Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(11): 105341, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37832873

ABSTRACT

The emergence of severe acute respiratory syndrome coronavirus 2, the causative agent of coronavirus disease 2019, has resulted in the largest pandemic in recent history. Current therapeutic strategies to mitigate this disease have focused on the development of vaccines and on drugs that inhibit the viral 3CL protease or RNA-dependent RNA polymerase enzymes. A less-explored and potentially complementary drug target is Nsp15, a uracil-specific RNA endonuclease that shields coronaviruses and other nidoviruses from mammalian innate immune defenses. Here, we perform a high-throughput screen of over 100,000 small molecules to identify Nsp15 inhibitors. We characterize the potency, mechanism, selectivity, and predicted binding mode of five lead compounds. We show that one of these, IPA-3, is an irreversible inhibitor that might act via covalent modification of Cys residues within Nsp15. Moreover, we demonstrate that three of these inhibitors (hexachlorophene, IPA-3, and CID5675221) block severe acute respiratory syndrome coronavirus 2 replication in cells at subtoxic doses. This study provides a pipeline for the identification of Nsp15 inhibitors and pinpoints lead compounds for further development against coronavirus disease 2019 and related coronavirus infections.


Subject(s)
Antiviral Agents , Endoribonucleases , SARS-CoV-2 , Viral Nonstructural Proteins , Antiviral Agents/pharmacology , Endoribonucleases/antagonists & inhibitors , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/drug effects
3.
EMBO Mol Med ; 10(2): 160-174, 2018 02.
Article in English | MEDLINE | ID: mdl-29246970

ABSTRACT

Highly active antiretroviral therapy (HAART) has improved the outlook for the HIV epidemic, but does not provide a cure. The proposed "shock-and-kill" strategy is directed at inducing latent HIV reservoirs, which may then be purged via boosted immune response or targeting infected cells. We describe five novel compounds that are capable of reversing HIV latency without affecting the general T-cell activation state. The new compounds exhibit synergy for reactivation of latent provirus with other latency-reversing agents (LRAs), in particular ingenol-3-angelate/PEP005. One compound, designated PH02, was efficient at reactivating viral transcription in several cell lines bearing reporter HIV-1 at different integration sites. Furthermore, it was capable of reversing latency in resting CD4+ T lymphocytes from latently infected aviremic patient cells on HAART, while producing minimal cellular toxicity. The combination of PH02 and PEP005 produces a strong synergistic effect for reactivation, as demonstrated through a quantitative viral outgrowth assay (qVOA), on CD4+ T lymphocytes from HIV-1-infected individuals. We propose that the PH02/PEP005 combination may represent an effective novel treatment for abrogating persistent HIV-1 infection.


Subject(s)
Diterpenes/pharmacology , HIV Infections/drug therapy , HIV-1/physiology , Virus Activation/drug effects , Virus Latency/drug effects , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Drug Synergism , HIV Infections/immunology , Humans , Lymphocyte Activation
4.
PLoS One ; 11(9): e0162384, 2016.
Article in English | MEDLINE | ID: mdl-27598782

