Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Assay Drug Dev Technol ; 11(5): 308-25, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23772552

ABSTRACT

Small ubiquitin-like modifier (SUMO) belongs to the family of ubiquitin-like proteins (Ubls) that can be reversibly conjugated to target-specific lysines on substrate proteins. Although covalently sumoylated products are readily detectible in gel-based assays, there has been little progress toward the development of robust quantitative sumoylation assay formats for the evaluation of large compound libraries. In an effort to identify inhibitors of ubiquitin carrier protein 9 (Ubc9)-dependent sumoylation, a high-throughput fluorescence polarization assay was developed, which allows detection of Lys-1201 sumoylation, corresponding to the major site of functional sumoylation within the transcriptional repressor trichorhino-phalangeal syndrome type I protein (TRPS1). A minimal hexapeptide substrate peptide, TMR-VVK1201TEK, was used in this assay format to afford high-throughput screening of the GlaxoSmithKline diversity compound collection. A total of 728 hits were confirmed but no specific noncovalent inhibitors of Ubc9 dependent trans-sumoylation were found. However, several diaminopyrimidine compounds were identified as inhibitors in the assay with IC50 values of 12.5 µM. These were further characterized to be competent substrates which were subject to sumoylation by SUMO-Ubc9 and which were competitive with the sumoylation of the TRPS1 peptide substrates.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Protein Interaction Mapping/methods , Spectrometry, Fluorescence/methods , Sumoylation/drug effects , Transcription Factors/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Binding Sites , Protein Binding , Repressor Proteins
2.
PLoS One ; 7(8): e43019, 2012.
Article in English | MEDLINE | ID: mdl-22952628

ABSTRACT

Proline-specific dipeptidyl peptidases (DPPs) are emerging targets for drug development. DPP4 inhibitors are approved in many countries, and other dipeptidyl peptidases are often referred to as DPP4 activity- and/or structure-homologues (DASH). Members of the DASH family have overlapping substrate specificities, and, even though they share low sequence identity, therapeutic or clinical cross-reactivity is a concern. Here, we report the structure of human DPP7 and its complex with a selective inhibitor Dab-Pip (L-2,4-diaminobutyryl-piperidinamide) and compare it with that of DPP4. Both enzymes share a common catalytic domain (α/ß-hydrolase). The catalytic pocket is located in the interior of DPP7, deep inside the cleft between the two domains. Substrates might access the active site via a narrow tunnel. The DPP7 catalytic triad is completely conserved and comprises Ser162, Asp418 and His443 (corresponding to Ser630, Asp708 and His740 in DPP4), while other residues lining the catalytic pockets differ considerably. The "specificity domains" are structurally also completely different exhibiting a ß-propeller fold in DPP4 compared to a rare, completely helical fold in DPP7. Comparing the structures of DPP7 and DPP4 allows the design of specific inhibitors and thus the development of less cross-reactive drugs. Furthermore, the reported DPP7 structures shed some light onto the evolutionary relationship of prolyl-specific peptidases through the analysis of the architectural organization of their domains.


Subject(s)
Dipeptidyl Peptidase 4/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/chemistry , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Proline/chemistry , Amino Acids/chemistry , Animals , Base Sequence , CHO Cells , Catalysis , Catalytic Domain , Cricetinae , Dimerization , Dipeptidyl Peptidase 4/chemistry , Evolution, Molecular , Humans , Insecta , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Substrate Specificity
3.
Chem Biol ; 18(2): 177-86, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21338916

ABSTRACT

c-Abl kinase activity is regulated by a unique mechanism involving the formation of an autoinhibited conformation in which the N-terminal myristoyl group binds intramolecularly to the myristoyl binding site on the kinase domain and induces the bending of the αI helix that creates a docking surface for the SH2 domain. Here, we report a small-molecule c-Abl activator, DPH, that displays potent enzymatic and cellular activity in stimulating c-Abl activation. Structural analyses indicate that DPH binds to the myristoyl binding site and prevents the formation of the bent conformation of the αI helix through steric hindrance, a mode of action distinct from the previously identified allosteric c-Abl inhibitor, GNF-2, that also binds to the myristoyl binding site. DPH represents the first cell-permeable, small-molecule tool compound for c-Abl activation.


