Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Am J Respir Cell Mol Biol ; 69(4): 422-440, 2023 10.
Article in English | MEDLINE | ID: mdl-37411041

ABSTRACT

Bleomycin-induced pulmonary fibrosis in mice mimics major hallmarks of idiopathic pulmonary fibrosis. Yet in this model, it spontaneously resolves over time. We studied molecular mechanisms of fibrosis resolution and lung repair, focusing on transcriptional and proteomic signatures and the effect of aging. Old mice showed incomplete and delayed lung function recovery 8 weeks after bleomycin instillation. This shift in structural and functional repair in old bleomycin-treated mice was reflected in a temporal shift in gene and protein expression. We reveal gene signatures and signaling pathways that underpin the lung repair process. Importantly, the downregulation of WNT, BMP, and TGFß antagonists Frzb, Sfrp1, Dkk2, Grem1, Fst, Fstl1, and Inhba correlated with lung function improvement. Those genes constitute a network with functions in stem cell pathways, wound, and pulmonary healing. We suggest that insufficient and delayed downregulation of those antagonists during fibrosis resolution in old mice explains the impaired regenerative outcome. Together, we identified signaling pathway molecules with relevance to lung regeneration that should be tested in-depth experimentally as potential therapeutic targets for pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Transcriptome , Mice , Animals , Transcriptome/genetics , Proteomics , Lung , Bleomycin , Mice, Inbred C57BL
2.
SLAS Discov ; 28(5): 240-246, 2023 07.
Article in English | MEDLINE | ID: mdl-37031752

ABSTRACT

Acoustic droplet ejection-open port interface-mass spectrometry (ADE-OPI-MS) is a novel label-free analytical technique, promising to become a versatile readout for high-throughput screening (HTS) applications. The recent introduction of ADE-OPI-MS devices to the laboratory equipment market, paired with their compatibility with laboratory automation platforms, should facilitate the adoption of this technology by a broader community. Towards this goal, instrument robustness in the context of HTS campaigns - where up to millions of samples in complex matrices are tested in a short time frame - represents a major challenge, which explains the absence of detailed literature reports on this subject. Here, we present the results of our first fully automated HTS campaign, based on the ADE-OPI-MS technology, aiming to identify inhibitors of a metabolic enzyme in a >1 million compound library. The report encompasses the assay development and validation steps, as well as the adaptation for HTS requirements, where refinement of the capillary cleaning concept was crucial for final success. Altogether, our study unequivocally demonstrates the applicability of the ADE-OPI-MS technology for HTS-based drug discovery.


Subject(s)
Drug Discovery , High-Throughput Screening Assays , High-Throughput Screening Assays/methods , Mass Spectrometry , Drug Discovery/methods , Acoustics , Automation, Laboratory
3.
Am J Physiol Lung Cell Mol Physiol ; 324(3): L245-L258, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36625483

ABSTRACT

The most common preclinical, in vivo model to study lung fibrosis is the bleomycin-induced lung fibrosis model in 2- to 3-mo-old mice. Although this model resembles key aspects of idiopathic pulmonary fibrosis (IPF), there are limitations in its predictability for the human disease. One of the main differences is the juvenile age of animals that are commonly used in experiments, resembling humans of around 20 yr. Because IPF patients are usually older than 60 yr, aging appears to play an important role in the pathogenesis of lung fibrosis. Therefore, we compared young (3 months) and old mice (21 months) 21 days after intratracheal bleomycin instillation. Analyzing lung transcriptomics (mRNAs and miRNAs) and proteomics, we found most pathways to be similarly regulated in young and old mice. However, old mice show imbalanced protein homeostasis as well as an increased inflammatory state in the fibrotic phase compared to young mice. Comparisons with published human transcriptomic data sets (GSE47460, GSE32537, and GSE24206) revealed that the gene signature of old animals correlates significantly better with IPF patients, and it also turned human healthy individuals better into "IPF patients" using an approach based on predictive disease modeling. Both young and old animals show similar molecular hallmarks of IPF in the bleomycin-induced lung fibrosis model, although old mice more closely resemble several features associated with IPF in comparison to young animals.


