1
|
Pierre N, Huynh-Thu VA, Baiwir D, Mazzucchelli G, Fléron M, Trzpiot L, Eppe G, De Pauw E, Laharie D, Satsangi J, Bossuyt P, Vuitton L, Vieujean S, Colombel JF, Meuwis MA, Louis E. External validation of serum biomarkers predicting short-term and mid/long-term relapse in patients with Crohn's disease stopping infliximab. Gut 2024; 73:1965-1973. [PMID: 39134391 DOI: 10.1136/gutjnl-2024-332648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/28/2024] [Indexed: 11/13/2024]
Abstract
OBJECTIVE In patients with Crohn's disease (CD) on combination therapy (infliximab and immunosuppressant) and stopping infliximab (cohort from the study of infliximab diSconTinuation in CrOhn's disease patients in stable Remission on combined therapy with Immunosuppressors (STORI)), the risk of short-term (≤6 months) and mid/long-term relapse (>6 months) was associated with distinct blood protein profiles. Our aim was to test the external validity of this finding in the SPARE cohort (A proSpective Randomized Controlled Trial comParing infliximAb-antimetabolites Combination Therapy to Anti-metabolites monotheRapy and Infliximab monothErapy in Crohn's Disease Patients in Sustained Steroid-free Remission on Combination Therapy). DESIGN In SPARE, patients with CD in sustained steroid-free clinical remission and on combination therapy were randomly allocated to three arms: continuing combination therapy, stopping infliximab or stopping immunosuppressant. In the baseline serum of the STORI and SPARE (arm stopping infliximab) cohorts, we studied 202 immune-related proteins. The proteins associated with time to relapse (univariable Cox model) were compared between STORI and SPARE. The discriminative ability of biomarkers (individually and combined in pairs) was evaluated by the c-statistic (concordance analysis) which was compared with C-reactive protein (CRP), faecal calprotectin and a previously validated model (CEASE). RESULTS In STORI and SPARE, distinct blood protein profiles were associated with the risk of short-term (eg, high level: CRP, haptoglobin, interleukin-6, C-type lectin domain family 4 member C) and mid/long-term relapse (eg, low level: Fms-related tyrosine kinase 3 ligand, kallistatin, fibroblast growth factor 2). At external validation, the top 10 biomarker pairs showed a higher c-statistic than the CEASE model, CRP and faecal calprotectin in predicting short-term (0.76-0.80 vs 0.74 vs 0.71 vs 0.69, respectively) and mid/long-term relapse (0.66-0.68 vs 0.61 vs 0.52 vs 0.59, respectively). CONCLUSION In patients with CD stopping infliximab, we confirm that the risk of short-term and mid/long-term relapse is associated with distinct blood protein profiles showing the potential to guide infliximab withdrawal. TRIAL REGISTRATION NUMBER NCT00571337 and NCT02177071.
Collapse
Affiliation(s)
- Nicolas Pierre
- Laboratory of Translational Gastroenterology, GIGA-institute, Liège University, Liège, Belgium
| | - Vân Anh Huynh-Thu
- Department of Electrical Engineering and Computer Science, Liège University, Liège, Belgium
| | | | - Gabriel Mazzucchelli
- Laboratory of Mass Spectrometry, MolSys Research Unit, Liège University, Liège, Belgium
| | | | - Lisette Trzpiot
- Laboratory of Mass Spectrometry, MolSys Research Unit, Liège University, Liège, Belgium
| | - Gauthier Eppe
- Laboratory of Mass Spectrometry, MolSys Research Unit, Liège University, Liège, Belgium
| | - Edwin De Pauw
- Laboratory of Mass Spectrometry, MolSys Research Unit, Liège University, Liège, Belgium
| | - David Laharie
- Service d'Hépato-gastroentérologie et oncologie digestive, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Jack Satsangi
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford, UK
| | - Peter Bossuyt
- Imelda GI Clinical Research Center, Imelda Hospital, Bonheiden, Belgium
| | - Lucine Vuitton
- Department of Gastroenterology, Besançon University Hospital, Besancon, France
- UMR 1098, Franche-Comté University, Besancon, France
| | - Sophie Vieujean
- Laboratory of Translational Gastroenterology, GIGA-institute, Liège University, Liège, Belgium
- Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| | - Jean-Frédéric Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marie-Alice Meuwis
- Laboratory of Translational Gastroenterology, GIGA-institute, Liège University, Liège, Belgium
- Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| | - Edouard Louis
- Laboratory of Translational Gastroenterology, GIGA-institute, Liège University, Liège, Belgium
- Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| |
Collapse
|
2
|
Ashkarran AA, Gharibi H, Sadeghi SA, Modaresi SM, Wang Q, Lin TJ, Yerima G, Tamadon A, Sayadi M, Jafari M, Lin Z, Ritz D, Kakhniashvili D, Guha A, Mofrad MRK, Sun L, Landry MP, Saei AA, Mahmoudi M. Small molecule modulation of protein corona for deep plasma proteome profiling. Nat Commun 2024; 15:9638. [PMID: 39511193 PMCID: PMC11544298 DOI: 10.1038/s41467-024-53966-z] [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: 03/14/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
The protein corona formed on nanoparticles (NPs) has potential as a valuable diagnostic tool for improving plasma proteome coverage. Here, we show that spiking small molecules, including metabolites, lipids, vitamins, and nutrients into plasma can induce diverse protein corona patterns on otherwise identical NPs, significantly enhancing the depth of plasma proteome profiling. The protein coronas on polystyrene NPs when exposed to plasma treated with an array of small molecules allows for the detection of 1793 proteins marking an 8.25-fold increase in the number of quantified proteins compared to plasma alone (218 proteins) and a 2.63-fold increase relative to the untreated protein corona (681 proteins). Furthermore, we discovered that adding 1000 µg/ml phosphatidylcholine could singularly enable the detection of 897 proteins. At this specific concentration, phosphatidylcholine selectively depletes the four most abundant plasma proteins, including albumin, thus reducing the dynamic range of plasma proteome and enabling the detection of proteins with lower abundance. Employing an optimized data-independent acquisition approach, the inclusion of phosphatidylcholine leads to the detection of 1436 proteins in a single plasma sample. Our molecular dynamics results reveal that phosphatidylcholine interacts with albumin via hydrophobic interactions, H-bonds, and water bridges. The addition of phosphatidylcholine also enables the detection of 337 additional proteoforms compared to untreated protein corona using a top-down proteomics approach. Given the critical role of plasma proteomics in biomarker discovery and disease monitoring, we anticipate the widespread adoption of this methodology for the identification and clinical translation of biomarkers.
Collapse
Affiliation(s)
- Ali Akbar Ashkarran
- Precision Health Program, Michigan State University, East Lansing, MI, USA
- Depatment of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Hassan Gharibi
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Qianyi Wang
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Teng-Jui Lin
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Ghafar Yerima
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Ali Tamadon
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Maryam Sayadi
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Maryam Jafari
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Zijin Lin
- Precision Health Program, Michigan State University, East Lansing, MI, USA
| | - Danilo Ritz
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - David Kakhniashvili
- Proteomics and Metabolomics Core Facility, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Avirup Guha
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Liangliang Sun
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Markita P Landry
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Amir Ata Saei
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI, USA.
- Depatment of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
3
|
Twigg CI, Perez JM, Ryu J, Hanson BK, Barrera Estrada VJ, Thomas SN. Evaluation of Serum Proteome Sample Preparation Methods to Support Clinical Proteomics Applications. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:2659-2669. [PMID: 39263706 PMCID: PMC11546599 DOI: 10.1021/jasms.4c00131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/24/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Serum contains several proteins that are associated with disease-related processes. Mass spectrometry (MS)-based proteomics approaches greatly facilitate serum protein biomarker development. However, the serum proteome complexity presents a technical challenge for the accurate, sensitive, and reproducible quantification of proteins by MS. Thus, efficient sample preparation methods are of critical importance for serum proteome analyses. In this study, we evaluated the technical performance of two serum proteome sample preparation methods using sera from patients with high-grade serous ovarian cancer and patients with benign nongynecological conditions with a goal of providing insight into their compatibility with clinical proteomics workflows. One method entailed the use of immobilized trypsin (SMART Digest Trypsin) with RapiGest SF, an acid-labile surfactant designed to enhance the in-solution enzymatic digestion of proteins. The other method incorporated a commercially available sample preparation kit, iST-BCT, which contains standardized reagents. Significantly higher protein sequence coverage, albeit with lower digestion efficiency, was obtained with the immobilized trypsin + RapiGest SF workflow, whereas the iST-BCT workflow was quicker and had marginally better reproducibility. Protein relative abundance analysis revealed that the serum proteomes clustered primarily by the sample processing workflow and secondarily by disease state. We conducted a time course study to determine whether differences in the relative abundance of diagnostic high-grade serous ovarian cancer serum protein biomarker candidates were biased according to the duration of enzymatic digestion. Our results highlight the importance of optimizing enzymatic digestion kinetics according to the peptide targets of interest while considering the sensitivity of the downstream analytical method utilized in clinical proteomics workflows designed to measure biomarkers.
Collapse
Affiliation(s)
- Carly
A. I. Twigg
- Department
of Laboratory Medicine and Pathology, University
of Minnesota School of Medicine, Minneapolis, Minnesota 55455, United States
| | - Jesenia M. Perez
- Microbiology,
Immunology, and Cancer Biology Graduate Program, University of Minnesota School of Medicine, Minneapolis, Minnesota 55455, United States
| | - Joohyun Ryu
- Department
of Laboratory Medicine and Pathology, University
of Minnesota School of Medicine, Minneapolis, Minnesota 55455, United States
| | - Benjamin K. Hanson
- Department
of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Stefani N. Thomas
- Department
of Laboratory Medicine and Pathology, University
of Minnesota School of Medicine, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
Zaarour L, Padula M, van Oorschot RAH, McNevin D. Mass spectrometry-based proteomics for source-level attribution after DNA extraction. Forensic Sci Int Genet 2024; 74:103168. [PMID: 39536553 DOI: 10.1016/j.fsigen.2024.103168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Biological traces recovered from crime scenes serve as vital evidence in forensic investigations. While DNA evidence is frequently used to address the sub-source level of the hierarchy of propositions, the biological source of the DNA can be highly probative at the source level. Current body fluid detection methods pose certain limitations, such as reports of false positive results from some of the presumptive and/or confirmatory tests in current use. These tests are also individual tests for the detection of one body fluid, meaning that if the sample is suspected to be a mixture of multiple body fluids, then different tests would need to be conducted to confirm the body fluid(s) present, which may exhaust small amounts of available biological trace. Proteomics applications for the identification of body fluids have been previously explored, and potential biomarkers indicative of body fluids discovered from liquid-chromatography tandem mass spectrometry (LC-MS/MS) methods have been reported. This work focuses on developing a mass spectrometry-based proteomics approach for the identification of body fluids by targeting discriminating peptide biomarkers from the non-DNA component left over after DNA extraction of samples. The non-DNA component is typically a waste product but with unappreciated evidential value. Our methodology for the purification of proteins from the post-DNA extraction waste includes an acetone precipitation and single-pot solid-phase-enhanced sample preparation (SP3) technique, microwave-assisted trypsin digestion, and LC-MS/MS analysis of the resultant peptides. Preliminary results from this proof-of-concept study include a list of potentially discriminating proteins and peptides for blood, saliva, and semen developed from the analysis of post-DNA extraction waste. Our method allows for multiple analytes to be targeted simultaneously from a DNA profiling waste stream and we anticipate that it could eventually be incorporated into standard forensic laboratory workflows.
Collapse
Affiliation(s)
- Layal Zaarour
- Centre for Forensic Science, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| | - Matthew Padula
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Roland A H van Oorschot
- Office of the Chief Forensic Scientist, Victoria Police Forensic Services Department, Macleod, Victoria, Australia; School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia
| | - Dennis McNevin
- Centre for Forensic Science, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
5
|
Gotti C, Roux-Dalvai F, Bérubé È, Lacombe-Rastoll A, Leclercq M, Jacob CC, Boissinot M, Martins C, Wijeratne NR, Bergeron MG, Droit A. LC-SRM Combined With Machine Learning Enables Fast Identification and Quantification of Bacterial Pathogens in Urinary Tract Infections. Mol Cell Proteomics 2024; 23:100832. [PMID: 39178943 PMCID: PMC11532907 DOI: 10.1016/j.mcpro.2024.100832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024] Open
Abstract
Urinary tract infections (UTIs) are a worldwide health problem. Fast and accurate detection of bacterial infection is essential to provide appropriate antibiotherapy to patients and to avoid the emergence of drug-resistant pathogens. While the gold standard requires 24 h to 48 h of bacteria culture prior to MALDI-TOF species identification, we propose a culture-free workflow, enabling bacterial identification and quantification in less than 4 h using 1 ml of urine. After rapid and automatable sample preparation, a signature of 82 bacterial peptides, defined by machine learning, was monitored in LC-MS, to distinguish the 15 species causing 84% of the UTIs. The combination of the sensitivity of the SRM mode on a triple quadrupole TSQ Altis instrument and the robustness of capillary flow enabled us to analyze up to 75 samples per day, with 99.2% accuracy on bacterial inoculations of healthy urines. We have also shown our method can be used to quantify the spread of the infection, from 8 × 104 to 3 × 107 CFU/ml. Finally, the workflow was validated on 45 inoculated urines and on 84 UTI-positive urine from patients, with respectively 93.3% and 87.1% of agreement with the culture-MALDI procedure at a level above 1 × 105 CFU/ml corresponding to an infection requiring antibiotherapy.
