortezomib-induced cell death to the accumulation of ASF1B, Myc, ODC1, Noxa, BNIP3, Gadd45alpha, p-SMC1A, SREBF1, and p53. Our results suggest that proteasome inhibition promotes cell death primarily by dysregulating Myc and polyamines, interfering with protein translation, and disrupting essential DNA damage repair pathways, leading to programmed cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Cell Death/drug effects , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , RNA, Small Interfering/genetics , Bortezomib , Cell Death/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Damage , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Ribosomes/drug effects , TOR Serine-Threonine Kinases , Transfection
9.
Mol Cancer Ther ; 6(1): 262-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17237285

ABSTRACT

Poh1 deubiquitinase activity is required for proteolytic processing of polyubiquitinated substrates by the 26S proteasome, linking deubiquitination to complete substrate degradation. Poh1 RNA interference (RNAi) in HeLa cells resulted in a reduction in cell viability and an increase in polyubiquitinated protein levels, supporting the link between Poh1 and the ubiquitin proteasome pathway. To more specifically test for any requirement of the zinc metalloproteinase motif of Poh1 to support cell viability and proteasome function, we developed a RNAi complementation strategy. Effects on cell viability and proteasome activity were assessed in cells with RNAi of endogenous Poh1 and induced expression of wild-type Poh1 or a mutant form of Poh1, in which two conserved histidines of the proposed catalytic site were replaced with alanines. We show that an intact zinc metalloproteinase motif is essential for cell viability and 26S proteasome function. As a required enzymatic component of the proteasome, Poh1 is an intriguing therapeutic drug target for cancer.


Subject(s)
Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Ubiquitin/metabolism , Amino Acid Motifs , Cell Survival , HeLa Cells , Humans , Mutant Proteins/metabolism , Proteasome Endopeptidase Complex/deficiency , RNA Interference , Trans-Activators/deficiency
10.
Mol Cancer Ther ; 5(12): 3052-61, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172407

ABSTRACT

Strains within the genus Salinospora have been shown to produce complex natural products having antibiotic and antiproliferative activities. The biochemical basis for the cytotoxic effects of salinosporamide A has been linked to its ability to inhibit the proteasome. Synthetically accessible salinosporamide A (ML858) was used to determine its biochemical and biological activities and to compare its effects with those of bortezomib. ML858 and bortezomib show time- and concentration-dependent inhibition of the proteasome in vitro. However, unlike bortezomib, which is a reversible inhibitor, ML858 covalently binds to the proteasome, resulting in the irreversible inhibition of 20S proteasome activity. ML858 was equipotent to bortezomib in cell-based reporter stabilization assays, but due to intramolecular instability is less potent in long-term assays. ML858 failed to maintain levels of proteasome inhibition necessary to achieve efficacy in tumor models responsive to bortezomib. Our results show that ML858 and bortezomib exhibit different kinetic and pharmacologic profiles and suggest that additional characterization of ML858 is warranted before its therapeutic potential can be fully appreciated.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Lactones/pharmacology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Pyrroles/pharmacology , Animals , Antineoplastic Agents/chemistry , Binding, Competitive , Boronic Acids/chemistry , Bortezomib , Drug Stability , Female , HT29 Cells , HeLa Cells , Humans , Lactones/chemistry , Mice , Mice, Nude , Mice, SCID , Protease Inhibitors/chemistry , Proteasome Endopeptidase Complex/metabolism , Pyrazines/chemistry , Pyrroles/chemistry , Xenograft Model Antitumor Assays
11.
Mol Biol Cell ; 14(5): 2071-87, 2003 May.
Article in English | MEDLINE | ID: mdl-12802076