Subject(s)
Bleomycin , Idiopathic Pulmonary Fibrosis , Humans , Mice , Animals , Bleomycin/pharmacology , Transcriptome , Proteomics , Lung/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Disease Models, Animal , Mice, Inbred C57BL
4.
Dis Model Mech ; 15(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34845494

ABSTRACT

Alterations in metabolic pathways were recently recognized as potential underlying drivers of idiopathic pulmonary fibrosis (IPF), translating into novel therapeutic targets. However, knowledge of metabolic and lipid regulation in fibrotic lungs is limited. To comprehensively characterize metabolic perturbations in the bleomycin mouse model of IPF, we analyzed the metabolome and lipidome by mass spectrometry. We identified increased tissue turnover and repair, evident by enhanced breakdown of proteins, nucleic acids and lipids and extracellular matrix turnover. Energy production was upregulated, including glycolysis, the tricarboxylic acid cycle, glutaminolysis, lactate production and fatty acid oxidation. Higher eicosanoid synthesis indicated inflammatory processes. Because the risk of IPF increases with age, we investigated how age influences metabolomic and lipidomic changes in the bleomycin-induced pulmonary fibrosis model. Surprisingly, except for cytidine, we did not detect any significantly differential metabolites or lipids between old and young bleomycin-treated lungs. Together, we identified metabolomic and lipidomic changes in fibrosis that reflect higher energy demand, proliferation, tissue remodeling, collagen deposition and inflammation, which might serve to improve diagnostic and therapeutic options for fibrotic lung diseases in the future.


Subject(s)
Bleomycin , Idiopathic Pulmonary Fibrosis , Animals , Bleomycin/adverse effects , Bleomycin/metabolism , Fibrosis , Lipidomics , Lung/pathology , Mice , Mice, Inbred C57BL
5.
SLAS Discov ; 26(8): 961-973, 2021 09.
Article in English | MEDLINE | ID: mdl-34308708

ABSTRACT

Acoustic droplet ejection (ADE)-open port interface (OPI)-mass spectrometry (MS) has recently been introduced as a versatile analytical method that combines fast and contactless acoustic sampling with sensitive and accurate electrospray ionization (ESI)-MS-based analyte detection. The potential of the technology to provide label-free measurements in subsecond analytical cycle times makes it an attractive option for high-throughput screening (HTS). Here, we report the first implementation of ADE-OPI-MS in a fully automated HTS environment, based on the example of a biochemical assay aiming at the identification of small-molecule inhibitors of the cyclic guanosine monophosphate-adenosine monophosphate (GMP-AMP) synthase (cGAS). First, we describe the optimization of the method to enable sensitive and accurate determination of enzyme activity and inhibition in miniaturized 1536-well microtiter plate format. Then we show both results from a validation single-concentration screen using a test set of 5500 compounds, and the subsequent concentration-response testing of selected hits in direct comparison with a previously established matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) readout. Finally, we present the development of an in-line OPI cleaning procedure aiming to match the instrument robustness required for large-scale HTS campaigns. Overall, this work points to critical method development parameters and provides guidance for the establishment of integrated ADE-OPI-MS as HTS-compatible technology for early drug discovery.


Subject(s)
Automation, Laboratory , Drug Discovery/methods , High-Throughput Screening Assays/methods , Mass Spectrometry/methods , Drug Discovery/standards , High-Throughput Screening Assays/standards , Humans , Mass Spectrometry/standards , Spectrometry, Mass, Electrospray Ionization/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
6.
Rapid Commun Mass Spectrom ; 35(12): e9096, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-33837598

