1
|
Metzler G, King RC, Fernández-Metzler C, Das A, Davis TG, Bajrami B, Bretschneider T, Kvaskoff D. Rapid Lipid Mediator Profiling by Convergence Chromatography-MS/MS. Methods Mol Biol 2025; 2855:341-354. [PMID: 39354317 DOI: 10.1007/978-1-0716-4116-3_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Bioactive lipid mediators derived from arachidonic acid constitute an attractive pool of metabolites that reflect cellular function and signaling, as well as potential biomarkers that may respond quantitatively to disease progression or pharmacological treatment. Their quantitative measurement in biological samples is complicated by the number of isomers that share common structural features, which are not easily distinguished by immunoassays or reverse phase chromatography-tandem mass spectrometry. Here, we present a method that enables the rapid analysis of a panel of over 25 biologically important eicosanoids in a 96-well format for cell culture supernatants, plasma, and organ tissues using convergence chromatography-tandem mass spectrometry to resolve these analytes of interest.
Collapse
Affiliation(s)
- Guille Metzler
- PharmaCadence Analytical Services, LLC, Hatfield, PA, USA
| | - Richard C King
- PharmaCadence Analytical Services, LLC, Hatfield, PA, USA
| | | | - Amitava Das
- Immunology and Respiratory Disease, Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - T Gregg Davis
- Immunology and Respiratory Disease, Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Besnik Bajrami
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Tom Bretschneider
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David Kvaskoff
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.
| |
Collapse
|
2
|
Medina A, Eon M, Mazzella N, Bonnineau C, Millan-Navarro D, Moreira A, Morin S, Creusot N. Sensitivity shift of the meta-metabolome and photosynthesis to the chemical stress in periphyton between months along one year and a half period: Case study of a terbuthylazine exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177681. [PMID: 39577586 DOI: 10.1016/j.scitotenv.2024.177681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Despite the knowledge of the effects of contaminants on periphyton, information is limited about their natural fluctuations in sensitivity to chemical stress between various months. In particular, the molecular and biochemical mechanisms associated with sensitivity of photosynthesis and its fluctuations remain poorly described. To tackle this lack of knowledge, meta-metabolomics offers a comprehensive picture of the sensitive molecular response preceding the physiological impact. This study aimed to describe changes in the sensitivity of periphyton to chemical stress at different months over one year and a half period, at both the physiological and molecular levels by measuring photosynthetic yield and meta-metabolome responses (targeted and untargeted approaches). Periphyton was colonized for four weeks and then exposed to a range of terbuthylazine concentrations (0.3-30 μg L-1) under controlled conditions for 4 h. Sensitivity was assessed by determining the benchmark doses for the meta-metabolome and photosynthesis, along with the cumulative distribution of aggregated metabolomics signals. The results showed a strong sensitivity shift in the meta-metabolome compared to a smaller shift in photosynthetic yield at different months. This study also confirmed the high sensitivity of the meta-metabolome, as most signals responded before photosynthesis. The annotation highlighted the discrepancies in the molecular response to TBA between the months in terms of metabolite classes (e.g. amino acids, alkaloids, and lipids), their sensitivity, and trends in common classes across months, and correlation to photosynthesis inhibition, notably oxylipins. Overall, this study highlights that the molecular response of the periphyton to chemical stress, and thus toxicity pathways, may differ between the months but can still lead to similar physiological responses.
Collapse
Affiliation(s)
- Arthur Medina
- INRAE Nouvelle-Aquitaine Bordeaux, UR EABX, 50 avenue de Verdun, Cestas 33612, France
| | - Melissa Eon
- INRAE Nouvelle-Aquitaine Bordeaux, UR EABX, 50 avenue de Verdun, Cestas 33612, France; Plateforme Bordeaux Metabolome, F-33140 Villenave d'Ornon, France
| | - Nicolas Mazzella
- INRAE Nouvelle-Aquitaine Bordeaux, UR EABX, 50 avenue de Verdun, Cestas 33612, France; Plateforme Bordeaux Metabolome, F-33140 Villenave d'Ornon, France
| | - Chloé Bonnineau
- INRAE Nouvelle-Aquitaine Bordeaux, UR EABX, 50 avenue de Verdun, Cestas 33612, France
| | - Débora Millan-Navarro
- INRAE Nouvelle-Aquitaine Bordeaux, UR EABX, 50 avenue de Verdun, Cestas 33612, France
| | - Aurelie Moreira
- INRAE Nouvelle-Aquitaine Bordeaux, UR EABX, 50 avenue de Verdun, Cestas 33612, France; Plateforme Bordeaux Metabolome, F-33140 Villenave d'Ornon, France
| | - Soizic Morin
- INRAE Nouvelle-Aquitaine Bordeaux, UR EABX, 50 avenue de Verdun, Cestas 33612, France
| | - Nicolas Creusot
- INRAE Nouvelle-Aquitaine Bordeaux, UR EABX, 50 avenue de Verdun, Cestas 33612, France; Plateforme Bordeaux Metabolome, F-33140 Villenave d'Ornon, France.
| |
Collapse
|
3
|
Varona M, Dobson DP, Napolitano JG, Thomas R, Ochoa JL, Russell DJ, Crittenden CM. High Resolution Ion Mobility Enables the Structural Characterization of Atropisomers of GDC-6036, a KRAS G12C Covalent Inhibitor. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:2586-2595. [PMID: 39051157 DOI: 10.1021/jasms.4c00103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
GDC-6036 is a covalent KRAS G12C inhibitor that demonstrates high potency and selectivity. Structurally, GDC-6036 consists of several motifs that make the analytical characterization of this molecule challenging, including a highly basic pyrrolidine motif bonded to a quinazoline ring via an ether bond and an atropisomeric carbon-carbon bond between functionalized pyridine and quinazoline groups. Structurally, the desired atropisomer was synthesized via an atroposelective Negishi coupling with very high yield. However, having a direct way to analyze and confirm the presence of the atropisomeric species remained challenging in routine analytical workflows. In this study, both variable temperature nuclear magnetic resonance (VT-NMR) and two different approaches of in-line ion mobility coupled to liquid chromatography mass spectrometry (LC-MS) workflows were evaluated for the characterization of GDC-6036 and its undesired atropisomer (Compound B) to support synthetic route development. Briefly, both VT-NMR and traveling wave ion mobility spectrometry (TWIMS) enabled by structures for lossless ion manipulation (SLIM) technology coupled to high resolution MS (HRMS) are able to elucidate the structures of the atropisomers in a complex mixture. Drift tube IMS (DTIMS) was also evaluated, but lacked the resolving power to demonstrate separation between the two species in a mixture, but did show slight differences in their arrival times when multiplexed and injected separately. The determined resolving power (Rp) by multiplexing the ions via DTIMS was 67.3 and 60.5 for GDC-6036 and Compound B, respectively, while the two peak resolving power (Rpp) was determined to be 0.41, indicating inadequate resolution between the two species. Alternatively, the SLIM-IM studies showed Rp of 103.8 and 99.4, with a Rpp of 2.64, indicating good separation between the atropisomers. Furthermore, the CCS/z for GDC-6036 and Compound B was determined to be 231.2 Å2/z and 235.0 Å2/z, respectively. Quantitative experiments demonstrate linearity (R2 >0.99) for both GDC-6036 and Compound B while maintaining separation via SLIM-IM. Spike recoveries of one atropisomer relative to the other yielded strong recoveries (98.7% to 102.5%) while maintaining reproducibility (<7% RSD). The study herein describes the analytical process for evaluating new technologies and strategies for implementation in routine biopharmaceutical characterization workflows.
Collapse
Affiliation(s)
- Marcelino Varona
- Synthetic Molecule Analytical Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel P Dobson
- Synthetic Molecule Analytical Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - José G Napolitano
- Synthetic Molecule Analytical Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rekha Thomas
- Synthetic Molecule Analytical Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jessica L Ochoa
- Synthetic Molecule Analytical Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - David J Russell
- Synthetic Molecule Analytical Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Christopher M Crittenden
- Synthetic Molecule Analytical Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
4
|
Ettedgui J, Yamamoto K, Blackman B, Koyasu N, Raju N, Vasalatiy O, Merkle H, Chekmenev EY, Goodson BM, Krishna MC, Swenson RE. In vivo Metabolic Sensing of Hyperpolarized [1- 13C]Pyruvate in Mice Using a Recyclable Perfluorinated Iridium Signal Amplification by Reversible Exchange Catalyst. Angew Chem Int Ed Engl 2024; 63:e202407349. [PMID: 38829568 PMCID: PMC11610520 DOI: 10.1002/anie.202407349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/05/2024]
Abstract
Real-time visualization of metabolic processes in vivo provides crucial insights into conditions like cancer and metabolic disorders. Metabolic magnetic resonance imaging (MRI), by amplifying the signal of pyruvate molecules through hyperpolarization, enables non-invasive monitoring of metabolic fluxes, aiding in understanding disease progression and treatment response. Signal Amplification By Reversible Exchange (SABRE) presents a simpler, cost-effective alternative to dissolution dynamic nuclear polarization, eliminating the need for expensive equipment and complex procedures. We present the first in vivo demonstration of metabolic sensing in a human pancreatic cancer xenograft model compared to healthy mice. A novel perfluorinated Iridium SABRE catalyst in a fluorinated solvent and methanol blend facilitated this breakthrough with a 1.2-fold increase in [1-13C]pyruvate SABRE hyperpolarization. The perfluorinated moiety allowed easy separation of the heavy-metal-containing catalyst from the hyperpolarized [1-13C]pyruvate target. The perfluorinated catalyst exhibited recyclability, maintaining SABRE-SHEATH activity through subsequent hyperpolarization cycles with minimal activity loss after the initial two cycles. Remarkably, the catalyst retained activity for at least 10 cycles, with a 3.3-fold decrease in hyperpolarization potency. This proof-of-concept study encourages wider adoption of SABRE hyperpolarized [1-13C]pyruvate MR for studying in vivo metabolism, aiding in diagnosing stages and monitoring treatment responses in cancer and other diseases.