ABSTRACT

Activation of the mitotic checkpoint by chemotherapeutic drugs such as taxol causes mammalian cells to arrest in mitosis and then undergo apoptosis. However, the biochemical basis of chemotherapeutic drug-induced cell death is unclear. Herein, we provide new evidence that both cell survival and cell death-signaling pathways are concomitantly activated during mitotic arrest by microtubule-interfering drugs. Treatment of HeLa cells with chemotherapeutic drugs activated both p38 mitogen-activated protein kinase (MAPK) and p21-activated kinase (PAK). p38 MAPK was necessary for chemotherapeutic drug-induced cell death because the p38 MAPK inhibitors SB203580 or SB202190 suppressed cell death. Dominant-active MKK6, a direct activator of p38 MAPK, also induced cell death by stimulating translocation of Bax from the cytosol to the mitochondria in a p38 MAPK-dependent manner. Dominant active PAK suppressed this MKK6-induced cell death. PAK seems to mediate cell survival by phosphorylating Bad, and inhibition of PAK in mitotically arrested cells reduced Bad phosphorylation and increased apoptosis. Our results suggest that therapeutic strategies that suppress PAK-mediated survival signals may improve the efficacy of current cancer chemotherapies by enhancing p38 MAPK-mediated cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Mitogen-Activated Protein Kinases/drug effects , Mitosis/drug effects , Nocodazole/pharmacology , Protein Serine-Threonine Kinases/drug effects , Proto-Oncogene Proteins c-bcl-2 , Cytoplasm/metabolism , HeLa Cells , Humans , Mitochondria/metabolism , Proto-Oncogene Proteins/metabolism , bcl-2-Associated X Protein , p21-Activated Kinases , p38 Mitogen-Activated Protein Kinases
12.
Br J Pharmacol ; 138(7): 1340-50, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12711635

ABSTRACT

1. We have examined possible mechanisms of cross-talk between the G(q/11)-linked M(3) muscarinic acetylcholine (mACh) receptor and the G(i/o)-linked M(2) mACh receptor by stable receptor coexpression in Chinese hamster ovary (CHO) cells. A number of second messenger (cyclic AMP, Ins(1,4,5)P(3)) and mitogen-activated protein kinase (ERK and JNK) responses stimulated by the mACh receptor agonist methacholine were examined in CHO-m2m3 cells and compared to those stimulated in CHO-m2 and CHO-m3 cell-lines, expressing comparable levels of M(2) or M(3) mACh receptors. 2. Based on comparisons between cell-lines and pertussis toxin (PTx) pretreatment to eliminate receptor-G(i/o) coupling, evidence was obtained for (i) an M(2) mACh receptor-mediated contribution to the predominantly M(3) mACh receptor-mediated Ins(1,4,5)P(3) response and (ii) a facilitation of the inhibitory effect of M(2) mACh receptor on forskolin-stimulated cyclic AMP accumulation by M(3) mACh receptor coactivation at low agonist concentrations (MCh 10(-9)-10(-6) M). 3. The most profound cross-talk effects were observed with respect to ERK activation. Thus, while MCh stimulated ERK activation in both CHO-m2 and CHO-m3 cells (pEC(50) values: 5.64+/-0.09 and 5.57+/-0.16, respectively), the concentration-effect relation was approx 50-fold left-shifted in CHO-m2m3 cells (pEC(50): 7.17+/-0.07). In addition, the ERK response was greater and more sustained in CHO-m2m3 cells. In contrast, only minor differences were seen in the time-courses and concentration-dependencies of JNK activation in CHO-m3 and CHO-m2m3 cells. 4. Costimulation of endogenous P2Y(2) purinoceptors also caused an approx 10-fold left-shift in the MCh-stimulated ERK response in CHO-m2 cells, suggesting that the G(q/11)/G(i/o) interaction to affect ERK activation is not specific to muscarinic receptors. 5. PTx pretreatment of cells had unexpected effects on ERK activation by MCh in both CHO-m2m3 and CHO-m3 cells. Thus, in CHO-m3 cells PTx pretreatment caused a marked left-shift in the MCh concentration-effect curve, while in PTx-treated CHO-m2m3 cells the maximal responsiveness was decreased, but the potency of MCh was only slightly affected. 6. The data presented here strongly suggest that cross-talk between M(2) and M(3) mACh receptors occurs at the level of both second messenger and ERK regulation. Further, these data provide novel insights into the involvement of G(i/o) proteins in both positive and negative modulation of ERK responses evoked by G protein-coupled receptors.