Subject(s)
Drug Discovery , Hydantoins/metabolism , Hydantoins/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Pyrazoles/metabolism , Pyrazoles/pharmacology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Enzyme Activation/drug effects , Hep G2 Cells , Humans , Hydantoins/chemistry , Models, Molecular , Molecular Sequence Data , Permeability , Phosphorylation/drug effects , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-crk/metabolism , Pyrazoles/chemistry
4.
J Biomol Screen ; 16(1): 53-64, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20938045

ABSTRACT

A 2-step kinase assay was developed and used in a high-throughput screen (HTS) of more than 1 million compounds in an effort to identify c-Abl tyrosine kinase activators. This assay employed a 2-step phosphorylation reaction: in the first step, purified recombinant c-Abl was activated by incubating with compound in the presence of adenosine triphosphate (ATP). In the second step, the TAMRA-labeled IMAP Abltide substrate was added to allow phosphorylation of the substrate to occur. The assay was calibrated such that inactive c-Abl protein was activated by ATP alone to a degree that it not only demonstrated a measurable c-Abl activity but also maintained a robust assay window for screening. The screen resulted in 8624 primary hits with >30% response. Further analysis showed that 1024 had EC(50) <10 µM with a max % response of >50%. These hits were structurally and chemically diverse with possibly different mechanisms for activating c-Abl. In addition, selective hits were shown to be cell permeable and were able to induce c-Abl activation as determined by In-Cell Western (ICW) analysis of HEK-MSRII cells transduced with BacMam virus expressing full-length c-Abl.


Subject(s)
Enzyme Activators/pharmacology , High-Throughput Screening Assays/methods , Proto-Oncogene Proteins c-abl/agonists , Proto-Oncogene Proteins c-abl/metabolism , Adenosine Triphosphate/metabolism , Baculoviridae/genetics , Biological Assay , Drug Discovery , Genetic Vectors/genetics , HEK293 Cells , Humans , Phosphorylation , Small Molecule Libraries/pharmacology , Transfection
5.
Protein Expr Purif ; 73(2): 167-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20457255

ABSTRACT

Phosphoinositide 3-kinases have been targeted for therapeutic research because they are key components of a cell signaling cascade controlling proliferation, growth, and survival. Direct activation of the PI3Kalpha pathway contributes to the development and progression of solid tumors in breast, endometrial, colon, ovarian, and gastric cancers. In the context of a drug discovery effort, the availability of a robust crystallographic system is a means to understand the subtle differences between ATP competitive inhibitor interactions with the active site and their selectivity against other PI3Kinase enzymes. To generate a suitable recombinant design for this purpose, a p85alpha-p110alpha fusion system was developed which enabled the expression and purification of a stoichiometrically homogeneous, constitutively active enzyme for structure determination with potent ATP competitive inhibitors (Raha et al., in preparation) [56]. This approach has yielded preparations with activity and inhibition characteristics comparable to those of the full-length PI3Kalpha from which X-ray diffracting crystals were grown with inhibitors bound in the active site.


Subject(s)
Class II Phosphatidylinositol 3-Kinases/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Adenosine Triphosphate/metabolism , Animals , Artificial Gene Fusion , Baculoviridae/metabolism , Binding Sites , Cells, Cultured , Class II Phosphatidylinositol 3-Kinases/chemistry , Class II Phosphatidylinositol 3-Kinases/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Drug Design , Inhibitory Concentration 50 , Models, Molecular , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Spodoptera/cytology , Spodoptera/metabolism , X-Ray Diffraction
6.
Protein Expr Purif ; 65(2): 251-60, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19297698

ABSTRACT

We describe here two strategies to produce biologically active chemokines with authentic N-terminal amino acid residues. The first involves producing the target chemokine with an N-terminal 6xHis-SUMO tag in Escherichia coli as inclusion bodies. The fusion protein is solubilized and purified with Ni-NTA-agarose in denaturing reagents. This is further followed by tag removal and refolding in a redox refolding buffer. The second approach involves expressing the target chemokine with an N-terminal 6xHis-Trx-SUMO tag in an engineered E. coli strain that facilitates formation of disulfide bonds in the cytoplasm. Following purification of the fusion protein via Ni-NTA and tag removal, the target chemokine is refolded without redox buffer and purified by reverse phase chromatography. Using the procedures, we have produced more than 15 biologically active chemokines, with a yield of up to 15 mg/L.


