1
|
van der Burgt YEM, Romijn FPHTM, Treep MM, Ruhaak LR, Cobbaert CM. Strategies to verify equimolar peptide release in mass spectrometry-based protein quantification exemplified for apolipoprotein(a). Clin Chem Lab Med 2025; 63:780-789. [PMID: 39450666 DOI: 10.1515/cclm-2024-0539] [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: 05/22/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVES Quantitative protein mass spectrometry (MS) is ideally suited for precision diagnostics and for reference standardization of protein analytes. At the Leiden Apolipoprotein Reference Laboratory we apply MS strategies to obtain detailed insight into the protein-to-peptide conversion in order to verify that quantifier peptides are not partly concealed in miscleaved protein backbone. METHODS Apolipoprotein(a) (apo(a)) was digested in a non-optimal manner to enhance the number of miscleaved peptides that were identified by high resolution liquid chromatography tandem-MS measurements. The protein-to-peptide conversion was carefully mapped with specific attention for miscleaved peptides that contain an apo(a) quantifier peptide. Four different isotopologues of each apo(a)-quantifier peptide were applied to evaluate linearity of internal peptide standards during measurement of specific real-life samples. RESULTS Two apo(a) quantifier peptides that were concealed in two different miscleaved peptides were included into a multiple reaction monitoring list in our targeted MS-based apo(a) quantifications to alert for potential protein digestion discrepancies. The presence of miscleaved peptides could be ruled out when applying our candidate reference measurement procedure (RMP) for apo(a) quantification. CONCLUSIONS These data further corroborate the validity of our apo(a) candidate RMP as higher order method for certification of commercial Lp(a) tests that is endorsed by the International Federation of Clinical Chemistry and Laboratory Medicine. MS-based molecular detection and quantification of heterogeneous apo(a) proteoforms will allow manufacturers' transitioning from confounded lipoprotein(a) [Lp(a)] mass levels into accurate molar apo(a) levels.
Collapse
Affiliation(s)
- Yuri E M van der Burgt
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| | - Fred P H T M Romijn
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| | - Maxim M Treep
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| |
Collapse
|
2
|
Orchard SE. What have Data Standards ever done for us? Mol Cell Proteomics 2025:100933. [PMID: 40024375 DOI: 10.1016/j.mcpro.2025.100933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
The Human Proteome Organization (HUPO) Proteomics Standards Initiative (PSI) has been successfully developing guidelines, data formats, and controlled vocabularies for both the field of molecular interaction and that of mass spectrometry for more than 20 years. This review explores some of the ways that the proteomics community has benefitted from the development of community standards and takes a look at some of the tools and resources that have been improved or developed as a result of the work of the HUPO-PSI.
Collapse
Affiliation(s)
- S E Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| |
Collapse
|
3
|
Zhang DE, He T, Shi T, Huang K, Peng A. Trends in the research and development of peptide drug conjugates: artificial intelligence aided design. Front Pharmacol 2025; 16:1553853. [PMID: 40083376 PMCID: PMC11903715 DOI: 10.3389/fphar.2025.1553853] [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: 12/31/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Peptide-drug conjugates (PDCs) represent an emerging class of targeted therapeutic agents that consist of small molecular drugs coupled to multifunctional peptides through cleavable or non-cleavable linkers. The principal advantage of PDCs lies in their capacity to deliver drugs to diseased tissues at increased local concentrations, thereby reducing toxicity and mitigating adverse effects by limiting damage to non-diseased tissues. Despite the increasing number of PDCs being developed for various diseases, their advancements remain relatively slow due to several development constraints, which include limited available peptides and linkers, narrow therapeutic applications, and incomplete evaluation and information platforms for PDCs. Marked by the recent Nobel Prize awarded to artificial intelligence (AI) and de novo protein design for "protein design and structure prediction," AI is playing an increasingly important role in drug discovery and development. In this review, we summarize the recent developments and limitations of PDCs, highlights the potential of AI in revolutionizing the design and evaluation of PDC.
Collapse
Affiliation(s)
- Dong-E Zhang
- The Third Hospital of Wuhan, Hubei University of Chinese Medicine, Wuhan, China
| | - Tong He
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyi Shi
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
- Tongji-RongCheng Biomedical Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anlin Peng
- The Third Hospital of Wuhan, Tongren Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Mugahid D, Lyon J, Demurjian C, Eolin N, Whittaker C, Godek M, Lauffenburger D, Fortune S, Levine S. A practical guide to FAIR data management in the age of multi-OMICS and AI. Front Immunol 2025; 15:1439434. [PMID: 39902035 PMCID: PMC11788310 DOI: 10.3389/fimmu.2024.1439434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/17/2024] [Indexed: 02/05/2025] Open
Abstract
Multi-cellular biological systems, including the immune system, are highly complex, dynamic, and adaptable. Systems biologists aim to understand such complexity at a quantitative level. However, these ambitious efforts are often limited by access to a variety of high-density intra-, extra- and multi-cellular measurements resolved in time and space and across a variety of perturbations. The advent of automation, OMICs and single-cell technologies now allows high dimensional multi-modal data acquisition from the same biological samples multiplexed at scale (multi-OMICs). As a result, systems biologists -theoretically- have access to more data than ever. However, the mathematical frameworks and computational tools needed to analyze and interpret such data are often still nascent, limiting the biological insights that can be obtained without years of computational method development and validation. More pressingly, much of the data sits in silos in formats that are incomprehensible to other scientists or machines limiting its value to the vaster scientific community, especially the computational biologists tasked with analyzing these vast amounts of data in more nuanced ways. With the rapid development and increasing interest in using artificial intelligence (AI) for the life sciences, improving how biologic data is organized and shared is more pressing than ever for scientific progress. Here, we outline a practical approach to multi-modal data management and FAIR sharing, which are in line with the latest US and EU funders' data sharing policies. This framework can help extend the longevity and utility of data by allowing facile use and reuse, accelerating scientific discovery in the biomedical sciences.
Collapse
Affiliation(s)
- Douaa Mugahid
- Department of Immunology and Infectious Diseases, T.H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Jared Lyon
- BioMicro Center, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Charlie Demurjian
- BioMicro Center, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Nathan Eolin
- BioMicro Center, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Charlie Whittaker
- BioMicro Center, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Mark Godek
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard, Cambridge, MA, United States
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Sarah Fortune
- Department of Immunology and Infectious Diseases, T.H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Stuart Levine
- BioMicro Center, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
5
|
Reddy PJ, Sun Z, Wippel HH, Baxter DH, Swearingen K, Shteynberg DD, Midha MK, Caimano MJ, Strle K, Choi Y, Chan AP, Schork NJ, Varela-Stokes AS, Moritz RL. Borrelia PeptideAtlas: A proteome resource of common Borrelia burgdorferi isolates for Lyme research. Sci Data 2024; 11:1313. [PMID: 39622905 PMCID: PMC11612207 DOI: 10.1038/s41597-024-04047-9] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
Lyme disease is caused by an infection with the spirochete Borrelia burgdorferi, and is the most common vector-borne disease in North America. B. burgdorferi isolates harbor extensive genomic and proteomic variability and further comparison of isolates is key to understanding the infectivity of the spirochetes and biological impacts of identified sequence variants. Here, we applied both transcriptome analysis and mass spectrometry-based proteomics to assemble peptide datasets of B. burgdorferi laboratory isolates B31, MM1, and the infective isolate B31-5A4, to provide a publicly available Borrelia PeptideAtlas. Included are total proteome, secretome, and membrane proteome identifications of the individual isolates. Proteomic data collected from 35 different experiment datasets, totaling 386 mass spectrometry runs, have identified 81,967 distinct peptides, which map to 1,113 proteins. The Borrelia PeptideAtlas covers 86% of the total B31 proteome of 1,291 protein sequences. The Borrelia PeptideAtlas is an extensible comprehensive peptide repository with proteomic information from B. burgdorferi isolates useful for Lyme disease research.
Collapse
Affiliation(s)
- Panga J Reddy
- Institute for Systems Biology, Seattle, Washington, USA
| | - Zhi Sun
- Institute for Systems Biology, Seattle, Washington, USA
| | | | | | | | | | - Mukul K Midha
- Institute for Systems Biology, Seattle, Washington, USA
| | | | - Klemen Strle
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Yongwook Choi
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Agnes P Chan
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | | | - Andrea S Varela-Stokes
- Tufts University Cummings School of Veterinary Medicine, Department of Comparative Pathobiology, Grafton, MA, 01536, USA
| | | |
Collapse
|
6
|
Basith S, Sangaraju VK, Manavalan B, Lee G. mHPpred: Accurate identification of peptide hormones using multi-view feature learning. Comput Biol Med 2024; 183:109297. [PMID: 39442438 DOI: 10.1016/j.compbiomed.2024.109297] [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: 08/20/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Peptide hormones were first used in medicine in the early 20th century, with the pivotal event being the isolation and purification of insulin in 1921. These hormones are integral to a sophisticated system that emerged early in evolution to regulate growth, development, and homeostasis. They serve as targeted signaling molecules that transfer specific information between cells and organs, ensuring coordinated and precise physiological responses. While experimental methods for identifying peptide hormones present challenges such as low abundance, stability issues, and complexity, computational methods offer promising alternatives. Advances in machine learning and bioinformatics have facilitated the prediction of peptide hormones, further enhancing their therapeutic potential. In this study, we explored three different computational frameworks for peptide hormone identification and determined that the meta-approach was the most suitable. Firstly, we evaluated the discriminative power of 26 feature descriptors using a series of baseline models and identified seven feature descriptors with high predictive potential. Through a systematic approach, we then selected the top 20 performing baseline models and integrated their predicted probabilities to train a meta-model, leveraging the strengths of multiple prediction strategies. Our final light gradient boosting-based meta-model, mHPpred, significantly outperformed the existing method, HOPPred, on both benchmarking and independent datasets. Notably, mHPpred also demonstrated superior performance compared to the hybrid and integrative framework approaches employed in this study. This superiority demonstrates the effectiveness of our multi-view feature learning strategy in capturing discriminative features and providing a more accurate prediction model for peptide hormones. mHPpred is publicly accessible at: https://balalab-skku.org/mHPpred.
Collapse
Affiliation(s)
- Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea.
| | - Vinoth Kumar Sangaraju
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Balachandran Manavalan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
7
|
Ran P, Wang Y, Li K, He S, Tan S, Lv J, Zhu J, Tang S, Feng J, Qin Z, Li Y, Huang L, Yin Y, Zhu L, Yang W, Ding C. STAVER: a standardized benchmark dataset-based algorithm for effective variation reduction in large-scale DIA-MS data. Brief Bioinform 2024; 25:bbae553. [PMID: 39504480 PMCID: PMC11540132 DOI: 10.1093/bib/bbae553] [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: 07/05/2024] [Revised: 09/12/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024] Open
Abstract
Mass spectrometry (MS)-based proteomics has become instrumental in comprehensively investigating complex biological systems. Data-independent acquisition (DIA)-MS, utilizing hybrid spectral library search strategies, allows for the simultaneous quantification of thousands of proteins, showing promise in enhancing protein identification and quantification precision. However, low-quality profiles can considerably undermine quantitative precision, resulting in inaccurate protein quantification. To tackle this challenge, we introduced STAVER, a novel algorithm that leverages standardized benchmark datasets to reduce non-biological variation in large-scale DIA-MS analyses. By eliminating unwanted noise in MS signals, STAVER significantly improved protein quantification precision, especially in hybrid spectral library searches. Moreover, we validated STAVER's robustness and applicability across multiple large-scale DIA datasets, demonstrating significantly enhanced precision and reproducibility of protein quantification. STAVER offers an innovative and effective approach for enhancing the quality of large-scale DIA proteomic data, facilitating cross-platform and cross-laboratory comparative analyses. This advancement significantly enhances the consistency and reliability of findings in clinical research. The complete package is available at https://github.com/Ran485/STAVER.