Collapse
Affiliation(s)
- Jessica Ettedgui
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, 9800 Medical Center Drive, Rockville, Maryland, 20850, United States
| | - Kazutoshi Yamamoto
- Center for Cancer Research, National Cancer Institute, Bethesda, 10 Center Drive Maryland, 20814, United States
| | - Burchelle Blackman
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, 9800 Medical Center Drive, Rockville, Maryland, 20850, United States
| | - Norikazu Koyasu
- Center for Cancer Research, National Cancer Institute, Bethesda, 10 Center Drive Maryland, 20814, United States
| | - Natarajan Raju
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, 9800 Medical Center Drive, Rockville, Maryland, 20850, United States
| | - Olga Vasalatiy
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, 9800 Medical Center Drive, Rockville, Maryland, 20850, United States
| | - Hellmut Merkle
- National Institute of Neurological Disorders and Stroke, Bethesda, 10 Center Drive Maryland, 20814, United States
| | - Eduard Y Chekmenev
- Department of Chemistry, Integrative Biosciences (Ibio), Karmanos Cancer Institute (KCI), Wayne State University, Detroit, Michigan, 48202, United States
| | - Boyd M Goodson
- School of Chemical & Biomolecular Sciences and Materials Technology Center, Southern Illinois University, Carbondale, Illinois, 62901, United States
| | - Murali C Krishna
- Center for Cancer Research, National Cancer Institute, Bethesda, 10 Center Drive Maryland, 20814, United States
| | - Rolf E Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, 9800 Medical Center Drive, Rockville, Maryland, 20850, United States
| |
Collapse
|
5
|
Garcia B, Becker CC, Weber L, Swarr GJ, Kido Soule MC, Apprill A, Kujawinski EB. Benzoyl Chloride Derivatization Advances the Quantification of Dissolved Polar Metabolites on Coral Reefs. J Proteome Res 2024; 23:2041-2053. [PMID: 38782401 PMCID: PMC11166142 DOI: 10.1021/acs.jproteome.4c00049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Extracellular chemical cues constitute much of the language of life among marine organisms, from microbes to mammals. Changes in this chemical pool serve as invisible signals of overall ecosystem health and disruption to this finely tuned equilibrium. In coral reefs, the scope and magnitude of the chemicals involved in maintaining reef equilibria are largely unknown. Processes involving small, polar molecules, which form the majority components of labile dissolved organic carbon, are often poorly captured using traditional techniques. We employed chemical derivatization with mass spectrometry-based targeted exometabolomics to quantify polar dissolved phase metabolites on five coral reefs in the U.S. Virgin Islands. We quantified 45 polar exometabolites, demonstrated their spatial variability, and contextualized these findings in terms of geographic and benthic cover differences. By comparing our results to previously published coral reef exometabolomes, we show the novel quantification of 23 metabolites, including central carbon metabolism compounds (e.g., glutamate) and novel metabolites such as homoserine betaine. We highlight the immense potential of chemical derivatization-based exometabolomics for quantifying labile chemical cues on coral reefs and measuring molecular level responses to environmental stressors. Overall, improving our understanding of the composition and dynamics of reef exometabolites is vital for effective ecosystem monitoring and management strategies.
Collapse
Affiliation(s)
- Brianna
M. Garcia
- Department of Marine Chemistry
and Geochemistry, Woods Hole Oceanographic
Institution, Woods
Hole, Massachusetts 02543, United States
| | | | - Laura Weber
- Department of Marine Chemistry
and Geochemistry, Woods Hole Oceanographic
Institution, Woods
Hole, Massachusetts 02543, United States
| | - Gretchen J. Swarr
- Department of Marine Chemistry
and Geochemistry, Woods Hole Oceanographic
Institution, Woods
Hole, Massachusetts 02543, United States
| | - Melissa C. Kido Soule
- Department of Marine Chemistry
and Geochemistry, Woods Hole Oceanographic
Institution, Woods
Hole, Massachusetts 02543, United States
| | - Amy Apprill
- Department of Marine Chemistry
and Geochemistry, Woods Hole Oceanographic
Institution, Woods
Hole, Massachusetts 02543, United States
| | - Elizabeth B. Kujawinski
- Department of Marine Chemistry
and Geochemistry, Woods Hole Oceanographic
Institution, Woods
Hole, Massachusetts 02543, United States
| |
Collapse
|
6
|
Soh WT, Roetschke HP, Cormican JA, Teo BF, Chiam NC, Raabe M, Pflanz R, Henneberg F, Becker S, Chari A, Liu H, Urlaub H, Liepe J, Mishto M. Protein degradation by human 20S proteasomes elucidates the interplay between peptide hydrolysis and splicing. Nat Commun 2024; 15:1147. [PMID: 38326304 PMCID: PMC10850103 DOI: 10.1038/s41467-024-45339-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 01/17/2024] [Indexed: 02/09/2024] Open
Abstract
If and how proteasomes catalyze not only peptide hydrolysis but also peptide splicing is an open question that has divided the scientific community. The debate has so far been based on immunopeptidomics, in vitro digestions of synthetic polypeptides as well as ex vivo and in vivo experiments, which could only indirectly describe proteasome-catalyzed peptide splicing of full-length proteins. Here we develop a workflow-and cognate software - to analyze proteasome-generated non-spliced and spliced peptides produced from entire proteins and apply it to in vitro digestions of 15 proteins, including well-known intrinsically disordered proteins such as human tau and α-Synuclein. The results confirm that 20S proteasomes produce a sizeable variety of cis-spliced peptides, whereas trans-spliced peptides are a minority. Both peptide hydrolysis and splicing produce peptides with well-defined characteristics, which hint toward an intricate regulation of both catalytic activities. At protein level, both non-spliced and spliced peptides are not randomly localized within protein sequences, but rather concentrated in hotspots of peptide products, in part driven by protein sequence motifs and proteasomal preferences. At sequence level, the different peptide sequence preference of peptide hydrolysis and peptide splicing suggests a competition between the two catalytic activities of 20S proteasomes during protein degradation.
Collapse
Affiliation(s)
- Wai Tuck Soh
- Research Group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Hanna P Roetschke
- Research Group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
- Centre for Inflammation Biology and Cancer Immunology & Peter Gorer Department of Immunobiology, King's College London, SE1 1UL, London, UK
- Research Group of Molecular Immunology, Francis Crick Institute, NW1 1AT, London, UK
| | - John A Cormican
- Research Group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Bei Fang Teo
- Centre for Inflammation Biology and Cancer Immunology & Peter Gorer Department of Immunobiology, King's College London, SE1 1UL, London, UK
- Research Group of Molecular Immunology, Francis Crick Institute, NW1 1AT, London, UK
- Immunology Programme, Life Sciences Institute; Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Nyet Cheng Chiam
- Research Group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Monika Raabe
- Research Group of Bioanalytical Mass Spectrometry, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Ralf Pflanz
- Research Group of Bioanalytical Mass Spectrometry, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Fabian Henneberg
- Department of Structural Dynamics, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Stefan Becker
- Department of NMR-based Structural Biology, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Ashwin Chari
- Research Group of Structural Biochemistry and Mechanisms, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute; Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Henning Urlaub
- Research Group of Bioanalytical Mass Spectrometry, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
- Institute of Clinical Chemistry, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Juliane Liepe
- Research Group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany.
| | - Michele Mishto
- Centre for Inflammation Biology and Cancer Immunology & Peter Gorer Department of Immunobiology, King's College London, SE1 1UL, London, UK.
- Research Group of Molecular Immunology, Francis Crick Institute, NW1 1AT, London, UK.
| |
Collapse
|
7
|
Cramer DAT, Yin V, Caval T, Franc V, Yu D, Wu G, Lloyd G, Langendorf C, Whisstock JC, Law RHP, Heck AJR. Proteoform-Resolved Profiling of Plasminogen Activation Reveals Novel Abundant Phosphorylation Site and Primary N-Terminal Cleavage Site. Mol Cell Proteomics 2024; 23:100696. [PMID: 38101751 PMCID: PMC10825491 DOI: 10.1016/j.mcpro.2023.100696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
Plasminogen (Plg), the zymogen of plasmin (Plm), is a glycoprotein involved in fibrinolysis and a wide variety of other physiological processes. Plg dysregulation has been implicated in a range of diseases. Classically, human Plg is categorized into two types, supposedly having different functional features, based on the presence (type I) or absence (type II) of a single N-linked glycan. Using high-resolution native mass spectrometry, we uncovered that the proteoform profiles of human Plg (and Plm) are substantially more extensive than this simple binary classification. In samples derived from human plasma, we identified up to 14 distinct proteoforms of Plg, including a novel highly stoichiometric phosphorylation site at Ser339. To elucidate the potential functional effects of these post-translational modifications, we performed proteoform-resolved kinetic analyses of the Plg-to-Plm conversion using several canonical activators. This conversion is thought to involve at least two independent cleavage events: one to remove the N-terminal peptide and another to release the active catalytic site. Our analyses reveal that these processes are not independent but are instead tightly regulated and occur in a step-wise manner. Notably, N-terminal cleavage at the canonical site (Lys77) does not occur directly from intact Plg. Instead, an activation intermediate corresponding to cleavage at Arg68 is initially produced, which only then is further processed to the canonical Lys77 product. Based on our results, we propose a refined categorization for human Plg proteoforms. In addition, we reveal that the proteoform profile of human Plg is more extensive than that of rat Plg, which lacks, for instance, the here-described phosphorylation at Ser339.