Collapse
Affiliation(s)
- Clarisse Gotti
- Computational Biology Laboratory, CHU de Québec - Université Laval Research Center, Québec City, Quebec, Canada; Proteomics Platform, CHU de Québec - Université Laval Research Center, Québec City, Quebec, Canada
| | - Florence Roux-Dalvai
- Computational Biology Laboratory, CHU de Québec - Université Laval Research Center, Québec City, Quebec, Canada; Proteomics Platform, CHU de Québec - Université Laval Research Center, Québec City, Quebec, Canada
| | - Ève Bérubé
- Centre de Recherche en Infectiologie de l'Université Laval, Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Quebec, Canada
| | - Antoine Lacombe-Rastoll
- Computational Biology Laboratory, CHU de Québec - Université Laval Research Center, Québec City, Quebec, Canada; Proteomics Platform, CHU de Québec - Université Laval Research Center, Québec City, Quebec, Canada
| | - Mickaël Leclercq
- Computational Biology Laboratory, CHU de Québec - Université Laval Research Center, Québec City, Quebec, Canada
| | | | - Maurice Boissinot
- Centre de Recherche en Infectiologie de l'Université Laval, Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Quebec, Canada
| | | | | | - Michel G Bergeron
- Centre de Recherche en Infectiologie de l'Université Laval, Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Quebec, Canada
| | - Arnaud Droit
- Computational Biology Laboratory, CHU de Québec - Université Laval Research Center, Québec City, Quebec, Canada; Proteomics Platform, CHU de Québec - Université Laval Research Center, Québec City, Quebec, Canada.
| |
Collapse
|
6
|
Flender D, Vilenne F, Adams C, Boonen K, Valkenborg D, Baggerman G. Exploring the dynamic landscape of immunopeptidomics: Unravelling posttranslational modifications and navigating bioinformatics terrain. MASS SPECTROMETRY REVIEWS 2024. [PMID: 39152539 DOI: 10.1002/mas.21905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024]
Abstract
Immunopeptidomics is becoming an increasingly important field of study. The capability to identify immunopeptides with pivotal roles in the human immune system is essential to shift the current curative medicine towards personalized medicine. Throughout the years, the field has matured, giving insight into the current pitfalls. Nowadays, it is commonly accepted that generalizing shotgun proteomics workflows is malpractice because immunopeptidomics faces numerous challenges. While many of these difficulties have been addressed, the road towards the ideal workflow remains complicated. Although the presence of Posttranslational modifications (PTMs) in the immunopeptidome has been demonstrated, their identification remains highly challenging despite their significance for immunotherapies. The large number of unpredictable modifications in the immunopeptidome plays a pivotal role in the functionality and these challenges. This review provides a comprehensive overview of the current advancements in immunopeptidomics. We delve into the challenges associated with identifying PTMs within the immunopeptidome, aiming to address the current state of the field.
Collapse
Affiliation(s)
- Daniel Flender
- Centre for Proteomics, University of Antwerp, Antwerpen, Belgium
- Health Unit, VITO, Mol, Belgium
| | - Frédérique Vilenne
- Health Unit, VITO, Mol, Belgium
- Data Science Institute, University of Hasselt, Hasselt, Belgium
| | - Charlotte Adams
- Department of Computer Science, University of Antwerp, Antwerp, Belgium
| | - Kurt Boonen
- Centre for Proteomics, University of Antwerp, Antwerpen, Belgium
- ImmuneSpec, Niel, Belgium
| | - Dirk Valkenborg
- Data Science Institute, University of Hasselt, Hasselt, Belgium
| | - Geert Baggerman
- Department of Computer Science, University of Antwerp, Antwerp, Belgium
- ImmuneSpec, Niel, Belgium
| |
Collapse
|
7
|
Basu R, Dambra R, Jiang D, Schätzlein SA, Njiyang S, Ashour J, Chiramel AI, Vigil A, Papov VV. Absolute quantification of viral proteins from pseudotyped VSV-GP using UPLC-MRM. Microbiol Spectr 2024; 12:e0365123. [PMID: 38916347 PMCID: PMC11302727 DOI: 10.1128/spectrum.03651-23] [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: 10/13/2023] [Accepted: 05/20/2024] [Indexed: 06/26/2024] Open
Abstract
The rapidly developing field of oncolytic virus (OV) therapy necessitates the development of new and improved analytical approaches for the characterization of the virus during production and development. Accurate monitoring and absolute quantification of viral proteins are crucial for OV product characterization and can facilitate the understanding of infection, immunogenicity, and development stages of viral replication. Targeted mass spectrometry methods like multiple reaction monitoring (MRM) offer a robust way to directly detect and quantify specific targeted proteins represented by surrogate peptides. We have leveraged the power of MRM by combining ultra-high performance liquid chromatography (UPLC) with a Sciex 6500 triple-stage quadrupole mass spectrometer to develop an assay that accurately and absolutely quantifies the structural proteins of a pseudotyped vesicular stomatitis virus (VSV) intended for use as a new biotherapeutic (designated hereafter as VSV-GP to differentiate it from native VSV). The new UPLC-MRM method provides absolute quantification with the use of heavy-labeled reference standard surrogate peptides. When added in known exact amounts to standards and samples, the reference standards normalize and account for any small perturbations during sample preparation and/or instrument performance, resulting in accurate and precise quantification. Because of the multiplexed nature of MRM, all targeted proteins are quantified at the same time. The optimized assay has been enhanced to quantify the ratios of the processed GP1 and GP2 proteins while simultaneously measuring any remaining or unprocessed form of the envelope protein GP complex (GPC; full-length GPC). IMPORTANCE The development of oncolytic viral therapy has gained considerable momentum in recent years. Vesicular stomatitis virus glycoprotein (VSV-GP) is a new biotherapeutic emerging in the oncolytic viral therapy platform. Novel analytical assays that can accurately and precisely quantify the viral proteins are a necessity for the successful development of viral vector as a biotherapeutic. We developed an ultra-high performance liquid chromatography multiple reaction monitoring-based assay to quantify the absolute concentrations of the different structural proteins of VSV-GP. The complete processing of GP complex (GPC) is a prerequisite for the infectivity of the virus. The assay extends the potential for quantifying full-length GPC, which provides an understanding of the processing of GPC (along with the quantification of GP1 and GP2 separately). We used this assay in tracking GPC processing in HEK-293-F production cell lines infected with VSV-GP.
Collapse
Affiliation(s)
- Rajeswari Basu
- Materials and Analytical Sciences, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, USA
| | - Richard Dambra
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut, USA
| | - Di Jiang
- Materials and Analytical Sciences, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, USA
| | - Sophia A. Schätzlein
- Therapeutic Virus Development Group, Virus Therapeutic Center, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Shu Njiyang
- Therapeutic Virus Development Group, Virus Therapeutic Center, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Joseph Ashour
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut, USA
| | - Abhilash I. Chiramel
- Therapeutic Virus Development Group, Virus Therapeutic Center, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Adam Vigil
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut, USA
| | - Vladimir V. Papov
- Materials and Analytical Sciences, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, USA
| |
Collapse
|
8
|
Liu L, Cai J, Yang K, Sun B, Liu W, Li Y, Hu H. Molecular beacon-peptide probe based double recycling amplification for multiplexed detection of serum exosomal microRNAs. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:5202-5211. [PMID: 38994818 DOI: 10.1039/d4ay00629a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Exosomal microRNAs (exomiRs) have been shown to play crucial roles as biomarkers for early detection and prognosis of cancer. However, simultaneous quantification of multiplex exomiRs is hindered by methods that require additional steps, such as labeling with fluorophores or gel visualization, which are susceptible to various factors. Herein, we developed a mass spectrometry-detectable and target-triggered method for multiplexed exomiR detection using three enzyme-based double recycling amplification in combination with well-designed molecular beacon-peptide (MBP) probes, called molecular beacon-peptide probe-based double recycling amplification (MBPDRA). MBP probes mediated the double recycling amplification reaction and were released as mass-detectable reporter peptides. In particular, the hybridization of the target microRNAs (miRNAs) with the stem-loop of the probe triggers two consecutive processes. The first cycle involved polymerase strand displacement amplification, leading to the production of complementary DNA (cycle I), and the second cycle encompassed the recycling exonuclease cleavage of the MBP probe (cycle II). Subsequently, excess probes were removed by interaction with streptavidin beads via biotin-streptavidin binding. The reporter peptides were released using trypsin and subsequently detected by mass spectrometry. Our method enables quantitative detection of multiple exomiRs with a dynamic range from 0.1 fM to 10 pM and a limit of quantification of 0.1 fM. Moreover, the proposed assay was successfully employed for quantification of three exomiRs, exmiR-21, exmiR-191, and exmiR-451a, in the sera of patients with pancreatic cancer. Based on these findings, we believe that the MBPDRA assay holds significant promise as a reliable method for quantifying multiple miRNAs in biomedical research and clinical diagnostics.
Collapse
Affiliation(s)
- Liang Liu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China.
| | - Junlong Cai
- Department of Clinical Trial Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Kun Yang
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China.
| | - Bo Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
| | - Yang Li
- Department of Blood Transfusion, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan, 430071, China.
| | - Hankun Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China.
| |
Collapse
|
9
|
Šprager E, Möller J, Lin Y, Reisinger V, Bratkovič T, Lunder M, Vašl J, Krajnc A. Identification of Acyl-Protein Thioesterase-1 as a Polysorbate-Degrading Host Cell Protein in a Monoclonal Antibody Formulation Using Activity-Based Protein Profiling. J Pharm Sci 2024; 113:2128-2139. [PMID: 38772451 DOI: 10.1016/j.xphs.2024.05.013] [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: 03/18/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/23/2024]
Abstract
Polysorbate (PS) degradation in monoclonal antibody (mAb) formulations poses a significant challenge in the biopharmaceutical industry. PS maintains protein stability during drug product's shelf life but is vulnerable to breakdown by low-abundance residual host cell proteins (HCPs), particularly hydrolytic enzymes such as lipases and esterases. In this study, we used activity-based protein profiling (ABPP) coupled with mass spectrometry to identify acyl-protein thioesterase-1 (APT-1) as a polysorbate-degrading HCP in one case of mAb formulation with stability problems. We validated the role of APT1 by matching the polysorbate degradation fingerprint in the mAb formulation with that of a recombinant APT1 protein. Furthermore, we found an agreement between APT1 levels and PS degradation rates in the mAb formulation, and we successfully halted PS degradation using APT1-specific inhibitors ML348 and ML211. APT1 was found to co-purify with a specific mAb via hitchhiking mechanism. Our work provides a streamlined approach to identifying critical HCPs in PS degradation, supporting quality-by-design principles in pharmaceutical development.
Collapse
Affiliation(s)
- Ernest Šprager
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia; Novartis Technical Research & Development, Biologics Technical Development Mengeš, Novartis Pharmaceutical Manufacturing LLC, Slovenia
| | - Jens Möller
- Novartis Technical Research & Development, Analytical Characterization, Novartis Pharmaceutical Manufacturing GmbH, Kundl, Austria
| | - Yuhsien Lin
- Novartis Technical Research & Development, Analytical Characterization, Novartis Pharmaceutical Manufacturing GmbH, Kundl, Austria
| | - Veronika Reisinger
- Novartis Technical Research & Development, Analytical Characterization, Novartis Pharmaceutical Manufacturing GmbH, Kundl, Austria
| | - Tomaž Bratkovič
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Mojca Lunder
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Jožica Vašl
- Novartis Technical Research & Development, Biologics Technical Development Mengeš, Novartis Pharmaceutical Manufacturing LLC, Slovenia
| | - Aleksander Krajnc
- Novartis Technical Research & Development, Biologics Technical Development Mengeš, Novartis Pharmaceutical Manufacturing LLC, Slovenia.
| |
Collapse
|
10
|
He Z, Zhou J, Dong C, Song C, Liao W, Xiong Y, Yang S. Machine learning and 4D-LFQ quantitative proteomic analysis explore the molecular mechanism of kidney stone formation. Heliyon 2024; 10:e34405. [PMID: 39114033 PMCID: PMC11305192 DOI: 10.1016/j.heliyon.2024.e34405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Background Nephrolithiasis, a common and chronic urological condition, exerts significant pressure on both the general public and society as a whole. The precise mechanisms of nephrolith formation remain inadequately comprehended. Nevertheless, the utilization of proteomics methods has not been employed to examine the development of renal calculi in order to efficiently hinder and manage the creation and reappearance of nephrolith. Nowadays, with the rapid development of proteomics techniques, more efficient and more accurate proteomics technique is utilized to uncover the mechanisms underlying diseases. The objective of this study was to investigate the possible alterations of HK-2 cells when exposed to varying amounts of calcium oxalate (CaOx). The aim was to understand the precise development of stone formation and recurrence, in order to find effective preventive and treatment methods. Methods To provide a complete view of the proteins involved in the development of nephrolithiasis, we utilized an innovative proteomics method called 4D-LFQ proteomic quantitative techniques. HK-2 cells were selected as our experimental subjects. Three groups (n = 3) of HK-2 cells were treated with intervention solutions containing 0 (negative control, NC), 1 mM, and 2 mM CaOx, respectively. For the proteins that showed differential expression, various analyses were conducted including examination of Gene Ontology (GO), Clusters of Orthologous Groups of proteins (KOG), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, enrichment analysis of protein domains, and hierarchical clustering analysis. The STRING database was used to identify the interaction network of the chosen proteins. Candidate proteins were validated using parallel reaction monitoring (PRM) in the end. Results All three groups verified the repeatability of samples. According to the results of 4D-LFQ proteomic quantitative analysis, there were 120, 262, and 81 differentially expressed proteins (DEPs) in the 1 mM-VS-NC, 2 mM-VS-NC, and 2 mM-VS-1mM conditions, respectively. According to GO annotation, the functional enrichment analysis indicates that the differentially expressed proteins (DEPs) were notably enriched in promoting cell migration and the extracellular matrix, among other functions. Analysis of enrichment, based on the KEGG pathway, revealed significant enrichment of DEPs in complement and coagulation cascades, as well as in ECM-receptor (extracellular matrix-receptor) interaction and other related pathways. 14 DEPs of great interest were selected as candidate proteins, including FN1, TFRC, ITGA3, FBN1, HYOU1, SPP1, HSPA5, COL6A1, MANF, HIP1R, JUP, AXL, CTNNB1 and DSG2.The data from PRM demonstrated the variation trend of 14 DEPs was identical as 4D-LFQ proteomic quantitative analysis. Conclusion Proteomics studies of CaOx-induced HK-2 cells using 4D-LFQ proteomic quantitative analysis and PRM may help to provide crucial potential target proteins and signaling pathways for elucidating the mechanism of nephrolithiasis and better treating nephrolithiasis.