ABSTRACT

RATIONALE: The low speed and low flexibility of most liquid chromatography/tandem mass spectrometry (LC/MS/MS) approaches in early drug discovery delay sample analysis from routine in vivo studies within the same day. A high-throughput platform for the rapid quantification of drug compounds in various in vivo assays was developed and established in routine bioanalysis. METHODS: Automated selection of an efficient and adequate LC method was realized by autonomous sample qualification for ultrafast batch gradients (9 s/sample) or for fast linear gradients (45 s/sample) if samples required chromatography. The hardware and software components of our Rapid and Integrated Analysis System (RIAS) were streamlined for increased analytical throughput via state-of-the-art automation while maintaining high analytical quality. RESULTS: Online decision-making was based on a quick assay suitability test (AST), based on a small and dedicated sample set evaluated by two different strategies. 84% of the acquired data points were within ±30% accuracy and 93% of the deviations between the lower limit of quantitation (LLOQ) values were ≤2-fold compared with standard LC/MS/MS systems. Speed, flexibility and overall automation significantly improved. CONCLUSIONS: The developed platform provided an analysis time of only 10 min (batch-mode) and 47 min (gradient-mode) per standard pharmacokinetic (PK) study (62 injections). Automation, data evaluation and results handling were optimized to pave the way for machine learning based on decision-making regarding the evaluation strategy of the AST.


Subject(s)
Chromatography, High Pressure Liquid/methods , Drug Discovery/methods , Machine Learning , Tandem Mass Spectrometry/methods , Automation , High-Throughput Screening Assays/methods , Limit of Detection , Pharmaceutical Preparations/analysis
7.
SLAS Discov ; 26(1): 44-57, 2021 01.
Article in English | MEDLINE | ID: mdl-33073664

ABSTRACT

Demonstration of in vitro target engagement for small-molecule ligands by measuring binding to a molecular target is an established approach in early drug discovery and a pivotal step in high-throughput screening (HTS)-based compound triaging. We describe the setup, evaluation, and application of a ligand binding assay platform combining automated affinity selection (AS)-based sample preparation and label-free matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) analysis. The platform enables mass spectrometry (MS)-based HTS for small-molecule target interactions from single-compound incubation mixtures and is embedded into a regular assay automation environment. Efficient separation of target-ligand complexes is achieved by in-plate size exclusion chromatography (SEC), and small-molecule ligands are subsequently identified by MALDI-TOF analysis. In contrast to alternative HTS-capable binding assay formats, MALDI-TOF AS-MS is capable of identifying orthosteric and allosteric ligands, as shown for the model system protein tyrosine phosphatase 1B (PTP1B), irrespective of protein function. Furthermore, determining relative binding affinities (RBAs) enabled ligand ranking in accordance with functional inhibition and reference data for PTP1B and a number of diverse protein targets. Finally, we present a validation screen of more than 23,000 compounds within 24 h, demonstrating the general applicability of the platform for the HTS-compatible assessment of protein-ligand interactions.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays/methods , Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Automation, Laboratory , Humans , Ligands
8.
Anal Chem ; 92(18): 12242-12249, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32786476

ABSTRACT

We present an acoustic ejection mass spectrometry (AEMS) setup for contactless electrospray ionization mass spectrometry (ESI-MS)-based sample injection at a sampling rate faster than current ESI and matrix-assisted laser desorption ionization (MALDI) techniques. For the direct transfer of samples out of 384-well plates into a modified ESI source, an open port interface (OPI) was combined with a modified acoustic droplet ejection (ADE) system. AEMS has the potential to eliminate bottlenecks known from classical MS approaches, such as speed, reproducibility, carryover, ion suppression, as well as sample preparation and consumption. This setup provided a drastically reduced transfer distance between OPI and ESI electrode for optimum throughput performance and broadens the scope of applications for this emerging technique. To simulate label-free applications of drug metabolism and pharmacokinetics (DMPK) analysis and high-throughput screening (HTS) campaigns, two stress tests were performed regarding ion suppression and system endurance in combination with minor sample preparation. The maximum sampling rate was 6 Hz for dextromethorphan and d3-dextrorphan (each 100 nM) for 1152 injections in 63 s at full width at half-maximum (FWHM) of 105 ms and a relative standard deviation (%RSD) of 7.7/7.5% without internal standard correction. Enzyme assay buffer and crude dog plasma caused signal suppression of 51/73% at %RSD of 5.7/6.7% (n = 120). An HTS endurance buffer was used for >25 000 injections with minor OPI pollution and constant signals (%RSD = 8.5%, FWHM of 177 ms ± 8.5%, n = 10 557). The optimized hardware and method setup resulted in high-throughput performance and enables further implementation in a fully automated platform for ESI-MS-based high-throughput screening.