ABSTRACT

The apolipoprotein E (APOE) gene is the most highly associated susceptibility locus for late onset Alzheimer's Disease (AD), and augmenting the beneficial physiological functions of apoE is a proposed therapeutic strategy. In a high throughput phenotypic screen for small molecules that enhance apoE secretion from human CCF-STTG1 astrocytoma cells, we show the chrysanthemic ester 82879 robustly increases expressed apoE up to 9.4-fold and secreted apoE up to 6-fold and is associated with increased total cholesterol in conditioned media. Compound 82879 is unique as structural analogues, including pyrethroid esters, show no effect on apoE expression or secretion. 82879 also stimulates liver x receptor (LXR) target genes including ATP binding cassette A1 (ABCA1), LXRα and inducible degrader of low density lipoprotein receptor (IDOL) at both mRNA and protein levels. In particular, the lipid transporter ABCA1 was increased by up to 10.6-fold upon 82879 treatment. The findings from CCF-STTG1 cells were confirmed in primary human astrocytes from three donors, where increased apoE and ABCA1 was observed along with elevated secretion of high-density lipoprotein (HDL)-like apoE particles. Nuclear receptor transactivation assays revealed modest direct LXR agonism by compound 82879, yet 10 µM of 82879 significantly upregulated apoE mRNA in mouse embryonic fibroblasts (MEFs) depleted of both LXRα and LXRß, demonstrating that 82879 can also induce apoE expression independent of LXR transactivation. By contrast, deletion of LXRs in MEFs completely blocked mRNA changes in ABCA1 even at 10 µM of 82879, indicating the ability of 82879 to stimulate ABCA1 expression is entirely dependent on LXR transactivation. Taken together, compound 82879 is a novel chrysanthemic ester capable of modulating apoE secretion as well as apoE-associated lipid metabolic pathways in astrocytes, which is structurally and mechanistically distinct from known LXR agonists.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Apolipoproteins E/genetics , Astrocytes/drug effects , Liver X Receptors/genetics , Pyrethrins/pharmacology , Receptors, LDL/genetics , ATP Binding Cassette Transporter 1/agonists , ATP Binding Cassette Transporter 1/metabolism , Animals , Apolipoproteins E/agonists , Apolipoproteins E/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Cell Line, Tumor , Esters , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , High-Throughput Screening Assays , Humans , Lipid Metabolism/drug effects , Liver X Receptors/agonists , Liver X Receptors/metabolism , Mice , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/agonists , Receptors, LDL/metabolism , Signal Transduction
5.
Eur J Med Chem ; 120: 64-73, 2016 Sep 14.
Article in English | MEDLINE | ID: mdl-27187859

ABSTRACT

The development of treatments for influenza that inhibit the M2 proton channel without being susceptible to the widespread resistance mechanisms associated with the adamantanes is an ongoing challenge. Using a yeast high-throughput yeast growth restoration assay designed to identify M2 channel inhibitors, a single screening hit was uncovered. This compound (3), whose structure was incorrectly identified in the literature, is an inhibitor with similar potency to amantadine against WT M2. A library of derivatives of 3 was prepared and activity against WT M2 and the two principal mutant strains (V27A and S31N) was assessed in the yeast assay. The best compounds were further evaluated in an antiviral plaque reduction assay using engineered WT, V27A and S31N M2 influenza A strains with otherwise identical genetic background. Compound 63 was found to inhibit all three virus strains in this cell based antiviral assay at micromolar concentrations, possibly through a mechanism other than M2 inhibition.


Subject(s)
Amantadine/pharmacology , Antiviral Agents/chemistry , Viral Matrix Proteins/antagonists & inhibitors , Amantadine/chemistry , Antiviral Agents/pharmacology , Humans , Influenza A virus/drug effects , Influenza A virus/genetics , Influenza, Human/drug therapy , Mutation , Protons , Small Molecule Libraries , Structure-Activity Relationship
6.
Bioorg Med Chem ; 23(13): 3237-47, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25979376

ABSTRACT

An efficient assay for monitoring the activity of the key autophagy-initiating enzyme ATG4B based on a small peptide substrate has been developed. A number of putative small fluorogenic peptide substrates were prepared and evaluated and optimized compounds showed reasonable rates of cleavage but required high enzyme concentrations which limited their value. A modified peptide substrate incorporating a less sterically demanding self-immolative element was designed and synthesized and was shown to have enhanced properties useful for evaluating inhibitors of ATG4B. Substrate cleavage was readily monitored and was linear for up to 4h but enzyme concentrations of about ten-fold higher were required compared to assays using protein substrate LC3 or analogs thereof (such as FRET-LC3). Several known inhibitors of ATG4B were evaluated using the small peptide substrate and gave IC50 values 3-7 fold higher than previously obtained values using the FRET-LC3 substrate.