Collapse
Affiliation(s)
- Dario A T Cramer
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands
| | - Victor Yin
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands
| | - Tomislav Caval
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands
| | - Vojtech Franc
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands
| | - Dingyi Yu
- Mass Spectrometry Facility, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Guojie Wu
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Gordon Lloyd
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Christopher Langendorf
- Mass Spectrometry Facility, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - James C Whisstock
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Ruby H P Law
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia.
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
8
|
Lee HJ, Jeon M, Seo Y, Kang I, Jeong W, Son J, Yi EC, Min H. Application of Skyline software for detecting prohibited substances in doping control analysis. PLoS One 2023; 18:e0295065. [PMID: 38051722 DOI: 10.1371/journal.pone.0295065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 11/14/2023] [Indexed: 12/07/2023] Open
Abstract
As the number of prohibited drugs has been progressively increasing and analytical methods for detecting such substances are renewed continuously for doping control, the need for more sensitive and accurate doping analysis has increased. To address the urgent need for high throughput and accurate analysis, liquid chromatography with tandem mass spectrometry is actively utilized in case of most of the newly designated prohibited substances. However, because all mass spectrometer vendors provide data processing software that is incapable of handling other instrumental data, it is difficult to cover all doping analysis procedures, from method development to result reporting, on one platform. Skyline is an open-source and vendor-neutral software program invented for the method development and data processing of targeted proteomics. Recently, the utilization of Skyline has been expanding for the quantitative analysis of small molecules and lipids. Herein, we demonstrated Skyline as a simple platform for unifying overall doping control, including the optimization of analytical methods, monitoring of data quality, discovery of suspected doping samples, and validation of analytical methods for detecting newly prohibited substances. For method optimization, we selected the optimal collision energies for 339 prohibited substances. Notably, 195 substances exhibited a signal intensity increase of >110% compared with the signal intensity of the original collision energy. All data related to method validation and quantitative analysis were efficiently visualized, extracted, or calculated using Skyline. Moreover, a comparison of the time consumed and the number of suspicious samples screened in the initial test procedure highlighted the advantages of using Skyline over the commercially available software TraceFinder in doping control.
Collapse
Affiliation(s)
- Hyeon-Jeong Lee
- Doping Control Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Yeongeon-dong, Jongno-gu, Seoul, Republic of Korea
| | - Mijin Jeon
- Doping Control Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul, Republic of Korea
| | - Yoondam Seo
- Doping Control Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul, Republic of Korea
| | - Inseon Kang
- Doping Control Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul, Republic of Korea
| | - Wooyeon Jeong
- Doping Control Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul, Republic of Korea
| | - Junghyun Son
- Doping Control Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul, Republic of Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Yeongeon-dong, Jongno-gu, Seoul, Republic of Korea
| | - Hophil Min
- Doping Control Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul, Republic of Korea
| |
Collapse
|
9
|
Macur K, Schissel A, Yu F, Lei S, Morsey B, Fox HS, Ciborowski P. Change of histone H3 lysine 14 acetylation stoichiometry in human monocyte derived macrophages as determined by MS-based absolute targeted quantitative proteomic approach: HIV infection and methamphetamine exposure. Clin Proteomics 2023; 20:48. [PMID: 37880620 PMCID: PMC10599040 DOI: 10.1186/s12014-023-09438-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/13/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Histones posttranslational modification represent an epigenetic mechanism that regulate gene expression and other cellular processes. Quantitative mass spectrometry used for the absolute quantification of such modifications provides further insight into cellular responses to extracellular insults such as infections or toxins. Methamphetamine (Meth), a drug of abuse, is affecting the overall function of the immune system. In this report, we developed, validated and applied a targeted, MS-based quantification assay to measure changes in histone H3 lysine 14 acetylation (H3K14Ac) during exposure of human primary macrophages to HIV-1 infection and/or Meth. METHODS The quantification assay was developed and validated to determine H3K14Ac stoichiometry in histones that were isolated from the nuclei of control (CIC) and exposed to Meth before (CIM) or/and after (MIM) HIV-infection human monocyte-derived macrophages (hMDM) of six donors. It was based on LC-MS/MS measurement using multiple reaction monitoring (MRM) acquisition of the unmodified and acetylated form of lysine K14 of histone H3 9KSTGGKAPR17 peptides and the corresponding stable isotope labeled (SIL) heavy peptide standards of the same sequences. The histone samples were propionylated (Poy) pre- and post- trypsin digestion so that the sequences of the monitored peptides were: K[Poy]STGGK[1Ac]APR, K[Poy]STGGK[1Ac]APR-heavy, K[Poy]STGGK[Poy]APR and K[Poy]STGGK[Poy]APR-heavy. The absolute amounts of the acetylated and unmodified peptides were determined by comparing to the abundances of their SIL standards, that were added to the samples in the known concentrations, and, then used for calculation of H3K14Ac stoichiometry in CIC, CIM and MIM hMDM. RESULTS The assay was characterized by LLOD of 0.106 fmol/µL and 0.204 fmol/µL for unmodified and acetylated H3 9KSTGGKAPR17 peptides, respectively. The LLOQ was 0.5 fmol/µL and the linear range of the assay was from 0.5 to 2500 fmol/µL. The absolute abundances of the quantified peptides varied between the donors and conditions, and so did the H3K14Ac stoichiometry. This was rather attributed to the samples nature itself, as the variability of their triplicate measurements was low. CONCLUSIONS The developed LC-MS/MS assay enabled absolute quantification of H3K14Ac in exposed to Meth HIV-infected hMDM. It can be further applied determination of this PTM stoichiometry in other studies on human primary macrophages.
Collapse
Affiliation(s)
- Katarzyna Macur
- Core Facility Laboratories, Intercollegiate Faculty of Biotechnology UG & MUG, University of Gdańsk, Gdańsk, Poland.
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Andrew Schissel
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shulei Lei
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brenda Morsey
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pawel Ciborowski
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
10
|
Qiao W, Chen Y, Zhong J, Madden BJ, Charlesworth CM, Martens YA, Liu CC, Knight J, Ikezu TC, Kurti A, Zhu Y, Meneses A, Rosenberg CL, Kuchenbecker LA, Vanmaele LK, Li F, Chen K, Shue F, Dacquel MV, Fryer J, Pandey A, Zhao N, Bu G. Trem2 H157Y increases soluble TREM2 production and reduces amyloid pathology. Mol Neurodegener 2023; 18:8. [PMID: 36721205 PMCID: PMC9890893 DOI: 10.1186/s13024-023-00599-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/19/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The rare p.H157Y variant of TREM2 (Triggering Receptor Expressed on Myeloid Cells 2) was found to increase Alzheimer's disease (AD) risk. This mutation is located at the cleavage site of TREM2 extracellular domain. Ectopic expression of TREM2-H157Y in HEK293 cells resulted in increased TREM2 shedding. However, the physiological outcomes of the TREM2 H157Y mutation remain unknown in the absence and presence of AD related pathologies. METHODS We generated a novel Trem2 H157Y knock-in mouse model through CRISPR/Cas9 technology and investigated the effects of Trem2 H157Y on TREM2 proteolytic processing, synaptic function, and AD-related amyloid pathologies by conducting biochemical assays, targeted mass spectrometry analysis of TREM2, hippocampal electrophysiology, immunofluorescent staining, in vivo micro-dialysis, and cortical bulk RNA sequencing. RESULTS Consistent with previous in vitro findings, Trem2 H157Y increases TREM2 shedding with elevated soluble TREM2 levels in the brain and serum. Moreover, Trem2 H157Y enhances synaptic plasticity without affecting microglial density and morphology, or TREM2 signaling. In the presence of amyloid pathology, Trem2 H157Y accelerates amyloid-β (Aβ) clearance and reduces amyloid burden, dystrophic neurites, and gliosis in two independent founder lines. Targeted mass spectrometry analysis of TREM2 revealed higher ratios of soluble to full-length TREM2-H157Y compared to wild-type TREM2, indicating that the H157Y mutation promotes TREM2 shedding in the presence of Aβ. TREM2 signaling was further found reduced in Trem2 H157Y homozygous mice. Transcriptomic profiling revealed that Trem2 H157Y downregulates neuroinflammation-related genes and an immune module correlated with the amyloid pathology. CONCLUSION Taken together, our findings suggest beneficial effects of the Trem2 H157Y mutation in synaptic function and in mitigating amyloid pathology. Considering the genetic association of TREM2 p.H157Y with AD risk, we speculate TREM2 H157Y in humans might increase AD risk through an amyloid-independent pathway, such as its effects on tauopathy and neurodegeneration which merit further investigation.
Collapse
Affiliation(s)
- Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Jun Zhong
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905 USA
| | - Benjamin J. Madden
- Medical Genome Facility, Proteomics Core, Mayo Clinic, Rochester, MN USA
| | | | - Yuka A. Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Joshua Knight
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | | | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Yiyang Zhu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Axel Meneses
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | | | | | - Lucy K. Vanmaele
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Fuyao Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Kai Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | | | - John Fryer
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ 85259 USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905 USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN USA
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104 India
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
- SciNeuro Pharmaceuticals, Rockville, MD 20805 USA
| |
Collapse
|
11
|
Multiplexed quantification of insulin and C-peptide by LC-MS/MS without the use of antibodies. J Mass Spectrom Adv Clin Lab 2022; 25:19-26. [PMID: 35734440 PMCID: PMC9207678 DOI: 10.1016/j.jmsacl.2022.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 11/30/2022] Open
Abstract
Simultaneous quantification of insulin and C-peptide without antibodies. Proteolysis with Glu-C permits sensitive and precise measurements. Calibration with certified reference material provides traceability. Relatively large bias when compared with a commercially available immunoassay.