Subject(s)
Chemokines/biosynthesis , Chemokines/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Chemokines/isolation & purification , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Oxidation-Reduction , Polymerase Chain Reaction , Protein Engineering , Protein Folding , Recombinant Fusion Proteins/isolation & purification
7.
Protein Expr Purif ; 60(2): 110-6, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18474430

ABSTRACT

Human Relaxin 2 is an insulin-related peptide hormone with a mass of 19,084 Da. The mRNA contains a number of arginine codons that are rarely used by Escherichia coli to produce highly expressed proteins. As a result, expressing this recombinant protein in E. coli is problematic. When human Relaxin 2 was expressed in E. coli BL21 (DE3), several forms of the protein were made. One species had the expected molecular weight (19,084 Da). A second species observed had a molecular weight of 21,244 Da. A third minor species had a molecular weight of 17,118 Da. These aberrant molecular weights can be explained as follows. First, a sequence CGA-AAA-AAG-AGA, containing the rare arginine codons CGA and AGA was the site of the +1 frameshift that generated the 21,244 Da species. Since there was a limited supply of this arginyl-tRNA, the peptidyl-tRNA moved +1 nucleotide to occupy the codon and resumed protein synthesis. Second, a -1 frameshift associated with 'slippery A' sequence XXA-AAA-AAG accounted for 10% of the product with a mass of 17,118 Da. Presumably, the shift to -1 also occurred because there was a paucity of the arginyl-tRNAArgucu. Introduction of a plasmid coding for the cognate tRNA for AGA and site directed mutagenesis prevented the formation of both frameshift species.


Subject(s)
Arginine/genetics , Codon , Escherichia coli/genetics , Frameshift Mutation , RNA, Transfer, Lys/genetics , Relaxin/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Fermentation , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Open Reading Frames , Relaxin/isolation & purification , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
8.
J Biomol Screen ; 12(8): 1050-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17989425

ABSTRACT

Tumor suppressor p53 is typically maintained at low levels in normal cells. In response to cellular stresses, such as DNA damage, p53 is stabilized and can stimulate responses leading to cell cycle arrest or apoptosis. Corresponding to its central role in preventing propagation of damaged cells, mutation or deletion of p53 is found in nearly 50% of all human tumors. Mdm2 (mouse-d-minute 2) and its human ortholog (hmdm2 or hdm2) catalyze the ubiquitination of p53, targeting it for degradation via the proteosome. Thus, the activity of mdm2 is inversely correlated with p53 levels. Based on this, inhibition of human mdm2 activity by a small-molecule therapeutic will lead to net stabilization of p53 and be the basis for development of a novel cancer therapeutic. Previous high-throughput screening assays of mdm2 measured the autoubiquitination activity of mdm2, which occurs in the absence of an acceptor substrate such as p53. The major drawback to this approach is that inhibitors of mdm2 autoubiquitination may lead to a net stabilization of mdm2 and thus have the opposite effect of inhibitors that interfere with p53 ubiquitination. The authors describe the development, validation, and execution of a high-throughput screening measuring the ubiquitination of p53 by mdm2, with p53 labeled with europium and the other substrate (Ub-UbcH5b) labeled with a Cy5 on the ubiquitin. After confirming that known inhibitors are detected with this assay, it was successfully automated and used to query >600,000 compounds from the GlaxoSmithKline collection for mdm2 inhibitors.