Collapse
Affiliation(s)
- Peng Ran
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Yunzhi Wang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Kai Li
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Shiman He
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Subei Tan
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Jiacheng Lv
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Jiajun Zhu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Shaoshuai Tang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Jinwen Feng
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Zhaoyu Qin
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Yan Li
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Lin Huang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Yanan Yin
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Lingli Zhu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Chen Ding
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi 830000, P. R. China
| |
Collapse
|
8
|
Thangudu RR, Holck M, Singhal D, Pilozzi A, Edwards N, Rudnick PA, Domagalski MJ, Chilappagari P, Ma L, Xin Y, Le T, Nyce K, Chaudhary R, Ketchum KA, Maurais A, Connolly B, Riffle M, Chambers MC, MacLean B, MacCoss MJ, McGarvey PB, Basu A, Otridge J, Casas-Silva E, Venkatachari S, Rodriguez H, Zhang X. NCI's Proteomic Data Commons: A Cloud-Based Proteomics Repository Empowering Comprehensive Cancer Analysis through Cross-Referencing with Genomic and Imaging Data. CANCER RESEARCH COMMUNICATIONS 2024; 4:2480-2488. [PMID: 39225545 PMCID: PMC11413857 DOI: 10.1158/2767-9764.crc-24-0243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/22/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Proteomics has emerged as a powerful tool for studying cancer biology, developing diagnostics, and therapies. With the continuous improvement and widespread availability of high-throughput proteomic technologies, the generation of large-scale proteomic data has become more common in cancer research, and there is a growing need for resources that support the sharing and integration of multi-omics datasets. Such datasets require extensive metadata including clinical, biospecimen, and experimental and workflow annotations that are crucial for data interpretation and reanalysis. The need to integrate, analyze, and share these data has led to the development of NCI's Proteomic Data Commons (PDC), accessible at https://pdc.cancer.gov. As a specialized repository within the NCI Cancer Research Data Commons (CRDC), PDC enables researchers to locate and analyze proteomic data from various cancer types and connect with genomic and imaging data available for the same samples in other CRDC nodes. Presently, PDC houses annotated data from more than 160 datasets across 19 cancer types, generated by several large-scale cancer research programs with cohort sizes exceeding 100 samples (tumor and associated normal when available). In this article, we review the current state of PDC in cancer research, discuss the opportunities and challenges associated with data sharing in proteomics, and propose future directions for the resource. SIGNIFICANCE The Proteomic Data Commons (PDC) plays a crucial role in advancing cancer research by providing a centralized repository of high-quality cancer proteomic data, enriched with extensive clinical annotations. By integrating and cross-referencing with complementary genomic and imaging data, the PDC facilitates multi-omics analyses, driving comprehensive insights, and accelerating discoveries across various cancer types.
Collapse
Affiliation(s)
| | | | | | | | - Nathan Edwards
- Georgetown University, Washington, District of Columbia.
| | | | | | | | - Lei Ma
- ICF, Rockville, Maryland.
| | - Yi Xin
- ICF, Rockville, Maryland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Esmeralda Casas-Silva
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Rockville, Maryland.
| | | | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, Maryland.
| | - Xu Zhang
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, Maryland.
| |
Collapse
|
9
|
Lundgren T, Clark PL, Champion MM. Fit for Purpose Approach To Evaluate Detection of Amino Acid Substitutions in Shotgun Proteomics. J Proteome Res 2024; 23:1263-1271. [PMID: 38478054 PMCID: PMC11003417 DOI: 10.1021/acs.jproteome.3c00730] [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: 11/03/2023] [Revised: 02/04/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
Amino acid substitutions (AASs) alter proteins from their genome-expected sequences. Accumulation of substitutions in proteins underlies numerous diseases and antibiotic mechanisms. Accurate global detection of AASs and their frequencies is crucial for understanding these mechanisms. Shotgun proteomics provides an untargeted method for measuring AASs but introduces biases when extrapolating from the genome to identify AASs. To characterize these biases, we created a "ground-truth" approach using the similarities betweenEscherichia coli and Salmonella typhimurium to model the complexity of AAS detection. Shotgun proteomics on mixed lysates generated libraries representing ∼100,000 peptide-spectra and 4161 peptide sequences with a single AAS and defined stoichiometry. Identifying S. typhimurium peptide-spectra with only the E. coli genome resulted in 64.1% correctly identified library peptides. Specific AASs exhibit variable identification efficiencies. There was no inherent bias from the stoichiometry of the substitutions. Short peptides and AASs localized near peptide termini had poor identification efficiency. We identify a new class of "scissor substitutions" that gain or lose protease cleavage sites. Scissor substitutions also had poor identification efficiency. This ground-truth AAS library reveals various sources of bias, which will guide the application of shotgun proteomics to validate AAS hypotheses.
Collapse
Affiliation(s)
- Taylor
J. Lundgren
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Patricia L. Clark
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre Dame, Indiana 46556, United States
- Department
of Chemical and Biomolecular Engineering, University of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Matthew M. Champion
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
10
|
Shome M, MacKenzie TMG, Subbareddy SR, Snyder MP. The Importance, Challenges, and Possible Solutions for Sharing Proteomics Data While Safeguarding Individuals' Privacy. Mol Cell Proteomics 2024; 23:100731. [PMID: 38331191 PMCID: PMC10915627 DOI: 10.1016/j.mcpro.2024.100731] [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: 08/14/2023] [Revised: 01/28/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024] Open
Abstract
Proteomics data sharing has profound benefits at the individual level as well as at the community level. While data sharing has increased over the years, mostly due to journal and funding agency requirements, the reluctance of researchers with regard to data sharing is evident as many shares only the bare minimum dataset required to publish an article. In many cases, proper metadata is missing, essentially making the dataset useless. This behavior can be explained by a lack of incentives, insufficient awareness, or a lack of clarity surrounding ethical issues. Through adequate training at research institutes, researchers can realize the benefits associated with data sharing and can accelerate the norm of data sharing for the field of proteomics, as has been the standard in genomics for decades. In this article, we have put together various repository options available for proteomics data. We have also added pros and cons of those repositories to facilitate researchers in selecting the repository most suitable for their data submission. It is also important to note that a few types of proteomics data have the potential to re-identify an individual in certain scenarios. In such cases, extra caution should be taken to remove any personal identifiers before sharing on public repositories. Data sets that will be useless without personal identifiers need to be shared in a controlled access repository so that only authorized researchers can access the data and personal identifiers are kept safe.
Collapse
Affiliation(s)
- Mahasish Shome
- Department of Genetics, Stanford University, Palo Alto, California, USA
| | - Tim M G MacKenzie
- Department of Genetics, Stanford University, Palo Alto, California, USA
| | | | - Michael P Snyder
- Department of Genetics, Stanford University, Palo Alto, California, USA.
| |
Collapse
|
11
|
Baumer ZT, Erber L, Jolley E, Lawrence S, Lin C, Murakami S, Perez V, Prall W, Schaening-Burgos C, Sylvia M, Chen S, Gregory BD. Defining the commonalities between post-transcriptional and post-translational modification communities. Trends Biochem Sci 2024; 49:185-188. [PMID: 37884411 DOI: 10.1016/j.tibs.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023]
Abstract
Post-transcriptional modifications of RNA (PRMs) and post-translational modifications of proteins (PTMs) are important regulatory mechanisms in biological processes and have many commonalities. However, the integration of these research areas is lacking. A recent discussion identified the priorities, areas of emphasis, and necessary technologies to advance and integrate these areas of study.
Collapse
Affiliation(s)
- Zachary T Baumer
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80305, USA
| | - Luke Erber
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66044, USA
| | - Elizabeth Jolley
- Department of Chemistry, Center for RNA Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Sheldon Lawrence
- Department of Biology, Oxford College of Emory University, Oxford, GA 30054, USA
| | - Chuwei Lin
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Shino Murakami
- Department of Pharmacology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Veronica Perez
- Horticultural Sciences Department, University of Florida, Gainesville, FL 32611, USA
| | - Wil Prall
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Megan Sylvia
- Intercollege Graduate Degree Program in Plant Biology, Department of Biology, and Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
| | - Sixue Chen
- Department of Biology, University of Mississippi, University, MS 38677, USA.
| | - Brian D Gregory
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
Shi K, Xiong Y, Wang Y, Deng Y, Wang W, Jing B, Gao X. PractiCPP: a deep learning approach tailored for extremely imbalanced datasets in cell-penetrating peptide prediction. Bioinformatics 2024; 40:btae058. [PMID: 38305405 PMCID: PMC11212486 DOI: 10.1093/bioinformatics/btae058] [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: 11/29/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/03/2024] Open
Abstract
MOTIVATION Effective drug delivery systems are paramount in enhancing pharmaceutical outcomes, particularly through the use of cell-penetrating peptides (CPPs). These peptides are gaining prominence due to their ability to penetrate eukaryotic cells efficiently without inflicting significant damage to the cellular membrane, thereby ensuring optimal drug delivery. However, the identification and characterization of CPPs remain a challenge due to the laborious and time-consuming nature of conventional methods, despite advances in proteomics. Current computational models, however, are predominantly tailored for balanced datasets, an approach that falls short in real-world applications characterized by a scarcity of known positive CPP instances. RESULTS To navigate this shortfall, we introduce PractiCPP, a novel deep-learning framework tailored for CPP prediction in highly imbalanced data scenarios. Uniquely designed with the integration of hard negative sampling and a sophisticated feature extraction and prediction module, PractiCPP facilitates an intricate understanding and learning from imbalanced data. Our extensive computational validations highlight PractiCPP's exceptional ability to outperform existing state-of-the-art methods, demonstrating remarkable accuracy, even in datasets with an extreme positive-to-negative ratio of 1:1000. Furthermore, through methodical embedding visualizations, we have established that models trained on balanced datasets are not conducive to practical, large-scale CPP identification, as they do not accurately reflect real-world complexities. In summary, PractiCPP potentially offers new perspectives in CPP prediction methodologies. Its design and validation, informed by real-world dataset constraints, suggest its utility as a valuable tool in supporting the acceleration of drug delivery advancements. AVAILABILITY AND IMPLEMENTATION The source code of PractiCPP is available on Figshare at https://doi.org/10.6084/m9.figshare.25053878.v1.