Introduction The measurement of insulin and C-peptide provides a valuable tool for the clinical evaluation of hypoglycemia. In research, these biomarkers are used together to better understand hyperinsulinemia, hepatic insulin clearance, and beta cell function. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is an attractive approach for the analysis of insulin and C-peptide because the platform is specific, can avoid certain limitations of immunoassays, and can be multiplexed. Previously described LC-MS/MS methods for the simultaneous quantification of insulin and C-peptide measure the intact analytes and most have relied on immunoaffinity enrichment. These approaches can be limited in terms of sensitivity and interference from auto-antibodies, respectively. We have developed a novel method that does not require antibodies and uses proteolytic digestion to yield readily ionizable proteotypic peptides that enables the sensitive, specific, and simultaneous quantitation of insulin and C-peptide. Methods Serum samples were precipitated with acetonitrile. Analytes were enriched using solid phase extraction and then digested with endoproteinase Glu-C. Surrogate peptides for insulin and C-peptide were analyzed using targeted LC-MS/MS. Results Inter-day imprecision was below 20 %CV and linearity was observed down to the lower limit of quantitation for both analytes (insulin = 0.09 ng/mL, C-peptide = 0.06 ng/mL). Comparison to a commercially available insulin immunoassay (Beckman Coulter UniCel DxI 600 Access) revealed a 30% bias between methods. Conclusion A novel LC-MS/MS method for the simultaneous analysis of insulin and C-peptide using Glu-C digestion was developed and evaluated. A detailed standard operating procedure is provided to help facilitate implementation in other laboratories.
Collapse
|
12
|
Taylor AL, Davis DE, Codreanu SG, Harrison FE, Sherrod SD, McLean JA. Targeted and Untargeted Mass Spectrometry Reveals the Impact of High-Fat Diet on Peripheral Amino Acid Regulation in a Mouse Model of Alzheimer's Disease. J Proteome Res 2021; 20:4405-4414. [PMID: 34382806 DOI: 10.1021/acs.jproteome.1c00344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent research regarding amino acid metabolism has shown that there may be a link between obesity and Alzheimer's disease (AD). This work reports a metabolomics study using targeted and untargeted mass spectrometry-based metabolomic strategies to investigate this link. Targeted hydrophilic interaction liquid chromatography-triple quadrupole mass spectrometry and untargeted reversed-phase liquid chromatography-high resolution tandem mass spectrometry assays were developed to analyze the metabolic changes that occur in AD and obesity. APPSwe/PS1ΔE9 (APP/PSEN1) transgenic mice (to represent familial or early-onset AD) and wild-type littermate controls were fed either a high-fat diet (HFD, 60% kcal from lard) or a low-fat diet (LFD, 10% kcal from lard) from 2 months of age or a reversal diet (HFD, followed by LFD from 9.5 months). For targeted analyses, we applied the guidelines outlined in the Clinical and Laboratory Standards Institute (CLSI) LC-MS C62-A document and the U.S. Food and Drug Administration (FDA) bioanalytical method validation guidance for industry to evaluate the figures of merit of the assays. Our targeted and untargeted metabolomics results suggest that numerous peripheral pathways, specifically amino acid metabolism and fatty acid metabolism, were significantly affected by AD and diet. Multiple amino acids (including alanine, glutamic acid, leucine, isoleucine, and phenylalanine), carnitines, and members of the fatty acid oxidation pathway were significantly increased in APP/PSEN1 mice on HFD compared to those on LFD. More substantial effects and changes were observed in the APP/PSEN1 mice than in the WT mice, suggesting that they were more sensitive to an HFD. These dysregulated peripheral pathways include numerous amino acid pathways and fatty acid beta oxidation and suggest that obesity combined with AD further enhances cognitive impairment, possibly through aggravated mitochondrial dysfunction. Furthermore, partial reversibility of many altered pathways was observed, which highlights that diet change can mitigate the metabolic effects of AD. The same trends in individual amino acids were observed in both strategies, highlighting the biological validity of the results.
Collapse
Affiliation(s)
- Amelia L Taylor
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Don E Davis
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Simona G Codreanu
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Fiona E Harrison
- Vanderbilt University Medical Center, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee 37235, United States
| | - Stacy D Sherrod
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - John A McLean
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
13
|
Davis DE, Leaptrot KL, Koomen DC, May JC, Cavalcanti GDA, Padilha MC, Pereira HMG, McLean JA. Multidimensional Separations of Intact Phase II Steroid Metabolites Utilizing LC-Ion Mobility-HRMS. Anal Chem 2021; 93:10990-10998. [PMID: 34319704 DOI: 10.1021/acs.analchem.1c02163] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The detection and unambiguous identification of anabolic-androgenic steroid metabolites are essential in clinical, forensic, and antidoping analyses. Recently, sulfate phase II steroid metabolites have received increased attention in steroid metabolism and drug testing. In large part, this is because phase II steroid metabolites are excreted for an extended time, making them a potential long-term chemical marker of choice for tracking steroid misuse in sports. Comprehensive analytical methods, such as liquid chromatography-tandem mass spectrometry (LC-MS/MS), have been used to detect and identify glucuronide and sulfate steroids in human urine with high sensitivity and reliability. However, LC-MS/MS identification strategies can be hindered by the fact that phase II steroid metabolites generate nonselective ion fragments across the different metabolite markers, limiting the confidence in metabolite identifications that rely on exact mass measurement and MS/MS information. Additionally, liquid chromatography-high-resolution mass spectrometry (LC-HRMS) is sometimes insufficient at fully resolving the analyte peaks from the sample matrix (commonly urine) chemical noise, further complicating accurate identification efforts. Therefore, we developed a liquid chromatography-ion mobility-high resolution mass spectrometry (LC-IM-HRMS) method to increase the peak capacity and utilize the IM-derived collision cross section (CCS) values as an additional molecular descriptor for increased selectivity and to improve identifications of intact steroid analyses at low concentrations.
Collapse
Affiliation(s)
- Don E Davis
- Department of Chemistry, Center for Innovative Technology, Institute of Chemical Biology, Institute for Integrative Biosystems Research and Education, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Katrina L Leaptrot
- Department of Chemistry, Center for Innovative Technology, Institute of Chemical Biology, Institute for Integrative Biosystems Research and Education, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - David C Koomen
- Department of Chemistry, Center for Innovative Technology, Institute of Chemical Biology, Institute for Integrative Biosystems Research and Education, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jody C May
- Department of Chemistry, Center for Innovative Technology, Institute of Chemical Biology, Institute for Integrative Biosystems Research and Education, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Gustavo de A Cavalcanti
- Brazilian Doping Control Laboratory (LBCD), Chemistry Institute, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Monica C Padilha
- Brazilian Doping Control Laboratory (LBCD), Chemistry Institute, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Henrique M G Pereira
- Brazilian Doping Control Laboratory (LBCD), Chemistry Institute, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - John A McLean
- Department of Chemistry, Center for Innovative Technology, Institute of Chemical Biology, Institute for Integrative Biosystems Research and Education, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
14
|
Gomez-Zepeda D, Frausto M, Nájera-González HR, Herrera-Estrella L, Ordaz-Ortiz JJ. Mass spectrometry-based quantification and spatial localization of small organic acid exudates in plant roots under phosphorus deficiency and aluminum toxicity. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2021; 106:1791-1806. [PMID: 33797826 DOI: 10.1111/tpj.15261] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/26/2021] [Indexed: 06/12/2023]
Abstract
Low-molecular-weight organic acid (OA) extrusion by plant roots is critical for plant nutrition, tolerance to cations toxicity, and plant-microbe interactions. Therefore, methodologies for the rapid and precise quantification of OAs are necessary to be incorporated in the analysis of roots and their exudates. The spatial location of root exudates is also important to understand the molecular mechanisms directing OA production and release into the rhizosphere. Here, we report the development of two complementary methodologies for OA determination, which were employed to evaluate the effect of inorganic ortho-phosphate (Pi) deficiency and aluminum toxicity on OA excretion by Arabidopsis roots. OA exudation by roots is considered a core response to different types of abiotic stress and for the interaction of roots with soil microbes, and for decades has been a target trait to produce plant varieties with increased capacities of Pi uptake and Al tolerance. Using targeted ultra-performance liquid chromatography coupled with high-resolution tandem mass spectrometry (UPLC-HRMS/MS), we achieved the quantification of six OAs in root exudates at sub-micromolar detection limits with an analysis time of less than 5 min per sample. We also employed targeted (MS/MS) matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) to detect the spatial location of citric and malic acid with high specificity in roots and exudates. Using these methods, we studied OA exudation in response to Al toxicity and Pi deficiency in Arabidopsis seedlings overexpressing genes involved in OA excretion. Finally, we show the transferability of the MALDI-MSI method by analyzing OA excretion in Marchantia polymorpha gemmalings subjected to Pi deficiency.