Collapse
Affiliation(s)
| | | | | | - Chao Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Wenbiao Liao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Yunhe Xiong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Sixing Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| |
Collapse
|
11
|
Lynch TL, Marin VL, McClure RA, Phipps C, Ronau JA, Rouhimoghadam M, Adams AM, Kandi S, Wolke ML, Shergalis AG, Potts GK, Nacham O, Richardson P, Kakavas SJ, Chhor G, Jenkins GJ, Woller KR, Warder SE, Vasudevan A, Reitsma JM. Quantitative Measurement of Rate of Targeted Protein Degradation. ACS Chem Biol 2024; 19:1604-1615. [PMID: 38980123 DOI: 10.1021/acschembio.4c00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Targeted protein degradation (TPD) is a therapeutic approach that leverages the cell's natural machinery to degrade targets instead of inhibiting them. This is accomplished by using mono- or bifunctional small molecules designed to induce the proximity of target proteins and E3 ubiquitin ligases, leading to ubiquitination and subsequent proteasome-dependent degradation of the target. One of the most significant attributes of the TPD approach is its proposed catalytic mechanism of action, which permits substoichiometric exposure to achieve the desired pharmacological effects. However, apart from one in vitro study, studies supporting the catalytic mechanism of degraders are largely inferred based on potency. A more comprehensive understanding of the degrader catalytic mechanism of action can help aspects of compound development. To address this knowledge gap, we developed a workflow for the quantitative measurement of the catalytic rate of degraders in cells. Comparing a selective and promiscuous BTK degrader, we demonstrate that both compounds function as efficient catalysts of BTK degradation, with the promiscuous degrader exhibiting faster rates due to its ability to induce more favorable ternary complexes. By leveraging computational modeling, we show that the catalytic rate is highly dynamic as the target is depleted from cells. Further investigation of the promiscuous kinase degrader revealed that the catalytic rate is a better predictor of optimal degrader activity toward a specific target compared to degradation magnitude alone. In summary, we present a versatile method for mapping the catalytic activity of any degrader for TPD in cells.
Collapse
Affiliation(s)
- Thomas L Lynch
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Violeta L Marin
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Ryan A McClure
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Colin Phipps
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Judith A Ronau
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Milad Rouhimoghadam
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Ashley M Adams
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Soumya Kandi
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Malerie L Wolke
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Andrea G Shergalis
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Gregory K Potts
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Omprakash Nacham
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Paul Richardson
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Stephan J Kakavas
- Target Enabling Technologies, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Gekleng Chhor
- Target Enabling Technologies, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Gary J Jenkins
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Kevin R Woller
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Scott E Warder
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Anil Vasudevan
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Justin M Reitsma
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| |
Collapse
|
12
|
Turko IV. Quantitative Analysis of Complement Membrane Attack Complex Proteins Associated with Extracellular Vesicles. Proteomes 2024; 12:21. [PMID: 39051239 PMCID: PMC11270256 DOI: 10.3390/proteomes12030021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
Extracellular vesicles (EVs) represent a universal mechanism of intercellular communication in normal and pathological conditions. There are reports showing the presence of complement proteins in EV preparations, specifically those that can form a membrane attack complex (MAC). In the present work, we have used a quantitative mass spectrometry method that allows for the measurement of multiple targeted proteins in one experimental run. The quantification of MAC-forming proteins, namely C5b, C6, C7, C8, and C9, in highly purified EVs from normal human plasma revealed the presence of MAC proteins at approximately equal stoichiometry that does not fit the expected stoichiometry of preformed MAC. We concluded that while MAC proteins can be associated with EVs from normal plasma and presumably can be delivered to the recipient cells, there is no evidence that the EVs carry preformed MAC.
Collapse
Affiliation(s)
- Illarion V Turko
- Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology, University of Maryland, Rockville, ML 20850, USA
| |
Collapse
|
13
|
Chiu KY, Ai Y, Tanim-Ai Hassan M, Li X, Gunawardena HP, Chen H. Standards-Free Absolute Quantitation of Oxidizable Glycopeptides by Coulometric Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1441-1450. [PMID: 38815255 DOI: 10.1021/jasms.4c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Currently, glycopeptide quantitation is mainly based on relative quantitation due to absolute quantitation requiring isotope-labeled or standard glycopeptides which may not be commercially available or are very costly and time consuming to synthesize. To address this grand challenge, coulometric mass spectrometry (CMS), based on the combination of electrochemistry (EC) and mass spectrometry (MS), was utilized to quantify electrochemically active glycopeptides without the need of using standard materials. In this study, we studied tyrosine-containing glycopeptides, NYIVGQPSS(β-GlcNAc)TGNL-OH and NYSVPSS(β-GlcNAc)TGNL-OH, and successfully quantified them directly with CMS with a discrepancy of less than 5% between the CMS measured amount and the theoretical amount. Taking one step further, we applied this approach to quantify glycopeptides generated from the digestion of NIST mAb, a monoclonal antibody reference material. Through HILIC column separation, five N297 glycopeptides resulting from NIST mAb tryptic digestion were successfully separated and quantified by CMS for an absolute amount without the use of any standard materials. This study indicates the potential utility of CMS for quantitative proteomics research.
Collapse
Affiliation(s)
- Kai-Yuan Chiu
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Yongling Ai
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Md Tanim-Ai Hassan
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Xuanwen Li
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Harsha P Gunawardena
- The Janssen Pharmaceutical Companies of Johnson & Johnson, Springhouse, Pennsylvania 19002, United States
| | - Hao Chen
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| |
Collapse
|
14
|
Pierre N, Huynh-Thu VA, Baiwir D, Vieujean S, Bequet E, Reenaers C, Van Kemseke C, Salée C, Massot C, Fléron M, Mazzucchelli G, Trzpiot L, Eppe G, De Pauw E, Louis E, Meuwis MA. Serum proteome signatures associated with ileal and colonic ulcers in Crohn's disease. J Proteomics 2024; 302:105199. [PMID: 38763457 DOI: 10.1016/j.jprot.2024.105199] [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: 03/08/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
At a clinical level, ileal and colonic Crohn's disease (CD) are considered as separate entities. These subphenotypes need to be better supported by biological data to develop personalised medicine in CD. To this end, we combined different technologies (proximity extension assay, selected reaction monitoring, and high-sensitivity turbidimetric immunoassay (hsCRP)) to measure 207 immune-related serum proteins in CD patients presenting no endoscopic lesions (endoscopic remission) (n = 23), isolated ileal ulcers (n = 17), or isolated colonic ulcers (n = 16). We showed that isolated ileal ulcers and isolated colonic ulcers were specifically associated with 6 and 18 serum proteins, respectively: (high level: JUN, CNTNAP2; low level: FCRL6, LTA, CLEC4A, NTF4); (high level: hsCRP, IL6, APCS, CFB, MBL2, IL7, IL17A, CCL19, CXCL10, CSF3, IL10, CLEC4G, MMP12, VEGFA; low level: CLEC3B, GSN, TNFSF12, TPSAB1). Isolated ileal ulcers and isolated colonic ulcers were detected by hsCRP with an area under the receiver operating characteristics curve of 0.64 (p-value = 0.07) and 0.77 (p-value = 0.001), respectively. We highlighted distinct serum proteome profiles associated with ileal and colonic ulcers in CD, this finding might support the development of therapeutics and biomarkers tailored to disease location. SIGNIFICANCE: Although ileal and colonic Crohn's disease present important clinical differences (eg, progression, response to treatment and reliability of biomarkers), these two entities are managed with the same therapeutic strategy. The biological specificities of ileal and colonic Crohn's disease need to be better characterised to develop more personalised approaches. The present study used robust technologies (selected reaction monitoring, proximity extension assays and turbidimetric immunoassay) to quantify precisely 207 serum immune-related proteins in three groups of Crohn's disease patients presenting: 1) no endoscopic lesions (endoscopic remission) (n = 23); 2) isolated ileal ulcers (n = 17); 3) isolated colonic ulcers (n = 16). We found distinct serum proteome signatures associated with ileal and colonic ulcers. Our findings could foster the development of biomarkers and treatments tailored to Crohn's disease location.
Collapse
Affiliation(s)
- Nicolas Pierre
- Laboratory of Translational Gastroenterology, GIGA-institute, University of Liege, Liege, Belgium.
| | - Vân Anh Huynh-Thu
- Department of Electrical Engineering and Computer Science, University of Liege, Liege, Belgium
| | | | - Sophie Vieujean
- Laboratory of Translational Gastroenterology, GIGA-institute, University of Liege, Liege, Belgium; Hepato-Gastroenterology and Digestive Oncology Department, Liege University Hospital, Liege, Belgium
| | - Emeline Bequet
- Laboratory of Translational Gastroenterology, GIGA-institute, University of Liege, Liege, Belgium; Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Liege University Hospital, Liege, Belgium
| | - Catherine Reenaers
- Hepato-Gastroenterology and Digestive Oncology Department, Liege University Hospital, Liege, Belgium
| | - Catherine Van Kemseke
- Hepato-Gastroenterology and Digestive Oncology Department, Liege University Hospital, Liege, Belgium
| | - Catherine Salée
- Laboratory of Translational Gastroenterology, GIGA-institute, University of Liege, Liege, Belgium
| | - Charlotte Massot
- Laboratory of Translational Gastroenterology, GIGA-institute, University of Liege, Liege, Belgium
| | | | - Gabriel Mazzucchelli
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liege, Liege, Belgium
| | - Lisette Trzpiot
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liege, Liege, Belgium
| | - Gauthier Eppe
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liege, Liege, Belgium
| | - Edwin De Pauw
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liege, Liege, Belgium
| | - Edouard Louis
- Laboratory of Translational Gastroenterology, GIGA-institute, University of Liege, Liege, Belgium; Hepato-Gastroenterology and Digestive Oncology Department, Liege University Hospital, Liege, Belgium
| | - Marie-Alice Meuwis
- Laboratory of Translational Gastroenterology, GIGA-institute, University of Liege, Liege, Belgium; Hepato-Gastroenterology and Digestive Oncology Department, Liege University Hospital, Liege, Belgium
| |
Collapse
|
15
|
Nam D, Ji M, Kang C, Kim H, Yang H, Bok KH, Bae J, Hong J, Lee SW. Wideband PRM: Highly Accurate and Sensitive Method for High-Throughput Targeted Proteomics. Anal Chem 2024; 96:10219-10227. [PMID: 38864836 DOI: 10.1021/acs.analchem.4c00518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Targeted mass spectrometry (MS) approaches, which are powerful methods for uniquely and confidently quantifying a specific panel of proteins in complex biological samples, play a crucial role in validating and clinically translating protein biomarkers discovered through global proteomic profiling. Common targeted MS methods, such as multiple reaction monitoring (MRM) and parallel-reaction monitoring (PRM), employ specific mass spectrometric technologies to quantify protein levels by comparing the transitions of surrogate endogenous (ENDO) peptides with those of stable isotope-labeled (SIL) peptide counterparts. These methods utilizing amino acid analyzed (AAA) SIL peptides warrant sensitive and precise measurements required for targeted MS assays. Compared with MRM, PRM provides higher experimental throughput by simultaneously acquiring all transitions of the target peptides and thereby compensates for different ion suppressions among transitions of a target peptide. However, PRM still suffers different ion suppressions between ENDO and SIL peptides due to spray instability, as the ENDO and SIL peptides were monitored at different liquid chromatography (LC) retention times. Here we introduce a new targeted MS method, termed wideband PRM (WBPRM), that is designed for high-throughput targeted MS analysis. WBPRM employs a wide isolation window for simultaneous fragmentation of both ENDO and SIL peptides along with multiplexed single ion monitoring (SIM) scans for enhanced MS sensitivity of the target peptides. Compared with PRM, WBPRM was demonstrated to provide increased sensitivity, precision, and reproducibility of quantitative measurements of target peptides with increased throughput, allowing more target peptide measurements in a shortened experiment time. WBPRM is a straightforward adaptation to a manufacturer-provided MS method, making it an easily implementable technique, particularly in complex biological samples where the demand for higher precision, sensitivity, and efficiency is paramount.
Collapse
Affiliation(s)
- Dowoon Nam
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Minyoung Ji
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Chaewon Kang
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Hokeun Kim
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Hyunju Yang
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Kwon Hee Bok
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Jingi Bae
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Jiwon Hong
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Sang-Won Lee
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
16
|
Whiteaker JR, Zhao L, Schoenherr RM, Huang D, Kennedy JJ, Ivey RG, Lin C, Lorentzen TD, Colantonio S, Caceres TW, Roberts RR, Knotts JG, Reading JJ, Perry CD, Garcia-Buntley SS, Bocik W, Hewitt SM, Paulovich AG. Characterization of an expanded set of assays for immunomodulatory proteins using targeted mass spectrometry. Sci Data 2024; 11:682. [PMID: 38918394 PMCID: PMC11199596 DOI: 10.1038/s41597-024-03467-x] [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: 02/01/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024] Open
Abstract
Immunotherapies are revolutionizing cancer care, but many patients do not achieve durable responses and immune-related adverse events are difficult to predict. Quantifying the hundreds of proteins involved in cancer immunity has the potential to provide biomarkers to monitor and predict tumor response. We previously developed robust, multiplexed quantitative assays for immunomodulatory proteins using targeted mass spectrometry, providing measurements that can be performed reproducibly and harmonized across laboratories. Here, we expand upon those efforts in presenting data from a multiplexed immuno-oncology (IO)-3 assay panel targeting 43 peptides representing 39 immune- and inflammation-related proteins. A suite of novel monoclonal antibodies was generated as assay reagents, and the fully characterized antibodies are made available as a resource to the community. The publicly available dataset contains complete characterization of the assay performance, as well as the mass spectrometer parameters and reagent information necessary for implementation of the assay. Quantification of the proteins will provide benefit to correlative studies in clinical trials, identification of new biomarkers, and improve understanding of the immune response in cancer.