Subject(s)
Biological Assay , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemistry , Fluorescent Dyes/chemical synthesis , Peptides/chemical synthesis , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Autophagy , Autophagy-Related Proteins , Cysteine Endopeptidases/genetics , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Humans , Microtubule-Associated Proteins/chemistry , Molecular Sequence Data , Peptides/chemistry , Proteolysis , Recombinant Fusion Proteins/genetics
7.
Mol Pain ; 11: 12, 2015 Mar 14.
Article in English | MEDLINE | ID: mdl-25889575

ABSTRACT

BACKGROUND: Cav3.2 channels facilitate nociceptive transmission and are upregulated in DRG neurons in response to nerve injury or peripheral inflammation. We reported that this enhancement of Cav3.2 currents in afferent neurons is mediated by deubiquitination of the channels by the deubiquitinase USP5, and that disrupting USP5/Cav3.2 channel interactions protected from inflammatory and neuropathic pain. RESULTS: Here we describe the development of a small molecule screening assay for USP5-Cav3.2 disruptors, and report on two hits of a ~5000 compound screen - suramin and the flavonoid gossypetin. In mouse models of inflammatory pain and neuropathic pain, both suramin and gossypetin produced dose-dependent and long-lasting mechanical anti-hyperalgesia that was abolished or greatly attenuated in Cav3.2 null mice. Suramin and Cav3.2/USP5 Tat-disruptor peptides were also tested in models of diabetic neuropathy and visceral pain, and provided remarkable protection. CONCLUSIONS: Overall, our findings provide proof of concept for a new class of analgesics that target T-type channel deubiquitination.


Subject(s)
Calcium Channels, T-Type/metabolism , Neuralgia/metabolism , Neurons, Afferent/metabolism , Suramin/pharmacology , Ubiquitin-Specific Proteases/metabolism , Analgesics/pharmacology , Animals , Ganglia, Spinal/physiopathology , Humans , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Inflammation/metabolism , Mice , Mice, Knockout , Neuralgia/physiopathology
8.
Mol Pharmacol ; 87(4): 606-16, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25597706

ABSTRACT

P2X4 is an ATP-gated nonselective cation channel highly permeable to calcium. There is increasing evidence that this homomeric purinoceptor, which is expressed in several neuronal and immune cell types, is involved in chronic pain and inflammation. The current paucity of unambiguous pharmacological tools available to interrogate or modulate P2X4 function led us to pursue the search for selective antagonists. In the high-throughput screen of a compound library, we identified the phenylurea BX430 (1-(2,6-dibromo-4-isopropyl-phenyl)-3-(3-pyridyl)urea, molecular weight = 413), with antagonist properties on human P2X4-mediated calcium uptake. Patch-clamp electrophysiology confirmed direct inhibition of P2X4 currents by extracellular BX430, with submicromolar potency (IC50 = 0.54 µM). BX430 is highly selective, having virtually no functional impact on all other P2X subtypes, namely, P2X1-P2X3, P2X5, and P2X7, at 10-100 times its IC50. Unexpected species differences were noticed, as BX430 is a potent antagonist of zebrafish P2X4 but has no effect on rat and mouse P2X4 orthologs. The concentration-response curve for ATP on human P2X4 in the presence of BX430 shows an insurmountable blockade, indicating a noncompetitive allosteric mechanism of action. Using a fluorescent dye uptake assay, we observed that BX430 also effectively suppresses ATP-evoked and ivermectin-potentiated membrane permeabilization induced by P2X4 pore dilation. Finally, in single-cell calcium imaging, we validated its selective inhibitory effects on native P2X4 channels at the surface of human THP-1 cells that were differentiated into macrophages. In summary, this ligand provides a novel molecular probe to assess the specific role of P2X4 in inflammatory and neuropathic conditions, where ATP signaling has been shown to be dysfunctional.


Subject(s)
Aminopyridines/pharmacology , Phenylurea Compounds/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X4/metabolism , Allosteric Regulation , Aminopyridines/chemistry , Animals , Calcium/metabolism , Databases, Chemical , HEK293 Cells , Humans , Mice , Patch-Clamp Techniques , Phenylurea Compounds/chemistry , Purinergic P2X Receptor Antagonists/chemistry , Rats , Receptors, Purinergic P2X4/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity , Zebrafish
9.
J Biomol Screen ; 19(10): 1372-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25117203