Subject(s)
Biological Assay/methods , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitination , Catalysis/drug effects , Europium/pharmacology , Fluorescence Resonance Energy Transfer , Humans , Inhibitory Concentration 50 , Proto-Oncogene Proteins c-mdm2/pharmacology , Reproducibility of Results , Time Factors , Titrimetry , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination/drug effects
9.
Protein Expr Purif ; 50(1): 102-10, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16893658

ABSTRACT

Interleukin 1beta (IL-1beta) is a potent stimulator of extracellular matrix degradation in models of osteoarthritis (OA). In contrast to bovine explant models which effectively respond to recombinant human IL-1beta, canine models are relatively refractory to human IL-1beta stimulation. Canine IL-1beta cDNA was cloned in order to produce a fully potent species matched preparation of IL-1beta for use specifically in canine models of OA. Established methods for the production of various orthologous IL-1beta proteins from different species are problematic due to the exquisite sensitivity of the mature IL-1beta product to N-terminal variations and the intrinsic technical challenges associated with producing an unmodified product. We have applied a seamless method of SUMO tagging and removal in order to produce a homogeneous unmodified preparation of canine IL-1beta from Escherichia coli which was found to be a potent inducer of aggrecanase activity in isolated canine articular chondrocytes. This method combines highly efficient aspects of seamless plasmid engineering, protein purification, and precise tag removal.


Subject(s)
Escherichia coli/metabolism , Interleukin-1beta/biosynthesis , Small Ubiquitin-Related Modifier Proteins/genetics , Amino Acid Sequence , Animals , Cattle , Cloning, Molecular , DNA, Complementary/genetics , Dogs , Gene Expression Regulation , Humans , Interleukin-1beta/genetics , Interleukin-1beta/isolation & purification , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
10.
Biochemistry ; 45(30): 9238-45, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16866370

ABSTRACT

Mdm2 negatively regulates p53 by inhibiting its transcriptional activity and promoting its degradation by functioning as an E3 ubiquitin ligase. The primary p53 binding site on mdm2 is located in its N-terminal domain. Through binding to p53 at its N-terminal transactivation domain, mdm2 directly blocks the transcriptional activation function of p53. We discovered that truncated mdm2 protein constructs without the N-terminal p53 binding domain are at least as active as full-length mdm2 in catalyzing p53 ubiquitination. Furthermore, the deletion of the central acidic domain significantly reduces the E3 ligase activity of mdm2 toward p53. We have also performed GST pull-down experiments to probe the direct binding of various mdm2 domain constructs toward full length p53 and found that mdm2 constructs without the N-terminal p53 binding domain retain the ability to bind to p53. Our kinetic and binding data localize the second p53 binding site between amino acids 211 and 361, including the acidic domain and the zinc finger region. Our work, consistent with other reports, suggests that the p53 tetramer interacts with at least two sites on mdm2. Although the interaction between the N-termini of mdm2 and p53 blocks the transactivation activity of p53, the interaction between the central domain of mdm2 and the core domain of p53 is critical for the ubiquitination and degradation of p53. This second mdm2-p53 interaction site represents an alternative target for small molecule modulators of the mdm2-p53 pathway.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism , Animals , Binding Sites/genetics , Cell Line , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Humans , Imidazoles/chemistry , Kinetics , Piperazines/chemistry , Protein Interaction Mapping , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins c-mdm2/genetics , Sequence Deletion , Spodoptera , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
11.
Proc Natl Acad Sci U S A ; 103(20): 7625-30, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16684877

ABSTRACT

Heat shock protein (Hsp)90 is emerging as an important therapeutic target for the treatment of cancer. Two analogues of the Hsp90 inhibitor geldanamycin are currently in clinical trials. Geldanamycin (GA) and its analogues have been reported to bind purified Hsp90 with low micromolar potency, in stark contrast to their low nanomolar antiproliferative activity in cell culture and their potent antitumor activity in animal models. Several models have been proposed to account for the approximately 100-fold-greater potency in cell culture, including that GA analogues bind with greater affinity to a five-protein Hsp90 complex than to Hsp90 alone. We have determined that GA and the fluorescent analogue BODIPY-GA (BDGA) both demonstrate slow, tight binding to purified Hsp90. BDGA, used to characterize the kinetics of ligand-Hsp90 interactions, was found to bind Hsp90alpha with k(off) = 2.5 x 10(-3) min(-1), t(1/2) = 4.6 h, and Ki* = 10 nM. It was found that BDGA binds to a functional multiprotein Hsp90 complex with kinetics and affinity identical to that of Hsp90 alone. Also, BDGA binds to Hsp90 from multiple cell lysates in a time-dependent manner with similar kinetics. Therefore, our results indicate that the high potency of GA in cell culture and in vivo can be accounted for by its time-dependent, tight binding to Hsp90 alone. In the broader context, these studies highlight the essentiality of detailed biochemical characterization of drug-target interactions for the effective translation of in vitro pharmacology to cellular and in vivo efficacy.