Collapse
Affiliation(s)
- Jeffrey R Whiteaker
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lei Zhao
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Regine M Schoenherr
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dongqing Huang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jacob J Kennedy
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Richard G Ivey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenwei Lin
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Travis D Lorentzen
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Simona Colantonio
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tessa W Caceres
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Rhonda R Roberts
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Joseph G Knotts
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Joshua J Reading
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Candice D Perry
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Sandra S Garcia-Buntley
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - William Bocik
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Stephen M Hewitt
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Amanda G Paulovich
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
17
|
Shin D, Kim Y, Park J, Kim Y. High-throughput Proteomics-Guided Biomarker Discovery of Hepatocellular Carcinoma. Biomed J 2024:100752. [PMID: 38901798 DOI: 10.1016/j.bj.2024.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Liver cancer stands as the fifth leading cause of cancer-related deaths globally. Hepatocellular carcinoma (HCC) comprises approximately 85%-90% of all primary liver malignancies. However, only 20-30% of HCC patients qualify for curative therapy, primarily due to the absence of reliable tools for early detection and prognosis of HCC. This underscores the critical need for molecular biomarkers for HCC management. Since proteins reflect disease status directly, proteomics has been utilized in biomarker developments for HCC. In particular, proteomics coupled with liquid chromatography-mass spectrometer (LC-MS) methods facilitate the process of discovering biomarker candidates for diagnosis, prognosis, and therapeutic strategies. In this work, we investigated LC-MS-based proteomics methods through recent reference reviews, with a particular focus on sample preparation and LC-MS methods appropriate for the discovery of HCC biomarkers and their clinical applications. We classified proteomics studies of HCC according to sample types, and we examined the coverage of protein biomarker candidates based on LC-MS methods in relation to study scales and goals. Comprehensively, we proposed protein biomarker candidates categorized by sample types and biomarker types for appropriate clinical use. In this review, we summarized recent LC-MS-based proteomics studies on HCC and proposed potential protein biomarkers. Our findings are expected to expand the understanding of HCC pathogenesis and enhance the efficiency of HCC diagnosis and prognosis, thereby contributing to improved patient outcomes.
Collapse
Affiliation(s)
- Dongyoon Shin
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea
| | - Yeongshin Kim
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Junho Park
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea; Department of Pharmacology, School of Medicine, CHA University, Seongnam, Republic of Korea.
| | - Youngsoo Kim
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
18
|
Liu W, Wang X, Yu H, Yan G, Shen S, Gao M, Zhang X. Integrated Platform for Large-Scale Quantitative Profiling of Phosphotyrosine Signaling Complexes Based on Cofractionation/Mass Spectrometry and Complex-Centric Algorithm. Anal Chem 2024; 96:9849-9858. [PMID: 38836774 DOI: 10.1021/acs.analchem.4c00285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The scarcity and dynamic nature of phosphotyrosine (pTyr)-modified proteins pose a challenge for researching protein complexes with pTyr modification, which are assembled through multiple protein-protein interactions. We developed an integrated complex-centric platform for large-scale quantitative profiling of pTyr signaling complexes based on cofractionation/mass spectrometry (CoFrac-MS) and a complex-centric algorithm. We initially constructed a trifunctional probe based on pTyr superbinder (SH2-S) for specifically binding and isolation of intact pTyr protein complexes. Then, the CoFrac-MS strategy was employed for the identification of pTyr protein complexes by integrating ion exchange chromatography in conjunction with data independent acquisition mass spectrometry. Furthermore, we developed a novel complex-centric algorithm for quantifying protein complexes based on the protein complex elution curve. Utilizing this algorithm, we effectively quantified 216 putative protein complexes. We further screened 21 regulated pTyr protein complexes related to the epidermal growth factor signal. Our study engenders a comprehensive framework for the intricate examination of pTyr protein complexes and presents, for the foremost occasion, a quantitative landscape delineating the composition of pTyr protein complexes in HeLa cells.
Collapse
Affiliation(s)
- Wei Liu
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Xuantang Wang
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Hailong Yu
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Guoquan Yan
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Mingxia Gao
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Xiangmin Zhang
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
19
|
Hsu WH, Cheng KW, Feng TH, Chen JY, Chen GY, Chen LY, Weng T, Hsu CC. Rapid Screening of New Psychoactive Substances Using pDART-QqQ-MS. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1370-1376. [PMID: 38652738 PMCID: PMC11157655 DOI: 10.1021/jasms.4c00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Drug abuse is a severe social problem worldwide. Particularly, the issue of new psychoactive substances (NPSs) have increasingly emerged. NPSs are structural or functional analogs of traditional illicit drugs, such as cocaine, cannabis, and amphetamine; these molecules provide the same or more severe neurological effects. Usually, immunoassays are utilized in the preliminary screening method. However, NPSs have poor detectability in commercially available immunoassay kits. Meanwhile, various chromatography combined with the mass spectrometry platform have been developed to quantify NPSs. Still, a significant amount of time and resources are required during these procedures. Therefore, we established a rapid analytical platform for NPSs employing paper-loaded direct analysis in real time triple quadrupole mass spectrometry (pDART-QqQ-MS). We implemented this platform for the semiquantitative analysis of forensic drug tests in urine. This platform significantly shrinks the analytical time of a single sample within 30 s and requires a low volume of the specimen. The platform can detect 21 NPSs in urine mixtures at a lower limit of qualification of concentration ranging from 20 to 75 nanograms per milliliter (ng mL-1) and is lower than the cutoff value of currently available immune-based devices for detecting multiple drugs (1000 ng mL-1). Urine samples from drug addicts have been collected to verify the platform's effectiveness. By combining efficiency and accuracy, our platform offers a promising solution for addressing the challenges posed by NPSs in drug abuse detection.
Collapse
Affiliation(s)
- Wei-Hsin Hsu
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Kai-Wen Cheng
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Tzu-Hsuan Feng
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Ju-Yu Chen
- Forensic
and Clinical Toxicology Center National Taiwan University College
of Medicine and National Taiwan University Hospital, Taipei 10051, Taiwan
| | - Guan-Yuan Chen
- Forensic
and Clinical Toxicology Center National Taiwan University College
of Medicine and National Taiwan University Hospital, Taipei 10051, Taiwan
- Department
and Graduate Institute of Forensic Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Lian-Yu Chen
- Institute
of Epidemiology and Preventive Medicine, National Taiwan University, Taipei 10051, Taiwan
- Kunming
Prevention and Control Center, Taipei City
Hospital, Taipei 108203, Taiwan
| | - Te−I Weng
- Forensic
and Clinical Toxicology Center National Taiwan University College
of Medicine and National Taiwan University Hospital, Taipei 10051, Taiwan
- Department
and Graduate Institute of Forensic Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Cheng-Chih Hsu
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Leeuwenhoek
Laboratories Co. Ltd., No. 71, Fanglan Rd, Taipei, 106038, Taiwan
| |
Collapse
|
20
|
Capuano A, D’Urso G, Gazzillo E, Lauro G, Chini MG, D’Auria MV, Ferraro MG, Iazzetti F, Irace C, Bifulco G, Casapullo A. Fatty Acid Synthase as Interacting Anticancer Target of the Terpenoid Myrianthic Acid Disclosed by MS-Based Proteomics Approaches. Int J Mol Sci 2024; 25:5918. [PMID: 38892106 PMCID: PMC11172900 DOI: 10.3390/ijms25115918] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
This research focuses on the target deconvolution of the natural compound myrianthic acid, a triterpenoid characterized by an ursane skeleton isolated from the roots of Myrianthus arboreus and from Oenothera maritima Nutt. (Onagraceae), using MS-based chemical proteomic techniques. Application of drug affinity responsive target stability (DARTS) and targeted-limited proteolysis coupled to mass spectrometry (t-LiP-MS) led to the identification of the enzyme fatty acid synthase (FAS) as an interesting macromolecular counterpart of myrianthic acid. This result, confirmed by comparison with the natural ursolic acid, was thoroughly investigated and validated in silico by molecular docking, which gave a precise picture of the interactions in the MA/FAS complex. Moreover, biological assays showcased the inhibitory activity of myrianthic acid against the FAS enzyme, most likely related to its antiproliferative activity towards tumor cells. Given the significance of FAS in specific pathologies, especially cancer, the myrianthic acid structural moieties could serve as a promising reference point to start the potential development of innovative approaches in therapy.
Collapse
Affiliation(s)
- Alessandra Capuano
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
- PhD Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Gilda D’Urso
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| | - Erica Gazzillo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
- PhD Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, C.da Fonte Lappone, 86090 Pesche, Italy
| | - Maria Valeria D’Auria
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy;
| | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy;
| | - Federica Iazzetti
- Biochem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (F.I.); (C.I.)
| | - Carlo Irace
- Biochem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (F.I.); (C.I.)
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| |
Collapse
|
21
|
Ross RB, Gadwa J, Yu J, Darragh LB, Knitz MW, Nguyen D, Olimpo NA, Abdelazeem KN, Nguyen A, Corbo S, Van Court B, Beynor J, Neupert B, Saviola AJ, D'Alessandro A, Karam SD. PPARα Agonism Enhances Immune Response to Radiotherapy While Dietary Oleic Acid Results in Counteraction. Clin Cancer Res 2024; 30:1916-1933. [PMID: 38363297 PMCID: PMC11061609 DOI: 10.1158/1078-0432.ccr-23-3433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/12/2024] [Accepted: 02/14/2024] [Indexed: 02/17/2024]
Abstract
PURPOSE Head and neck cancer (HNC) improvements are stagnant, even with advances in immunotherapy. Our previous clinical trial data show that altered fatty acid (FA) metabolism correlates with outcome. We hypothesized that pharmacologic and dietary modulation of FA catabolism will affect therapeutic efficacy. EXPERIMENTAL DESIGN We performed in vivo and in vitro experiments using PPARα agonism with fenofibrate (FF) or high oleic acid diets (OAD) with radiotherapy, generating metabolomic, proteomic, stable isotope tracing, extracellular flux analysis, and flow-cytometric data to investigate these alterations. RESULTS FF improved antitumor efficacy of high dose per fraction radiotherapy in HNC murine models, whereas the OAD reversed this effect. FF-treated mice on the control diet had evidence of increased FA catabolism. Stable isotope tracing showed less glycolytic utilization by ex vivo CD8+ T cells. Improved efficacy correlated with intratumoral alterations in eicosanoid metabolism and downregulated mTOR and CD36. CONCLUSIONS Metabolic intervention with increased FA catabolism improves the efficacy of HNC therapy and enhances antitumoral immune response.
Collapse
Affiliation(s)
- Richard Blake Ross
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Jacob Gadwa
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Justin Yu
- Department of Otolaryngology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Laurel B. Darragh
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Michael W. Knitz
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Diemmy Nguyen
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Nicholas A. Olimpo
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Khalid N.M. Abdelazeem
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
- Radiation Biology Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Alexander Nguyen
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Sophia Corbo
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Benjamin Van Court
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Jessica Beynor
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Brooke Neupert
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Anthony J. Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Sana D. Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Center, Aurora, Colorado
- Department of Immunology, University of Colorado Anschutz Medical Center, Aurora, Colorado
| |
Collapse
|
22
|
Son A, Kim W, Lee W, Park J, Kim H. Applicability of selected reaction monitoring for precise screening tests. Expert Rev Proteomics 2024; 21:237-246. [PMID: 38697802 DOI: 10.1080/14789450.2024.2350975] [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: 10/27/2023] [Accepted: 03/27/2024] [Indexed: 05/05/2024]
Abstract
INTRODUCTION The proactive identification of diseases through screening tests has long been endorsed as a means to preempt symptomatic onset. However, such screening endeavors are fraught with complications, such as diagnostic inaccuracies, procedural risks, and patient unease during examinations. These challenges are amplified when screenings for multiple diseases are administered concurrently. Selected Reaction Monitoring (SRM) offers a unique advantage, allowing for the high-throughput quantification of hundreds of analytes with minimal interferences. AREAS COVERED Our research posits that SRM-based assays, traditionally tailored for single-disease biomarker profiling, can be repurposed for multi-disease screening. This innovative approach has the potential to substantially alleviate time, labor, and cost demands on healthcare systems and patients alike. Nonetheless, there are formidable methodological hurdles to overcome. These include difficulties in detecting low-abundance proteins and the risk of model overfitting due to the multiple functionalities of single proteins across different disease spectrums - issues especially pertinent in blood-based assays where detection sensitivity is constrained. As we move forward, technological strides in sample preparation, online extraction, throughput, and automation are expected to ameliorate these limitations. EXPERT OPINION The maturation of mass spectrometry's integration into clinical laboratories appears imminent, positioning it as an invaluable asset for delivering highly sensitive, reproducible, and precise diagnostic results.