Collapse
Affiliation(s)
- David Gomez-Zepeda
- Unidad de Genómica Avanzada, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Irapuato, 36824, Mexico
| | - Moises Frausto
- Unidad de Genómica Avanzada, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Irapuato, 36824, Mexico
- Institute of Genomics for Crop Abiotic Stress Tolerance, Department of Plant and Soil Science, Texas Tech University, Lubbock, TX, 79409, USA
| | - Héctor-Rogelio Nájera-González
- Unidad de Genómica Avanzada, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Irapuato, 36824, Mexico
| | - Luis Herrera-Estrella
- Unidad de Genómica Avanzada, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Irapuato, 36824, Mexico
- Institute of Genomics for Crop Abiotic Stress Tolerance, Department of Plant and Soil Science, Texas Tech University, Lubbock, TX, 79409, USA
| | - José-Juan Ordaz-Ortiz
- Unidad de Genómica Avanzada, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Irapuato, 36824, Mexico
| |
Collapse
|
15
|
Widner B, Kido Soule MC, Ferrer-González FX, Moran MA, Kujawinski EB. Quantification of Amine- and Alcohol-Containing Metabolites in Saline Samples Using Pre-extraction Benzoyl Chloride Derivatization and Ultrahigh Performance Liquid Chromatography Tandem Mass Spectrometry (UHPLC MS/MS). Anal Chem 2021; 93:4809-4817. [PMID: 33689314 DOI: 10.1021/acs.analchem.0c03769] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Dissolved metabolites serve as nutrition, energy, and chemical signals for microbial systems. However, the full scope and magnitude of these processes in marine systems are unknown, largely due to insufficient methods, including poor extraction of small, polar compounds using common solid-phase extraction resins. Here, we utilized pre-extraction derivatization and ultrahigh performance liquid chromatography electrospray ionization tandem mass spectrometry (UHPLC-ESI-MS/MS) to detect and quantify targeted dissolved metabolites in seawater and saline culture media. Metabolites were derivatized with benzoyl chloride by their primary and secondary amine and alcohol functionalities and quantified using stable isotope-labeled internal standards (SIL-ISs) produced from 13C6-labeled benzoyl chloride. We optimized derivatization, extraction, and sample preparation for field and culture samples and evaluated matrix-derived biases. We have optimized this quantitative method for 73 common metabolites, of which 50 cannot be quantified without derivatization due to low extraction efficiencies. Of the 73 metabolites, 66 were identified in either culture media or seawater and 45 of those were quantified. This derivatization method is sensitive (detection limits = pM to nM), rapid (∼5 min per sample), and high throughput.
Collapse
Affiliation(s)
- Brittany Widner
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, United States
| | - Melissa C Kido Soule
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, United States
| | | | - Mary Ann Moran
- Department of Marine Sciences, University of Georgia, Athens, Georgia 30602, United States
| | - Elizabeth B Kujawinski
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, United States
| |
Collapse
|
16
|
Fu J, Luo Y, Mou M, Zhang H, Tang J, Wang Y, Zhu F. Advances in Current Diabetes Proteomics: From the Perspectives of Label- free Quantification and Biomarker Selection. Curr Drug Targets 2021; 21:34-54. [PMID: 31433754 DOI: 10.2174/1389450120666190821160207] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Due to its prevalence and negative impacts on both the economy and society, the diabetes mellitus (DM) has emerged as a worldwide concern. In light of this, the label-free quantification (LFQ) proteomics and diabetic marker selection methods have been applied to elucidate the underlying mechanisms associated with insulin resistance, explore novel protein biomarkers, and discover innovative therapeutic protein targets. OBJECTIVE The purpose of this manuscript is to review and analyze the recent computational advances and development of label-free quantification and diabetic marker selection in diabetes proteomics. METHODS Web of Science database, PubMed database and Google Scholar were utilized for searching label-free quantification, computational advances, feature selection and diabetes proteomics. RESULTS In this study, we systematically review the computational advances of label-free quantification and diabetic marker selection methods which were applied to get the understanding of DM pathological mechanisms. Firstly, different popular quantification measurements and proteomic quantification software tools which have been applied to the diabetes studies are comprehensively discussed. Secondly, a number of popular manipulation methods including transformation, pretreatment (centering, scaling, and normalization), missing value imputation methods and a variety of popular feature selection techniques applied to diabetes proteomic data are overviewed with objective evaluation on their advantages and disadvantages. Finally, the guidelines for the efficient use of the computationbased LFQ technology and feature selection methods in diabetes proteomics are proposed. CONCLUSION In summary, this review provides guidelines for researchers who will engage in proteomics biomarker discovery and by properly applying these proteomic computational advances, more reliable therapeutic targets will be found in the field of diabetes mellitus.
Collapse
Affiliation(s)
- Jianbo Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hongning Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jing Tang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| |
Collapse
|
17
|
Varela-Rodríguez H, Abella-Quintana DG, Espinal-Centeno A, Varela-Rodríguez L, Gomez-Zepeda D, Caballero-Pérez J, García-Medel PL, Brieba LG, Ordaz-Ortiz JJ, Cruz-Ramirez A. Functional Characterization of the Lin28/let-7 Circuit During Forelimb Regeneration in Ambystoma mexicanum and Its Influence on Metabolic Reprogramming. Front Cell Dev Biol 2020; 8:562940. [PMID: 33330447 PMCID: PMC7710800 DOI: 10.3389/fcell.2020.562940] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 10/27/2020] [Indexed: 12/31/2022] Open
Abstract
The axolotl (Ambystoma mexicanum) is a caudate amphibian, which has an extraordinary ability to restore a wide variety of damaged structures by a process denominated epimorphosis. While the origin and potentiality of progenitor cells that take part during epimorphic regeneration are known to some extent, the metabolic changes experienced and their associated implications, remain unexplored. However, a circuit with a potential role as a modulator of cellular metabolism along regeneration is that formed by Lin28/let-7. In this study, we report two Lin28 paralogs and eight mature let-7 microRNAs encoded in the axolotl genome. Particularly, in the proliferative blastema stage amxLin28B is more abundant in the nuclei of blastemal cells, while the microRNAs amx-let-7c and amx-let-7a are most downregulated. Functional inhibition of Lin28 factors increase the levels of most mature let-7 microRNAs, consistent with an increment of intermediary metabolites of the Krebs cycle, and phenotypic alterations in the outgrowth of the blastema. In summary, we describe the primary components of the Lin28/let-7 circuit and their function during axolotl regeneration, acting upstream of metabolic reprogramming events.
Collapse
Affiliation(s)
- Hugo Varela-Rodríguez
- Molecular and Developmental Complexity Group, Unidad de Genómica Avanzada (LANGEBIO), Centro de Investigación y de Estudios Avanzados del IPN, Guanajuato, Mexico
| | - Diana G Abella-Quintana
- Molecular and Developmental Complexity Group, Unidad de Genómica Avanzada (LANGEBIO), Centro de Investigación y de Estudios Avanzados del IPN, Guanajuato, Mexico
| | - Annie Espinal-Centeno
- Molecular and Developmental Complexity Group, Unidad de Genómica Avanzada (LANGEBIO), Centro de Investigación y de Estudios Avanzados del IPN, Guanajuato, Mexico
| | | | - David Gomez-Zepeda
- Mass Spectrometry and Metabolomics Laboratory, Unidad de Genómica Avanzada (LANGEBIO), Centro de Investigación y de Estudios Avanzados del IPN, Guanajuato, Mexico
| | - Juan Caballero-Pérez
- Molecular and Developmental Complexity Group, Unidad de Genómica Avanzada (LANGEBIO), Centro de Investigación y de Estudios Avanzados del IPN, Guanajuato, Mexico
| | - Paola L García-Medel
- Structural Biochemistry Group, Unidad de Genómica Avanzada (LANGEBIO), Centro de Investigación y de Estudios Avanzados del IPN, Guanajuato, Mexico
| | - Luis G Brieba
- Structural Biochemistry Group, Unidad de Genómica Avanzada (LANGEBIO), Centro de Investigación y de Estudios Avanzados del IPN, Guanajuato, Mexico
| | - José J Ordaz-Ortiz
- Mass Spectrometry and Metabolomics Laboratory, Unidad de Genómica Avanzada (LANGEBIO), Centro de Investigación y de Estudios Avanzados del IPN, Guanajuato, Mexico
| | - Alfredo Cruz-Ramirez
- Molecular and Developmental Complexity Group, Unidad de Genómica Avanzada (LANGEBIO), Centro de Investigación y de Estudios Avanzados del IPN, Guanajuato, Mexico
| |
Collapse
|
18
|
Luo ZH, Liu ZW, Mao Y, Shu R, Fu LC, Yang RY, Hu YJ, Shen XL. Cajanolactone A, a stilbenoid from cajanus cajan, prevents ovariectomy-induced obesity and liver steatosis in mice fed a regular diet. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153290. [PMID: 32777485 DOI: 10.1016/j.phymed.2020.153290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/03/2020] [Accepted: 07/23/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Visceral obesity and fatty liver are prevalent in postmenopausal women. The stilbene-rich extract of Cajanus cajan (L.) Millsp. has been reported to prevent ovariectomy-induced and diet-induced weight gain in animal models, and stilbenoids from C. cajan are thought to have the potential to prevent postmenopausal obesity and fatty liver. PURPOSE Cajanolactone A (CLA) is the main stilbenoid from C. cajan with osteoblastogenic promoting activity. This study investigated the potential of CLA to prevent postmenopausal obesity and fatty liver. Underlying mechanisms were also investigated. METHOD Ovariectomized C57BL/6 mice fed a regular diet were used as mimics of postmenopausal women and given 10, 20, or 40 mg/kg/d of CLA, 0.1 mg/kg/d of estradiol valerate (EV, positive control), or vehicle (OVX) orally for 16 weeks. Mice of the same age subjected to a sham operation were used as control (Sham). Body weights were recorded every 2 weeks for 16 weeks. Body compositions were analyzed via micro-CT. Serum levels of lipids, adipocytokines and aminotransferases were measured using the relevant kits. mRNA levels of genes of interest were detected by RT-qPCR. Proteomic study of perigonadal white adipose tissue (pWAT) was performed using tandem-mass-tags-based proteomic technology combined with Parallel-Reaction-Monitoring (PRM) validation. RESULTS CLA showed potential equivalent to that of EV to prevent ovariectomy-induced overweight, obesity, dyslipidemia, liver steatosis and liver dysfunction, but did not prevent uterine atrophy. In the liver, CLA significantly inhibited ovariectomy-induced upregulation in expression of lipogenic genes SREBP-1c and ChREBP, and stimulated the mRNA expression of apolipoprotein B gene ApoB. In pWAT, CLA reversed, or partially reversed ovariectomy-induced downregulation in the expression of a number of metabolism- and mitochondrial-function-related proteins, including Ndufa3, Pcx, Pdhb, Acly, Acaca, Aldh2, Aacs and Echs1. In addition, ovariectomy-inhibited mRNA expression of Pdhb, Aacs, Acsm5, Echs1, and Aldh2 genes in pWAT was also reversed. CONCLUSION CLA was demonstrated to be a potential non-estrogen-like drug candidate for prevention of postmenopausal obesity and fatty liver. The underlying mechanism might involve the inhibition of lipogenesis and promotion of triglycerides output in the liver, and the promotion of metabolism and mitochondrial functions of visceral white adipose tissue.