Subject(s)
Acoustics , Cytochrome P-450 Enzyme System/blood , Dextromethorphan/analysis , Dextrorphan/analysis , High-Throughput Screening Assays , Animals , Cytochrome P-450 Enzyme System/metabolism , Dogs , Electrodes , Female , High-Throughput Screening Assays/instrumentation , Male , Particle Size , Spectrometry, Mass, Electrospray Ionization/instrumentation , Time Factors
9.
Sci Rep ; 10(1): 1314, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992752

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a major cause of liver fibrosis with increasing prevalence worldwide. Currently there are no approved drugs available. The development of new therapies is difficult as diagnosis and staging requires biopsies. Consequently, predictive plasma biomarkers would be useful for drug development. Here we present a multi-omics approach to characterize the molecular pathophysiology and to identify new plasma biomarkers in a choline-deficient L-amino acid-defined diet rat NASH model. We analyzed liver samples by RNA-Seq and proteomics, revealing disease relevant signatures and a high correlation between mRNA and protein changes. Comparison to human data showed an overlap of inflammatory, metabolic, and developmental pathways. Using proteomics analysis of plasma we identified mainly secreted proteins that correlate with liver RNA and protein levels. We developed a multi-dimensional attribute ranking approach integrating multi-omics data with liver histology and prior knowledge uncovering known human markers, but also novel candidates. Using regression analysis, we show that the top-ranked markers were highly predictive for fibrosis in our model and hence can serve as preclinical plasma biomarkers. Our approach presented here illustrates the power of multi-omics analyses combined with plasma proteomics and is readily applicable to human biomarker discovery.


Subject(s)
Biomarkers , Genomics , Liver Diseases/etiology , Liver Diseases/metabolism , Proteomics , Animals , Chronic Disease , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Genomics/methods , Liver Diseases/diagnosis , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Phenotype , Proteome , Proteomics/methods
10.
SLAS Discov ; 25(4): 372-383, 2020 04.
Article in English | MEDLINE | ID: mdl-31583948

ABSTRACT

Comprehensive and unbiased detection methods are a prerequisite for high-throughput screening (HTS) campaigns within drug discovery research. Label-free matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry (MS) has been introduced as an HTS-compatible readout for biochemical test systems to support the drug discovery process. So far, reported HTS applications were based on surface-modified systems or proof-of-concept studies. We present the utilization of a MALDI-TOF-based screening platform to identify inhibitors of human cyclic GMP-AMP synthase (cGAS), a mediator of innate immune response whose aberration has been causally correlated to a number of inflammatory disorders. In this context, the development and validation of a MALDI-TOF-based activity assay is reported to demonstrate fast, robust, and accurate detection of chemical cGAS inhibition by direct quantification of the physiological reaction product cyclic GMP-ATP (cGAMP). Results from a screen of a diverse library of more than 1 million small molecules in 1536-well format against the catalytic cGAS activity are presented with excellent assay performance and data quality. Identified hits were qualified in dose-response experiments and confirmed by RapidFire-MS measurements. Conclusively, the presented data provide the first proof of applicability of direct automated MALDI-TOF MS as a readout strategy for large-scale drug discovery HTS campaigns.