Collapse
Affiliation(s)
- Ahrum Son
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Woojin Kim
- Department of Bio-AI convergence Chungnam National University,Daejeon, South Korea
| | - Wonseok Lee
- Department of Bio-AI convergence Chungnam National University,Daejeon, South Korea
| | - Jongham Park
- Department of Bio-AI convergence Chungnam National University,Daejeon, South Korea
| | - Hyunsoo Kim
- Department of Bio-AI convergence Chungnam National University,Daejeon, South Korea
- Department of Convergent Bioscience and Informatics, Chungnam National University, Daejeon, Republic of Korea
- SCICS, Daejeon, Republic of Korea
| |
Collapse
|
23
|
Zhang Q, Liu L. Novel insights into small open reading frame-encoded micropeptides in hepatocellular carcinoma: A potential breakthrough. Cancer Lett 2024; 587:216691. [PMID: 38360139 DOI: 10.1016/j.canlet.2024.216691] [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: 10/23/2023] [Revised: 01/13/2024] [Accepted: 01/27/2024] [Indexed: 02/17/2024]
Abstract
Traditionally, non-coding RNAs (ncRNAs) are regarded as a class of RNA transcripts that lack encoding capability; however, advancements in technology have revealed that some ncRNAs contain small open reading frames (sORFs) that are capable of encoding micropeptides of approximately 150 amino acids in length. sORF-encoded micropeptides (SEPs) have emerged as intriguing entities in hepatocellular carcinoma (HCC) research, shedding light on this previously unexplored realm. Recent studies have highlighted the regulatory functions of SEPs in the occurrence and progression of HCC. Some SEPs exhibit inhibitory effects on HCC, but others facilitate its development. This discovery has revolutionized the landscape of HCC research and clinical management. Here, we introduce the concept and characteristics of SEPs, summarize their associations with HCC, and elucidate their carcinogenic mechanisms in HCC metabolism, signaling pathways, cell proliferation, and metastasis. In addition, we propose a step-by-step workflow for the investigation of HCC-associated SEPs. Lastly, we discuss the challenges and prospects of applying SEPs in the diagnosis and treatment of HCC. This review aims to facilitate the discovery, optimization, and clinical application of HCC-related SEPs, inspiring the development of early diagnostic, individualized, and precision therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Qiangnu Zhang
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), 518020, Shenzhen, China
| | - Liping Liu
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), 518020, Shenzhen, China.
| |
Collapse
|
24
|
Carvalho SB, Profit L, Krishnan S, Gomes RA, Alexandre BM, Clavier S, Hoffman M, Brower K, Gomes-Alves P. SWATH-MS as a strategy for CHO host cell protein identification and quantification supporting the characterization of mAb purification platforms. J Biotechnol 2024; 384:1-11. [PMID: 38340900 DOI: 10.1016/j.jbiotec.2024.02.001] [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: 10/18/2023] [Revised: 01/17/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
Host cell proteins (HCPs) are process-related impurities expressed by the host cells during biotherapeutics' manufacturing, such as monoclonal antibodies (mAbs). Some challenging HCPs evade clearance during the downstream processing and can be co-purified with the molecule of interest, which may impact product stability, efficacy, and safety. Therefore, HCP content is a critical quality attribute to monitor and quantify across the bioprocess. Here we explored a mass spectrometry (MS)-based proteomics tool, the sequential window acquisition of all theoretical fragment-ion spectra (SWATH) strategy, as an orthogonal method to traditional ELISA. The SWATH workflow was applied for high-throughput individual HCP identification and quantification, supporting characterization of a mAb purification platform. The design space of HCP clearance of two polishing resins was evaluated through a design of experiment study. Absolute quantification of high-risk HCPs was achieved (reaching 1.8 and 4.2 ppm limits of quantification, for HCP A and B respectively) using HCP-specific synthetic heavy labeled peptide calibration curves. Profiling of other HCPs was also possible using an average calibration curve (using labeled peptides from different HCPs). The SWATH approach is a powerful tool for HCP assessment during bioprocess development enabling simultaneous monitoring and quantification of different individual HCPs and improving process understanding of their clearance.
Collapse
Affiliation(s)
- Sofia B Carvalho
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras 2780-901, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. Da República, Oeiras 2780-157, Portugal
| | - Ludivine Profit
- Mammalian Platform, Global CMC Development, Sanofi R&D, Vitry-sur-Seine, France
| | - Sushmitha Krishnan
- Mammalian Platform, Global CMC Development, Sanofi R&D, Framingham, MA, USA
| | - Ricardo A Gomes
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras 2780-901, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. Da República, Oeiras 2780-157, Portugal
| | - Bruno M Alexandre
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras 2780-901, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. Da República, Oeiras 2780-157, Portugal
| | - Severine Clavier
- BioAnalytics, Global CMC Development, Sanofi R&D, Vitry-sur-Seine, France
| | - Michael Hoffman
- Mammalian Platform, Global CMC Development, Sanofi R&D, Framingham, MA, USA
| | - Kevin Brower
- Mammalian Platform, Global CMC Development, Sanofi R&D, Framingham, MA, USA.
| | - Patrícia Gomes-Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras 2780-901, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. Da República, Oeiras 2780-157, Portugal.
| |
Collapse
|
25
|
Huang CF, Kline JT, Negrão F, Robey MT, Toby TK, Durbin KR, Fellers RT, Friedewald JJ, Levitsky J, Abecassis MMI, Melani RD, Kelleher NL, Fornelli L. Targeted Quantification of Proteoforms in Complex Samples by Proteoform Reaction Monitoring. Anal Chem 2024; 96:3578-3586. [PMID: 38354049 PMCID: PMC11008684 DOI: 10.1021/acs.analchem.3c05578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Existing mass spectrometric assays used for sensitive and specific measurements of target proteins across multiple samples, such as selected/multiple reaction monitoring (SRM/MRM) or parallel reaction monitoring (PRM), are peptide-based methods for bottom-up proteomics. Here, we describe an approach based on the principle of PRM for the measurement of intact proteoforms by targeted top-down proteomics, termed proteoform reaction monitoring (PfRM). We explore the ability of our method to circumvent traditional limitations of top-down proteomics, such as sensitivity and reproducibility. We also introduce a new software program, Proteoform Finder (part of ProSight Native), specifically designed for the easy analysis of PfRM data. PfRM was initially benchmarked by quantifying three standard proteins. The linearity of the assay was shown over almost 3 orders of magnitude in the femtomole range, with limits of detection and quantification in the low femtomolar range. We later applied our multiplexed PfRM assay to complex samples to quantify biomarker candidates in peripheral blood mononuclear cells (PBMCs) from liver-transplanted patients, suggesting their possible translational applications. These results demonstrate that PfRM has the potential to contribute to the accurate quantification of protein biomarkers for diagnostic purposes and to improve our understanding of disease etiology at the proteoform level.
Collapse
Affiliation(s)
- Che-Fan Huang
- Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
| | - Jake T Kline
- School of Biological Sciences, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Fernanda Negrão
- Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
| | - Matthew T Robey
- Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
- Proteinaceous, Inc., Evanston, Illinois 60201, United States
| | - Timothy K Toby
- Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
| | - Kenneth R Durbin
- Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
- Proteinaceous, Inc., Evanston, Illinois 60201, United States
| | - Ryan T Fellers
- Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
- Proteinaceous, Inc., Evanston, Illinois 60201, United States
| | - John J Friedewald
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Josh Levitsky
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Michael M I Abecassis
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Rafael D Melani
- Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
| | - Neil L Kelleher
- Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
| | - Luca Fornelli
- School of Biological Sciences, University of Oklahoma, Norman, Oklahoma 73019, United States
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
26
|
Wang H, Ni X, Clark N, Randall K, Boeglin L, Chivukula S, Woo C, DeRosa F, Sun G. Absolute quantitation of human wild-type DNAI1 protein in lung tissue using a nanoLC-PRM-MS-based targeted proteomics approach coupled with immunoprecipitation. Clin Proteomics 2024; 21:8. [PMID: 38311768 PMCID: PMC10840268 DOI: 10.1186/s12014-024-09453-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/20/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Dynein axonemal intermediate chain 1 protein (DNAI1) plays an essential role in cilia structure and function, while its mutations lead to primary ciliary dyskinesia (PCD). Accurate quantitation of DNAI1 in lung tissue is crucial for comprehensive understanding of its involvement in PCD, as well as for developing the potential PCD therapies. However, the current protein quantitation method is not sensitive enough to detect the endogenous level of DNAI1 in complex biological matrix such as lung tissue. METHODS In this study, a quantitative method combining immunoprecipitation with nanoLC-MS/MS was developed to measure the expression level of human wild-type (WT) DNAI1 protein in lung tissue. To our understanding, it is the first immunoprecipitation (IP)-MS based method for absolute quantitation of DNAI1 protein in lung tissue. The DNAI1 quantitation was achieved through constructing a standard curve with recombinant human WT DNAI1 protein spiked into lung tissue matrix. RESULTS This method was qualified with high sensitivity and accuracy. The lower limit of quantitation of human DNAI1 was 4 pg/mg tissue. This assay was successfully applied to determine the endogenous level of WT DNAI1 in human lung tissue. CONCLUSIONS The results clearly demonstrate that the developed assay can accurately quantitate low-abundance WT DNAI1 protein in human lung tissue with high sensitivity, indicating its high potential use in the drug development for DNAI1 mutation-caused PCD therapy.
Collapse
Affiliation(s)
- Hui Wang
- Translate Bio, a Sanofi Company, Lexington, MA, 02421, USA.
| | - Xiaoyan Ni
- Translate Bio, a Sanofi Company, Lexington, MA, 02421, USA
| | - Nicholas Clark
- Translate Bio, a Sanofi Company, Lexington, MA, 02421, USA
| | | | - Lianne Boeglin
- Translate Bio, a Sanofi Company, Lexington, MA, 02421, USA
| | | | - Caroline Woo
- Translate Bio, a Sanofi Company, Lexington, MA, 02421, USA
| | - Frank DeRosa
- Translate Bio, a Sanofi Company, Lexington, MA, 02421, USA
| | - Gang Sun
- Translate Bio, a Sanofi Company, Lexington, MA, 02421, USA.
| |
Collapse
|
27
|
Morretta E, Capuano A, D’Urso G, Voli A, Mozzicafreddo M, Di Gaetano S, Capasso D, Sala M, Scala MC, Campiglia P, Piccialli V, Casapullo A. Identification of Mortalin as the Main Interactor of Mycalin A, a Poly-Brominated C-15 Acetogenin Sponge Metabolite, by MS-Based Proteomics. Mar Drugs 2024; 22:52. [PMID: 38393023 PMCID: PMC10890321 DOI: 10.3390/md22020052] [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: 12/07/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Mycalin A (MA) is a polybrominated C-15 acetogenin isolated from the marine sponge Mycale rotalis. Since this substance displays a strong antiproliferative bioactivity towards some tumour cells, we have now directed our studies towards the elucidation of the MA interactome through functional proteomic approaches, (DARTS and t-LIP-MS). DARTS experiments were performed on Hela cell lysates with the purpose of identifying MA main target protein(s); t-LiP-MS was then applied for an in-depth investigation of the MA-target protein interaction. Both these techniques exploit limited proteolysis coupled with MS analysis. To corroborate LiP data, molecular docking studies were performed on the complexes. Finally, biological and SPR analysis were conducted to explore the effect of the binding. Mortalin (GRP75) was identified as the MA's main interactor. This protein belongs to the Hsp70 family and has garnered significant attention due to its involvement in certain forms of cancer. Specifically, its overexpression in cancer cells appears to hinder the pro-apoptotic function of p53, one of its client proteins, because it becomes sequestered in the cytoplasm. Our research, therefore, has been focused on the possibility that MA might prevent this sequestration, promoting the re-localization of p53 to the nucleus and facilitating the apoptosis of tumor cells.
Collapse
Affiliation(s)
- Elva Morretta
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (E.M.); (A.C.); (G.D.); (A.V.); (M.S.); (M.C.S.); (P.C.)
| | - Alessandra Capuano
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (E.M.); (A.C.); (G.D.); (A.V.); (M.S.); (M.C.S.); (P.C.)
- PhD Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Italy
| | - Gilda D’Urso
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (E.M.); (A.C.); (G.D.); (A.V.); (M.S.); (M.C.S.); (P.C.)
| | - Antonia Voli
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (E.M.); (A.C.); (G.D.); (A.V.); (M.S.); (M.C.S.); (P.C.)
- PhD Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Italy
| | - Matteo Mozzicafreddo
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy;
| | - Sonia Di Gaetano
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy;
| | - Domenica Capasso
- Department of Physics, Ettore Pancini, University of Naples Federico II, Via Cintia 21, 80126 Naples, Italy;
| | - Marina Sala
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (E.M.); (A.C.); (G.D.); (A.V.); (M.S.); (M.C.S.); (P.C.)
| | - Maria Carmina Scala
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (E.M.); (A.C.); (G.D.); (A.V.); (M.S.); (M.C.S.); (P.C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (E.M.); (A.C.); (G.D.); (A.V.); (M.S.); (M.C.S.); (P.C.)
| | - Vincenzo Piccialli
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, 80126 Naples, Italy
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (E.M.); (A.C.); (G.D.); (A.V.); (M.S.); (M.C.S.); (P.C.)
| |
Collapse
|
28
|
Sakson R, Beedgen L, Bernhard P, Alp KM, Lübbehusen N, Röth R, Niesler B, Luzarowski M, Shevchuk O, Mayer MP, Thiel C, Ruppert T. Targeted Proteomics Reveals Quantitative Differences in Low-Abundance Glycosyltransferases of Patients with Congenital Disorders of Glycosylation. Int J Mol Sci 2024; 25:1191. [PMID: 38256263 PMCID: PMC10816918 DOI: 10.3390/ijms25021191] [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: 12/23/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Protein glycosylation is an essential post-translational modification in all domains of life. Its impairment in humans can result in severe diseases named congenital disorders of glycosylation (CDGs). Most of the glycosyltransferases (GTs) responsible for proper glycosylation are polytopic membrane proteins that represent challenging targets in proteomics. We established a multiple reaction monitoring (MRM) assay to comprehensively quantify GTs involved in the processes of N-glycosylation and O- and C-mannosylation in the endoplasmic reticulum. High robustness was achieved by using an enriched membrane protein fraction of isotopically labeled HEK 293T cells as an internal protein standard. The analysis of primary skin fibroblasts from eight CDG type I patients with impaired ALG1, ALG2, and ALG11 genes, respectively, revealed a substantial reduction in the corresponding protein levels. The abundance of the other GTs, however, remained unchanged at the transcript and protein levels, indicating that there is no fail-safe mechanism for the early steps of glycosylation in the endoplasmic reticulum. The established MRM assay was shared with the scientific community via the commonly used open source Skyline software environment, including Skyline Batch for automated data analysis. We demonstrate that another research group could easily reproduce all analysis steps, even while using different LC-MS hardware.