ABSTRACT

Mutations of DNA repair pathways contribute to tumorigenesis and provide a therapeutic target for synthetic lethal interactions in tumor cells. Given that tyrosyl-DNA phosphodiesterase 1 (Tdp1) repairs stalled topoisomerase-I DNA complexes, we hypothesized that inhibition of Tdp1 has synthetic lethal effects in some cancers. To test this, we screened tumor arrays for Tdp1 expression and observed that Tdp1 is expressed in many tumors, including more than 90% of human breast tumors. Subsequent chemical screening identified putative Tdp1 inhibitors. Treatment of control human mammary epithelial cells and the breast cancer cell line MCF-7 with compound CD00509 preferentially sensitized MCF-7 cells to camptothecin and decreased cell proliferation 25% more than camptothecin treatment alone. This suggests that CD00509 specifically targeted Tdp1 in vitro, and CD00509 increased the sensitivity of wild-type murine embryonic fibroblasts (MEFs) to camptothecin to a degree comparable to that of Tdp1(-/-) MEFs. In addition, consistent with poly ADP-ribose polymerase-1 (PARP-1) collaborating with Tdp1 in DNA repair, combined Tdp1 and PARP-1 inhibition was more detrimental to MCF-7 cells than either treatment alone, whereas the combination was not additively harmful to control mammary cells. We conclude that targeting Tdp1 in anticancer therapy preferentially enhances the sensitivity of some breast cancer cells to camptothecin and may be an effective adjuvant for breast cancer therapy.


Subject(s)
High-Throughput Screening Assays/methods , Neoplasms/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Pyrans/pharmacology , Thiobarbiturates/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Computer Simulation , DNA Damage/drug effects , Female , Gene Knockdown Techniques , Histones/metabolism , Humans , In Vitro Techniques , MCF-7 Cells/drug effects , Mice , Molecular Docking Simulation , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Tissue Array Analysis , Topoisomerase I Inhibitors/pharmacology
10.
Bioorg Med Chem Lett ; 24(15): 3537-40, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24948562

ABSTRACT

High throughput screening of a pre-fractionated natural product library identified 11 active fractions showing ApoE modulation activity. Mass-directed fractionation of one active crude extract from the Australian marine sponge Callyspongia sp. resulted in the isolation of 13 metabolites, including three new bromotyrosine derivatives, callyspongic acid (1), 3,5-dibromo-4-methoxyphenylpyruvic acid (2), N-acetyl-3-bromo-4-hydroxylphenylethamine (3), and ten known compounds (4-13). The structure elucidation of compounds 1-3 was based on their 1D and 2D NMR and MS spectroscopic data. 3,5-Dibromo-4-methoxyphenylpyruvic acid (2) showed weak activity in increasing the apolipoprotein E secretion from human CCF-STTG1 cells at the concentration of 40 µM.


Subject(s)
Apolipoproteins E/metabolism , Callyspongia/chemistry , Tyrosine/analogs & derivatives , Animals , Australia , Callyspongia/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship , Tyrosine/chemistry , Tyrosine/metabolism , Tyrosine/pharmacology
11.
Assay Drug Dev Technol ; 12(3): 176-89, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24735444

ABSTRACT

The cysteine protease ATG4B plays a role in key steps of the autophagy process and is of interest as a potential therapeutic target. At an early step, ATG4B cleaves proLC3 isoforms to form LC3-I for subsequent lipidation to form LC3-II and autophagosome membrane insertion. ATG4B also cleaves phosphatidylethanolamine (PE) from LC3-II to regenerate LC3-I, enabling its recycling for further membrane biogenesis. Here, we report several novel assays for monitoring the enzymatic activity of ATG4B. An assay based on mass spectrometric analysis and quantification of cleavage of the substrate protein LC3-B was developed and, while useful for mechanistic studies, was not suitable for high throughput screening (HTS). A doubly fluorescent fluorescence resonance energy transfer (FRET) ligand YFP-LC3B-EmGFP (FRET-LC3) was constructed and shown to be an excellent substrate for ATG4B with rates of cleavage similar to that for LC3B itself. A HTS assay to identify candidate inhibitors of ATG4B utilizing FRET-LC3 as a substrate was developed and validated with a satisfactory Z' factor and high signal-to-noise ratio suitable for screening small molecule libraries. Pilot screens of the 1,280-member library of pharmacologically active compounds (LOPAC(™)) and a 3,481-member library of known drugs (KD2) gave hit rates of 0.6% and 0.5% respectively, and subsequent titrations confirmed ATG4B inhibitory activity for three compounds, both in the FRET and mass spectrometry assays. The FRET- and mass spectrometry-based assays we have developed will allow for both HTS for inhibitors of ATG4B and mechanistic approaches to study inhibition of a major component of the autophagy pathway.