Subject(s)
DNA/genetics , High-Throughput Screening Assays , Nucleotidyltransferases/antagonists & inhibitors , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Cytosol/enzymology , DNA/drug effects , Drug Discovery , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Humans , Nucleotidyltransferases/genetics , Small Molecule Libraries/pharmacology
11.
SLAS Discov ; 24(7): 766-777, 2019 08.
Article in English | MEDLINE | ID: mdl-31059309

ABSTRACT

Microbial-dependent trimethylamine (TMA) generation from dietary precursors such as choline was recently linked to cardiovascular diseases (CVDs) as well as chronic kidney disease (CKD). Inhibition of TMA-generating enzymes in gut bacteria would be an innovative approach to treat these diseases. The potential to accurately quantify secreted TMA levels highlights the capacity of mass spectrometry (MS) for tracking microbial TMA-lyase activity. However, high-throughput screening (HTS) by conventional MS instrumentation is hampered by limited sample throughput. Recent advancement in liquid handling and instrumentation of matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS provides an HTS-compatible MS technology. The deciphering of enzymatic reactions using this label-free readout has been successfully applied but has thus far been limited to peptide/protein-centric activity assays. Here, we demonstrate the versatile applicability of MALDI-TOF by tracking a small molecule within a highly complex sample background. The key to success for this concept was chemical derivatization of the target molecule enabling quantitative assessment of microbial TMA formation. Further, its potential was demonstrated in a side-by-side comparison to RapidFire-MS in a primary screen and subsequent dose-response experiments. Overall, the established assay enables the screening for microbial TMA-lyase inhibitors and serves as a proof of concept for the applicability of MALDI-TOF for demanding assay concepts per se.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Lyases/antagonists & inhibitors , Methylamines/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Humans
12.
SLAS Technol ; 24(4): 386-393, 2019 08.
Article in English | MEDLINE | ID: mdl-30698995

ABSTRACT

Label-free in vitro potency assays are an emerging field in drug discovery to enable more physiological conditions, to improve the readout quality, and to save time. For this approach mass spectrometry (MS) is a powerful technology to directly follow physiological processes. The speed of this methodology, however, was for a long time not compatible with chemiluminescence- or fluorescence-based assays. Recent advances in matrix-assisted laser desorption/ionization (MALDI) instrumentation paved the way for high-throughput MS analysis of label-free assays for large compound libraries, whereas electrospray ionization (ESI)-based mass spectrometers equipped with RapidFire autosamplers were limited to medium throughput. Here we present a technological advancement of the RapidFire device to enable cycle times of 2.5 s per sample. This newly developed BLAZE-mode substantially boosted the ESI-MS analysis speed, providing an alternative technology for label-free high-throughput screening.


Subject(s)
Automation, Laboratory/methods , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Spectrometry, Mass, Electrospray Ionization/methods , Automation, Laboratory/instrumentation , High-Throughput Screening Assays/instrumentation
13.
J Med Chem ; 62(1): 306-316, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30207464

ABSTRACT

Protein tyrosine phosphatase non-receptor type 5 (PTPN5, STEP) is a brain specific phosphatase that regulates synaptic function and plasticity by modulation of N-methyl-d-aspartate receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking. Dysregulation of STEP has been linked to neurodegenerative and neuropsychiatric diseases, highlighting this enzyme as an attractive therapeutic target for drug discovery. Selective targeting of STEP with small molecules has been hampered by high conservation of the active site among protein tyrosine phosphatases. We report the discovery of the first small molecule allosteric activator for STEP that binds to the phosphatase domain. Allosteric binding is confirmed by both X-ray and 15N NMR experiments, and specificity has been demonstrated by an enzymatic test cascade. Molecular dynamics simulations indicate stimulation of enzymatic activity by a long-range allosteric mechanism. To allow the scientific community to make use of this tool, we offer to provide the compound in the course of an open innovation initiative.