Collapse
Affiliation(s)
- Roman Sakson
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
- Heidelberg Biosciences International Graduate School (HBIGS), Heidelberg University, 69120 Heidelberg, Germany
| | - Lars Beedgen
- Center for Child and Adolescent Medicine, Department Pediatrics I, Heidelberg University, 69120 Heidelberg, Germany
| | - Patrick Bernhard
- Institute for Surgical Pathology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - K. Merve Alp
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Nicole Lübbehusen
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Ralph Röth
- nCounter Core Facility, Institute of Human Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Beate Niesler
- nCounter Core Facility, Institute of Human Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Marcin Luzarowski
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Olga Shevchuk
- Department of Immunodynamics, Institute of Experimental Immunology and Imaging, University Hospital Essen, 45147 Essen, Germany
| | - Matthias P. Mayer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Christian Thiel
- Center for Child and Adolescent Medicine, Department Pediatrics I, Heidelberg University, 69120 Heidelberg, Germany
| | - Thomas Ruppert
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| |
Collapse
|
29
|
Ceccacci S, Corsi L, Spinelli L, Caroli C, Marani M, Anceschi L, Mozzicafreddo M, Pellati F, Monti MC. A label free chemoproteomic-based platform to disclose cannabidiol molecular mechanism of action on chronic myelogenous leukemia cancer cells. Heliyon 2024; 10:e24196. [PMID: 38268604 PMCID: PMC10806336 DOI: 10.1016/j.heliyon.2024.e24196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/26/2024] Open
Abstract
The discovery of the interactome of cannabidiol (CBD), a non-psychoactive cannabinoid from Cannabis sativa L., has been here performed on chronic myelogenous leukemia cancer cells, using an optimized chemo-proteomic stage, which links Drug Affinity Responsive Target Stability with Limited Proteolysis Multiple Reaction Monitoring approaches. The obtained results showed the ability of CBD to target simultaneously some potential protein partners, corroborating its well-known poly-pharmacology activity. In human chronic myelogenous leukemia K562 cancer cells, the most fascinating protein partner was identified as the 116 kDa U5 small nuclear ribonucleoprotein element called EFTUD2, which fits with the spliceosome complex. The binding mode of this oncogenic protein with CBD was clarified using mass spectrometry-based and in silico analysis.
Collapse
Affiliation(s)
- Sara Ceccacci
- Department of Pharmacy, Università di Salerno, Via Giovanni Paolo II, 84084, Fisciano, Italy
- PhD Program in Drug Discovery and Development, Department of Pharmacy, Università di Salerno, Via Giovanni Paolo II, 84084, Fisciano, Italy
| | - Lorenzo Corsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 103/287, 41125, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy
| | - Lucio Spinelli
- Department of Pharmacy, Università di Salerno, Via Giovanni Paolo II, 84084, Fisciano, Italy
- Department of Pharmacy, Università degli Studi di Napoli ‘Federico II’, Via Domenico Montesano, 49, 80131, Napoli, Italy
| | - Clarissa Caroli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 103/287, 41125, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy
| | - Matilde Marani
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 103/287, 41125, Modena, Italy
| | - Lisa Anceschi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 103/287, 41125, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy
| | - Matteo Mozzicafreddo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Federica Pellati
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 103/287, 41125, Modena, Italy
| | - Maria Chiara Monti
- Department of Pharmacy, Università di Salerno, Via Giovanni Paolo II, 84084, Fisciano, Italy
| |
Collapse
|
30
|
Capuano A, D’Urso G, Aliberti M, Ruggiero D, Terracciano S, Festa C, Tosco A, Chini MG, Lauro G, Bifulco G, Casapullo A. Chemoproteomics Reveals USP5 (Ubiquitin Carboxyl-Terminal Hydrolase 5) as Promising Target of the Marine Polyketide Gracilioether A. Mar Drugs 2024; 22:41. [PMID: 38248666 PMCID: PMC10817451 DOI: 10.3390/md22010041] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Mass spectrometry-based chemical proteomic approaches using limited proteolysis have become a powerful tool for the identification and analysis of the interactions between a small molecule (SM) and its protein target(s). Gracilioether A (GeA) is a polyketide isolated from a marine sponge, for which we aimed to trace the interactome using this strategy. DARTS (Drug Affinity Responsive Target Stability) and t-LiP-MS (targeted-Limited Proteolysis-Mass Spectrometry) represented the main techniques used in this study. DARTS was applied on HeLa cell lysate for the identification of the GeA target proteins, and t-LiP-MS was employed to investigate the protein's regions involved in the binding with GeA. The results were complemented through the use of binding studies using Surface Plasmon Resonance (SPR) and in silico molecular docking experiments. Ubiquitin carboxyl-terminal hydrolase 5 (USP5) was identified as a promising target of GeA, and the interaction profile of the USP5-GeA complex was explained. USP5 is an enzyme involved in the pathway of protein metabolism through the disassembly of the polyubiquitin chains on degraded proteins into ubiquitin monomers. This activity is connected to different cellular functions concerning the maintenance of chromatin structure and receptors and the degradation of abnormal proteins and cancerogenic progression. On this basis, this structural information opens the way to following studies focused on the definition of the biological potential of Gracilioether A and the rational development of novel USP5 inhibitors based on a new structural skeleton.
Collapse
Affiliation(s)
- Alessandra Capuano
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; (A.C.); (G.D.); (M.A.); (D.R.); (S.T.); (A.T.); (G.L.); (G.B.)
- PhD Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Salerno, Italy
| | - Gilda D’Urso
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; (A.C.); (G.D.); (M.A.); (D.R.); (S.T.); (A.T.); (G.L.); (G.B.)
| | - Michela Aliberti
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; (A.C.); (G.D.); (M.A.); (D.R.); (S.T.); (A.T.); (G.L.); (G.B.)
- PhD Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Salerno, Italy
| | - Dafne Ruggiero
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; (A.C.); (G.D.); (M.A.); (D.R.); (S.T.); (A.T.); (G.L.); (G.B.)
| | - Stefania Terracciano
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; (A.C.); (G.D.); (M.A.); (D.R.); (S.T.); (A.T.); (G.L.); (G.B.)
| | - Carmen Festa
- Dipartimento di Farmacia, University of Napoli “Federico II”, Via Domenico Montesano 49, 80131 Napoli, Italy;
| | - Alessandra Tosco
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; (A.C.); (G.D.); (M.A.); (D.R.); (S.T.); (A.T.); (G.L.); (G.B.)
| | - Maria Giovanna Chini
- Dipartimento di Bioscienze e Territorio, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy;
| | - Gianluigi Lauro
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; (A.C.); (G.D.); (M.A.); (D.R.); (S.T.); (A.T.); (G.L.); (G.B.)
| | - Giuseppe Bifulco
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; (A.C.); (G.D.); (M.A.); (D.R.); (S.T.); (A.T.); (G.L.); (G.B.)
| | - Agostino Casapullo
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy; (A.C.); (G.D.); (M.A.); (D.R.); (S.T.); (A.T.); (G.L.); (G.B.)
| |
Collapse
|
31
|
Ryu J, Boylan KLM, Twigg CAI, Evans R, Skubitz APN, Thomas SN. Quantification of putative ovarian cancer serum protein biomarkers using a multiplexed targeted mass spectrometry assay. Clin Proteomics 2024; 21:1. [PMID: 38172678 PMCID: PMC10762856 DOI: 10.1186/s12014-023-09447-4] [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: 08/04/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Ovarian cancer is the most lethal gynecologic malignancy in women, and high-grade serous ovarian cancer (HGSOC) is the most common subtype. Currently, no clinical test has been approved by the FDA to screen the general population for ovarian cancer. This underscores the critical need for the development of a robust methodology combined with novel technology to detect diagnostic biomarkers for HGSOC in the sera of women. Targeted mass spectrometry (MS) can be used to identify and quantify specific peptides/proteins in complex biological samples with high accuracy, sensitivity, and reproducibility. In this study, we sought to develop and conduct analytical validation of a multiplexed Tier 2 targeted MS parallel reaction monitoring (PRM) assay for the relative quantification of 23 putative ovarian cancer protein biomarkers in sera. METHODS To develop a PRM method for our target peptides in sera, we followed nationally recognized consensus guidelines for validating fit-for-purpose Tier 2 targeted MS assays. The endogenous target peptide concentrations were calculated using the calibration curves in serum for each target peptide. Receiver operating characteristic (ROC) curves were analyzed to evaluate the diagnostic performance of the biomarker candidates. RESULTS We describe an effort to develop and analytically validate a multiplexed Tier 2 targeted PRM MS assay to quantify candidate ovarian cancer protein biomarkers in sera. Among the 64 peptides corresponding to 23 proteins in our PRM assay, 24 peptides corresponding to 16 proteins passed the assay validation acceptability criteria. A total of 6 of these peptides from insulin-like growth factor-binding protein 2 (IBP2), sex hormone-binding globulin (SHBG), and TIMP metalloproteinase inhibitor 1 (TIMP1) were quantified in sera from a cohort of 69 patients with early-stage HGSOC, late-stage HGSOC, benign ovarian conditions, and healthy (non-cancer) controls. Confirming the results from previously published studies using orthogonal analytical approaches, IBP2 was identified as a diagnostic biomarker candidate based on its significantly increased abundance in the late-stage HGSOC patient sera compared to the healthy controls and patients with benign ovarian conditions. CONCLUSIONS A multiplexed targeted PRM MS assay was applied to detect candidate diagnostic biomarkers in HGSOC sera. To evaluate the clinical utility of the IBP2 PRM assay for HGSOC detection, further studies need to be performed using a larger patient cohort.
Collapse
Affiliation(s)
- Joohyun Ryu
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Kristin L M Boylan
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Carly A I Twigg
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Richard Evans
- Clinical and Translational Research Institute, University of Minnesota, Minneapolis, MN, USA
| | - Amy P N Skubitz
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Stefani N Thomas
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA.
| |
Collapse
|
32
|
Whiteaker JR, Zhao L, Kennedy JJ, Ivey RG, Paulovich AG. Targeted Mass Spectrometry for Quantification of Receptor Tyrosine Kinase Signaling. Methods Mol Biol 2024; 2823:253-267. [PMID: 39052225 DOI: 10.1007/978-1-0716-3922-1_16] [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] [Indexed: 07/27/2024]
Abstract
Targeted proteomics enables sensitive and specific quantification of proteins and post-translational modifications. By coupling peptide immunoaffinity enrichment with targeted mass spectrometry, we have developed the methodology for multiplexed quantification of proteins and phosphosites involved in the RAS/MAPK signaling network. The method uses anti-peptide antibodies to enrich analytes and heavy stable isotope-labeled internal standards, spiked in at known concentrations. The enriched peptides are directly measured by multiple-reaction monitoring (MRM), a well-characterized quantitative mass spectrometry-based method. The analyte (light) peptide response is measured relative to the heavy standard. The method described provides quantitative measurements of phospho-signaling and is generally applicable to other phosphopeptides and sample types.
Collapse
Affiliation(s)
| | - Lei Zhao
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | | | | |
Collapse
|
33
|
Degliesposti G. Probing Protein Complexes Composition, Stoichiometry, and Interactions by Peptide-Based Mass Spectrometry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 3234:41-57. [PMID: 38507199 DOI: 10.1007/978-3-031-52193-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
The characterization of a protein complex by mass spectrometry can be conducted at different levels. Initial steps regard the qualitative composition of the complex and subunit identification. After that, quantitative information such as stoichiometric ratios and copy numbers for each subunit in a complex or super-complex is acquired. Peptide-based LC-MS/MS offers a wide number of methods and protocols for the characterization of protein complexes. This chapter concentrates on the applications of peptide-based LC-MS/MS for the qualitative, quantitative, and structural characterization of protein complexes focusing on subunit identification, determination of stoichiometric ratio and number of subunits per complex as well as on cross-linking mass spectrometry and hydrogen/deuterium exchange as methods for the structural investigation of the biological assemblies.
Collapse
|
34
|
Solovyeva EM, Utzinger S, Vissières A, Mitchelmore J, Ahrné E, Hermes E, Poetsch T, Ronco M, Bidinosti M, Merkl C, Serluca FC, Fessenden J, Naumann U, Voshol H, Meyer AS, Hoersch S. Integrative Proteogenomics for Differential Expression and Splicing Variation in a DM1 Mouse Model. Mol Cell Proteomics 2024; 23:100683. [PMID: 37993104 PMCID: PMC10770608 DOI: 10.1016/j.mcpro.2023.100683] [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: 07/31/2022] [Revised: 09/02/2023] [Accepted: 11/17/2023] [Indexed: 11/24/2023] Open
Abstract
Dysregulated mRNA splicing is involved in the pathogenesis of many diseases including cancer, neurodegenerative diseases, and muscular dystrophies such as myotonic dystrophy type 1 (DM1). Comprehensive assessment of dysregulated splicing on the transcriptome and proteome level has been methodologically challenging, and thus investigations have often been targeting only few genes. Here, we performed a large-scale coordinated transcriptomic and proteomic analysis to characterize a DM1 mouse model (HSALR) in comparison to wild type. Our integrative proteogenomics approach comprised gene- and splicing-level assessments for mRNAs and proteins. It recapitulated many known instances of aberrant mRNA splicing in DM1 and identified new ones. It enabled the design and targeting of splicing-specific peptides and confirmed the translation of known instances of aberrantly spliced disease-related genes (e.g., Atp2a1, Bin1, Ryr1), complemented by novel findings (Flnc and Ywhae). Comparative analysis of large-scale mRNA and protein expression data showed quantitative agreement of differentially expressed genes and splicing patterns between disease and wild type. We hence propose this work as a suitable blueprint for a robust and scalable integrative proteogenomic strategy geared toward advancing our understanding of splicing-based disorders. With such a strategy, splicing-based biomarker candidates emerge as an attractive and accessible option, as they can be efficiently asserted on the mRNA and protein level in coordinated fashion.