Subject(s)
CHO Cells/metabolism , Receptor Cross-Talk/physiology , Receptor, Muscarinic M2/physiology , Receptor, Muscarinic M3/physiology , Receptors, Cell Surface/physiology , Second Messenger Systems/physiology , Animals , Cricetinae , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/physiology , Mutation , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M3/genetics
13.
Biochem J ; 373(Pt 1): 71-9, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12650640

ABSTRACT

The proteasome is a multisubunit proteolytic enzyme comprising activator complexes bound to the 20 S catalytic core. The functions of the proteasomal activator (PA) 700 in ubiquitin/ATP-dependent protein degradation and of the PA28 alpha/beta activators in antigen presentation are well defined. However, the function of a third PA, PA28 gamma, remains elusive. We now show that mitogen-activated protein kinase (MAPK)/extracellular-signal-regulated kinase (ERK) kinase kinase 3 (MEKK3), a MAPK kinase kinase (MAPKKK) involved in MAPK kinase 7 (MKK7)-c-Jun N-terminal kinase ('JNK') and MKK6-p38 signalling, can bind PA28 gamma but not PA28 alpha. In contrast, B-Raf, a MAPKKK specific for the MAPK/ERK kinase ('MEK')-ERK module, binds PA28 gamma and alpha. The PA28 gamma-binding domain of MEKK3 is located within its N-terminal regulatory domain (amino acids 1-178). Expression of MEKK3 in Cos-7 cells led to an increase in endogenous and co-expressed PA28 gamma protein levels, whereas kinase-deficient MEKK3 had no effect on PA28 gamma expression. Furthermore, in vitro assays indicated that PA28 gamma was a MEKK3 substrate. MEKK3 represents the first protein kinase capable of binding and phosphorylating a PA, and provides a potential mechanism to link stress-activated protein kinase signalling with the PA28 gamma-dependent proteasome.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Nuclear Proteins/metabolism , Animals , Autoantigens , Binding Sites , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , MAP Kinase Kinase Kinase 3 , MAP Kinase Kinase Kinases/chemistry , MAP Kinase Kinase Kinases/genetics , Mitosis , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Mutagenesis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , PC12 Cells , Phosphorylation , Polymerase Chain Reaction , Proteasome Endopeptidase Complex , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
14.
J Neurochem ; 83(5): 1139-53, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12437585

ABSTRACT

The coupling of the group I metabotropic glutamate receptors, mGlu1a and mGlu5a, to the extracellular signal-regulated protein kinase (ERK) pathway has been studied in Chinese hamster ovary cell-lines where receptor expression is under inducible control. Both mGlu receptors stimulated comparable, robust and agonist concentration-dependent ERK activations in the CHO cell-lines. The mGlu1a receptor-mediated ERK response was almost completely attenuated by pertussis toxin (PTx) pretreatment, whereas the mGlu5a-ERK response, and the phosphoinositide response to activation of either receptor, was PTx-insensitive. mGlu1a and mGlu5a receptor coupling to ERK occurred via mechanisms independent of phosphoinositide 3-kinase activity and intracellular and/or extracellular Ca2+ concentration. While acute treatment with a protein kinase C (PKC) inhibitor did not attenuate agonist-stimulated ERK activation, down-regulation of PKCs by phorbol ester treatment for 24 h did attenuate both mGlu1a and mGlu5a receptor-mediated responses. Further, inhibition of Src non-receptor tyrosine kinase activity by PP1 attenuated the ERK response generated by both receptor subtypes, but only mGlu1a receptor-ERK activation was attenuated by PDGF receptor tyrosine kinase inhibitor AG1296. These findings demonstrate that, although expressed in a common cell background, these closely related mGlu receptors utilize different G proteins to cause ERK activation and may recruit different tyrosine kinases to facilitate this response.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Female , Humans , Hydrolysis/drug effects , Mitogen-Activated Protein Kinases/drug effects , Ovary/cytology , Ovary/drug effects , Ovary/metabolism , Pertussis Toxin/pharmacology , Phosphatidylinositols/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Quisqualic Acid/pharmacology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors
15.
Nat Cell Biol ; 4(11): 901-6, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12402047