Subject(s)
Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Small Molecule Libraries/chemistry , Allosteric Regulation , Allosteric Site , Animals , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Mice , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Small Molecule Libraries/metabolism
14.
SLAS Technol ; 24(2): 209-221, 2019 04.
Article in English | MEDLINE | ID: mdl-30074850

ABSTRACT

Label-free, mass spectrometric (MS) deciphering of enzymatic reactions by direct analysis of substrate-to-product conversion provides the next step toward more physiological relevant assays within drug discovery campaigns. Reduced risk of suffering from compound interference combined with diminished necessity for tailored signal mediators emphasizes the valuable role of label-free readouts. However, MS-based detection has not hitherto met high-throughput screening (HTS) requirements because of the lack of HTS-compatible sample introduction. In the present study, we report on a fully automated liquid-handling concept built in-house to concatenate biochemical assays with matrix-assisted laser desorption/ionization time-of-flight closing this technological gap. The integrated reformatting from 384- to 1536-well format enables cycle times of 0.6 s/sample for automated spotting and 0.4 s/sample for MS analysis, matching the requirements of HTS compatibility. In-depth examination of spotting quality, quantification accuracy, and instrument robustness together with the implementation of a protein tyrosine phosphatase 1B (PTP1B) inhibitor screening (4896 compounds) demonstrate the potential of the heavily inquired HTS integration of the label-free MS readout. Overall, the presented data demonstrate that the introduced automation concept makes label-free MS-based readouts accessible for HTS within drug discovery campaigns but also in other research areas requiring ultrafast MS-based detection.


Subject(s)
Drug Discovery/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Drug Discovery/instrumentation , Drug Evaluation, Preclinical/instrumentation , High-Throughput Screening Assays/instrumentation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/instrumentation
15.
SLAS Discov ; 23(6): 561-573, 2018 07.
Article in English | MEDLINE | ID: mdl-29466676

ABSTRACT

Label-free, mass spectrometric (MS) detection is an emerging technology in the field of drug discovery. Unbiased deciphering of enzymatic reactions is a proficient advantage over conventional label-based readouts suffering from compound interference and intricate generation of tailored signal mediators. Significant evolvements of matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS, as well as associated liquid handling instrumentation, triggered extensive efforts in the drug discovery community to integrate the comprehensive MS readout into the high-throughput screening (HTS) portfolio. Providing speed, sensitivity, and accuracy comparable to those of conventional, label-based readouts, combined with merits of MS-based technologies, such as label-free parallelized measurement of multiple physiological components, emphasizes the advantages of MALDI-TOF for HTS approaches. Here we describe the assay development for the identification of protein tyrosine phosphatase 1B (PTP1B) inhibitors. In the context of this precious drug target, MALDI-TOF was integrated into the HTS environment and cross-compared with the well-established AlphaScreen technology. We demonstrate robust and accurate IC50 determination with high accordance to data generated by AlphaScreen. Additionally, a tailored MALDI-TOF assay was developed to monitor compound-dependent, irreversible modification of the active cysteine of PTP1B. Overall, the presented data proves the promising perspective for the integration of MALDI-TOF into drug discovery campaigns.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , High-Throughput Screening Assays/methods
16.
SLAS Discov ; 22(4): 425-432, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28328321

ABSTRACT

Autotaxin (ATX) is a promising drug target for the treatment of several diseases, such as cancer and fibrosis. ATX hydrolyzes lysophosphatidyl choline (LPC) into bioactive lysophosphatidic acid (LPA). The potency of ATX inhibitors can be readily determined by using fluorescence-based LPC derivatives. While such assays are ultra-high throughput, they are prone to false positives compared to assays based on natural LPC. Here we report the development of ultrafast mass spectrometry-based ATX assays enabling the measurement of data points within 13 s, which is 10 times faster than classic liquid chromatography-mass spectrometry. To this end, we set up a novel in vitro and whole-blood assay. We demonstrate that the potencies determined with these assays are in good agreement with the in vivo efficacy and that the whole-blood assay has the best predictive power. This high-throughput label-free approach paired with the translatable data quality is highly attractive for appropriate guidance of medicinal chemists for constructing strong structure-activity relationships.