Collapse
Affiliation(s)
- Kexin Shi
- Syneron Technology, Guangzhou 510000, China
- Individualized Interdisciplinary Program (Data Science and Analytics), The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | | | - Yu Wang
- Syneron Technology, Guangzhou 510000, China
| | - Yifan Deng
- Syneron Technology, Guangzhou 510000, China
| | - Wenjia Wang
- Data Science and Analytics Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511400, Guangdong, China
| | - Bingyi Jing
- Department of Statistics and Data Science, Southern University of Science and Technology, Shenzhen 518000, China
| | - Xin Gao
- Syneron Technology, Guangzhou 510000, China
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
- Computational Bioscience Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Wang XY, Xu YM, Lau ATY. Proteogenomics in Cancer: Then and Now. J Proteome Res 2023; 22:3103-3122. [PMID: 37725793 DOI: 10.1021/acs.jproteome.3c00196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
For years, the paths of sequencing technologies and mass spectrometry have occurred in isolation, with each developing its own unique culture and expertise. These two technologies are crucial for inspecting complementary aspects of the molecular phenotype across the central dogma. Integrative multiomics strives to bridge the analysis gap among different fields to complete more comprehensive mechanisms of life events and diseases. Proteogenomics is one integrated multiomics field. Here in this review, we mainly summarize and discuss three aspects: workflow of proteogenomics, proteogenomics applications in cancer research, and the SWOT (Strengths, Weaknesses, Opportunities, Threats) analysis of proteogenomics in cancer research. In conclusion, proteogenomics has a promising future as it clarifies the functional consequences of many unannotated genomic abnormalities or noncanonical variants and identifies driver genes and novel therapeutic targets across cancers, which would substantially accelerate the development of precision oncology.
Collapse
Affiliation(s)
- Xiu-Yun Wang
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| |
Collapse
|
16
|
Yeom S, Nam D, Bok KH, Kwon HK, Kim S, Lee SW, Kim HJ. Synthesis of S-Carbamidomethyl Cysteine and Its Use for Quantification of Cysteinyl Peptides by Targeted Proteomics. Anal Chem 2023; 95:14413-14420. [PMID: 37707799 DOI: 10.1021/acs.analchem.3c02768] [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: 09/15/2023]
Abstract
Proteomics has played a central role in the identification of reliable disease biomarkers, which are the basis of precision medicine, a promising approach for tackling recalcitrant diseases such as cancer, that elude conventional treatments. Among proteomic methodologies, targeted proteomics employing stable isotope-labeled (SIL) internal standards is particularly suited for the clinical translation of biomarker information owing to its high throughput and accuracy in the quantitative analysis of patient-derived proteomes. Using SIL internal standards ensures the utmost level of confidence in detection and precision in targeted MS experiments. For successfully establishing assays based on targeted proteomics, it is crucial to secure broad coverage when selecting the SIL standard peptide panel. However, cysteinyl peptides have often been excluded because of cysteine's high chemical reactivity. To address this limitation, a new cysteine building block was developed by incorporating a sulfhydryl group configured with an S-carbamidomethyl group, which is commonly used in proteome sampling. This compound was found to be chemically stable and applicable to a variety of solid-phase peptide synthesis (SPPS) campaigns. Furthermore, a direct comparison of the synthesized SIL peptides and tryptic endogenous peptides demonstrated the potential utility of an SPPS flow based on the new cysteine building block for improving the success of targeted proteomic applications.
Collapse
Affiliation(s)
- Suyeon Yeom
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Dowoon Nam
- 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
| | - Hye Kyeong Kwon
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| | - Seungwoo Kim
- 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
| | - Hak Joong Kim
- Department of Chemistry and Center for ProteoGenome Research, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
17
|
Ribeiro HC, Zandonadi FDS, Sussulini A. An overview of metabolomic and proteomic profiling in bipolar disorder and its clinical value. Expert Rev Proteomics 2023; 20:267-280. [PMID: 37830362 DOI: 10.1080/14789450.2023.2267756] [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: 07/03/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION Bipolar disorder (BD) is a complex psychiatric disease characterized by alternating mood episodes. As for any other psychiatric illness, currently there is no biochemical test that is able to support diagnosis or therapeutic decisions for BD. In this context, the discovery and validation of biomarkers are interesting strategies that can be achieved through proteomics and metabolomics. AREAS COVERED In this descriptive review, a literature search including original articles and systematic reviews published in the last decade was performed with the objective to discuss the results of BD proteomic and metabolomic profiling analyses and indicate proteins and metabolites (or metabolic pathways) with potential clinical value. EXPERT OPINION A large number of proteins and metabolites have been reported as potential BD biomarkers; however, most studies do not reach biomarker validation stages. An effort from the scientific community should be directed toward the validation of biomarkers and the development of simplified bioanalytical techniques or protocols to determine them in biological samples, in order to translate proteomic and metabolomic findings into clinical routine assays.
Collapse
Affiliation(s)
- Henrique Caracho Ribeiro
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas(UNICAMP), Campinas, SP, Brazil
| | - Flávia da Silva Zandonadi
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas(UNICAMP), Campinas, SP, Brazil
| | - Alessandra Sussulini
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas(UNICAMP), Campinas, SP, Brazil
- Instituto Nacional de Ciência e Tecnologia de Bioanalítica (INCTBio), Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
18
|
Reddy PJ, Sun Z, Wippel HH, Baxter D, Swearingen K, Shteynberg DD, Midha MK, Caimano MJ, Strle K, Choi Y, Chan AP, Schork NJ, Moritz RL. Borrelia PeptideAtlas: A proteome resource of common Borrelia burgdorferi isolates for Lyme research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545244. [PMID: 37398146 PMCID: PMC10312716 DOI: 10.1101/2023.06.16.545244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Lyme disease, caused by an infection with the spirochete Borrelia burgdorferi, is the most common vector-borne disease in North America. B. burgdorferi strains harbor extensive genomic and proteomic variability and further comparison is key to understanding the spirochetes infectivity and biological impacts of identified sequence variants. To achieve this goal, both transcript and mass spectrometry (MS)-based proteomics was applied to assemble peptide datasets of laboratory strains B31, MM1, B31-ML23, infective isolates B31-5A4, B31-A3, and 297, and other public datasets, to provide a publicly available Borrelia PeptideAtlas http://www.peptideatlas.org/builds/borrelia/. Included is information on total proteome, secretome, and membrane proteome of these B. burgdorferi strains. Proteomic data collected from 35 different experiment datasets, with a total of 855 mass spectrometry runs, identified 76,936 distinct peptides at a 0.1% peptide false-discovery-rate, which map to 1,221 canonical proteins (924 core canonical and 297 noncore canonical) and covers 86% of the total base B31 proteome. The diverse proteomic information from multiple isolates with credible data presented by the Borrelia PeptideAtlas can be useful to pinpoint potential protein targets which are common to infective isolates and may be key in the infection process.
Collapse
Affiliation(s)
| | - Zhi Sun
- Institute for Systems Biology, Seattle, Washington, USA
| | | | - David Baxter
- Institute for Systems Biology, Seattle, Washington, USA
| | | | | | | | | | - Klemen Strle
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Yongwook Choi
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Agnes P. Chan
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | | | | |
Collapse
|
19
|
Hadisurya M, Li L, Kuwaranancharoen K, Wu X, Lee ZC, Alcalay RN, Padmanabhan S, Tao WA, Iliuk A. Quantitative proteomics and phosphoproteomics of urinary extracellular vesicles define putative diagnostic biosignatures for Parkinson's disease. COMMUNICATIONS MEDICINE 2023; 3:64. [PMID: 37165152 PMCID: PMC10172329 DOI: 10.1038/s43856-023-00294-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/27/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene have been recognized as genetic risk factors for Parkinson's disease (PD). However, compared to cancer, fewer genetic mutations contribute to the cause of PD, propelling the search for protein biomarkers for early detection of the disease. METHODS Utilizing 138 urine samples from four groups, healthy individuals (control), healthy individuals with G2019S mutation in the LRRK2 gene (non-manifesting carrier/NMC), PD individuals without G2019S mutation (idiopathic PD/iPD), and PD individuals with G2019S mutation (LRRK2 PD), we applied a proteomics strategy to determine potential diagnostic biomarkers for PD from urinary extracellular vesicles (EVs). RESULTS After efficient isolation of urinary EVs through chemical affinity followed by mass spectrometric analyses of EV peptides and enriched phosphopeptides, we identify and quantify 4476 unique proteins and 2680 unique phosphoproteins. We detect multiple proteins and phosphoproteins elevated in PD EVs that are known to be involved in important PD pathways, in particular the autophagy pathway, as well as neuronal cell death, neuroinflammation, and formation of amyloid fibrils. We establish a panel of proteins and phosphoproteins as novel candidates for disease biomarkers and substantiate the biomarkers using machine learning, ROC, clinical correlation, and in-depth network analysis. Several putative disease biomarkers are further partially validated in patients with PD using parallel reaction monitoring (PRM) and immunoassay for targeted quantitation. CONCLUSIONS These findings demonstrate a general strategy of utilizing biofluid EV proteome/phosphoproteome as an outstanding and non-invasive source for a wide range of disease exploration.
Collapse
Affiliation(s)
- Marco Hadisurya
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Li Li
- Tymora Analytical Operations, West Lafayette, IN, 47906, USA
| | | | - Xiaofeng Wu
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Zheng-Chi Lee
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
- West Lafayette Junior/Senior High School, West Lafayette, IN, 47906, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Shalini Padmanabhan
- The Michael J. Fox Foundation for Parkinson's Research, New York City, NY, 10163, USA
| | - W Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA.
- Tymora Analytical Operations, West Lafayette, IN, 47906, USA.
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA.
| | - Anton Iliuk
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA.
- Tymora Analytical Operations, West Lafayette, IN, 47906, USA.
| |
Collapse
|
20
|
Prakash A, García-Seisdedos D, Wang S, Kundu DJ, Collins A, George N, Moreno P, Papatheodorou I, Jones AR, Vizcaíno JA. Integrated View of Baseline Protein Expression in Human Tissues. J Proteome Res 2023; 22:729-742. [PMID: 36577097 PMCID: PMC9990129 DOI: 10.1021/acs.jproteome.2c00406] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The availability of proteomics datasets in the public domain, and in the PRIDE database, in particular, has increased dramatically in recent years. This unprecedented large-scale availability of data provides an opportunity for combined analyses of datasets to get organism-wide protein abundance data in a consistent manner. We have reanalyzed 24 public proteomics datasets from healthy human individuals to assess baseline protein abundance in 31 organs. We defined tissue as a distinct functional or structural region within an organ. Overall, the aggregated dataset contains 67 healthy tissues, corresponding to 3,119 mass spectrometry runs covering 498 samples from 489 individuals. We compared protein abundances between different organs and studied the distribution of proteins across these organs. We also compared the results with data generated in analogous studies. Additionally, we performed gene ontology and pathway-enrichment analyses to identify organ-specific enriched biological processes and pathways. As a key point, we have integrated the protein abundance results into the resource Expression Atlas, where they can be accessed and visualized either individually or together with gene expression data coming from transcriptomics datasets. We believe this is a good mechanism to make proteomics data more accessible for life scientists.