Subject(s)
Antibiotics, Antineoplastic , HSP90 Heat-Shock Proteins , Quinones , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Benzoquinones , Boron Compounds/chemistry , Boron Compounds/metabolism , Cells, Cultured , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic , Protein Binding , Quinones/chemistry , Quinones/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
12.
J Med Chem ; 48(18): 5644-7, 2005 Sep 08.
Article in English | MEDLINE | ID: mdl-16134930

ABSTRACT

Inhibitors of human methionine aminopeptidase type 2 (hMetAP2) are of interest as potential treatments for cancer. A new class of small molecule reversible inhibitors of hMetAP2 was discovered and optimized, the 4-aryl-1,2,3-triazoles. Compound 24, a potent inhibitor of cobalt-activated hMetAP2, also inhibits human and mouse endothelial cell growth. Using a mouse matrigel model, this reversible hMetAP2 inhibitor was also shown to inhibit angiogenesis in vivo.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Angiogenesis Inhibitors/chemical synthesis , Metalloendopeptidases/antagonists & inhibitors , Triazoles/chemical synthesis , Aminopeptidases/chemistry , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Binding Sites , Biological Availability , Cell Proliferation/drug effects , Cells, Cultured , Cobalt/metabolism , Collagen , Crystallography, X-Ray , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Enzyme Activation , Humans , Laminin , Metalloendopeptidases/chemistry , Mice , Models, Molecular , Molecular Structure , Proteoglycans , Rats , Structure-Activity Relationship , Triazoles/chemistry , Triazoles/pharmacology
14.
J Pharmacol Exp Ther ; 307(2): 720-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12975486

ABSTRACT

Histone deacetylases (HDACs) represent an expanding family of protein modifying-enzymes that play important roles in cell proliferation, chromosome remodeling, and gene transcription. We have previously shown that recombinant human HDAC8 can be expressed in bacteria and retain its catalytic activity. To further explore the catalytic activity of HDACs, we expressed two additional human class I HDACs, HDAC1 and HDAC3, in baculovirus. Recombinant HDAC1 and HDAC3 fusion proteins remained soluble and catalytically active and were purified to near homogeneity. Interestingly, trichostatin (TSA) was found to be a potent inhibitor for all three HDACs (IC50 value of approximately 0.1-0.3 microM), whereas another HDAC inhibitor MS-27-275 (N-(2-aminophenyl)-4-[N-(pyridin-3-methyloxycarbonyl)-aminomethyl]benzamide) preferentially inhibited HDAC1 (IC50 value of approximately 0.3 microM) versus HDAC3 (IC50 value of approximately 8 microM) and had no inhibitory activity toward HDAC8 (IC50 value >100 microM). MS-27-275 as well as TSA increased histone H4 acetylation, induced apoptosis in the human colon cancer cell line SW620, and activated the simian virus 40 early promoter. HDAC1 protein was more abundantly expressed in SW620 cells compared with that of HDAC3 and HDAC8. Using purified recombinant HDAC proteins, we identified several novel HDAC inhibitors that preferentially inhibit HDAC1 or HDAC8. These inhibitors displayed distinct properties in inducing histone acetylation and reporter gene expression. These results suggest selective HDAC inhibitors could be identified using recombinantly expressed HDACs and that HDAC1 may be a promising therapeutic target for designing HDAC inhibitors for proliferative diseases such as cancer.


Subject(s)
Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Pyridines/pharmacology , Cloning, Molecular , Colonic Neoplasms/pathology , Drug Interactions , Gene Expression , Histone Deacetylase 1 , Histone Deacetylases , Humans , Protein Isoforms/antagonists & inhibitors , Recombinant Proteins/antagonists & inhibitors , Tumor Cells, Cultured
15.
Bioorg Med Chem Lett ; 13(8): 1483-6, 2003 Apr 17.
Article in English | MEDLINE | ID: mdl-12668017

ABSTRACT

In our continuing efforts to identify small molecule vitronectin receptor antagonists, we have discovered a series of phenylbutyrate derivatives, exemplified by 16, which have good potency and excellent oral bioavailability (approximately 100% in rats). This new series is derived conceptually from opening of the seven-membered ring of SB-265123.