Subject(s)
Cysteine Endopeptidases/chemistry , Drug Evaluation, Preclinical/methods , Fluorescence Recovery After Photobleaching/methods , Fluorescent Dyes/chemistry , Mass Spectrometry/methods , Autophagy-Related Proteins , Cysteine Endopeptidases/analysis , Enzyme Activation , Substrate Specificity
12.
PLoS One ; 8(2): e55271, 2013.
Article in English | MEDLINE | ID: mdl-23383318

ABSTRACT

The M2 proton channel of the influenza A virus is the target of the anti-influenza drugs amantadine and rimantadine. The effectiveness of these drugs has been dramatically limited by the rapid spread of drug resistant mutations, mainly at sites S31N, V27A and L26F in the pore of the channel. Despite progress in designing inhibitors of V27A and L26F M2, there are currently no drugs targeting these mutated channels in clinical trials. Progress in developing new drugs has been hampered by the lack of a robust assay with sufficient throughput for discovery of new active chemotypes among chemical libraries and sufficient sensitivity to provide the SAR data essential for their improvement and development as drugs. In this study we adapted a yeast growth restoration assay, in which expression of the M2 channel inhibits yeast growth and exposure to an M2 channel inhibitor restores growth, into a robust and sensitive high-throughput screen for M2 channel inhibitors. A screen of over 250,000 pure chemicals and semi-purified fractions from natural extracts identified 21 active compounds comprising amantadine, rimantadine, 13 related adamantanes and 6 non-adamantanes. Of the non-adamantanes, hexamethylene amiloride and a triazine derivative represented new M2 inhibitory chemotypes that also showed antiviral activity in a plaque reduction assay. Of particular interest is the fact that the triazine derivative was not sufficiently potent for detection as an inhibitor in the traditional two electrode voltage clamp assay for M2 channel activity, but its discovery in the yeast assay led to testing of analogues of which one was as potent as amantadine.


Subject(s)
Antiviral Agents/isolation & purification , Drug Discovery/methods , High-Throughput Screening Assays/methods , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/genetics , Antiviral Agents/pharmacology , Mutation, Missense/genetics , Patch-Clamp Techniques , Sensitivity and Specificity , Yeasts/drug effects , Yeasts/growth & development
13.
ACS Chem Biol ; 4(2): 139-44, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19175306

ABSTRACT

A marine natural product extract library has been screened with a functional cell-based G-protein coupled receptor assay to find compounds capable of binding the human cannabinoid receptors CB1 and CB2. The methanol extract of the marine sponge Dasychalina fragilis collected in Papua New Guinea was active in the assay. Bioassay-guided fractionation of the extract identified the phosphorylated sterol sulfate haplosamate A (1) as a cannabinoid receptor agonist. The high water solubility of haplosamate A (1) allowed exploration of its binding interactions with the human cannabinoid receptors in whole insect cells by means of saturation transfer double-difference NMR spectroscopy. This technique confirmed that haplosamate A (1) binds selectively to these receptors.


Subject(s)
Porifera/chemistry , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Sterols , Animals , Binding, Competitive , Biological Assay , Cell Line, Transformed , Humans , Insecta , Magnetic Resonance Spectroscopy , Papua New Guinea , Sterols/chemistry , Sterols/isolation & purification , Tissue Extracts
14.
Biochemistry ; 47(35): 9098-106, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18690710

