151
|
van den Broek I, Mastali M, Mouapi K, Bystrom C, Bairey Merz CN, Van Eyk JE. Quality Control and Outlier Detection of Targeted Mass Spectrometry Data from Multiplex Protein Panels. J Proteome Res 2020; 19:2278-2293. [PMID: 32270680 DOI: 10.1021/acs.jproteome.9b00854] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Increased throughput as well as increased multiplexing of liquid chromatography coupled to selected reaction monitoring mass spectrometry (LC-SRM-MS) assays for protein quantification challenges routine data analysis. Despite the measurement of multiple transitions from multiple peptides, for clinical applications a single (quantifier) transition from one (quantifier) signature peptide is used to represent the protein quantity with most data used solely to validate the quantifier result. To support the generation of reliable protein results from multiplexed LC-SRM-MS assays with large sample numbers, we developed a data analysis process for quality control and outlier detection using data from an 11-protein multiplex LC-SRM-MS method for dried blood samples (195 492 chromatographic peaks from 1481 samples * 11 proteins * 2 peptides * 3 transitions * 2 isotopologues). The 2-tiered data analysis process detects outliers for ion transition ratio, peptide ratio, and % difference between duplicates, applying less stringent criteria to samples with a small % difference between duplicates (Tier 1) and more stringent criteria to samples with unassessed or a large % difference between duplicates (Tier 2). After manual peak review, 1127 samples (76%) were selected based on the sample quality. The data analysis process thereafter automatically selected quantifier transitions/peptides, removed quality control failures and outliers (8%), averaged duplicates, and generated a comprehensive report listing 6085 quality controlled protein-level results. The proposed data analysis process serves as a starting point toward standardized data analysis of multiplexed LC-SRM-MS protein assays.
Collapse
Affiliation(s)
- Irene van den Broek
- Cedars-Sinai Precision Biomarker Laboratories, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, 9th Floor, Los Angeles, California 90048, United States
| | - Mitra Mastali
- Cedars-Sinai Precision Biomarker Laboratories, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, 9th Floor, Los Angeles, California 90048, United States.,Advanced Clinical BioSystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Kelly Mouapi
- Advanced Clinical BioSystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Cory Bystrom
- Cedars-Sinai Precision Biomarker Laboratories, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, 9th Floor, Los Angeles, California 90048, United States
| | - C Noel Bairey Merz
- Barbra Streisand Women's Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Jennifer E Van Eyk
- Cedars-Sinai Precision Biomarker Laboratories, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, 9th Floor, Los Angeles, California 90048, United States.,Advanced Clinical BioSystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Barbra Streisand Women's Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| |
Collapse
|
152
|
van der Burgt YEM, Siliakus KM, Cobbaert CM, Ruhaak LR. HILIC-MRM-MS for Linkage-Specific Separation of Sialylated Glycopeptides to Quantify Prostate-Specific Antigen Proteoforms. J Proteome Res 2020; 19:2708-2716. [PMID: 32142289 PMCID: PMC8280738 DOI: 10.1021/acs.jproteome.0c00050] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
![]()
Elevated serum prostate-specific
antigen (PSA) levels in body fluids
may indicate prostate cancer (PCa), but it is noted that the clinical
performance is rather poor. Specificity and sensitivity values of
20 and 94% at a cutoff value of 4.1 ng/mL, respectively, result in
overdiagnosis and unnecessary interventions. Previous exploratory
studies have indicated that the glycosylation of PSA potentially leads
to improved PCa diagnosis based on qualitative analyses. However,
the applied methods are not suited for a quantitative evaluation or
implementation in a medical laboratory. Therefore, in this proof-of-principle
study, we have evaluated the use of hydrophilic interaction liquid
chromatography (HILIC) in combination with targeted quantitative mass
spectrometry for the sialic acid linkage-specific analysis of PSA
glyco-proteoforms based on either trypsin or ArgC peptides. The efficiency
of PSA proteolysis was optimized as well as the glycopeptide separation
conditions (buffer type, strength, and pH). The HILIC-based analysis
of PSA glyco-proteoforms presented here has the potential for the
clinical validation of patient cohorts. The method shows the feasibility
of the use of a HILIC stationary phase for the separation of isomeric
glycopeptides to detect specific glyco-proteoforms. This is the first
step toward the development and evaluation of PSA glyco-proteoforms
for use in a clinical chemistry setting aiming for improved PCa diagnosis
or screening.