Collapse
Affiliation(s)
- Elizaveta M Solovyeva
- Research Informatics, Biomedical Research at Novartis, Basel, Switzerland; V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow, Russia.
| | - Stephan Utzinger
- Diseases of Aging and Regenerative Medicine, Biomedical Research at Novartis, Basel, Switzerland
| | | | - Joanna Mitchelmore
- Diseases of Aging and Regenerative Medicine, Biomedical Research at Novartis, Basel, Switzerland
| | - Erik Ahrné
- Discovery Sciences, Biomedical Research at Novartis, Basel, Switzerland
| | - Erwin Hermes
- Discovery Sciences, Biomedical Research at Novartis, Basel, Switzerland
| | - Tania Poetsch
- Discovery Sciences, Biomedical Research at Novartis, Basel, Switzerland
| | - Marie Ronco
- Diseases of Aging and Regenerative Medicine, Biomedical Research at Novartis, Basel, Switzerland
| | - Michael Bidinosti
- Diseases of Aging and Regenerative Medicine, Biomedical Research at Novartis, Basel, Switzerland
| | - Claudia Merkl
- Diseases of Aging and Regenerative Medicine, Biomedical Research at Novartis, Basel, Switzerland
| | - Fabrizio C Serluca
- Research Informatics, Biomedical Research at Novartis, Cambridge, Massachusetts, USA
| | - James Fessenden
- Neurodegenerative Diseases, Biomedical Research at Novartis, Cambridge, Massachusetts, USA
| | - Ulrike Naumann
- Discovery Sciences, Biomedical Research at Novartis, Basel, Switzerland
| | - Hans Voshol
- Discovery Sciences, Biomedical Research at Novartis, Basel, Switzerland
| | - Angelika S Meyer
- Diseases of Aging and Regenerative Medicine, Biomedical Research at Novartis, Basel, Switzerland
| | - Sebastian Hoersch
- Research Informatics, Biomedical Research at Novartis, Basel, Switzerland.
| |
Collapse
|
35
|
Bianco M, Ventura G, Calvano CD, Losito I, Cataldi TRI. Food allergen detection by mass spectrometry: From common to novel protein ingredients. Proteomics 2023; 23:e2200427. [PMID: 37691088 DOI: 10.1002/pmic.202200427] [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: 05/30/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023]
Abstract
Food allergens are molecules, mainly proteins, that trigger immune responses in susceptible individuals upon consumption even when they would otherwise be harmless. Symptoms of a food allergy can range from mild to acute; this last effect is a severe and potentially life-threatening reaction. The European Union (EU) has identified 14 common food allergens, but new allergens are likely to emerge with constantly changing food habits. Mass spectrometry (MS) is a promising alternative to traditional antibody-based assays for quantifying multiple allergenic proteins in complex matrices with high sensitivity and selectivity. Here, the main allergenic proteins and the advantages and drawbacks of some MS acquisition protocols, such as multiple reaction monitoring (MRM) and data-dependent analysis (DDA) for identifying and quantifying common allergenic proteins in processed foodstuffs are summarized. Sections dedicated to novel foods like microalgae and insects as new sources of allergenic proteins are included, emphasizing the significance of establishing stable marker peptides and validated methods using database searches. The discussion involves the in-silico digestion of allergenic proteins, providing insights into their potential impact on immunogenicity. Finally, case studies focussing on microalgae highlight the value of MS as an effective analytical tool for ensuring regulatory compliance throughout the food control chain.
Collapse
Affiliation(s)
- Mariachiara Bianco
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Giovanni Ventura
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
- Centro interdipartimentale SMART, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Cosima D Calvano
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
- Centro interdipartimentale SMART, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Ilario Losito
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
- Centro interdipartimentale SMART, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Tommaso R I Cataldi
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
- Centro interdipartimentale SMART, Università degli Studi di Bari Aldo Moro, Bari, Italy
| |
Collapse
|
36
|
Hocking B, Armstrong A, Mann DJ. Covalent fragment libraries in drug discovery-Design, synthesis, and screening methods. PROGRESS IN MEDICINAL CHEMISTRY 2023; 62:105-146. [PMID: 37981350 DOI: 10.1016/bs.pmch.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
As the development of drugs with a covalent mode of action is becoming increasingly popular, well-validated covalent fragment-based drug discovery (FBDD) methods have been comparatively slow to keep up with the demand. In this chapter the principles of covalent fragment reactivity, library design, synthesis, and screening methods are explored in depth, focussing on literature examples with direct applications to practical covalent fragment library design and screening. Further, questions about the future of the field are explored and potential useful advances are proposed.
Collapse
Affiliation(s)
- Brad Hocking
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Alan Armstrong
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, United Kingdom
| | - David J Mann
- Department of Life Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
37
|
Farkona S, Pastrello C, Konvalinka A. Proteomics: Its Promise and Pitfalls in Shaping Precision Medicine in Solid Organ Transplantation. Transplantation 2023; 107:2126-2142. [PMID: 36808112 DOI: 10.1097/tp.0000000000004539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Solid organ transplantation is an established treatment of choice for end-stage organ failure. However, all transplant patients are at risk of developing complications, including allograft rejection and death. Histological analysis of graft biopsy is still the gold standard for evaluation of allograft injury, but it is an invasive procedure and prone to sampling errors. The past decade has seen an increased number of efforts to develop minimally invasive procedures for monitoring allograft injury. Despite the recent progress, limitations such as the complexity of proteomics-based technology, the lack of standardization, and the heterogeneity of populations that have been included in different studies have hindered proteomic tools from reaching clinical transplantation. This review focuses on the role of proteomics-based platforms in biomarker discovery and validation in solid organ transplantation. We also emphasize the value of biomarkers that provide potential mechanistic insights into the pathophysiology of allograft injury, dysfunction, or rejection. Additionally, we forecast that the growth of publicly available data sets, combined with computational methods that effectively integrate them, will facilitate a generation of more informed hypotheses for potential subsequent evaluation in preclinical and clinical studies. Finally, we illustrate the value of combining data sets through the integration of 2 independent data sets that pinpointed hub proteins in antibody-mediated rejection.
Collapse
Affiliation(s)
- Sofia Farkona
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Chiara Pastrello
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute University Health Network, Toronto, ON, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Ana Konvalinka
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| |
Collapse
|
38
|
Quinn C, Rico MC, Merali C, Barrero CA, Perez-Leal O, Mischley V, Karanicolas J, Friedman SL, Merali S. Secreted folate receptor γ drives fibrogenesis in metabolic dysfunction-associated steatohepatitis by amplifying TGFβ signaling in hepatic stellate cells. Sci Transl Med 2023; 15:eade2966. [PMID: 37756380 DOI: 10.1126/scitranslmed.ade2966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 08/16/2023] [Indexed: 09/29/2023]
Abstract
Hepatic fibrosis is the primary determinant of mortality in patients with metabolic dysfunction-associated steatohepatitis (MASH). Transforming growth factor-β (TGFβ), a master profibrogenic cytokine, is a promising therapeutic target that has not yet been translated into an effective therapy in part because of liabilities associated with systemic TGFβ antagonism. We have identified that soluble folate receptor γ (FOLR3), which is expressed in humans but not in rodents, is a secreted protein that is elevated in the livers of patients with MASH but not in those with metabolic dysfunction-associated steatotic liver disease, those with type II diabetes, or healthy individuals. Global proteomics showed that FOLR3 was the most highly significant MASH-specific protein and was positively correlated with increasing fibrosis stage, consistent with stimulation of activated hepatic stellate cells (HSCs), which are the key fibrogenic cells in the liver. Exposure of HSCs to exogenous FOLR3 led to elevated extracellular matrix (ECM) protein production, an effect synergistically potentiated by TGFβ1. We found that FOLR3 interacts with the serine protease HTRA1, a known regulator of TGFBR, and activates TGFβ signaling. Administration of human FOLR3 to mice induced severe bridging fibrosis and an ECM pattern resembling human MASH. Our study thus uncovers a role of FOLR3 in enhancing fibrosis.
Collapse
Affiliation(s)
- Connor Quinn
- Temple University School of Pharmacy, Philadelphia, PA 19140 USA
| | - Mario C Rico
- Temple University School of Pharmacy, Philadelphia, PA 19140 USA
| | - Carmen Merali
- Temple University School of Pharmacy, Philadelphia, PA 19140 USA
| | - Carlos A Barrero
- Temple University School of Pharmacy, Philadelphia, PA 19140 USA
| | - Oscar Perez-Leal
- Temple University School of Pharmacy, Philadelphia, PA 19140 USA
| | - Victoria Mischley
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
- Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - John Karanicolas
- Temple University School of Pharmacy, Philadelphia, PA 19140 USA
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Salim Merali
- Temple University School of Pharmacy, Philadelphia, PA 19140 USA
| |
Collapse
|
39
|
Sohaei D, Thebault S, Avery LM, Batruch I, Lam B, Xu W, Saadeh RS, Scarisbrick IA, Diamandis EP, Prassas I, Freedman MS. Cerebrospinal fluid camk2a levels at baseline predict long-term progression in multiple sclerosis. Clin Proteomics 2023; 20:33. [PMID: 37644477 PMCID: PMC10466840 DOI: 10.1186/s12014-023-09418-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/28/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) remains a highly unpredictable disease. Many hope that fluid biomarkers may contribute to better stratification of disease, aiding the personalisation of treatment decisions, ultimately improving patient outcomes. OBJECTIVE The objective of this study was to evaluate the predictive value of CSF brain-specific proteins from early in the disease course of MS on long term clinical outcomes. METHODS In this study, 34 MS patients had their CSF collected and stored within 5 years of disease onset and were then followed clinically for at least 15 years. CSF concentrations of 64 brain-specific proteins were analyzed in the 34 patient CSF, as well as 19 age and sex-matched controls, using a targeted liquid-chromatography tandem mass spectrometry approach. RESULTS We identified six CSF brain-specific proteins that significantly differentiated MS from controls (p < 0.05) and nine proteins that could predict disease course over the next decade. CAMK2A emerged as a biomarker candidate that could discriminate between MS and controls and could predict long-term disease progression. CONCLUSION Targeted approaches to identify and quantify biomarkers associated with MS in the CSF may inform on long term MS outcomes. CAMK2A may be one of several candidates, warranting further exploration.
Collapse
Affiliation(s)
- Dorsa Sohaei
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Simon Thebault
- Department of Medicine, The Ottawa Hospital, 01 Smyth Road, Box 601, Ottawa, ON, K1H 8L6, Canada
- The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Lisa M Avery
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Department of Biostatistics, The Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - Ihor Batruch
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Brian Lam
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, United States of America
| | - Wei Xu
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Department of Biostatistics, The Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - Rubah S Saadeh
- Department of Physical Medicine and Rehabilitation, Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Isobel A Scarisbrick
- Department of Physical Medicine and Rehabilitation, Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Mount Sinai Hospital, Joseph & Wolf Lebovic Ctr, 60 Murray St [Box 32]; Flr 6 - Rm L6-201, Toronto, ON, M5T 3L9, Canada.
- Laboratory Medicine Program, University Health Network, Toronto, Canada.
| | - Mark S Freedman
- Department of Medicine, The Ottawa Hospital, 01 Smyth Road, Box 601, Ottawa, ON, K1H 8L6, Canada.
- The Ottawa Hospital Research Institute, Ottawa, Canada.
| |
Collapse
|
40
|
Yeo J, Kang J, Kim H, Moon C. A Critical Overview of HPLC-MS-Based Lipidomics in Determining Triacylglycerol and Phospholipid in Foods. Foods 2023; 12:3177. [PMID: 37685110 PMCID: PMC10486615 DOI: 10.3390/foods12173177] [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: 07/02/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/10/2023] Open
Abstract
With the current advancement in mass spectrometry (MS)-based lipidomics, the knowledge of lipidomes and their diverse roles has greatly increased, enabling a deeper understanding of the action of bioactive lipid molecules in plant- and animal-based foods. This review provides in-depth information on the practical use of MS techniques in lipidomics, including lipid extraction, adduct formation, MS analysis, data processing, statistical analysis, and bioinformatics. Moreover, this contribution demonstrates the effectiveness of MS-based lipidomics for identifying and quantifying diverse lipid species, especially triacylglycerols and phospholipids, in foods. Further, it summarizes the wide applications of MS-based lipidomics in food science, such as for assessing food processing methods, detecting food adulteration, and measuring lipid oxidation in foods. Thus, MS-based lipidomics may be a useful method for identifying the action of individual lipid species in foods.
Collapse
Affiliation(s)
- JuDong Yeo
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea; (J.K.); (H.K.); (C.M.)
| | | | | | | |
Collapse
|
41
|
Park J, Wilkins C, Avtonomov D, Hong J, Back S, Kim H, Shulman N, MacLean BX, Lee SW, Kim S. Targeted proteomics data interpretation with DeepMRM. CELL REPORTS METHODS 2023; 3:100521. [PMID: 37533638 PMCID: PMC10391571 DOI: 10.1016/j.crmeth.2023.100521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/18/2023] [Accepted: 06/15/2023] [Indexed: 08/04/2023]
Abstract
Targeted proteomics is widely utilized in clinical proteomics; however, researchers often devote substantial time to manual data interpretation, which hinders the transferability, reproducibility, and scalability of this approach. We introduce DeepMRM, a software package based on deep learning algorithms for object detection developed to minimize manual intervention in targeted proteomics data analysis. DeepMRM was evaluated on internal and public datasets, demonstrating superior accuracy compared with the community standard tool Skyline. To promote widespread adoption, we have incorporated a stand-alone graphical user interface for DeepMRM and integrated its algorithm into the Skyline software package as an external tool.
Collapse
Affiliation(s)
| | | | | | - Jiwon Hong
- Department of Chemistry, Center for Proteogenomic Research, Korea University, Seoul 02841, Republic of Korea
| | - Seunghoon Back
- Department of Chemistry, Center for Proteogenomic Research, Korea University, Seoul 02841, Republic of Korea
| | - Hokeun Kim
- Department of Chemistry, Center for Proteogenomic Research, Korea University, Seoul 02841, Republic of Korea
| | - Nicholas Shulman
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Brendan X. MacLean
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Sang-Won Lee
- Department of Chemistry, Center for Proteogenomic Research, Korea University, Seoul 02841, Republic of Korea
| | - Sangtae Kim
- Bertis Bioscience, Inc., San Diego, CA 92121, USA
| |
Collapse
|
42
|
Révész Á, Hevér H, Steckel A, Schlosser G, Szabó D, Vékey K, Drahos L. Collision energies: Optimization strategies for bottom-up proteomics. MASS SPECTROMETRY REVIEWS 2023; 42:1261-1299. [PMID: 34859467 DOI: 10.1002/mas.21763] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 06/07/2023]
Abstract
Mass-spectrometry coupled to liquid chromatography is an indispensable tool in the field of proteomics. In the last decades, more and more complex and diverse biochemical and biomedical questions have arisen. Problems to be solved involve protein identification, quantitative analysis, screening of low abundance modifications, handling matrix effect, and concentrations differing by orders of magnitude. This led the development of more tailored protocols and problem centered proteomics workflows, including advanced choice of experimental parameters. In the most widespread bottom-up approach, the choice of collision energy in tandem mass spectrometric experiments has outstanding role. This review presents the collision energy optimization strategies in the field of proteomics which can help fully exploit the potential of MS based proteomics techniques. A systematic collection of use case studies is then presented to serve as a starting point for related further scientific work. Finally, this article discusses the issue of comparing results from different studies or obtained on different instruments, and it gives some hints on methodology transfer between laboratories based on measurement of reference species.