ABSTRACT

RS1, also known as retinoschisin, is an extracellular discoidin domain-containing protein that has been implicated in maintaining the cellular organization and synaptic structure of the vertebrate retina. Mutations in the gene encoding RS1 are responsible for X-linked retinoschisis, a retinal degenerative disease characterized by the splitting of the retinal cell layers and visual impairment. To better understand the role of RS1 in retinal cell biology and X-linked retinoschisis, we have studied the interaction of wild-type and mutant RS1 with various carbohydrates coupled to agarose supports. RS1 bound efficiently to galactose-agarose and to a lesser extent lactose-agarose, but not agarose, N-acetylgalactosamine-agarose, N-acetylglucosamine-agarose, mannose-agarose, or heparin-agarose. RS1 cysteine mutants (C59S/C223S and C59S/C223S/C40S) which prevent disulfide-linked octamer formation exhibited little if any binding to galactose-agarose. The disease-causing R141H mutant bound galactose-agarose at levels similar to that of wild-type RS1, whereas the R141S mutant resulted in a marked reduction in the level of galactose-agarose binding. RS1 bound to galactose-agarose could be effectively displaced by incubation with isopropyl beta- d-1-thiogalactopyranoside (IPTG). This property was used as a basis to develop an efficient purification procedure. Anion exchange and galactose affinity chromatography was used to purify RS1 from the culture media of stably transformed Sf21 insect cells that express and secrete RS1. This cell expression and protein purification method should prove useful in the isolation of RS1 for detailed structure-function studies.


Subject(s)
Eye Proteins/isolation & purification , Eye Proteins/metabolism , Galactose/metabolism , Lectins/chemistry , Protozoan Proteins/chemistry , Animals , Binding Sites , Cells, Cultured , Discoidins , Eye Proteins/chemistry , Humans , Protein Structure, Tertiary
15.
Drug Metab Dispos ; 36(6): 991-4, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18322072

ABSTRACT

The anthracycline drugs are important for the treatment of a number of malignancies; however, their clinical use is associated with dose-dependent severe chronic cardiotoxicity. Although the mechanism for this side effect has not yet been identified, the alcohol metabolites formed during daunorubicin (DAUN) and doxorubicin (DOX) therapies have been implicated. The alcohol metabolites of DAUN and DOX, daunorubicinol (DAUNol) and doxorubicinol (DOXol), respectively, are generated through reduction of the C-13 carbonyl function, which is reportedly mediated by members of the aldo-keto reductase and carbonyl reductase families of proteins. In our search for potential biomarkers for the occurrence of this side effect, we examined the activity of recombinant aldo-keto reductase enzymes, aldo-keto reductase (AKR) 1A1 and AKR1C2, with DAUN and DOX as substrates. Using purified histidine-tagged recombinant proteins and the direct measurement of metabolite formation with a high-performance liquid chromatography-fluorescence assay, we did not observe DAUNol or DOXol generation in vitro by AKR1C2, whereas AKR1A1 did catalyze the reduction reactions. DAUNol was generated by AKR1A1 at a rate of 1.71 +/- 0.09 nmol/min/mg protein, and a low level of DOXol was produced by AKR1A1; however, it was below the limits of quantification for the method. These data suggest that the generation of DAUNol or DOXol by AKR1C2 metabolism in vivo is unlikely to occur during anthracycline treatment.


Subject(s)
Alcohol Oxidoreductases/metabolism , Antibiotics, Antineoplastic/metabolism , Daunorubicin/metabolism , Doxorubicin/metabolism , Hydroxysteroid Dehydrogenases/metabolism , Recombinant Proteins/metabolism , Alcohol Oxidoreductases/genetics , Aldehyde Reductase , Aldo-Keto Reductases , Chromatography, High Pressure Liquid , Daunorubicin/analogs & derivatives , Doxorubicin/analogs & derivatives , Fluorescence , Humans , Hydroxysteroid Dehydrogenases/genetics , Recombinant Proteins/genetics
16.
Drug Metab Dispos ; 36(5): 904-10, 2008 May.
Article in English | MEDLINE | ID: mdl-18276838