ABSTRACT

cAMP is involved in a wide variety of cellular processes that were thought to be mediated by protein kinase A (PKA). However, cAMP also directly regulates Epac1 and Epac2, guanine nucleotide-exchange factors (GEFs) for the small GTPases Rap1 and Rap2 (refs 2,3). Unfortunately, there is an absence of tools to discriminate between PKA- and Epac-mediated effects. Therefore, through rational drug design we have developed a novel cAMP analogue, 8-(4-chloro-phenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate (8CPT-2Me-cAMP), which activates Epac, but not PKA, both in vitro and in vivo. Using this analogue, we tested the widespread model that Rap1 mediates cAMP-induced regulation of the extracellular signal-regulated kinase (ERK). However, both in cell lines in which cAMP inhibits growth-factor-induced ERK activation and in which cAMP activates ERK, 8CPT-2Me-cAMP did not affect ERK activity. Moreover, in cell lines in which cAMP activates ERK, inhibition of PKA and Ras, but not Rap1, abolished cAMP-mediated ERK activation. We conclude that cAMP-induced regulation of ERK and activation of Rap1 are independent processes.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , Cyclic AMP/metabolism , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/metabolism , Mitogen-Activated Protein Kinases/metabolism , rap1 GTP-Binding Proteins/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , In Vitro Techniques , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction , Time Factors , Transfection
16.
Biochem J ; 367(Pt 2): 549-59, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12126481

ABSTRACT

We have assessed the growth response of Chinese-hamster ovary (CHO) cells to activation of recombinantly expressed G-protein-coupled muscarinic M(2) or M(3) acetylcholine receptors (AChRs). We show that activation of these receptors leads to divergent growth responses: M(2) AChR activation causes an increase in DNA synthesis, whereas M(3) AChR activation causes a dramatic decrease in DNA synthesis. We have characterized the M(3) AChR-mediated growth inhibition and show that it involves a G(1) phase cell-cycle arrest. Further analysis of this arrest indicates that it involves an increase in expression of the cyclin-dependent kinase (CDK) inhibitor, p21(Cip1/Waf1) (where Cip1 is CDK-interacting protein 1 and Waf1 is wild-type p53-associated fragment 1), in response to M(3) AChR activation. This increase in protein expression leads to an increase in p21(Cip1/Waf1) association with CDK2, a decrease in CDK2 activity and an accumulation of hypophosphorylated retinoblastoma protein. The increased p21(Cip1/Waf1) expression is due, at least in part, to an increase in p21(Cip1/Waf1) mRNA, and receptor-mediated changes in phosphorylation of c-Jun provide a mechanism to account for this p21(Cip1/Waf1) transcriptional regulation. Evaluation of the extracellular signal-regulated protein kinase and c-Jun N-terminal kinase activities has shown striking differences in the profiles of activation of these mitogen-activated protein kinases by the M(2) and M(3) AChRs, and their potential involvement in mediating growth arrest by the M(3) AChR is discussed.


Subject(s)
CDC2-CDC28 Kinases , Cyclins/metabolism , G1 Phase/physiology , Receptors, Muscarinic/metabolism , Animals , CHO Cells , Carbachol/pharmacology , Cell Division/drug effects , Cell Division/physiology , Cholinergic Agonists/pharmacology , Cricetinae , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , DNA/biosynthesis , G1 Phase/drug effects , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptor, Muscarinic M2 , Receptor, Muscarinic M3 , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinoblastoma Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...