Collapse
Affiliation(s)
- Zhuo-Hui Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, PR China
| | - Zhi-Wen Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, PR China
| | - Yu Mao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, PR China
| | - Rong Shu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, PR China
| | - Lin-Chun Fu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, PR China
| | - Rui-Yi Yang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, PR China
| | - Ying-Jie Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, PR China.
| | - Xiao-Ling Shen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, PR China.
| |
Collapse
|
19
|
Ferrer-González FX, Widner B, Holderman NR, Glushka J, Edison AS, Kujawinski EB, Moran MA. Resource partitioning of phytoplankton metabolites that support bacterial heterotrophy. ISME JOURNAL 2020; 15:762-773. [PMID: 33097854 PMCID: PMC8027193 DOI: 10.1038/s41396-020-00811-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 11/09/2022]
Abstract
The communities of bacteria that assemble around marine microphytoplankton are predictably dominated by Rhodobacterales, Flavobacteriales, and families within the Gammaproteobacteria. Yet whether this consistent ecological pattern reflects the result of resource-based niche partitioning or resource competition requires better knowledge of the metabolites linking microbial autotrophs and heterotrophs in the surface ocean. We characterized molecules targeted for uptake by three heterotrophic bacteria individually co-cultured with a marine diatom using two strategies that vetted the exometabolite pool for biological relevance by means of bacterial activity assays: expression of diagnostic genes and net drawdown of exometabolites, the latter detected with mass spectrometry and nuclear magnetic resonance using novel sample preparation approaches. Of the more than 36 organic molecules with evidence of bacterial uptake, 53% contained nitrogen (including nucleosides and amino acids), 11% were organic sulfur compounds (including dihydroxypropanesulfonate and dimethysulfoniopropionate), and 28% were components of polysaccharides (including chrysolaminarin, chitin, and alginate). Overlap in phytoplankton-derived metabolite use by bacteria in the absence of competition was low, and only guanosine, proline, and N-acetyl-D-glucosamine were predicted to be used by all three. Exometabolite uptake pattern points to a key role for ecological resource partitioning in the assembly marine bacterial communities transforming recent photosynthate.
Collapse
Affiliation(s)
| | - Brittany Widner
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, MA, 02543, USA
| | - Nicole R Holderman
- Department of Biochemistry and Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - John Glushka
- Department of Biochemistry and Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Arthur S Edison
- Department of Biochemistry and Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Elizabeth B Kujawinski
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, MA, 02543, USA
| | - Mary Ann Moran
- Department of Marine Sciences, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
20
|
Davis DE, Sherrod SD, Gant-Branum RL, Colby JM, McLean JA. Targeted Strategy to Analyze Antiepileptic Drugs in Human Serum by LC-MS/MS and LC-Ion Mobility-MS. Anal Chem 2020; 92:14648-14656. [PMID: 33047601 PMCID: PMC10103591 DOI: 10.1021/acs.analchem.0c03172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Routine small-molecule analysis is challenging owing to the need for high selectivity and/or low limits of quantification. This work reports a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method to quantify 14 antiepileptic drugs (AEDs) in human serum. For the optimized LC-MS/MS method described herein, we applied the guidelines outlined in the Clinical and Laboratory Standards Institute (CLSI) LC-MS C62-A document and the U.S. Food and Drug Administration (FDA) Bioanalytical Method Validation Guidance for Industry to evaluate the quality of the assay. In these studies, AED linearity, analyte recovery, matrix effects, precision, and accuracy were assessed. Using liquid chromatography-drift tube ion mobility-mass spectrometry (LC-DTIM-MS), a qualitative method was also used to increase confidence in AED identification using accurate mass and collision cross section (CCS) measurements. The LC-DTIM-MS method was also used to assess the ability of drift tube CCS measurements to aid in the separation and identification of AED structural isomers and other AEDs. These data show that another dimension of information, namely CCS measurements, provides an orthogonal dimension of structural information needed for AED analysis. Multiplexed AED measurements using LC-MS/MS and LC-DTIM-MS have the potential to enable better optimization of dosing owing to the high precision capabilities available in these types of analytical studies. Taken together, these data also show the ability to increase confidence in small-molecule identification and quantification using these analytical technologies.
Collapse
Affiliation(s)
- Don E Davis
- Center for Innovative Technology, Department of Chemistry, Institute of Chemical Biology, Institute for Integrative Biosystems Research and Education, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Stacy D Sherrod
- Center for Innovative Technology, Department of Chemistry, Institute of Chemical Biology, Institute for Integrative Biosystems Research and Education, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Randi L Gant-Branum
- Center for Innovative Technology, Department of Chemistry, Institute of Chemical Biology, Institute for Integrative Biosystems Research and Education, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jennifer M Colby
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37235, United States
| | - John A McLean
- Center for Innovative Technology, Department of Chemistry, Institute of Chemical Biology, Institute for Integrative Biosystems Research and Education, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
21
|
Richard VR, Zahedi RP, Eintracht S, Borchers CH. An LC-MRM assay for the quantification of metanephrines from dried blood spots for the diagnosis of pheochromocytomas and paragangliomas. Anal Chim Acta 2020; 1128:140-148. [PMID: 32825898 PMCID: PMC7346842 DOI: 10.1016/j.aca.2020.06.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/23/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022]
Abstract
The quantitation of metanephrine (MN), normetanephrine (NMN), and 3-methoxytyramine (3-MT) – referred to as metanephrines -- by LC-MS/MS is the gold-standard for screening for pheochromocytoma and paragangliomas (PPGLs), tumours of the adrenal gland and the peripheral nervous system. An assay for metanephrines from dried blood spots (DBSs) would be of high clinical utility as it simplifies sample collection, enables remote sampling, and could increase compliance with the clinical recommendation for supine sampling. Moreover, DBS sampling facilitates the measurement of blood-derived metanephrines in pediatric patients – where DBSs are well-established – in order to diagnose neuroblastomas. Here, we adapted an established derivatization-based LC-MRM-MS assay for plasma catecholamines, and optimized the sample extraction, LC, and MS parameters to produce a fast, sensitive, and robust method for the measurement of metanephrines from DBSs, including 3-methoxytyramine. The DBS samples were excised, derivatized with phenyl isothiocyanate (PITC) on-spot, extracted, and measured by LC-MRM-MS. To validate assay suitability and performance, we assessed the linearity, precision, accuracy, recovery, and matrix effects of the method, and determined the stability of metanephrines in DBSs under different storage conditions. Assay performance for NMN, MN, and 3-MT was sufficient for quantitation from a single DBS within a linear range from 40 to 2000 pg/mL. MN and NMN were stable in DBSs for 2 weeks, whereas 3-MT was stable for one week regardless of storage temperature. Altogether, this work represents the first quantitative LC-MS/MS method for metanephrines from DBSs and provides a novel opportunity for the diagnosis of PPGLs and neuroblastomas in the future. Development and validation of an assay for metanephrines from dried blood spots. Method utilizes on-spot derivatization followed by LC-MRM-MS based quantitation. Method demonstrated high degree of sensitivity, precision, and accuracy. High relevance for the clinical diagnosis of pheochromocytomas and paragangliomas.
Collapse
Affiliation(s)
- Vincent R Richard
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada; Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow, Russia.
| | - René P Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada; Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow, Russia.
| | - Shaun Eintracht
- Department of Diagnostic Medicine, Jewish General Hospital, McGill University, Montreal, QC, Canada.
| | - Christoph H Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada; Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow, Russia; Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, QC, Canada.
| |
Collapse
|
22
|
Ayton S, Janelidze S, Roberts B, Palmqvist S, Kalinowski P, Diouf I, Belaidi AA, Stomrud E, Bush AI, Hansson O. Acute phase markers in CSF reveal inflammatory changes in Alzheimer's disease that intersect with pathology, APOE ε4, sex and age. Prog Neurobiol 2020; 198:101904. [PMID: 32882319 DOI: 10.1016/j.pneurobio.2020.101904] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 01/31/2023]
Abstract
It is unknown how neuroinflammation may feature in the etiology of Alzheimer's disease (AD). We profiled acute phase response (APR) proteins (α1-antitrypsin, α1-antichymotrypsin, ceruloplasmin, complement C3, ferritin, α-fibrinogen, β-fibrinogen, γ-fibrinogen, haptoglobin, hemopexin) in CSF of 1291 subjects along the clinical and biomarker spectrum of AD to investigate the association between inflammatory changes, disease outcomes, and demographic variables. Subjects were stratified by Aβ42/t-tau as well as the following clinical diagnoses: cognitively normal (CN); subjective cognitive decline (SCD); mild cognitive impairment (MCI); and AD dementia. In separate multiple regressions (adjusting for diagnosis, age, sex, APOE-ε4) of each APR protein and a composite of all APR proteins, CSF Aβ42/t-tau status was associated with elevated ferritin, but not any other APR protein in CN and SCD subjects. Rather, the APR was elevated along with symptomatic progression (CN < SCD < MCI < AD), and this was elevation was mediated by CSF p-tau181. APOE ε4 status did not affect levels of any APR proteins in CSF, while these were elevated in males and with increased age. The performance of the APR in predicting clinical diagnosis was influenced by APOE ε4 status, sex, and age. These data provide new insight into inflammatory changes in AD and how this intersects with pathology changes and patient demographics.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Blaine Roberts
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Pawel Kalinowski
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Ibrahima Diouf
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Abdel A Belaidi
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia.