Subject(s)
Integrin alphaVbeta3/antagonists & inhibitors , Phenylbutyrates/pharmacology , Phenylbutyrates/pharmacokinetics , Acetates/chemistry , Administration, Oral , Aminopyridines/chemistry , Animals , Biological Availability , Cell Adhesion/drug effects , Cell Line , Half-Life , Humans , Phenylbutyrates/chemistry , Rats
16.
Protein Expr Purif ; 27(2): 279-92, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12597888

ABSTRACT

Interleukin-18 (IL-18) is activated and released from immune effector cells to stimulate acquired and innate immune responses involving T and natural killer (NK) cells. The release of IL-18 from mammalian cells is linked to its proteolytic activation by caspases including interleukin 1 converting enzyme (ICE). The absence of a signal peptide sequence and the requirement for coupled activation and cellular release have presented challenges for the large-scale recombinant production of IL-18. In this study, we have explored methods for the direct production of authentic human IL-18 toward the development of a large-scale production system. Expression of mature IL-18 directly in Escherichia coli with a methionine initiating codon leads to the production of MetIL-18 that is dramatically less potent in bioassays than IL-18 produced as a pro-peptide and activated in vitro. To produce an authentic IL-18, we have devised a bicistronic expression system for the coupled transcription and translation of ProIL-18 with caspase-1 (ICE) or caspase-4 (ICE-rel II, TX, ICH-2). Mature IL-18 with an authentic N-terminus was produced and has a biological activity and potency comparable to that of in vitro processed mature IL-18. Optimization of this system for the maximal production yields can be accomplished by modulating the temperature, to affect the rate of caspase activation and to favor the accumulation of ProIL-18, prior to its proteolytic processing by activated caspase. The effect of temperature is particularly profound for the caspase-4 co-expression process, enabling optimized production levels of over 150 mg/L in shake flasks at 25 degrees C. An alternative bicistronic expression design utilizing a precise ubiquitin IL-18 fusion, processed by co-expressed ubiquitinase, was also successfully used to generate fully active IL-18, thereby demonstrating that the pro-sequence of IL-18 is not required for recombinant IL-18 production.


Subject(s)
Interleukin-18/biosynthesis , Interleukin-18/chemistry , Amino Acid Sequence , Base Sequence , Biological Assay , Caspase 1/metabolism , Caspases/metabolism , Caspases, Initiator , Codon , Cysteine/metabolism , DNA, Complementary/metabolism , Dithionitrobenzoic Acid/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Escherichia coli/metabolism , Gene Library , Humans , Interleukin-18/metabolism , Methionine/chemistry , Molecular Sequence Data , Plasmids/metabolism , Protein Biosynthesis , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Sulfhydryl Reagents/pharmacology , Temperature , Time Factors , Transcription, Genetic , Ubiquitin/metabolism
17.
Cancer Lett ; 188(1-2): 127-40, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12406558

ABSTRACT

Histone deacetylase (HDAC) inhibitors such as trichostatin (TSA) and butyrate have been shown to inhibit cancer cell proliferation, induce apoptosis and regulate the expression of genes involved in cell cycle. Although the precise mechanism underlying HDAC inhibitor-induced cell growth arrest is not fully understood, induction of cell cycle related genes such as p21(cip/waf), is thought to be important. Here we showed that in the SW620 human colon cancer cell line, TSA and butyrate induced the growth arrest and DNA damage gene 45alpha (GADD45alpha) and GADD45beta. Furthermore, GADD45beta and p21(cip/waf) messenger RNA were induced in the absence of protein synthesis, indicating that both genes were immediate target genes for TSA. Cyclohexamide and TSA super-induced the expression of GADD45alpha and beta, but not p21(cip/waf). Interestingly while mitogen-activated kinase (MEK) inhibitor PD98059 and p38 kinase inhibitor SB242235 were unable to affect GADD45 induction, two serine/threonine protein kinase inhibitors (H7 and H8) as well as curcumin completely blocked the super-induction. Concomitant to the inhibition of GADD45 induction, H7 and H8 also blocked TSA-induced apoptosis. Taken together, these results suggest that GADD45 induction may play important role in TSA-induced cellular effects.