Collapse
Affiliation(s)
- Ananth Prakash
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom.,Open Targets, Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - David García-Seisdedos
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Shengbo Wang
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Deepti Jaiswal Kundu
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Andrew Collins
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, LiverpoolL69 7ZB, United Kingdom
| | - Nancy George
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Pablo Moreno
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Irene Papatheodorou
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom.,Open Targets, Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Andrew R Jones
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, LiverpoolL69 7ZB, United Kingdom
| | - Juan Antonio Vizcaíno
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom.,Open Targets, Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| |
Collapse
|
21
|
Mohammed Y, Goodlett D, Borchers CH. Bioinformatics Tools and Knowledgebases to Assist Generating Targeted Assays for Plasma Proteomics. Methods Mol Biol 2023; 2628:557-577. [PMID: 36781806 DOI: 10.1007/978-1-0716-2978-9_32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
In targeted proteomics experiments, selecting the appropriate proteotypic peptides as surrogate for the target protein is a crucial pre-acquisition step. This step is largely a bioinformatics exercise that involves integrating information on the peptides and proteins and using various software tools and knowledgebases. We present here a few resources that automate and simplify the selection process to a great degree. These tools and knowledgebases were developed primarily to streamline targeted proteomics assay development and include PeptidePicker, PeptidePickerDB, MRMAssayDB, MouseQuaPro, and PeptideTracker. We have used these tools to develop and document thousands of targeted proteomics assays, many of them for plasma proteins with focus on human and mouse. An important aspect in all these resources is the integrative approach on which they are based. Using these tools in the first steps of designing a singleplexed or multiplexed targeted proteomic experiment can reduce the necessary experimental steps tremendously. All the tools and knowledgebases we describe here are Web-based and freely accessible so scientists can query the information conveniently from the browser. This chapter provides an overview of these software tools and knowledgebases, their content, and how to use them for targeted plasma proteomics. We further demonstrate how to use them with the results of the HUPO Human Plasma Proteome Project to produce a new database of 3.8 k targeted assays for known human plasma proteins. Upon experimental validation, these assays should help in the further quantitative characterizing of the plasma proteome.
Collapse
Affiliation(s)
- Yassene Mohammed
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, ZA, Netherlands. .,University of Victoria - Genome BC Proteomics Centre, Victoria, BC, Canada. .,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.
| | - David Goodlett
- University of Victoria - Genome BC Proteomics Centre, Victoria, BC, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.,University of Gdansk, International Centre for Cancer Vaccine Science, Gdansk, Poland
| | - Christoph H Borchers
- Proteomics Centre, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.,Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, QC, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, Canada.,Department of Pathology, McGill University, Montreal, QC, Canada
| |
Collapse
|
22
|
Deutsch EW, Bandeira N, Perez-Riverol Y, Sharma V, Carver J, Mendoza L, Kundu DJ, Wang S, Bandla C, Kamatchinathan S, Hewapathirana S, Pullman B, Wertz J, Sun Z, Kawano S, Okuda S, Watanabe Y, MacLean B, MacCoss M, Zhu Y, Ishihama Y, Vizcaíno J. The ProteomeXchange consortium at 10 years: 2023 update. Nucleic Acids Res 2023; 51:D1539-D1548. [PMID: 36370099 PMCID: PMC9825490 DOI: 10.1093/nar/gkac1040] [Citation(s) in RCA: 326] [Impact Index Per Article: 163.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/20/2022] [Accepted: 10/23/2022] [Indexed: 11/13/2022] Open
Abstract
Mass spectrometry (MS) is by far the most used experimental approach in high-throughput proteomics. The ProteomeXchange (PX) consortium of proteomics resources (http://www.proteomexchange.org) was originally set up to standardize data submission and dissemination of public MS proteomics data. It is now 10 years since the initial data workflow was implemented. In this manuscript, we describe the main developments in PX since the previous update manuscript in Nucleic Acids Research was published in 2020. The six members of the Consortium are PRIDE, PeptideAtlas (including PASSEL), MassIVE, jPOST, iProX and Panorama Public. We report the current data submission statistics, showcasing that the number of datasets submitted to PX resources has continued to increase every year. As of June 2022, more than 34 233 datasets had been submitted to PX resources, and from those, 20 062 (58.6%) just in the last three years. We also report the development of the Universal Spectrum Identifiers and the improvements in capturing the experimental metadata annotations. In parallel, we highlight that data re-use activities of public datasets continue to increase, enabling connections between PX resources and other popular bioinformatics resources, novel research and also new data resources. Finally, we summarise the current state-of-the-art in data management practices for sensitive human (clinical) proteomics data.
Collapse
Affiliation(s)
| | - Nuno Bandeira
- Center for Computational Mass Spectrometry, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
- Dept. Computer Science and Engineering, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Yasset Perez-Riverol
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | | | - Jeremy J Carver
- Center for Computational Mass Spectrometry, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
- Dept. Computer Science and Engineering, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Luis Mendoza
- Institute for Systems Biology, Seattle WA 98109, USA
| | - Deepti J Kundu
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Shengbo Wang
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Chakradhar Bandla
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Selvakumar Kamatchinathan
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Suresh Hewapathirana
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Benjamin S Pullman
- Center for Computational Mass Spectrometry, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
- Dept. Computer Science and Engineering, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Julie Wertz
- Center for Computational Mass Spectrometry, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
- Dept. Computer Science and Engineering, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Zhi Sun
- Institute for Systems Biology, Seattle WA 98109, USA
| | - Shin Kawano
- Faculty of Contemporary Society, Toyama University of International Studies, Toyama 930-1292, Japan
- Database Center for Life Science (DBCLS), Joint Support-Center for Data Science Research, Research Organization of Information and Systems, Chiba 277-0871, Japan
- School of Frontier Engineering, Kitasato University, Sagamihara 252-0373, Japan
| | - Shujiro Okuda
- Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yu Watanabe
- Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | | | | | - Yunping Zhu
- Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Juan Antonio Vizcaíno
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| |
Collapse
|
23
|
Cox J. Prediction of peptide mass spectral libraries with machine learning. Nat Biotechnol 2023; 41:33-43. [PMID: 36008611 DOI: 10.1038/s41587-022-01424-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/11/2022] [Indexed: 01/21/2023]
Abstract
The recent development of machine learning methods to identify peptides in complex mass spectrometric data constitutes a major breakthrough in proteomics. Longstanding methods for peptide identification, such as search engines and experimental spectral libraries, are being superseded by deep learning models that allow the fragmentation spectra of peptides to be predicted from their amino acid sequence. These new approaches, including recurrent neural networks and convolutional neural networks, use predicted in silico spectral libraries rather than experimental libraries to achieve higher sensitivity and/or specificity in the analysis of proteomics data. Machine learning is galvanizing applications that involve large search spaces, such as immunopeptidomics and proteogenomics. Current challenges in the field include the prediction of spectra for peptides with post-translational modifications and for cross-linked pairs of peptides. Permeation of machine-learning-based spectral prediction into search engines and spectrum-centric data-independent acquisition workflows for diverse peptide classes and measurement conditions will continue to push sensitivity and dynamic range in proteomics applications in the coming years.
Collapse
Affiliation(s)
- Jürgen Cox
- Computational Systems Biochemistry Research Group, Max-Planck Institute of Biochemistry, Martinsried, Germany.
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway.
| |
Collapse
|
24
|
Mou M, Pan Z, Lu M, Sun H, Wang Y, Luo Y, Zhu F. Application of Machine Learning in Spatial Proteomics. J Chem Inf Model 2022; 62:5875-5895. [PMID: 36378082 DOI: 10.1021/acs.jcim.2c01161] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Spatial proteomics is an interdisciplinary field that investigates the localization and dynamics of proteins, and it has gained extensive attention in recent years, especially the subcellular proteomics. Numerous evidence indicate that the subcellular localization of proteins is associated with various cellular processes and disease progression. Mass spectrometry (MS)-based and imaging-based experimental approaches have been developed to acquire large-scale spatial proteomic data. To allow the reliable analysis of increasingly complex spatial proteomics data, machine learning (ML) methods have been widely used in both MS-based and imaging-based spatial proteomic data analysis pipelines. Here, we comprehensively survey the applications of ML in spatial proteomics from following aspects: (1) data resources for spatial proteome are comprehensively introduced; (2) the roles of different ML algorithms in data analysis pipelines are elaborated; (3) successful applications of spatial proteomics and several analytical tools integrating ML methods are presented; (4) challenges existing in modern ML-based spatial proteomics studies are discussed. This review provides guidelines for researchers seeking to apply ML methods to analyze spatial proteomic data and can facilitate insightful understanding of cell biology as well as the future research in medical and drug discovery communities.
Collapse
Affiliation(s)
- Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ziqi Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingkun Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huaicheng Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
25
|
de Melo ALL, Linder A, Sundfeldt K, Lindquist D, Hedman H. Single-molecule array assay reveals the prognostic impact of plasma LRIG1 in ovarian carcinoma. Acta Oncol 2022; 61:1425-1433. [PMID: 36326616 DOI: 10.1080/0284186x.2022.2140016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Ovarian carcinoma is the eighth most common cause of cancer death in women worldwide. The disease is predominantly diagnosed at a late stage. This contributes to high recurrence rates, eventually leading to the development of treatment-resistant disease. Leucine-rich repeats and immunoglobulin-like domains protein 1 (LRIG1) is a transmembrane protein that functions as a tumor suppressor and regulator of growth factor signaling. LRIG1 levels have not been investigated in human plasma previously. MATERIALS AND METHODS A quantitative LRIG1-specific single molecule array assay was developed and validated. LRIG1 levels were quantified in plasma samples from 486 patients with suspicious ovarian masses. RESULTS Among women with ovarian carcinoma, LRIG1 levels were significantly elevated compared to women with benign or borderline type tumors. High LRIG1 plasma levels were associated with worse overall survival and shorter disease-free survival both in the group of all malignant cases and among the stage 3 cases only. LRIG1 was an independent prognostic factor in patients with stage 3 ovarian carcinoma. CONCLUSION LRIG1 plasma levels were elevated in patients with ovarian carcinoma, and high levels were associated with poor prognosis, suggesting that LRIG1 might be an etiologic factor and a potentially useful biomarker in ovarian carcinoma.
Collapse
Affiliation(s)
| | - Anna Linder
- Sahlgrenska Center for Cancer research, Department of Gynecology and Obstetrics, Institute of clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin Sundfeldt
- Sahlgrenska Center for Cancer research, Department of Gynecology and Obstetrics, Institute of clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - David Lindquist
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Håkan Hedman
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| |
Collapse
|
26
|
Surrogate peptide selection and internal standardization for accurate quantification of endogenous proteins. Bioanalysis 2022; 14:949-961. [PMID: 36017716 DOI: 10.4155/bio-2022-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Relative quantification techniques have dominated the field of proteomics. However, biomarker discovery, mathematical model development and studies on transporter-mediated drug disposition still need absolute quantification of proteins. The quality of data of trace-level protein quantification is solely dependent on the specific selection of surrogate peptides. Selection of surrogate peptides has a major impact on the accuracy of the method. In this article, the advanced approaches for selection of surrogate peptides, which can provide absolute quantification of the proteins are discussed. In addition, internal standardization, which accounts for variations in the quantitation process to achieve absolute protein quantification is discussed.