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
23
|
Hellhake S, Meckelmann SW, Empl MT, Rentmeister K, Wißdorf W, Steinberg P, Schmitz OJ, Benter T, Schebb NH. Non-targeted and targeted analysis of oxylipins in combination with charge-switch derivatization by ion mobility high-resolution mass spectrometry. Anal Bioanal Chem 2020; 412:5743-5757. [PMID: 32699965 PMCID: PMC7413910 DOI: 10.1007/s00216-020-02795-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/15/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022]
Abstract
Eicosanoids and other oxylipins play an important role in mediating inflammation as well as other biological processes. For the investigation of their biological role(s), comprehensive analytical methods are necessary, which are able to provide reliable identification and quantification of these compounds in biological matrices. Using charge-switch derivatization with AMPP (N-(4-aminomethylphenyl)pyridinium chloride) in combination with liquid chromatography ion mobility quadrupole time-of-flight mass spectrometry (LC-IM-QTOF-MS), we developed a non-target approach to analyze oxylipins in plasma, serum, and cells. The developed workflow makes use of an ion mobility resolved fragmentation to pinpoint derivatized molecules based on the cleavage of AMPP, which yields two specific fragment ions. This allows a reliable identification of known and unknown eicosanoids and other oxylipins. We characterized the workflow using 52 different oxylipins and investigated their fragmentation patterns and ion mobilities. Limits of detection ranged between 0.2 and 10.0 nM (1.0-50 pg on column), which is comparable with other state-of-the-art methods using LC triple quadrupole (QqQ) MS. Moreover, we applied this strategy to analyze oxylipins in different biologically relevant matrices, as cultured cells, human plasma, and serum. Graphical abstract.
Collapse
Affiliation(s)
- Stefan Hellhake
- School of Mathematics and Natural Sciences, University of Wuppertal, Gauss-Str. 20, 42119, Wuppertal, Germany
| | - Sven W Meckelmann
- Applied Analytical Chemistry & Teaching and Research Center for Separation, University of Duisburg-Essen, Universitätsstr. 5-7, 45141, Essen, Germany
| | - Michael T Empl
- Institute for Food Toxicology, University of Veterinary Medicine Hannover, Bünteweg 2, 30559, Hannover, Germany
| | - Kristina Rentmeister
- Applied Analytical Chemistry & Teaching and Research Center for Separation, University of Duisburg-Essen, Universitätsstr. 5-7, 45141, Essen, Germany
| | - Walter Wißdorf
- School of Mathematics and Natural Sciences, University of Wuppertal, Gauss-Str. 20, 42119, Wuppertal, Germany
| | - Pablo Steinberg
- Institute for Food Toxicology, University of Veterinary Medicine Hannover, Bünteweg 2, 30559, Hannover, Germany
| | - Oliver J Schmitz
- Applied Analytical Chemistry & Teaching and Research Center for Separation, University of Duisburg-Essen, Universitätsstr. 5-7, 45141, Essen, Germany
| | - Thorsten Benter
- School of Mathematics and Natural Sciences, University of Wuppertal, Gauss-Str. 20, 42119, Wuppertal, Germany
| | - Nils Helge Schebb
- School of Mathematics and Natural Sciences, University of Wuppertal, Gauss-Str. 20, 42119, Wuppertal, Germany.
| |
Collapse
|
24
|
Vogeser M, Stone JA. A suggested standard for validation of LC-MS/MS based analytical series in diagnostic laboratories. CLINICAL MASS SPECTROMETRY (DEL MAR, CALIF.) 2020; 16:25-32. [PMID: 34820517 PMCID: PMC8600988 DOI: 10.1016/j.clinms.2020.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/03/2020] [Accepted: 02/25/2020] [Indexed: 11/25/2022]
Abstract
•Policies applied to confirm LC-MS/MS-based results differ between laboratories.•We suggest a systematic approach for validation of individual diagnostic series.•An individual series validation plan can be established with a checklist.
Collapse
Affiliation(s)
- Michael Vogeser
- Hospital of the University of Munich (LMU), Institute of Laboratory Medicine, Munich, Germany
| | - Judith A. Stone
- University of California, San Francisco Medical Center, Laboratory Medicine, Parnassus Chemistry, San Francisco, CA, USA
| |
Collapse
|
25
|
Adams KJ, Pratt B, Bose N, Dubois LG, St John-Williams L, Perrott KM, Ky K, Kapahi P, Sharma V, MacCoss MJ, Moseley MA, Colton CA, MacLean BX, Schilling B, Thompson JW. Skyline for Small Molecules: A Unifying Software Package for Quantitative Metabolomics. J Proteome Res 2020; 19:1447-1458. [PMID: 31984744 DOI: 10.1021/acs.jproteome.9b00640] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Vendor-independent software tools for quantification of small molecules and metabolites are lacking, especially for targeted analysis workflows. Skyline is a freely available, open-source software tool for targeted quantitative mass spectrometry method development and data processing with a 10 year history supporting six major instrument vendors. Designed initially for proteomics analysis, we describe the expansion of Skyline to data for small molecule analysis, including selected reaction monitoring, high-resolution mass spectrometry, and calibrated quantification. This fundamental expansion of Skyline from a peptide-sequence-centric tool to a molecule-centric tool makes it agnostic to the source of the molecule while retaining Skyline features critical for workflows in both peptide and more general biomolecular research. The data visualization and interrogation features already available in Skyline, such as peak picking, chromatographic alignment, and transition selection, have been adapted to support small molecule data, including metabolomics. Herein, we explain the conceptual workflow for small molecule analysis using Skyline, demonstrate Skyline performance benchmarked against a comparable instrument vendor software tool, and present additional real-world applications. Further, we include step-by-step instructions on using Skyline for small molecule quantitative method development and data analysis on data acquired with a variety of mass spectrometers from multiple instrument vendors.
Collapse
Affiliation(s)
- Kendra J Adams
- Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina 27701, United States.,Department of Neurology, Duke University, Durham, North Carolina 27710, United States
| | - Brian Pratt
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Neelanjan Bose
- Buck Institute for Research on Aging, Novato, California 94945, United States
| | - Laura G Dubois
- Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina 27701, United States
| | - Lisa St John-Williams
- Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina 27701, United States
| | - Kevin M Perrott
- Buck Institute for Research on Aging, Novato, California 94945, United States
| | - Karina Ky
- University of California San Francisco, San Francisco, California 94143, United States
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, California 94945, United States
| | - Vagisha Sharma
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - M Arthur Moseley
- Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina 27701, United States
| | - Carol A Colton
- Department of Neurology, Duke University, Durham, North Carolina 27710, United States
| | - Brendan X MacLean
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Birgit Schilling
- Buck Institute for Research on Aging, Novato, California 94945, United States
| | - J Will Thompson
- Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina 27701, United States.,Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| | | |
Collapse
|
26
|
Arima N, Sasaki Y, Lee LH, Zhang H, Figueiredo JL, Mlynarchik AK, Qiao J, Yamada I, Higashi H, Ha AH, Halu A, Mizuno K, Singh SA, Yamazaki Y, Aikawa M. Multiorgan Systems Study Reveals Igfbp7 as a Suppressor of Gluconeogenesis after Gastric Bypass Surgery. J Proteome Res 2020; 19:129-143. [PMID: 31661273 DOI: 10.1021/acs.jproteome.9b00441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Roux-en-Y gastric bypass (RYGB) surgery reduces weight in obese patients. A marked decrease in blood glucose levels occurs before weight loss; however, key molecules that improve the glycemic profile remain largely unknown. Using a murine RYGB surgery model, we performed multiorgan proteomics and bioinformatics to monitor the proteins and molecular pathways that change in this early glycemic response. Multiplexed proteomic kinetics data analysis revealed that the Roux limb, biliopancreatic limb, liver, and pancreas each exhibited unique temporal and molecular responses to the RYGB surgery. In addition, protein-protein network analysis indicated that the changes to the microbial environment in the intestine may play a crucial role in the beneficial effects of RYGB surgery. Furthermore, insulin-like growth factor binding protein 7 (Igfbp7) was identified as an early induced protein in the Roux limb. Known secretory properties of Igfbp7 prompted us to further investigate its role as a remote organ regulator of glucose metabolism. Igfbp7 overexpression decreased blood glucose levels in diet-induced obese mice and attenuated gluconeogenic gene expression in the liver. Secreted Igfbp7 appeared to mediate these beneficial effects. These results demonstrate that organs responded differentially to RYGB surgery and indicate that Igfbp7 may play an important role in improving blood glucose levels.