Collapse
Affiliation(s)
- Yuri E M van der Burgt
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Kasper M Siliakus
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
153
|
Chen Q, Jiang Y, Ren Y, Ying M, Lu B. Peptide Selection for Accurate Targeted Protein Quantification via a Dimethylation High-Resolution Mass Spectrum Strategy with a Peptide Release Kinetic Model. ACS OMEGA 2020; 5:3809-3819. [PMID: 32149207 PMCID: PMC7057324 DOI: 10.1021/acsomega.9b02002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 02/06/2020] [Indexed: 06/10/2023]
Abstract
A crucial step in accurate targeted protein quantification using targeted proteomics is to determine optimal proteotypic peptides representing targeted proteins. In this study, a workflow of peptide selection to determine proteotypic peptides using a dimethylation high-resolution mass spectrum strategy with a peptide release kinetic model was investigated and applied in peptide selection of bovine serum albumin. After specificity, digestibility, recovery, and stability evaluation of tryptic peptides in bovine serum albumin, the optimal proteotypic peptide was selected as LVNELTEFAK. The quantification method using LVNELTEFAK gave a linear range of 1-100 ppm with the coefficient greater than 0.9990, and the detection limit of bovine serum albumin in milk was 0.78 mg/kg. Compared with the proteotypic peptides selected by Skyline, the method showed a better performance in method validation. The workflow exhibited high comprehensiveness and efficiency in peptide selection, facilitating accurate targeted protein quantification in the food matrix, which lack protein standards.
Collapse
Affiliation(s)
- Qi Chen
- National
Engineering Laboratory of Intelligent Food Technology and Equipment,
Key Laboratory for Agro-Products Postharvest Handling of Ministry
of Agriculture, Key Laboratory for Agro-Products Nutritional Evaluation
of Ministry of Agriculture, Zhejiang Key Laboratory for Agro-Food
Processing, Fuli Institute of Food Science, College of Biosystems
Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- Ningbo
Research Institute, Zhejiang University, Ningbo 315100, China
| | - Yirong Jiang
- National
Engineering Laboratory of Intelligent Food Technology and Equipment,
Key Laboratory for Agro-Products Postharvest Handling of Ministry
of Agriculture, Key Laboratory for Agro-Products Nutritional Evaluation
of Ministry of Agriculture, Zhejiang Key Laboratory for Agro-Food
Processing, Fuli Institute of Food Science, College of Biosystems
Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- Ningbo
Research Institute, Zhejiang University, Ningbo 315100, China
| | - Yiping Ren
- Yangtze
Delta Region Institute of Tsinghua University, Jiaxing 314006, China
| | - Meirong Ying
- Zhejiang
Grain and Oil Product Quality Inspection Center, Hangzhou 310012, China
| | - Baiyi Lu
- National
Engineering Laboratory of Intelligent Food Technology and Equipment,
Key Laboratory for Agro-Products Postharvest Handling of Ministry
of Agriculture, Key Laboratory for Agro-Products Nutritional Evaluation
of Ministry of Agriculture, Zhejiang Key Laboratory for Agro-Food
Processing, Fuli Institute of Food Science, College of Biosystems
Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- Ningbo
Research Institute, Zhejiang University, Ningbo 315100, China
| |
Collapse
|
154
|
Venkatesh A, Aggarwal S, Kumar S, Rajyaguru S, Kumar V, Bankar S, Shastri J, Patankar S, Srivastava S. Comprehensive proteomics investigation of P. vivax-infected human plasma and parasite isolates. BMC Infect Dis 2020; 20:188. [PMID: 32122317 PMCID: PMC7053139 DOI: 10.1186/s12879-020-4885-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
Background In recent times, Plasmodium vivax (P. vivax) has become a serious threat to public health due to its ability to cause severe infection with fatal outcomes. Its unique biology makes it resilient to control measures that are otherwise effective against P. falciparum. A deeper understanding of P. vivax biology and pathogenesis is, therefore, essential for developing the right control strategies. Proteomics of P. falciparum has been helpful in studying disease biology and elucidating molecular mechanisms involved in the development of disease. However, unlike P. falciparum, proteomics data for P. vivax infection is minimal due to the absence of a continuous culture system. The dependence on clinical samples and animal models has drastically limited P. vivax research, creating critical knowledge gaps in our understanding of