Collapse
|
27
|
Walzer M, García-Seisdedos D, Prakash A, Brack P, Crowther P, Graham RL, George N, Mohammed S, Moreno P, Papatheodorou I, Hubbard SJ, Vizcaíno JA. Implementing the reuse of public DIA proteomics datasets: from the PRIDE database to Expression Atlas. Sci Data 2022; 9:335. [PMID: 35701420 PMCID: PMC9197839 DOI: 10.1038/s41597-022-01380-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 05/12/2022] [Indexed: 11/14/2022] Open
Abstract
The number of mass spectrometry (MS)-based proteomics datasets in the public domain keeps increasing, particularly those generated by Data Independent Acquisition (DIA) approaches such as SWATH-MS. Unlike Data Dependent Acquisition datasets, the re-use of DIA datasets has been rather limited to date, despite its high potential, due to the technical challenges involved. We introduce a (re-)analysis pipeline for public SWATH-MS datasets which includes a combination of metadata annotation protocols, automated workflows for MS data analysis, statistical analysis, and the integration of the results into the Expression Atlas resource. Automation is orchestrated with Nextflow, using containerised open analysis software tools, rendering the pipeline readily available and reproducible. To demonstrate its utility, we reanalysed 10 public DIA datasets from the PRIDE database, comprising 1,278 SWATH-MS runs. The robustness of the analysis was evaluated, and the results compared to those obtained in the original publications. The final expression values were integrated into Expression Atlas, making SWATH-MS experiments more widely available and combining them with expression data originating from other proteomics and transcriptomics datasets.
Collapse
Affiliation(s)
- Mathias Walzer
- European Molecular Biology Laboratory, EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, United Kingdom.
| | - David García-Seisdedos
- European Molecular Biology Laboratory, EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Ananth Prakash
- European Molecular Biology Laboratory, EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Paul Brack
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Peter Crowther
- Melandra Limited, 16 Brook Road, Urmston, Manchester, M41 5RY, United Kingdom
| | - Robert L Graham
- School of Biological Sciences, Chlorine Gardens, Queen's University Belfast, Belfast, BT9 5DL, United Kingdom
| | - Nancy George
- European Molecular Biology Laboratory, EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Suhaib Mohammed
- European Molecular Biology Laboratory, EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Pablo Moreno
- European Molecular Biology Laboratory, EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Irene Papatheodorou
- European Molecular Biology Laboratory, EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Simon J Hubbard
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Juan Antonio Vizcaíno
- European Molecular Biology Laboratory, EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, United Kingdom.
| |
Collapse
|
28
|
Li H, Li T, Wang Y, Zhang S, Sheng H, Fu L. Liquid chromatography coupled to tandem mass spectrometry for comprehensive quantification of crustacean tropomyosin and arginine kinase in food matrix. Food Control 2022. [DOI: 10.1016/j.foodcont.2022.109137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
29
|
Nissa MU, Reddy PJ, Pinto N, Sun Z, Ghosh B, Moritz RL, Goswami M, Srivastava S. The PeptideAtlas of a widely cultivated fish Labeo rohita: A resource for the Aquaculture Community. Sci Data 2022; 9:171. [PMID: 35418183 PMCID: PMC9008064 DOI: 10.1038/s41597-022-01259-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 03/11/2022] [Indexed: 11/09/2022] Open
Abstract
Labeo rohita (Rohu) is one of the most important fish species produced in world aquaculture. Integrative omics research provides a strong platform to understand the basic biology and translate this knowledge into sustainable solutions in tackling disease outbreak, increasing productivity and ensuring food security. Mass spectrometry-based proteomics has provided insights to understand the biology in a new direction. Very little proteomics work has been done on 'Rohu' limiting such resources for the aquaculture community. Here, we utilised an extensive mass spectrometry based proteomic profiling data of 17 histologically normal tissues, plasma and embryo of Rohu to develop an open source PeptideAtlas. The current build of "Rohu PeptideAtlas" has mass-spectrometric evidence for 6015 high confidence canonical proteins at 1% false discovery rate, 2.9 million PSMs and ~150 thousand peptides. This is the first open-source proteomics repository for an aquaculture species. The 'Rohu PeptideAtlas' would promote basic and applied aquaculture research to address the most critical challenge of ensuring nutritional security for a growing population.
Collapse
Affiliation(s)
- Mehar Un Nissa
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | | | - Nevil Pinto
- Central Institute of Fisheries Education, Indian Council of Agricultural Research, Versova, Mumbai, Maharashtra, 400061, India
| | - Zhi Sun
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Biplab Ghosh
- Regional Centre for Biotechnology, Faridabad, 121001, India
| | | | - Mukunda Goswami
- Central Institute of Fisheries Education, Indian Council of Agricultural Research, Versova, Mumbai, Maharashtra, 400061, India.
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India.
| |
Collapse
|
30
|
Emwas AH, Szczepski K, Al-Younis I, Lachowicz JI, Jaremko M. Fluxomics - New Metabolomics Approaches to Monitor Metabolic Pathways. Front Pharmacol 2022; 13:805782. [PMID: 35387341 PMCID: PMC8977530 DOI: 10.3389/fphar.2022.805782] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/24/2022] [Indexed: 12/18/2022] Open
Abstract
Fluxomics is an innovative -omics research field that measures the rates of all intracellular fluxes in the central metabolism of biological systems. Fluxomics gathers data from multiple different -omics fields, portraying the whole picture of molecular interactions. Recently, fluxomics has become one of the most relevant approaches to investigate metabolic phenotypes. Metabolic flux using 13C-labeled molecules is increasingly used to monitor metabolic pathways, to probe the corresponding gene-RNA and protein-metabolite interaction networks in actual time. Thus, fluxomics reveals the functioning of multi-molecular metabolic pathways and is increasingly applied in biotechnology and pharmacology. Here, we describe the main fluxomics approaches and experimental platforms. Moreover, we summarize recent fluxomic results in different biological systems.
Collapse
Affiliation(s)
- Abdul-Hamid Emwas
- King Abdullah University of Science and Technology, Core Labs, Thuwal, Saudi Arabia
| | - Kacper Szczepski
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Inas Al-Younis
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Sciences & Engineering Division (BESE), Thuwal, Saudi Arabia
| | - Joanna Izabela Lachowicz
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| |
Collapse
|
31
|
Balbo Pogliano C, Ceppi I, Giovannini S, Petroulaki V, Palmer N, Uliana F, Gatti M, Kasaciunaite K, Freire R, Seidel R, Altmeyer M, Cejka P, Matos J. The CDK1-TOPBP1-PLK1 axis regulates the Bloom's syndrome helicase BLM to suppress crossover recombination in somatic cells. SCIENCE ADVANCES 2022; 8:eabk0221. [PMID: 35119917 PMCID: PMC8816346 DOI: 10.1126/sciadv.abk0221] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
Bloom's syndrome is caused by inactivation of the BLM helicase, which functions with TOP3A and RMI1-2 (BTR complex) to dissolve recombination intermediates and avoid somatic crossing-over. We show here that crossover avoidance by BTR further requires the activity of cyclin-dependent kinase-1 (CDK1), Polo-like kinase-1 (PLK1), and the DDR mediator protein TOPBP1, which act in the same pathway. Mechanistically, CDK1 phosphorylates BLM and TOPBP1 and promotes the interaction of both proteins with PLK1. This is amplified by the ability of TOPBP1 to facilitate phosphorylation of BLM at sites that stimulate both BLM-PLK1 and BLM-TOPBP1 binding, creating a positive feedback loop that drives rapid BLM phosphorylation at the G2-M transition. In vitro, BLM phosphorylation by CDK/PLK1/TOPBP1 stimulates the dissolution of topologically linked DNA intermediates by BLM-TOP3A. Thus, we propose that the CDK1-TOPBP1-PLK1 axis enhances BTR-mediated dissolution of recombination intermediates late in the cell cycle to suppress crossover recombination and curtail genomic instability.
Collapse
Affiliation(s)
| | - Ilaria Ceppi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), 6500 Bellinzona, Switzerland
| | - Sara Giovannini
- Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
| | - Vasiliki Petroulaki
- Department of Chromosome Biology, Max Perutz Labs, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Nathan Palmer
- Department of Chromosome Biology, Max Perutz Labs, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Federico Uliana
- Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
| | - Marco Gatti
- Department of Molecular Mechanisms of Disease, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Kristina Kasaciunaite
- Peter Debye Institute for Soft Matter Physics, Universität Leipzig, 04103 Leipzig, Germany
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias–FIISC, Ofra s/n, 38320 La Laguna, Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife, Spain
- Universidad Fernando Pessoa Canarias, 35450 Las Palmas de Gran Canaria, Spain
| | - Ralf Seidel
- Peter Debye Institute for Soft Matter Physics, Universität Leipzig, 04103 Leipzig, Germany
| | - Matthias Altmeyer
- Department of Molecular Mechanisms of Disease, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Petr Cejka
- Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), 6500 Bellinzona, Switzerland
| | - Joao Matos
- Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
- Department of Chromosome Biology, Max Perutz Labs, University of Vienna, Vienna Biocenter, Vienna, Austria
| |
Collapse
|
32
|
Balotf S, Wilson R, Tegg RS, Nichols DS, Wilson CR. Shotgun Proteomics as a Powerful Tool for the Study of the Proteomes of Plants, Their Pathogens, and Plant-Pathogen Interactions. Proteomes 2022; 10:5. [PMID: 35225985 PMCID: PMC8883913 DOI: 10.3390/proteomes10010005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/31/2022] Open
Abstract
The interaction between plants and pathogenic microorganisms is a multifaceted process mediated by both plant- and pathogen-derived molecules, including proteins, metabolites, and lipids. Large-scale proteome analysis can quantify the dynamics of proteins, biological pathways, and posttranslational modifications (PTMs) involved in the plant-pathogen interaction. Mass spectrometry (MS)-based proteomics has become the preferred method for characterizing proteins at the proteome and sub-proteome (e.g., the phosphoproteome) levels. MS-based proteomics can reveal changes in the quantitative state of a proteome and provide a foundation for understanding the mechanisms involved in plant-pathogen interactions. This review is intended as a primer for biologists that may be unfamiliar with the diverse range of methodology for MS-based shotgun proteomics, with a focus on techniques that have been used to investigate plant-pathogen interactions. We provide a summary of the essential steps required for shotgun proteomic studies of plants, pathogens and plant-pathogen interactions, including methods for protein digestion, identification, separation, and quantification. Finally, we discuss how protein PTMs may directly participate in the interaction between a pathogen and its host plant.