Subject(s)
Antigens, Differentiation/genetics , Butyrates/pharmacology , Cell Cycle Proteins , Colonic Neoplasms/genetics , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Nuclear Proteins/genetics , RNA, Messenger/biosynthesis , Blotting, Northern , Blotting, Western , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Cycloheximide/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Protein Synthesis Inhibitors/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
18.
J Biol Chem ; 277(48): 46609-15, 2002 Nov 29.
Article in English | MEDLINE | ID: mdl-12244092

ABSTRACT

Chk1 is a serine-threonine kinase that plays an important role in the DNA damage response, including G(2)/M cell cycle control. UCN-01 (7-hydroxystaurosporine), currently in clinical trials, has recently been shown to be a potent Chk1 inhibitor that abrogates the G(2)/M checkpoint induced by DNA-damaging agents. To understand the structural basis of Chk1 inhibition by UCN-01, we determined the crystal structure of the Chk1 kinase domain in complex with UCN-01. Chk1 structures with staurosporine and its analog SB-218078 were also determined. All three compounds bind in the ATP-binding pocket of Chk1, producing only slight changes in the protein conformation. Selectivity of UCN-01 toward Chk1 over cyclin-dependent kinases can be explained by the presence of a hydroxyl group in the lactam moiety interacting with the ATP-binding pocket. Hydrophobic interactions and hydrogen-bonding interactions were observed in the structures between UCN-01 and the Chk1 kinase domain. The high structural complementarity of these interactions is consistent with the potency and selectivity of UCN-01.


Subject(s)
Alkaloids/pharmacology , Protein Kinase Inhibitors , Alkaloids/chemistry , Amino Acid Sequence , Checkpoint Kinase 1 , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Kinases/chemistry , Staurosporine/analogs & derivatives , Structure-Activity Relationship
19.
J Immunother ; 25 Suppl 1: S20-7, 2002.
Article in English | MEDLINE | ID: mdl-12048347

ABSTRACT

Interleukin (IL)-18 has profound antitumor activity when administered at high doses as a single agent for prolonged periods in BALB/c mice bearing late, well-established MOPC-315 tumors. Management with a qD x 27 schedule resulted in regression of tumors in all animals receiving 5 mg/kg/d. A protracted daily management regimen appears to be necessary to induce regression in this advanced tumor model. Biologic markers were assessed and appear to be potentially useful in evaluating the immunologic and antitumor activity of IL-18. The biomarkers of IL-18's immunologic activity include, but are not limited to, IL-1alpha, IL-2, IL-8, IL-10, IL-12, IL-13, interferon-gamma, tumor necrosis factor-alpha, and granulocyte-macrophage colony-stimulating factor. The profile of these circulating cytokines and their expression levels at baseline, and after IL-18 delivery, can be measured in the serum, as well as from splenocytes of mice or human peripheral blood mononuclear cells derived from either normal subjects or patients with cancer. We compared IL-18 and IL-12 alone or in combination for their ability to induce cytokine production and natural killer cytolytic activity. Our data support the notion that IL-18 induces a predominantly Th1 response, and that the mechanism of IL-18 activity differs from that of IL-12. The biologic activity of IL-18 management revealed by increases in serum levels of cytokines and enhancement of natural killer cytolytic activity will be useful as clinical trials initiate in 2002. Expression of interferon-gamma and granulocyte-macrophage colony-stimulating factor serum levels correlates directly over a broad dose escalation with the level of IL-18. Therefore, this provides a convenient pharmacodynamic reference to the biologic response to IL-18 that may serve to guide the conduct of clinical trials.


Subject(s)
Cytokines/metabolism , Immunity, Cellular/drug effects , Interleukin-12/pharmacology , Interleukin-18/pharmacology , Plasmacytoma/drug therapy , Th1 Cells/drug effects , Animals , Cytokines/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Female , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Recombination, Genetic , Reference Values , Sensitivity and Specificity , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...