ABSTRACT

Aldo-keto reductases (AKRs) are a class of NADPH-dependent oxidoreductases that have been linked to metabolism of the anthracyclines doxorubicin (DOX) and daunorubicin (DAUN). Although widely used, cardiotoxicity continues to be a serious side effect that may be linked to metabolites or reactive intermediates generated in their metabolism. In this study we examine the little known effects of nonsynonymous single nucleotide polymorphisms of human AKR1A1 on the metabolism of these drugs to their alcohol metabolites. Expressed and purified from bacteria using affinity chromatography, the AKR1A1 protein with a single histidine (6x-His) tag exhibited the greatest activity using two test substrates: p-nitrobenzaldehyde (5.09 +/- 0.16 micromol/min/mg of purified protein) and DL-glyceraldehyde (1.24 +/- 0.17 micromol/min/mg). These activities are in agreement with published literature values of nontagged human AKR1A1. The 6x-His-tagged AKR1A1 wild type and allelic variants, E55D and N52S, were subsequently examined for metabolic activity using DAUN and DOX. The tagged variants showed significantly reduced activities (1.10 +/- 0.42 and 0.72 +/- 0.47 nmol of daunorubicinol (DAUNol) formed/min/mg of purified protein for E55D and N52S, respectively) compared with the wild type (2.34 +/- 0.71 nmol/min/mg). The wild type and E55D variant metabolized DOX to doxorubicinol (DOXol); however, the levels fell below the limit of quantitation (25 nM). The N52S variant yielded no detectable DOXol. A kinetic analysis of the DAUN reductase activities revealed that both amino acid substitutions lead to reduced substrate affinity, measured as significant increases in the measured K(m) for the reduction reaction by AKR1A1. Hence, it is possible that these allelic variants can act as genetic biomarkers for the clinical development of DAUN-induced cardiotoxicity.


Subject(s)
Aldehyde Reductase/metabolism , Antibiotics, Antineoplastic/metabolism , Daunorubicin/metabolism , Recombinant Proteins/metabolism , Aldehyde Reductase/genetics , Alleles , Biomarkers/metabolism , Doxorubicin/metabolism , Genetic Variation , Humans , Recombinant Proteins/genetics
17.
Mol Cell Proteomics ; 6(9): 1574-88, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17510048

ABSTRACT

Translational research is progressing toward combined genomics and proteomics analyses of small and precious samples. In our analyses of spinal cord material, we systematically evaluated disruption and extraction techniques to determine an optimum process for the coupled analysis of RNA and protein from a single 5-mm segment of tissue. Analyses of these distinct molecular species were performed using microarrays and high resolution two-dimensional gels, respectively. Comparison of standard homogenization with automated frozen disruption (AFD) identified negligible differences in the relative abundance of genes (44) with all genes identified by either process. Analysis on either the Affymetrix or Applied Biosystems Inc. gene array platforms provided good correlations between the extraction techniques. In contrast, the AFD technique enabled identification of more unique proteins from spinal cord tissue than did standard homogenization. Furthermore use of an optimized CHAPS/urea extraction provided better protein recovery, as shown by quantitative two-dimensional gel analyses, than did solvent precipitation during TRIzol-based RNA extraction. Thus, AFD of tissue samples followed by protein and RNA isolation from separate aliquots of the frozen powdered sample is the most effective route to ensure full, quantitative analyses of both molecular entities.


Subject(s)
Genomics/methods , Proteomics/methods , Spinal Cord/metabolism , Animals , Automation , Electrophoresis, Gel, Two-Dimensional , Guanidines/pharmacology , Male , Models, Biological , Oligonucleotide Array Sequence Analysis , Phenols/pharmacology , Protein Biosynthesis , Proteome , RNA/chemistry , Rats , Rats, Sprague-Dawley
18.
Biochem Cell Biol ; 84(2): 148-56, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16609695

ABSTRACT

The lysosomal hydrolase, glucocerebrosidase (GBA), catalyses the penultimate step in the breakdown of membrane glycosphingolipids. An inherited deficiency of this enzyme activity leads to the onset of Gaucher disease, the most common lysosomal storage disorder. Affected individuals range from adults with hepatosplenomegaly, haematological complications, and bone pain (type 1 disease) to children and neonates with severe neuronopathy leading to neurological degradation and premature death (type 2 and type 3 disease). Enzyme replacement therapy has become the standard of treatment for type I Gaucher disease but remains an expensive option, in part because of the cost of recombinant enzyme production using mammalian cell culture. Using a nonlytic integrative plasmid expression system, we have successfully produced active human GBA in stable transformed Sf9 (Spodoptera frugiperda) cells. Both the 39 and 19 amino acid native GBA signal sequences were capable of endoplasmic reticulum targeting, which led to secretion of the recombinant protein, although approximately 30% more enzyme was produced using the longer signal sequence. The secreted product was purified to apparent electrophoretic homogeneity using hydrophobic interaction chromatography and found to be produced in a fully glycosylated and a hypoglycosylated form, both of which cross-reacted with a human GBA-specific monoclonal antibody. The pH optimum (at pH 5.5) for activity of the recombinant enzyme was as expected for human GBA using the artificial substrate 4-methyl-umbelliferyl-beta-D-glycopyranoside. With initial nonoptimized expression levels estimated at 10-15 mg/L using small-scale batch cultures, stable transformed insect cells could provide a viable alternative system for the heterologous production of human GBA when grown under optimized perfusion culture conditions.