Collapse
Affiliation(s)
- Sadegh Balotf
- New Town Research Laboratories, Tasmanian Institute of Agriculture, University of Tasmania, New Town, TAS 7008, Australia; (S.B.); (R.S.T.)
| | - Richard Wilson
- Central Science Laboratory, University of Tasmania, Hobart, TAS 7001, Australia;
| | - Robert S. Tegg
- New Town Research Laboratories, Tasmanian Institute of Agriculture, University of Tasmania, New Town, TAS 7008, Australia; (S.B.); (R.S.T.)
| | - David S. Nichols
- Central Science Laboratory, University of Tasmania, Hobart, TAS 7001, Australia;
| | - Calum R. Wilson
- New Town Research Laboratories, Tasmanian Institute of Agriculture, University of Tasmania, New Town, TAS 7008, Australia; (S.B.); (R.S.T.)
| |
Collapse
|
33
|
Perez-Riverol Y, Bai J, Bandla C, García-Seisdedos D, Hewapathirana S, Kamatchinathan S, Kundu D, Prakash A, Frericks-Zipper A, Eisenacher M, Walzer M, Wang S, Brazma A, Vizcaíno J. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res 2022; 50:D543-D552. [PMID: 34723319 PMCID: PMC8728295 DOI: 10.1093/nar/gkab1038] [Citation(s) in RCA: 3742] [Impact Index Per Article: 1247.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
The PRoteomics IDEntifications (PRIDE) database (https://www.ebi.ac.uk/pride/) is the world's largest data repository of mass spectrometry-based proteomics data. PRIDE is one of the founding members of the global ProteomeXchange (PX) consortium and an ELIXIR core data resource. In this manuscript, we summarize the developments in PRIDE resources and related tools since the previous update manuscript was published in Nucleic Acids Research in 2019. The number of submitted datasets to PRIDE Archive (the archival component of PRIDE) has reached on average around 500 datasets per month during 2021. In addition to continuous improvements in PRIDE Archive data pipelines and infrastructure, the PRIDE Spectra Archive has been developed to provide direct access to the submitted mass spectra using Universal Spectrum Identifiers. As a key point, the file format MAGE-TAB for proteomics has been developed to enable the improvement of sample metadata annotation. Additionally, the resource PRIDE Peptidome provides access to aggregated peptide/protein evidences across PRIDE Archive. Furthermore, we will describe how PRIDE has increased its efforts to reuse and disseminate high-quality proteomics data into other added-value resources such as UniProt, Ensembl and Expression Atlas.
Collapse
Affiliation(s)
- Yasset Perez-Riverol
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Jingwen Bai
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Chakradhar Bandla
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - David García-Seisdedos
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Suresh Hewapathirana
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Selvakumar Kamatchinathan
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Deepti J Kundu
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Ananth Prakash
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Anika Frericks-Zipper
- Ruhr University Bochum, Medical Faculty, Medizinisches Proteom-Center, D-44801 Bochum, Germany
- Ruhr University Bochum, Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, 44801 Bochum, Germany
| | - Martin Eisenacher
- Ruhr University Bochum, Medical Faculty, Medizinisches Proteom-Center, D-44801 Bochum, Germany
- Ruhr University Bochum, Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, 44801 Bochum, Germany
| | - Mathias Walzer
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Shengbo Wang
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Alvis Brazma
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Juan Antonio Vizcaíno
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| |
Collapse
|
34
|
La Ferlita A, Alaimo S, Ferro A, Pulvirenti A. Pathway Analysis for Cancer Research and Precision Oncology Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:143-161. [DOI: 10.1007/978-3-030-91836-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Saha B, Chhatriya B, Pramanick S, Goswami S. Bioinformatic Analysis and Integration of Transcriptome and Proteome Results Identify Key Coding and Noncoding Genes Predicting Malignancy in Intraductal Papillary Mucinous Neoplasms of the Pancreas. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1056622. [PMID: 34790815 PMCID: PMC8592698 DOI: 10.1155/2021/1056622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/07/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Intraductal papillary mucinous neoplasms (IPMNs) are precursor lesions of pancreatic ductal adenocarcinoma (PDAC). IPMNs are generally associated with high risk of developing malignancy and therefore need to be diagnosed and assessed accurately, once detected. Existing diagnostic methods are inadequate, and identification of efficient biomarker capable of detecting high-risk IPMNs is necessitated. Moreover, the mechanism of development of malignancy in IPMNs is also elusive. METHODS Gene expression meta-analysis conducted using 12 low-risk IPMN and 23 high-risk IPMN tissue samples. We have also listed all the altered miRNAs and long noncoding RNAs (lncRNAs), identified their target genes, and performed pathway analysis. We further enlisted cyst fluid proteins detected to be altered in high-risk or malignant IPMNs and compared them with fraction of differentially expressed genes secreted into cyst fluid. RESULTS Our meta-analysis identified 270 upregulated and 161 downregulated genes characteristically altered in high-risk IPMNs. We further identified 61 miRNAs and 14 lncRNAs and their target genes and key pathways contributing towards understanding of the gene regulation during the progression of the disease. Most importantly, we have detected 12 genes altered significantly both in cystic lesions and cyst fluid. CONCLUSION Our study reports, for the first time, a meta-analysis identifying key changes in gene expression between low-risk and high-risk IPMNs and also explains the regulatory aspect through construction of a miRNA-lncRNA-mRNA interaction network. The 12-gene-signature could function as potential biomarker in cyst fluid for detection of IPMN with a high risk of developing malignancy.
Collapse
Affiliation(s)
- Barsha Saha
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | | | | | - Srikanta Goswami
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| |
Collapse
|
36
|
Vitorino R, Choudhury M, Guedes S, Ferreira R, Thongboonkerd V, Sharma L, Amado F, Srivastava S. Peptidomics and proteogenomics: background, challenges and future needs. Expert Rev Proteomics 2021; 18:643-659. [PMID: 34517741 DOI: 10.1080/14789450.2021.1980388] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION With available genomic data and related information, it is becoming possible to better highlight mutations or genomic alterations associated with a particular disease or disorder. The advent of high-throughput sequencing technologies has greatly advanced diagnostics, prognostics, and drug development. AREAS COVERED Peptidomics and proteogenomics are the two post-genomic technologies that enable the simultaneous study of peptides and proteins/transcripts/genes. Both technologies add a remarkably large amount of data to the pool of information on various peptides associated with gene mutations or genome remodeling. Literature search was performed in the PubMed database and is up to date. EXPERT OPINION This article lists various techniques used for peptidomic and proteogenomic analyses. It also explains various bioinformatics workflows developed to understand differentially expressed peptides/proteins and their role in disease pathogenesis. Their role in deciphering disease pathways, cancer research, and biomarker discovery using biofluids is highlighted. Finally, the challenges and future requirements to overcome the current limitations for their effective clinical use are also discussed.
Collapse
Affiliation(s)
- Rui Vitorino
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.,Laqv/requimte, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Manisha Choudhury
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Powai, India
| | - Sofia Guedes
- Laqv/requimte, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Rita Ferreira
- Laqv/requimte, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Francisco Amado
- Laqv/requimte, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Powai, India
| |
Collapse
|
37
|
Quantification of Changes in Protein Expression Using SWATH Proteomics. Methods Mol Biol 2021. [PMID: 34236656 DOI: 10.1007/978-1-0716-1641-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Sequential Window Acquisition of all THeoretical fragment ion spectra (SWATH) is a data independent acquisition mode used to accurately quantify thousands of proteins in a biological sample in a single run. It exploits fast scanning hybrid mass spectrometers to combine accuracy, reproducibility and sensitivity. This method requires the use of ion libraries, a sort of databases of spectral and chromatographic information about the proteins to be quantified. In this chapter, a typical workflow of SWATH experiment is described, from the sample preparation to the analysis of proteomics data.
Collapse
|
38
|
Belvedere R, Morretta E, Pessolano E, Novizio N, Tosco A, Porta A, Whiteford J, Perretti M, Filippelli A, Monti MC, Petrella A. Mesoglycan exerts its fibrinolytic effect through the activation of annexin A2. J Cell Physiol 2021; 236:4926-4943. [PMID: 33284486 DOI: 10.1002/jcp.30207] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Mesoglycan is a drug based on a mixture of glycosaminoglycans mainly used for the treatment of blood vessel diseases acting as antithrombotic and profibrinolytic drugs. Besides the numerous clinical studies, there is no information about its function on the fibrinolytic cascade. Here, we have elucidated the mechanism of action by which mesoglycan induces the activation of plasmin from endothelial cells. Surprisingly, by a proteomic analysis, we found that, following mesoglycan treatment, these cells show a notable amount of annexin A2 (ANXA2) at the plasma membrane. This protein has been widely associated with fibrinolysis and appears able to move to the membrane when phosphorylated. In our model, this translocation has proven to enhance cell migration, invasion, and angiogenesis. Furthermore, the interaction of mesoglycan with syndecan 4 (SDC4), a coreceptor belonging to the class of heparan sulfate proteoglycans, represents the upstream event of the ANXA2 behavior. Indeed, the activation of SDC4 triggers the motility of endothelial cells culminating in angiogenesis. Interestingly, mesoglycan can induce the release of plasmin in endothelial cell supernatants only in the presence of ANXA2. This evaluation suggests that mesoglycan triggers the formation of a chain mechanism starting from the activation of SDC4, and the related cascade of events, including src complex and PKCα activation, promoting the phosphorylation of ANXA2 and its translocation to plasma membrane. This indicates a connection among mesoglycan, SDC4-(PKCα-src), and ANXA2 which, in turn, links the tissue plasminogen activator bringing it closer to plasminogen. This latter is so cleaved to release the plasmin and degrade fibrin sleeves.
Collapse
Affiliation(s)
| | - Elva Morretta
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Emanuela Pessolano
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nunzia Novizio
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Alessandra Tosco
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - James Whiteford
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Amelia Filippelli
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi (SA), Italy
| | | | | |
Collapse
|
39
|
Sá JDO, Trino LD, Oliveira AK, Lopes AFB, Granato DC, Normando AGC, Santos ES, Neves LX, Carnielli CM, Paes Leme AF. Proteomic approaches to assist in diagnosis and prognosis of oral cancer. Expert Rev Proteomics 2021; 18:261-284. [PMID: 33945368 DOI: 10.1080/14789450.2021.1924685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Oral squamous cell carcinoma (OSCC) ranks among the top 10 leading causes of cancer worldwide, with 5-year survival rate of about 50%, high lymph node metastasis, and relapse rates. The OSCC diagnosis, prognosis, and treatment are mostly based on the clinical TNM classification. There is an urgent need for the discovery of biomarkers and therapeutic targets to assist in the clinical decision-making process.Areas covered: We summarize proteomic studies of the OSCC tumor, immune microenvironment, potential liquid biopsy sites, and post-translational modifications trying to retrieve information in the discovery and verification or (pre)validation phases. The search strategy was based on the combination of MeSH terms and expert refinement.Expert opinion: Untargeted combined with targeted proteomics are strategies that provide reliable and reproducible quantitation of proteins and are the methods of choice of many groups worldwide. Undoubtedly, proteomics has been contributing to the understanding of OSCC progression and uncovers potential candidates as biomarker or therapeutic targets. Nevertheless, none of these targets are available in the clinical practice yet. The scientific community needs to overcome the limitations by investing in robust experimental designs to strengthen the value of the findings, leveraging the translation of knowledge, and further supporting clinical decisions.