Subject(s)
Enzyme Inhibitors/blood , High-Throughput Screening Assays , Lysophosphatidylcholines/blood , Lysophospholipids/blood , Mass Spectrometry/methods , Phosphoric Diester Hydrolases/blood , Animals , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Haplorhini , Humans , Hydrolysis , Lysophosphatidylcholines/chemistry , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/chemistry , Rats , Rats, Wistar , Recombinant Proteins/blood
17.
Bioanalysis ; 8(18): 1903-17, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27524289

ABSTRACT

AIM: The kynurenine (KYN) pathway is implicated in diseases such as cancer, psychiatric, neurodegenerative and autoimmune disorders. Measurement of KYN metabolite levels will help elucidating the involvement of the KYN pathway in the disease pathology and inform drug development. METHODOLOGY: Samples of plasma, cerebrospinal fluid or brain tissue were spiked with deuterated internal standards, processed and analyzed by LC-MS/MS; analytes were chromatographically separated by gradient elution on a C18 reversed phase analytical column without derivatization. CONCLUSION: We established an LC-MS/MS method to measure 11 molecules, namely tryptophan, KYN, 3-OH-KYN, 3-OH-anthranilic acid, quinolinic acid, picolinic acid, kynurenic acid, xanthurenic acid, serotonin, dopamine and neopterin within 5.5 min, with sufficient sensitivity to quantify these molecules in small sample volumes of plasma, cerebrospinal fluid and brain tissue.


Subject(s)
Brain/metabolism , Kynurenine/blood , Kynurenine/cerebrospinal fluid , Neopterin/blood , Neopterin/cerebrospinal fluid , Tryptophan/blood , Tryptophan/cerebrospinal fluid , Animals , Chromatography, High Pressure Liquid/methods , Humans , Kynurenine/analogs & derivatives , Kynurenine/metabolism , Mice, Inbred C57BL , Neopterin/metabolism , Quinolinic Acid/blood , Quinolinic Acid/cerebrospinal fluid , Quinolinic Acid/metabolism , Signal Transduction , Tandem Mass Spectrometry/methods , Tryptophan/metabolism , ortho-Aminobenzoates/blood , ortho-Aminobenzoates/cerebrospinal fluid , ortho-Aminobenzoates/metabolism
18.
Brain Behav Immun ; 54: 59-72, 2016 May.
Article in English | MEDLINE | ID: mdl-26724575

ABSTRACT

Psychosocial stress is a major risk factor for mood and anxiety disorders, in which excessive reactivity to aversive events/stimuli is a major psychopathology. In terms of pathophysiology, immune-inflammation is an important candidate, including high blood and brain levels of metabolites belonging to the kynurenine pathway. Animal models are needed to study causality between psychosocial stress, immune-inflammation and hyper-reactivity to aversive stimuli. The present mouse study investigated effects of psychosocial stress as chronic social defeat (CSD) versus control-handling (CON) on: Pavlovian tone-shock fear conditioning, activation of the kynurenine pathway, and efficacy of a specific inhibitor (IDOInh) of the tryptophan-kynurenine catabolising enzyme indoleamine 2,3-dioxygenase (IDO1), in reversing CSD effects on the kynurenine pathway and fear. CSD led to excessive fear learning and memory, whilst repeated oral escitalopram (antidepressant and anxiolytic) reversed excessive fear memory, indicating predictive validity of the model. CSD led to higher blood levels of TNF-α, IFN-γ, kynurenine (KYN), 3-hydroxykynurenine (3-HK) and kynurenic acid, and higher KYN and 3-HK in amygdala and hippocampus. CSD was without effect on IDO1 gene or protein expression in spleen, ileum and liver, whilst increasing liver TDO2 gene expression. Nonetheless, oral IDOInh reduced blood and brain levels of KYN and 3-HK in CSD mice to CON levels, and we therefore infer that CSD increases IDO1 activity by increasing its post-translational activation. Furthermore, repeated oral IDOInh reversed excessive fear memory in CSD mice to CON levels. IDOInh reversal of CSD-induced hyper-activity in the kynurenine pathway and fear system contributes significantly to the evidence for a causal pathway between psychosocial stress, immune-inflammation and the excessive fearfulness that is a major psychopathology in stress-related neuropsychiatric disorders.