Subject(s)
Glucosylceramidase/biosynthesis , Animals , Base Sequence , Cell Line , DNA, Complementary/genetics , Gaucher Disease/drug therapy , Gaucher Disease/enzymology , Gaucher Disease/genetics , Glucosylceramidase/analysis , Glucosylceramidase/genetics , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Protein Sorting Signals/genetics , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Spodoptera , Transformation, Genetic
19.
Cytotechnology ; 44(3): 93-102, 2004 Mar.
Article in English | MEDLINE | ID: mdl-19003232

ABSTRACT

Factor Xa is a serine protease, whose high selectivity can be used to cleave protein tags from recombinant proteins. A fusion protein comprised of a self-activating form of factor X linked to a cellulose-binding module, saCBMFX, was produced in a stable transformed Sf9 insect cell line. The activity of the insect cell produced saCBMFX was higher than the equivalent mammalian cell produced material. A 1.5 l batch fermentation reached a maximum cell concentration of 1.6 x 10(7) cells ml(-1) and a final saCBMFX concentration of 4 mg l(-1). The production of saCBMFX by this cell line was also analyzed in a 1.5 l perfusion system using an ultrasonic filter as a cell-retention device for flow rates up to 3.5 l day(-1). The cell-retention efficiency of an air backflush mode of acoustic filter operation was greater than 95% and eliminated the need to pump the relatively shear sensitive insect cells. In the perfusion system over 4 x 10(7) Sf9 cells ml(-1) were obtained with a viability greater than 80%. With a doubling of viable cell concentration from 1.5 to 3 x 10(7) cells ml(-1) the saCBMFX production rate was doubled to 6 mg l(-1) day(-1). The saCBMFX volumetric productivity of the perfusion system was higher than the batch fermentations (0.6 mg l(-1) day(-1)) by an order of magnitude.

20.
Anal Biochem ; 320(1): 88-103, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12895473

ABSTRACT

Insect cells are an underexplored resource for functional G-protein-coupled receptor (GPCR) assays, despite a strong record in biochemical (binding) assays. Here we describe the use of vectors capable of creating stably transformed insect cell lines to generate a cell-based functional GPCR assay. This assay employs the luminescent photoprotein aequorin and the promiscuous G-protein subunit Galpha16 and is broadly applicable to human GPCRs. We demonstrate that the assay can quantitate ligand concentration-activity relationships for seven different human GPCRs, can differentiate between partial and full agonists, and can determine rank order potencies for both agonists and antagonists that match those seen with other assay systems. Human Galpha16 improves signal strength but is not required for activity with some receptors. The coexpression of human and bovine betagamma subunits and/or phospholipase Cbeta makes no difference to agonist efficacy or potency. Two different receptors expressed in the same cell line respond to their specific agonists, and two different cell lines (Sf9 and High 5) are able to functionally detect the same expressed GPCR. Sf9 cells have the capability to produce fully functional human receptors, allied to a low background of endogenous receptors, and so are a valuable system for investigating orphan GPCRs and receptor dimerization.


Subject(s)
Biological Assay/methods , Cell Line, Transformed , Imidazoles , Moths/cytology , Receptors, G-Protein-Coupled/metabolism , Aequorin/analysis , Animals , Cattle , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gq-G11/analysis , Heterotrimeric GTP-Binding Proteins/analysis , Heterotrimeric GTP-Binding Proteins/physiology , Humans , Ligands , Pyrazines/pharmacology , Receptors, Dopamine D1/agonists , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...