the disease. This study describes an in-depth proteomics analysis of P. vivax-infected human plasma and parasite isolates, to understand parasite biology, pathogenesis, and to identify new diagnostic targets for P. vivax malaria. Methods A mass-spectrometry- (MS) based proteomics approach (Q Exactive) was applied to analyze human plasma and parasite isolates from vivax malaria patients visiting a primary health centre in India. Additionally, a targeted proteomics assay was standardized for validating unique peptides of most recurring parasite proteins. Results Thirty-eight P. vivax proteins were detected in human plasma with high confidence. Several glycolytic enzymes were found along with hypothetical, cytoskeletal, ribosomal, and nuclear proteins. Additionally, 103 highly abundant P. vivax proteins were detected in parasite isolates. This represents the highest number of parasite proteins to be reported from clinical samples so far. Interestingly, five of these; three Plasmodium exported proteins (PVX_003545, PVX_003555 and PVX_121935), a hypothetical protein (PVX_083555) and Pvstp1 (subtelomeric transmembrane protein 1, PVX_094303) were found in both plasma and parasite isolates. Conclusions A parasite proteomics investigation is essential to understand disease pathobiology and design novel interventions. Control strategies against P. vivax also depend on early diagnosis. This work provides deeper insights into the biology of P. vivax by identifying proteins expressed by the parasite during its complex life-cycle within the human host. The study also reports antigens that may be explored as diagnostic candidates.
Collapse
Affiliation(s)
- Apoorva Venkatesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Shalini Aggarwal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Swati Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Srushti Rajyaguru
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Vipin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Sheetal Bankar
- Department of Microbiology, T. N. Medical College and BYL Nair Hospital, Mumbai, India
| | - Jayanthi Shastri
- Department of Microbiology, T. N. Medical College and BYL Nair Hospital, Mumbai, India
| | - Swati Patankar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| |
Collapse
|
155
|
Eshghi A, Pistawka AJ, Liu J, Chen M, Sinclair NJT, Hardie DB, Elliott M, Chen L, Newman R, Mohammed Y, Borchers CH. Concentration Determination of >200 Proteins in Dried Blood Spots for Biomarker Discovery and Validation. Mol Cell Proteomics 2020; 19:540-553. [PMID: 31896676 PMCID: PMC7050112 DOI: 10.1074/mcp.tir119.001820] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/06/2019] [Indexed: 12/26/2022] Open
Abstract
The use of protein biomarkers as surrogates for clinical endpoints requires extensive multilevel validation including development of robust and sensitive assays for precise measurement of protein concentration. Multiple reaction monitoring (MRM) is a well-established mass-spectrometric method that can be used for reproducible protein-concentration measurements in biological specimens collected via microsampling. The dried blood spot (DBS) microsampling technique can be performed non-invasively without the expertise of a phlebotomist, and can enhance analyte stability which facilitate the application of this technique in retrospective studies while providing lower storage and shipping costs, because cold-chain logistics can be eliminated. Thus, precise, sensitive, and multiplexed methods for measuring protein concentrations in DBSs can be used for de novo biomarker discovery and for biomarker quantification or verification experiments. To achieve this goal, MRM assays were developed for multiplexed concentration measurement of proteins in DBSs.The lower limit of quantification (LLOQ) was found to have a median total coefficient of variation (CV) of 18% for 245 proteins, whereas the median LLOQ was 5 fmol of peptide injected on column, and the median inter-day CV over 4 days for measuring endogenous protein concentration was 8%. The majority (88%) of the assays displayed parallelism, whereas the peptide standards remained stable throughout the assay workflow and after exposure to multiple freeze-thaw cycles. For 190 proteins, the measured protein concentrations remained stable in DBS stored at ambient laboratory temperature for up to 2 months. Finally, the developed assays were used to measure the concentration ranges for 200 proteins in twenty same sex, same race and age matched individuals.