Subject(s)
Brain/metabolism , Citalopram/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Kynurenine/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Animals , Antidepressive Agents, Second-Generation/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Brain/enzymology , Fear/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenic Acid/metabolism , Kynurenine/analogs & derivatives , Kynurenine/blood , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Stress, Psychological/enzymology , Stress, Psychological/psychology , Tryptophan/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Brain Behav Immun ; 50: 125-140, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26173174

ABSTRACT

The similarity between sickness behavior syndrome (SBS) in infection and autoimmune disorders and certain symptoms in major depressive disorder (MDD), and the high co-morbidity of autoimmune disorders and MDD, constitutes some of the major evidence for the immune-inflammation hypothesis of MDD. CD40 ligand-CD40 immune-activation is important in host response to infection and in development of autoimmunity. Mice given a single intra-peritoneal injection of CD40 agonist antibody (CD40AB) develop SBS for 2-3days characterized by weight loss and increased sleep, effects that are dependent on the cytokine, tumor necrosis factor (TNF). Here we report that CD40AB also induces behavioral effects that extend beyond acute SBS and co-occur with but are not mediated by kynurenine pathway activation and recovery. CD40AB led to decreased saccharin drinking (days 1-7) and decreased Pavlovian fear conditioning (days 5-6), and was without effect on physical fatigue (day 5). These behavioral effects co-occurred with increased plasma and brain levels of kynurenine and its metabolites (days 1-7/8). Co-injection of TNF blocker etanercept with CD40AB prevented each of SBS, reduced saccharin drinking, and kynurenine pathway activation in plasma and brain. Repeated oral administration of a selective indoleamine 2,3-dioxygenase (IDO) inhibitor blocked activation of the kynurenine pathway but was without effect on SBS and saccharin drinking. This study provides novel evidence that CD40-TNF activation induces deficits in saccharin drinking and Pavlovian fear learning and activates the kynurenine pathway, and that CD40-TNF activation of the kynurenine pathway is not necessary for induction of the acute or extended SBS effects.


Subject(s)
CD40 Antigens/immunology , CD40 Ligand/immunology , Illness Behavior/physiology , Kynurenine/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Behavior, Animal/drug effects , CD40 Antigens/agonists , CD40 Ligand/metabolism , Conditioning, Psychological/drug effects , Depressive Disorder, Major/immunology , Depressive Disorder, Major/metabolism , Drinking Behavior/drug effects , Fear/drug effects , Illness Behavior/drug effects , Kynurenine/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/immunology , Signal Transduction/drug effects , Syndrome , Tumor Necrosis Factor-alpha/metabolism
20.
J Biomol Screen ; 16(3): 370-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21335598

ABSTRACT

Over the past decade, drug discovery programs have started to address the optimization of key ADME properties already at an early stage of the process. Hence, analytical chemists have been confronted with tremendously rising sample numbers and have had to develop methodologies accelerating quantitative liquid chromatography/tandem mass spectrometry (LC/MS/MS). This article focuses on the application of a generic and fully automated LC/MS/MS, named Rapid and Integrated Analysis System (RIAS), as a high-throughput platform for the rapid quantification of drug-like compounds in various in vitro ADME assays. Previous efforts were dedicated to the setup and feasibility study of a workflow-integrated platform combining a modified high-throughput liquid handling LC/MS/MS system controlled by a customized software interface and a customized data-processing and reporting tool. Herein the authors present an extension of this previously developed basic application to a broad set of ADME screening campaigns, covering CYP inhibition, Caco-2, and PAMPA assays. The platform is capable of switching automatically between various ADME assays, performs MS compound optimization if required, and provides a speed of 8 s from sample to sample, independently of the type of ADME assay. Quantification and peak review are adopted to the high-throughput environment and tested against a standard HPLC-ESI technology.


Subject(s)
Chromatography, Liquid , Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Tandem Mass Spectrometry , Caco-2 Cells , High-Throughput Screening Assays/instrumentation , High-Throughput Screening Assays/methods , Humans , Pharmaceutical Preparations/analysis , Software , Systems Integration
SELECTION OF CITATIONS
SEARCH DETAIL
...