Collapse
Affiliation(s)
- Naoaki Arima
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | - Yusuke Sasaki
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | | | | | | | | | | | - Iwao Yamada
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | - Hideyuki Higashi
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | | | | | - Ken Mizuno
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | | | - Yukiyoshi Yamazaki
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | - Masanori Aikawa
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health , Moscow , Russian Federation , 119146
| |
Collapse
|
27
|
Crews BO, Drees JC, Greene DN. Data-driven quality assurance to prevent erroneous test results. Crit Rev Clin Lab Sci 2019:1-15. [PMID: 31680585 DOI: 10.1080/10408363.2019.1678567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Increasing laboratory automation and efficiency requires quality assurance (QA) approaches to ensure that reported results are precise and accurate. Prerequisites for designing optimal QA strategies include an in-depth understanding of the laboratory processes, the expected results, and of the mechanisms that can cause erroneous results. Oftentimes, a laboratory's own data, extracted from the laboratory information system, electronic medical record, and/or clinical data warehouse are necessary to master the aforementioned requirements. Data-driven QA utilizes retrospective and/or prospective laboratory results to minimize errors in the clinical laboratory due to pre-analytical or analytical vulnerabilities. Additionally, exploitation of this data may improve result interpretation. The objective of this review is to illustrate specific examples of data-driven QA approaches for several areas of the clinical laboratory and for different phases of the testing cycle.
Collapse
Affiliation(s)
- Bridgit O Crews
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| | - Julia C Drees
- The Permanente Medical Group, Kaiser Permanente Northern California Regional Laboratories, Berkeley, CA, USA
| | | |
Collapse
|
28
|
Automation of chromatographic peak review and order to result data transfer in a clinical mass spectrometry laboratory. Clin Chim Acta 2019; 498:84-89. [DOI: 10.1016/j.cca.2019.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 11/18/2022]
|
29
|
Oranzi NR, Lei J, Kemperman RHJ, Chouinard CD, Holmquist B, Garrett TJ, Yost RA. Rapid Quantitation of 25-Hydroxyvitamin D2 and D3 in Human Serum Using Liquid Chromatography/Drift Tube Ion Mobility-Mass Spectrometry. Anal Chem 2019; 91:13555-13561. [DOI: 10.1021/acs.analchem.9b02683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Nicholas R. Oranzi
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, Florida 32611, United States
| | - Jiajun Lei
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, Florida 32611, United States
| | - Robin H. J. Kemperman
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, Florida 32611, United States
| | - Christopher D. Chouinard
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, Florida 32901, United States
| | | | - Timothy J. Garrett
- Department of Pathology, University of Florida, Gainesville, Florida 32610, United States
| | - Richard A. Yost
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, Florida 32611, United States
- Department of Pathology, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
30
|
Schaller TH, Foster MW, Thompson JW, Spasojevic I, Normantaite D, Moseley MA, Sanchez-Perez L, Sampson JH. Pharmacokinetic Analysis of a Novel Human EGFRvIII:CD3 Bispecific Antibody in Plasma and Whole Blood Using a High-Resolution Targeted Mass Spectrometry Approach. J Proteome Res 2019; 18:3032-3041. [PMID: 31267741 DOI: 10.1021/acs.jproteome.9b00145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Bispecific single chain antibody fragments (bi-scFv) represent an emerging class of biotherapeutics. We recently developed a fully human bi-scFv (EGFRvIII:CD3 bi-scFv) with the goal of redirecting CD3-expressing T cells to recognize and destroy malignant, EGFRvIII-expressing glioma. In mice, we showed that EGFRvIII:CD3 bi-scFv effectively treats orthotopic patient-derived malignant glioma and syngeneic glioblastoma. Here, we developed a targeted assay for pharmacokinetic (PK) analysis of EGFRvIII:CD3 bi-scFv, a necessary step in the drug development process. Using microflow liquid chromatography coupled to a high resolution parallel reaction monitoring mass spectrometry, and data analysis in Skyline, we developed a bottom-up proteomic assay for quantification of EGFRvIII:CD3 bi-scFv in both plasma and whole blood. Importantly, a protein calibrator, along with stable isotope-labeled EGFRvIII:CD3 bi-scFv protein, were used for absolute quantification. A PK analysis in a CD3 humanized mouse revealed that EGFRvIII:CD3 bi-scFv in plasma and whole blood has an initial half-life of ∼8 min and a terminal half-life of ∼2.5 h. Our results establish a sensitive, high-throughput assay for direct quantification of EGFRvIII:CD3 bi-scFv without the need for immunoaffinity enrichment. Moreover, these pharmacokinetic parameters will guide drug optimization and dosing regimens in future IND-enabling and phase I studies of EGFRvIII:CD3 bi-scFv.
Collapse
Affiliation(s)
- Teilo H Schaller
- Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , North Carolina , United States.,Department of Neurosurgery , Duke University Medical Center , Durham , North Carolina , United States.,Department of Pathology , Duke University Medical Center , Durham , North Carolina , United States
| | - Matthew W Foster
- Duke Proteomics and Metabolomics Shared Resource, Duke Center for Genomic and Computational Biology , Duke University , Durham , North Carolina , United States
| | - J Will Thompson
- Duke Proteomics and Metabolomics Shared Resource, Duke Center for Genomic and Computational Biology , Duke University , Durham , North Carolina , United States
| | - Ivan Spasojevic
- Duke Cancer Institute PK/PD Core Laboratory , Durham , North Carolina , United States.,Department of Medicine , Duke University School of Medicine , Durham , North Carolina , United States
| | - Deimante Normantaite
- Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , North Carolina , United States
| | - M Arthur Moseley
- Duke Proteomics and Metabolomics Shared Resource, Duke Center for Genomic and Computational Biology , Duke University , Durham , North Carolina , United States
| | - Luis Sanchez-Perez
- Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , North Carolina , United States.,Department of Neurosurgery , Duke University Medical Center , Durham , North Carolina , United States
| | - John H Sampson
- Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , North Carolina , United States.,Department of Neurosurgery , Duke University Medical Center , Durham , North Carolina , United States.,Department of Pathology , Duke University Medical Center , Durham , North Carolina , United States
| |
Collapse
|
31
|
Zhou Q, Andersson R, Hu D, Bauden M, Kristl T, Sasor A, Pawłowski K, Pla I, Hilmersson KS, Zhou M, Lu F, Marko-Varga G, Ansari D. Quantitative proteomics identifies brain acid soluble protein 1 (BASP1) as a prognostic biomarker candidate in pancreatic cancer tissue. EBioMedicine 2019; 43:282-294. [PMID: 30982764 PMCID: PMC6557784 DOI: 10.1016/j.ebiom.2019.04.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/27/2019] [Accepted: 04/03/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pancreatic cancer is a heterogenous disease with a poor prognosis. This study aimed to discover and validate prognostic tissue biomarkers in pancreatic cancer using a mass spectrometry (MS) based proteomics approach. METHODS Global protein sequencing of fresh frozen pancreatic cancer and healthy pancreas tissue samples was conducted by MS to discover potential protein biomarkers. Selected candidate proteins were further verified by targeted proteomics using parallel reaction monitoring (PRM). The expression of biomarker candidates was validated by immunohistochemistry in a large tissue microarray (TMA) cohort of 141 patients with resectable pancreatic cancer. Kaplan-Meier and Cox proportional hazard modelling was used to investigate the prognostic utility of candidate protein markers. FINDINGS In the initial MS-discovery phase, 165 proteins were identified as potential biomarkers. In the subsequent MS-verification phase, a panel of 45 candidate proteins was verified by the development of a PRM assay. Brain acid soluble protein 1 (BASP1) was identified as a new biomarker candidate for pancreatic cancer possessing largely unknown biological and clinical functions and was selected for further analysis. Importantly, bioinformatic analysis indicated that BASP1 interacts with Wilms tumour protein (WT1) in pancreatic cancer. TMA-based immunohistochemistry analysis showed that BASP1 was an independent predictor of prolonged survival (HR 0.468, 95% CI 0.257-0.852, p = .013) and predicted favourable response to adjuvant chemotherapy, whereas WT1 indicated a worsened survival (HR 1.636, 95% CI 1.083-2.473, p = .019) and resistance to chemotherapy. Interaction analysis showed that patients with negative BASP1 and high WT1 expression had the poorest outcome (HR 3.536, 95% CI 1.336-9.362, p = .011). INTERPRETATION We here describe an MS-based proteomics platform for developing biomarkers for pancreatic cancer. Bioinformatic analysis and clinical data from our study suggest that BASP1 and its putative interaction partner WT1 can be used as biomarkers for predicting outcomes in pancreatic cancer patients.
Collapse
Affiliation(s)
- Qimin Zhou
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden; The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Roland Andersson
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Dingyuan Hu
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Monika Bauden
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Theresa Kristl
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Agata Sasor
- Department of Pathology, Skåne University Hospital, Lund, Sweden
| | - Krzysztof Pawłowski
- Department of Experimental Design and Bioinformatics, Warsaw University of Life Sciences, Warsaw, Poland; Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Indira Pla
- Department of Biomedical Engineering, Clinical Protein Science and Imaging, Lund University, Lund, Sweden
| | - Katarzyna Said Hilmersson
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Mengtao Zhou
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fan Lu
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - György Marko-Varga
- Department of Biomedical Engineering, Clinical Protein Science and Imaging, Lund University, Lund, Sweden
| | - Daniel Ansari
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
32
|
Abstract
This article describes the need for, stratifies the complexity of, and proposes detailed lists of training competencies for diagnostic laboratory personnel using quantitative liquid chromatography-tandem mass spectrometry (LC-MS/MS) for patient care. Although quantitative LC-MS/MS is evolving toward greater automation with less need for technical expertise, gaps remain in resources for training and assessment.
Collapse
|