Collapse
Affiliation(s)
- Jamile De Oliveira Sá
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil.,Departamento De Diagnóstico Oral, Faculdade De Odontologia De Piracicaba, Universidade Estadual De Campinas (UNICAMP), Piracicaba, Brazil
| | - Luciana Daniele Trino
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Ana Karina Oliveira
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Ariane Fidelis Busso Lopes
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Daniela Campos Granato
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Ana Gabriela Costa Normando
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil.,Departamento De Diagnóstico Oral, Faculdade De Odontologia De Piracicaba, Universidade Estadual De Campinas (UNICAMP), Piracicaba, Brazil
| | - Erison Santana Santos
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil.,Departamento De Diagnóstico Oral, Faculdade De Odontologia De Piracicaba, Universidade Estadual De Campinas (UNICAMP), Piracicaba, Brazil
| | - Leandro Xavier Neves
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Carolina Moretto Carnielli
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Adriana Franco Paes Leme
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| |
Collapse
|
40
|
Christopher JA, Stadler C, Martin CE, Morgenstern M, Pan Y, Betsinger CN, Rattray DG, Mahdessian D, Gingras AC, Warscheid B, Lehtiö J, Cristea IM, Foster LJ, Emili A, Lilley KS. Subcellular proteomics. NATURE REVIEWS. METHODS PRIMERS 2021; 1:32. [PMID: 34549195 PMCID: PMC8451152 DOI: 10.1038/s43586-021-00029-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/15/2021] [Indexed: 12/11/2022]
Abstract
The eukaryotic cell is compartmentalized into subcellular niches, including membrane-bound and membrane-less organelles. Proteins localize to these niches to fulfil their function, enabling discreet biological processes to occur in synchrony. Dynamic movement of proteins between niches is essential for cellular processes such as signalling, growth, proliferation, motility and programmed cell death, and mutations causing aberrant protein localization are associated with a wide range of diseases. Determining the location of proteins in different cell states and cell types and how proteins relocalize following perturbation is important for understanding their functions, related cellular processes and pathologies associated with their mislocalization. In this Primer, we cover the major spatial proteomics methods for determining the location, distribution and abundance of proteins within subcellular structures. These technologies include fluorescent imaging, protein proximity labelling, organelle purification and cell-wide biochemical fractionation. We describe their workflows, data outputs and applications in exploring different cell biological scenarios, and discuss their main limitations. Finally, we describe emerging technologies and identify areas that require technological innovation to allow better characterization of the spatial proteome.
Collapse
Affiliation(s)
- Josie A. Christopher
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Charlotte Stadler
- Department of Protein Sciences, Karolinska Institutet, Science for Life Laboratory, Solna, Sweden
| | - Claire E. Martin
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Marcel Morgenstern
- Institute of Biology II, Biochemistry and Functional Proteomics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Yanbo Pan
- Department of Oncology and Pathology, Karolinska Institutet, Science for Life Laboratory, Solna, Sweden
| | - Cora N. Betsinger
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - David G. Rattray
- Department of Biochemistry & Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Diana Mahdessian
- Department of Protein Sciences, Karolinska Institutet, Science for Life Laboratory, Solna, Sweden
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Bettina Warscheid
- Institute of Biology II, Biochemistry and Functional Proteomics, Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS and CIBSS Signaling Research Centers, University of Freiburg, Freiburg, Germany
| | - Janne Lehtiö
- Department of Oncology and Pathology, Karolinska Institutet, Science for Life Laboratory, Solna, Sweden
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Leonard J. Foster
- Department of Biochemistry & Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew Emili
- Center for Network Systems Biology, Boston University, Boston, MA, USA
| | - Kathryn S. Lilley
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| |
Collapse
|
41
|
Sengupta A, Naresh G, Mishra A, Parashar D, Narad P. Proteome analysis using machine learning approaches and its applications to diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 127:161-216. [PMID: 34340767 DOI: 10.1016/bs.apcsb.2021.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
With the tremendous developments in the fields of biological and medical technologies, huge amounts of data are generated in the form of genomic data, images in medical databases or as data on protein sequences, and so on. Analyzing this data through different tools sheds light on the particulars of the disease and our body's reactions to it, thus, aiding our understanding of the human health. Most useful of these tools is artificial intelligence and deep learning (DL). The artificially created neural networks in DL algorithms help extract viable data from the datasets, and further, to recognize patters in these complex datasets. Therefore, as a part of machine learning, DL helps us face all the various challenges that come forth during protein prediction, protein identification and their quantification. Proteomics is the study of such proteins, their structures, features, properties and so on. As a form of data science, Proteomics has helped us progress excellently in the field of genomics technologies. One of the major techniques used in proteomics studies is mass spectrometry (MS). However, MS is efficient with analysis of large datasets only with the added help of informatics approaches for data analysis and interpretation; these mainly include machine learning and deep learning algorithms. In this chapter, we will discuss in detail the applications of deep learning and various algorithms of machine learning in proteomics.
Collapse
Affiliation(s)
- Abhishek Sengupta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - G Naresh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Astha Mishra
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Diksha Parashar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Priyanka Narad
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India.
| |
Collapse
|
42
|
Jackson TD, Hock DH, Fujihara KM, Palmer CS, Frazier AE, Low YC, Kang Y, Ang CS, Clemons NJ, Thorburn DR, Stroud DA, Stojanovski D. The TIM22 complex mediates the import of sideroflexins and is required for efficient mitochondrial one-carbon metabolism. Mol Biol Cell 2021; 32:475-491. [PMID: 33476211 PMCID: PMC8101445 DOI: 10.1091/mbc.e20-06-0390] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Acylglycerol kinase (AGK) is a mitochondrial lipid kinase that contributes to protein biogenesis as a subunit of the TIM22 complex at the inner mitochondrial membrane. Mutations in AGK cause Sengers syndrome, an autosomal recessive condition characterized by congenital cataracts, hypertrophic cardiomyopathy, skeletal myopathy, and lactic acidosis. We mapped the proteomic changes in Sengers patient fibroblasts and AGKKO cell lines to understand the effects of AGK dysfunction on mitochondria. This uncovered down-regulation of a number of proteins at the inner mitochondrial membrane, including many SLC25 carrier family proteins, which are predicted substrates of the complex. We also observed down-regulation of SFXN proteins, which contain five transmembrane domains, and show that they represent a novel class of TIM22 complex substrate. Perturbed biogenesis of SFXN proteins in cells lacking AGK reduces the proliferative capabilities of these cells in the absence of exogenous serine, suggesting that dysregulation of one-carbon metabolism is a molecular feature in the biology of Sengers syndrome.
Collapse
Affiliation(s)
- Thomas D Jackson
- Department of Biochemistry and Pharmacology and The Bio21 Molecular Science and Biotechnology Institute
| | - Daniella H Hock
- Department of Biochemistry and Pharmacology and The Bio21 Molecular Science and Biotechnology Institute
| | - Kenji M Fujihara
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3010, Australia.,Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Catherine S Palmer
- Department of Biochemistry and Pharmacology and The Bio21 Molecular Science and Biotechnology Institute
| | - Ann E Frazier
- Murdoch Children's Research Institute and.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Yau C Low
- Murdoch Children's Research Institute and.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Yilin Kang
- Department of Biochemistry and Pharmacology and The Bio21 Molecular Science and Biotechnology Institute
| | - Ching-Seng Ang
- Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nicholas J Clemons
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3010, Australia.,Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - David R Thorburn
- Murdoch Children's Research Institute and.,Victorian Clinical Genetics Services Royal Children's Hospital, Melbourne, Victoria 3052, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - David A Stroud
- Department of Biochemistry and Pharmacology and The Bio21 Molecular Science and Biotechnology Institute
| | - Diana Stojanovski
- Department of Biochemistry and Pharmacology and The Bio21 Molecular Science and Biotechnology Institute
| |
Collapse
|
43
|
Gao Y, Ping L, Duong D, Zhang C, Dammer EB, Li Y, Chen P, Chang L, Gao H, Wu J, Xu P. Mass-Spectrometry-Based Near-Complete Draft of the Saccharomyces cerevisiae Proteome. J Proteome Res 2021; 20:1328-1340. [PMID: 33443437 DOI: 10.1021/acs.jproteome.0c00721] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Proteomics approaches designed to catalogue all open reading frames (ORFs) under a defined set of growth conditions of an organism have flourished in recent years. However, no proteome has been sequenced completely so far. Here, we generate the largest yeast proteome data set, including 5610 identified proteins, using a strategy based on optimized sample preparation and high-resolution mass spectrometry. Among the 5610 identified proteins, 94.1% are core proteins, which achieves near-complete coverage of the yeast ORFs. Comprehensive analysis of missing proteins showed that proteins are missed mainly due to physical properties. A review of protein abundance shows that our proteome encompasses a uniquely broad dynamic range. Additionally, these values highly correlate with mRNA abundance, implying a high level of accuracy, sensitivity, and precision. We present examples of how the data could be used, including reannotating gene localization, providing expression evidence of pseudogenes. Our near-complete yeast proteome data set will be a useful and important resource for further systematic studies.
Collapse
Affiliation(s)
- Yuan Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China
| | - Lingyan Ping
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China
| | - Duc Duong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China.,Center for Neurodegenerative Diseases, Emory Proteomics Service Center, and Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Chengpu Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China
| | - Eric B Dammer
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China.,Center for Neurodegenerative Diseases, Emory Proteomics Service Center, and Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Yanchang Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China
| | - Peiru Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China
| | - Lei Chang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China
| | - Huiying Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China
| | - Junzhu Wu
- School of Basic Medical Science, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, School of Medicine, Wuhan University, Wuhan 430072, P. R. China
| | - Ping Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, P. R. China.,School of Basic Medical Science, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, School of Medicine, Wuhan University, Wuhan 430072, P. R. China.,Anhui Medical University, Hefei 230032, P. R. China.,Hebei Province Key Lab of Research and Application on Microbial Diversity, College of Life Sciences, Hebei University, Baoding, Hebei 071002, China
| |
Collapse
|
44
|
Hackett WE, Zaia J. Calculating Glycoprotein Similarities From Mass Spectrometric Data. Mol Cell Proteomics 2021; 20:100028. [PMID: 32883803 PMCID: PMC8724611 DOI: 10.1074/mcp.r120.002223] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/24/2020] [Accepted: 09/03/2020] [Indexed: 12/23/2022] Open
Abstract
Complex protein glycosylation occurs through biosynthetic steps in the secretory pathway that create macro- and microheterogeneity of structure and function. Required for all life forms, glycosylation diversifies and adapts protein interactions with binding partners that underpin interactions at cell surfaces and pericellular and extracellular environments. Because these biological effects arise from heterogeneity of structure and function, it is necessary to measure their changes as part of the quest to understand nature. Quite often, however, the assumption behind proteomics that posttranslational modifications are discrete additions that can be modeled using the genome as a template does not apply to protein glycosylation. Rather, it is necessary to quantify the glycosylation distribution at each glycosite and to aggregate this information into a population of mature glycoproteins that exist in a given biological system. To date, mass spectrometric methods for assigning singly glycosylated peptides are well-established. But it is necessary to quantify glycosylation heterogeneity accurately in order to gauge the alterations that occur during biological processes. The task is to quantify the glycosylated peptide forms as accurately as possible and then apply appropriate bioinformatics algorithms to the calculation of micro- and macro-similarities. In this review, we summarize current approaches for protein quantification as they apply to this glycoprotein similarity problem.