Collapse
Affiliation(s)
- Ágnes Révész
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Helga Hevér
- Chemical Works of Gedeon Richter Plc, Budapest, Hungary
| | - Arnold Steckel
- Department of Analytical Chemistry, MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Institute of Chemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Gitta Schlosser
- Department of Analytical Chemistry, MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Institute of Chemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Dániel Szabó
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Károly Vékey
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - László Drahos
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
43
|
Nimer RM, Abdel Rahman AM. Recent advances in proteomic-based diagnostics of cystic fibrosis. Expert Rev Proteomics 2023; 20:151-169. [PMID: 37766616 DOI: 10.1080/14789450.2023.2258282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 07/06/2023] [Indexed: 09/29/2023]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetic disease characterized by thick and sticky mucus accumulation, which may harm numerous internal organs. Various variables such as gene modifiers, environmental factors, age of diagnosis, and CF transmembrane conductance regulator (CFTR) gene mutations influence phenotypic disease diversity. Biomarkers that are based on genomic information may not accurately represent the underlying mechanism of the disease as well as its lethal complications. Therefore, recent advancements in mass spectrometry (MS)-based proteomics may provide deep insights into CF mechanisms and cellular functions by examining alterations in the protein expression patterns from various samples of individuals with CF. AREAS COVERED We present current developments in MS-based proteomics, its application, and findings in CF. In addition, the future roles of proteomics in finding diagnostic and prognostic novel biomarkers. EXPERT OPINION Despite significant advances in MS-based proteomics, extensive research in a large cohort for identifying and validating diagnostic, prognostic, predictive, and therapeutic biomarkers for CF disease is highly needed.
Collapse
Affiliation(s)
- Refat M Nimer
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Anas M Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genome Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
44
|
Rathore D, Marino MJ, Nita-Lazar A. Omics and systems view of innate immune pathways. Proteomics 2023; 23:e2200407. [PMID: 37269203 DOI: 10.1002/pmic.202200407] [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: 02/14/2023] [Revised: 04/16/2023] [Accepted: 05/23/2023] [Indexed: 06/04/2023]
Abstract
Multiomics approaches to studying systems biology are very powerful techniques that can elucidate changes in the genomic, transcriptomic, proteomic, and metabolomic levels within a cell type in response to an infection. These approaches are valuable for understanding the mechanisms behind disease pathogenesis and how the immune system responds to being challenged. With the emergence of the COVID-19 pandemic, the importance and utility of these tools have become evident in garnering a better understanding of the systems biology within the innate and adaptive immune response and for developing treatments and preventative measures for new and emerging pathogens that pose a threat to human health. In this review, we focus on state-of-the-art omics technologies within the scope of innate immunity.
Collapse
Affiliation(s)
- Deepali Rathore
- Functional Cellular Networks Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew J Marino
- Functional Cellular Networks Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Aleksandra Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
45
|
Yildiz P, Ozcan S. A single protein to multiple peptides: Investigation of protein-peptide correlations using targeted alpha-2-macroglobulin analysis. Talanta 2023; 265:124878. [PMID: 37392709 DOI: 10.1016/j.talanta.2023.124878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/30/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Recent advances in proteomics technologies have enabled the analysis of thousands of proteins in a high-throughput manner. Mass spectrometry (MS) based proteomics uses a peptide-centric approach where biological samples undergo specific proteolytic digestion and then only unique peptides are used for protein identification and quantification. Considering the fact that a single protein may have multiple unique peptides and a number of different forms, it becomes essential to understand dynamic protein-peptide relationships to ensure robust and reliable peptide-centric protein analysis. In this study, we investigated the correlation between protein concentration and corresponding unique peptide responses under a conventional proteolytic digestion condition. Protein-peptide correlation, digestion efficiency, matrix-effect, and concentration-effect were evaluated. Twelve unique peptides of alpha-2-macroglobulin (A2MG) were monitored using a targeted MS approach to acquire insights into protein-peptide dynamics. Although the peptide responses were reproducible between replicates, protein-peptide correlation was moderate in protein standards and low in complex matrices. The results suggest that reproducible peptide signal could be misleading in clinical studies and a peptide selection could dramatically change the outcome at protein level. This is the first study investigating quantitative protein-peptide correlations in biological samples using all unique peptides representing the same protein and opens a discussion on peptide-based proteomics.
Collapse
Affiliation(s)
- Pelin Yildiz
- Department of Chemistry, Middle East Technical University (METU), 06800, Ankara, Turkiye; Nanografi Nanotechnology Co, Middle East Technical University (METU) Technopolis, 06531, Ankara, Turkiye
| | - Sureyya Ozcan
- Department of Chemistry, Middle East Technical University (METU), 06800, Ankara, Turkiye; Cancer Systems Biology Laboratory (CanSyL), Middle East Technical University (METU), 06800, Ankara, Turkiye.
| |
Collapse
|
46
|
Ahn HS, Lee Y, Kim HM, Ju S, Lee S, Jeong HG, Park SJ, Park KH, Lee J, Lee JY, Woo SJ, Lee C. Multiplexed plasma protein classifiers for the diagnosis of age-related macular degeneration. Clin Transl Med 2023; 13:e1307. [PMID: 37317693 DOI: 10.1002/ctm2.1307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023] Open
Affiliation(s)
- Hee-Sung Ahn
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | | | - Hyeong Min Kim
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Shinyeong Ju
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Seonjeong Lee
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Hyeon-Gyo Jeong
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Sang Jun Park
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Kyu Hyung Park
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Junyeop Lee
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joo Yong Lee
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Se Joon Woo
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Cheolju Lee
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- RetiMark Co. Ltd, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| |
Collapse
|
47
|
Gurdo N, Volke DC, McCloskey D, Nikel PI. Automating the design-build-test-learn cycle towards next-generation bacterial cell factories. N Biotechnol 2023; 74:1-15. [PMID: 36736693 DOI: 10.1016/j.nbt.2023.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/15/2023] [Accepted: 01/22/2023] [Indexed: 02/04/2023]
Abstract
Automation is playing an increasingly significant role in synthetic biology. Groundbreaking technologies, developed over the past 20 years, have enormously accelerated the construction of efficient microbial cell factories. Integrating state-of-the-art tools (e.g. for genome engineering and analytical techniques) into the design-build-test-learn cycle (DBTLc) will shift the metabolic engineering paradigm from an almost artisanal labor towards a fully automated workflow. Here, we provide a perspective on how a fully automated DBTLc could be harnessed to construct the next-generation bacterial cell factories in a fast, high-throughput fashion. Innovative toolsets and approaches that pushed the boundaries in each segment of the cycle are reviewed to this end. We also present the most recent efforts on automation of the DBTLc, which heralds a fully autonomous pipeline for synthetic biology in the near future.
Collapse
Affiliation(s)
- Nicolás Gurdo
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens, Lyngby, Denmark
| | - Daniel C Volke
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens, Lyngby, Denmark
| | - Douglas McCloskey
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens, Lyngby, Denmark
| | - Pablo Iván Nikel
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens, Lyngby, Denmark.
| |
Collapse
|
48
|
Sun B, Liu J, Cai P, Wu J, Liu W, Hu H, Liu L. Aptamer-based sample purification for mass spectrometric quantification of trastuzumab in human serum. Talanta 2023; 257:124349. [PMID: 36827940 DOI: 10.1016/j.talanta.2023.124349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023]
Abstract
In this study, we developed a simple liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay to quantify trastuzumab in human serum using aptamers for sample purification. Trastuzumab was extracted from serum samples using the capture probe based on its aptamer CH1S-3, followed by reduction, alkylation, trypsin digestion, and quantification using LC-MS/MS. Additionally, a unique peptide, FTISADTSK, was employed as a surrogate peptide and quantified, and *FTISADTSK (13C915N-labeled phenylalanine) was used as an internal standard to minimize variability in detection among the samples. The detection range for this method was 0.5-250 μg/mL, with a high correlation coefficient (r2 > 0.99). The intra- and inter-day precision (%CV, the coefficient of variation) of the quality control samples was less than 12.7%, and the accuracy (%bias) was below 8.64%. After optimization and verification, this assay was used to determine trastuzumab levels in clinical human serum samples. The results indicated that the trastuzumab concentrations had an approximate 4-fold difference among ten patients (range: 11.80-41.90 μg/mL). This study provides a novel approach for the accurate and quantitative monitoring of the mAb-trastuzumab.
Collapse
Affiliation(s)
- Bo Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Jiuyang Liu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Pei Cai
- School of Pharmacy, Wuhan University, Wuhan, 430071, China
| | - Jianhua Wu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China.
| | - Hankun Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China; Hubei Micro-explore Innovative Pharmaceutical Research Co., Ltd, Wuhan, 430074, China.
| | - Liang Liu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
49
|
Watson CM, Dammer EB, Ping L, Duong DM, Modeste E, Carter EK, Johnson ECB, Levey AI, Lah JJ, Roberts BR, Seyfried NT. Quantitative Mass Spectrometry Analysis of Cerebrospinal Fluid Protein Biomarkers in Alzheimer's Disease. Sci Data 2023; 10:261. [PMID: 37160957 PMCID: PMC10170100 DOI: 10.1038/s41597-023-02158-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with cerebrospinal fluid (CSF) β-amyloid (Aβ), total Tau, and phosphorylated Tau (pTau) providing the most sensitive and specific biomarkers for diagnosis. However, these diagnostic biomarkers do not reflect the complex changes in AD brain beyond amyloid (A) and Tau (T) pathologies. Here, we report a selected reaction monitoring mass spectrometry (SRM-MS) method with isotopically labeled standards for relative protein quantification in CSF. Biomarker positive (AT+) and negative (AT-) CSF pools were used as quality controls (QCs) to assess assay precision. We detected 62 peptides (51 proteins) with an average coefficient of variation (CV) of ~13% across 30 QCs and 133 controls (cognitively normal, AT-), 127 asymptomatic (cognitively normal, AT+) and 130 symptomatic AD (cognitively impaired, AT+). Proteins that could distinguish AT+ from AT- individuals included SMOC1, GDA, 14-3-3 proteins, and those involved in glycolysis. Proteins that could distinguish cognitive impairment were mainly neuronal proteins (VGF, NPTX2, NPTXR, and SCG2). This demonstrates the utility of SRM-MS to quantify CSF protein biomarkers across stages of AD.
Collapse
Affiliation(s)
- Caroline M Watson
- Department of Neurology, Emory University School of Medicine, Atlanta, USA
| | - Eric B Dammer
- Department of Neurology, Emory University School of Medicine, Atlanta, USA
| | - Lingyan Ping
- Department of Neurology, Emory University School of Medicine, Atlanta, USA
| | - Duc M Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, USA
| | - Erica Modeste
- Department of Neurology, Emory University School of Medicine, Atlanta, USA
| | - E Kathleen Carter
- Department of Biochemistry, Emory University School of Medicine, Atlanta, USA
| | - Erik C B Johnson
- Department of Neurology, Emory University School of Medicine, Atlanta, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, USA.
| | - James J Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, USA.
| | - Blaine R Roberts
- Department of Neurology, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, USA
| | - Nicholas T Seyfried
- Department of Neurology, Emory University School of Medicine, Atlanta, USA.
- Department of Biochemistry, Emory University School of Medicine, Atlanta, USA.
| |
Collapse
|
50
|
Maxwell CB, Sandhu JK, Cao TH, McCann GP, Ng LL, Jones DJL. The Edge Effect in High-Throughput Proteomics: A Cautionary Tale. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023. [PMID: 37155737 DOI: 10.1021/jasms.3c00035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
In order for mass spectrometry to continue to grow as a platform for high-throughput clinical and translational research, careful consideration must be given to quality control by ensuring that the assay performs reproducibly and accurately and precisely. In particular, the throughput required for large cohort clinical validation in biomarker discovery and diagnostic screening has driven the growth of multiplexed targeted liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) assays paired with sample preparation and analysis in multiwell plates. However, large scale MS-based proteomics studies are often plagued by batch effects: sources of technical variation in the data, which can arise from a diverse array of sources such as sample preparation batches, different reagent lots, or indeed MS signal drift. These batch effects can confound the detection of true signal differences, resulting in incorrect conclusions being drawn about significant biological effects or lack thereof. Here, we present an intraplate batch effect termed the edge effect arising from temperature gradients in multiwell plates, commonly reported in preclinical cell culture studies but not yet reported in a clinical proteomics setting. We present methods herein to ameliorate the phenomenon including proper assessment of heating techniques for multiwell plates and incorporation of surrogate standards, which can normalize for intraplate variation.
Collapse
Affiliation(s)
- Colleen B Maxwell
- The Leicester van Geest MultiOmics Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, United Kingdom
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Jatinderpal K Sandhu
- The Leicester van Geest MultiOmics Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, United Kingdom
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Thong H Cao
- The Leicester van Geest MultiOmics Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, United Kingdom
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Gerry P McCann
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Leong L Ng
- The Leicester van Geest MultiOmics Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, United Kingdom
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Donald J L Jones
- The Leicester van Geest MultiOmics Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, United Kingdom
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
- Leicester Cancer Research Centre, RKCSB, University of Leicester, Leicester LE2 7LX, United Kingdom
| |
Collapse
|