Collapse
Affiliation(s)
- William E Hackett
- Bioinformatics Program, Boston University, Boston, Massachusetts, USA
| | - Joseph Zaia
- Bioinformatics Program, Boston University, Boston, Massachusetts, USA; Department of Biochemistry, Boston University, Boston, Massachusetts, USA.
| |
Collapse
|
45
|
Kopczynski D, Hentschel A, Coman C, Schebb NH, Hornemann T, Mashek DG, Hartung NM, Shevchuk O, Schött HF, Lorenz K, Torta F, Burla B, Zahedi RP, Sickmann A, Kreutz MR, Ejsing CS, Medenbach J, Ahrends R. Simple Targeted Assays for Metabolic Pathways and Signaling: A Powerful Tool for Targeted Proteomics. Anal Chem 2020; 92:13672-13676. [PMID: 32865986 PMCID: PMC7586293 DOI: 10.1021/acs.analchem.0c02793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
![]()
We
introduce STAMPS, a pathway-centric web service for the development
of targeted proteomics assays. STAMPS guides the user by providing
several intuitive interfaces for a rapid and simplified method design.
Applying our curated framework to signaling and metabolic pathways,
we reduced the average assay development time by a factor of ∼150
and revealed that the insulin signaling is actively controlled by
protein abundance changes in insulin-sensitive and -resistance states.
Although at the current state STAMPS primarily contains mouse data,
it was designed for easy extension with additional organisms.
Collapse
Affiliation(s)
- Dominik Kopczynski
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Strasse 6b, 44227 Dortmund, Germany
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Strasse 6b, 44227 Dortmund, Germany
| | - Cristina Coman
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Strasse 6b, 44227 Dortmund, Germany.,Department of Analytical Chemistry, University of Vienna, Währinger Strasse 38, 1090 Vienna, Austria
| | - Nils Helge Schebb
- Institute for Food Toxicology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, 30173 Hannover, Germany.,Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gauss-Strasse 20, 42119 Wuppertal, Germany
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zürich, Wagistrasse 14 Schlieren, 8952 Zürich, Switzerland
| | - Douglas G Mashek
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, 401 East River Parkway, Minneapolis, Minnesota 55455, United States.,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55455, United States
| | - Nicole M Hartung
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gauss-Strasse 20, 42119 Wuppertal, Germany
| | - Olga Shevchuk
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Strasse 6b, 44227 Dortmund, Germany
| | - Hans-Frieder Schött
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Strasse 6b, 44227 Dortmund, Germany.,Singapore Lipidomics Incubator (SLING), Department of Biochemistry, YLL School of Medicine, National University of Singapore, 28 Medical Drive, #03-03, Singapore 117456, Singapore
| | - Kristina Lorenz
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Strasse 6b, 44227 Dortmund, Germany
| | - Federico Torta
- Singapore Lipidomics Incubator (SLING), Department of Biochemistry, YLL School of Medicine, National University of Singapore, 28 Medical Drive, #03-03, Singapore 117456, Singapore
| | - Bo Burla
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, 28 Medical Drive, #03-03, Singapore 117456, Singapore
| | - René P Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, 3755 Cote Sainte Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Strasse 6b, 44227 Dortmund, Germany.,Medizinische Fakultät, Medizinisches Proteom-Center (MPC), Ruhr-Universität Bochum, Gesundheitscampus 4, 44801 Bochum, Germany.,Department of Chemistry, College of Physical Sciences, University of Aberdeen, Meston Walk, Aberdeen AB24 3UE, Scotland, United Kingdom
| | - Michael R Kreutz
- Leibniz Group "Dendritic Organelles and Synaptic Function", University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology, ZMNH, Martinistrasse 52, 20251 Hamburg, Germany.,RG Neuroplasticity, Leibniz Institute for Neurobiology, Brenneckestraße 6, 39120 Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Christer S Ejsing
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M DK-5230, Denmark.,Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Jan Medenbach
- Institute of Biochemistry I, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Strasse 6b, 44227 Dortmund, Germany.,Department of Analytical Chemistry, University of Vienna, Währinger Strasse 38, 1090 Vienna, Austria
| |
Collapse
|
46
|
Affiliation(s)
- Hayden Wilkinson
- NIBRT GlycoScience Group, National Institute for Bioprocessing, Research and Training, Blackrock, Dublin, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
- UCD School of Medicine, College of Health and Agricultural Science, University College Dublin, Dublin, Ireland
| | - Radka Saldova
- NIBRT GlycoScience Group, National Institute for Bioprocessing, Research and Training, Blackrock, Dublin, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
- UCD School of Medicine, College of Health and Agricultural Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
47
|
Szymkowicz L, Wilson DJ, James DA. Development of a targeted nanoLC-MS/MS method for quantitation of residual toxins from Bordetella pertussis. J Pharm Biomed Anal 2020; 188:113395. [DOI: 10.1016/j.jpba.2020.113395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/29/2022]
|
48
|
Campos TL, Korhonen PK, Hofmann A, Gasser RB, Young ND. Combined use of feature engineering and machine-learning to predict essential genes in Drosophila melanogaster. NAR Genom Bioinform 2020; 2:lqaa051. [PMID: 33575603 PMCID: PMC7671374 DOI: 10.1093/nargab/lqaa051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/05/2020] [Accepted: 07/04/2020] [Indexed: 12/17/2022] Open
Abstract
Characterizing genes that are critical for the survival of an organism (i.e. essential) is important to gain a deep understanding of the fundamental cellular and molecular mechanisms that sustain life. Functional genomic investigations of the vinegar fly, Drosophila melanogaster, have unravelled the functions of numerous genes of this model species, but results from phenomic experiments can sometimes be ambiguous. Moreover, the features underlying gene essentiality are poorly understood, posing challenges for computational prediction. Here, we harnessed comprehensive genomic-phenomic datasets publicly available for D. melanogaster and a machine-learning-based workflow to predict essential genes of this fly. We discovered strong predictors of such genes, paving the way for computational predictions of essentiality in less-studied arthropod pests and vectors of infectious diseases.
Collapse
Affiliation(s)
- Tulio L Campos
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Pasi K Korhonen
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas Hofmann
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Neil D Young
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
49
|
Leuchsenring AB, Karlsson C, Bundgaard L, Malmström J, Heegaard PMH. Targeted mass spectrometry for Serum Amyloid A (SAA) isoform profiling in sequential blood samples from experimentally Staphylococcus aureus infected pigs. J Proteomics 2020; 227:103904. [PMID: 32702520 DOI: 10.1016/j.jprot.2020.103904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/19/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022]
Abstract
Serum amyloid A (SAA) is a well-described acute phase protein induced during the acute phase response (APR) to infection. Four isoform specific genes are found in most mammals. Depending on species, SAA3 and SAA4 are generally preferentially expressed extrahepatically whereas SAA1 and SAA2 are hepatic isoforms dominating the SAA serum pool. Little is known about how specific infections affect the serum SAA isoform profile, as SAA isoform discriminating antibodies are not generally available. An antibody independent, quantitative targeted MS method (Selected Reaction Monitoring, SRM) based on available information on porcine SAA isoform genes was developed and used to profile SAA in serum samples from pigs experimentally infected with Staphylococcus aureus (Sa). While results suggest SAA2 as the main circulating porcine SAA isoform, induced around 10 times compared to non-infected controls, total SAA serum concentrations reached only around 4 μg/mL, much lower than established previously by immunoassays. This might suggest that SAA isoform variants not detected by the SRM method might be present in porcine serum. The assay allows monitoring host responses to experimental infections, infectious diseases and inflammation states in the pig at an unprecedented level of detail. It can also be used in a non-calibrated (relative quantification) format. SIGNIFICANCE: We developed an SRM MS method which for the first time allowed the specific quantification of each of the circulating porcine SAA isoforms (SAA2, SAA3, SAA4). It was found that SAA2 is the dominating circulating isoform of SAA in the pig and that, during the acute phase response to Sa infection SAA2, SAA3 and SAA4 are induced approx. 10, 15 and 2 times, respectively. Absolute levels of the isoforms as determined by SRM MS were much lower than reported previously for total SAA quantified by immunosassays, suggesting the existence of hitherto non-described SAA variants. SRM MS holds great promise for the study of the basic biology of SAA isoforms with the potential to study an even broader range of SAA variants.
Collapse
Affiliation(s)
- Anna Barslund Leuchsenring
- Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Lyngby, Denmark
| | - Christofer Karlsson
- Department of Clinical Sciences, Lund, Division of Infection Medicine, Lund University, BMC, Lund, Sweden
| | - Louise Bundgaard
- Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Lyngby, Denmark
| | - Johan Malmström
- Department of Clinical Sciences, Lund, Division of Infection Medicine, Lund University, BMC, Lund, Sweden
| | - Peter M H Heegaard
- Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
50
|
Hock DH, Reljic B, Ang CS, Muellner-Wong L, Mountford HS, Compton AG, Ryan MT, Thorburn DR, Stroud DA. HIGD2A is Required for Assembly of the COX3 Module of Human Mitochondrial Complex IV. Mol Cell Proteomics 2020; 19:1145-1160. [PMID: 32317297 PMCID: PMC7338084 DOI: 10.1074/mcp.ra120.002076] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Assembly factors play a critical role in the biogenesis of mitochondrial respiratory chain complexes I-IV where they assist in the membrane insertion of subunits, attachment of co-factors, and stabilization of assembly intermediates. The major fraction of complexes I, III and IV are present together in large molecular structures known as respiratory chain supercomplexes. Several assembly factors have been proposed as required for supercomplex assembly, including the hypoxia inducible gene 1 domain family member HIGD2A. Using gene-edited human cell lines and extensive steady state, translation and affinity enrichment proteomics techniques we show that loss of HIGD2A leads to defects in the de novo biogenesis of mtDNA-encoded COX3, subsequent accumulation of complex IV intermediates and turnover of COX3 partner proteins. Deletion of HIGD2A also leads to defective complex IV activity. The impact of HIGD2A loss on complex IV was not altered by growth under hypoxic conditions, consistent with its role being in basal complex IV assembly. Although in the absence of HIGD2A we show that mitochondria do contain an altered supercomplex assembly, we demonstrate it to harbor a crippled complex IV lacking COX3. Our results redefine HIGD2A as a classical assembly factor required for building the COX3 module of complex IV.
Collapse
Affiliation(s)
- Daniella H Hock
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Boris Reljic
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Ching-Seng Ang
- Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, Victoria, Australia
| | - Linden Muellner-Wong
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Hayley S Mountford
- Brain and Mitochondrial Research, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Alison G Compton
- Brain and Mitochondrial Research, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael T Ryan
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - David R Thorburn
- Brain and Mitochondrial Research, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia; Mitochondrial Laboratory, Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - David A Stroud
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|