Collapse
Affiliation(s)
- Azad Eshghi
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada.
| | - Adam J Pistawka
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada
| | - Jun Liu
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Michael Chen
- Island Medical Program, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Nicholas J T Sinclair
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada
| | - Darryl B Hardie
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada
| | - Monica Elliott
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada
| | - Lei Chen
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada
| | - Rachael Newman
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada
| | - Yassene Mohammed
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada; Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Christoph H Borchers
- University of Victoria - Genome BC Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada; Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec H3T 1E2, Canada; Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, Quebec H3T 1E2, Canada; Department of Data Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Skolkovo Innovation Center, Nobel St., Moscow143026, Russia.
| |
Collapse
|
156
|
Swatkoski SJ, Croley TR. Screening of Processed Foods for Transgenic Proteins from Genetically Engineered Plants Using Targeted Mass Spectrometry. Anal Chem 2020; 92:3455-3462. [PMID: 31961133 DOI: 10.1021/acs.analchem.9b05577] [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: 11/29/2022]
Abstract
Screening of food products for the presence of material from genetically engineered (GE) plants is typically done using deoxyribonucleic acid (DNA)-based methods to detect the presence of transgenic DNA. In this study, we have demonstrated the feasibility of using targeted mass spectrometry (MS) to detect a protein expressed by transgenic DNA to confirm the presence of GE plant material in processed foods. Scheduled parallel reaction monitoring (sPRM) was used to detect the enzyme, 5-enolpyruvulshikimate-3-phosphate synthase, from Agrobacterium sp. strain CP4 (CP4 EPSPS), which confers glyphosate tolerance in transgenic crops. Five CP4 EPSPS surrogate peptides and their corresponding retention times identified via data-dependent LC/MS/MS analysis of a glyphosate-tolerant soybean certified reference material, GTS 40-3-2, were used to develop the sPRM assay. The assay was used to screen four soy-based infant formulas, four corn-based cereals, corn tortilla chips, and cornmeal for the presence of CP4 EPSPS. At least four of the five selected surrogate peptides were detected in nine of the products analyzed, suggesting that targeted MS can serve as a complementary analytical method to DNA-based methods for the detection of material from GE plants in processed foods.
Collapse
Affiliation(s)
- Stephen J Swatkoski
- Center for Food Safety and Applied Nutrition , U.S. Food and Drug Administration , 5001 Campus Drive , College Park , Maryland 20740 , United States
| | - Timothy R Croley
- Center for Food Safety and Applied Nutrition , U.S. Food and Drug Administration , 5001 Campus Drive , College Park , Maryland 20740 , United States
| |
Collapse
|
157
|
Song Z, Pearce MC, Jiang Y, Yang L, Goodall C, Miranda CL, Milovancev M, Bracha S, Kolluri SK, Maier CS. Delineation of hypoxia-induced proteome shifts in osteosarcoma cells with different metastatic propensities. Sci Rep 2020; 10:727. [PMID: 31959767 PMCID: PMC6971036 DOI: 10.1038/s41598-019-56878-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone cancer in children and young adults. Solid tumors are characterized by intratumoral hypoxia, and hypoxic cells are associated with the transformation to aggressive phenotype and metastasis. The proteome needed to support an aggressive osteosarcoma cell phenotype remains largely undefined. To link metastatic propensity to a hypoxia-induced proteotype, we compared the protein profiles of two isogenic canine OS cell lines, POS (low metastatic) and HMPOS (highly metastatic), under normoxia and hypoxia. Label-free shotgun proteomics was applied to comprehensively characterize the hypoxia-responsive proteome profiles in the OS cell phenotypes. Hypothesis-driven parallel reaction monitoring was used to validate the differential proteins observed in the shotgun data and to monitor proteins of which we expected to exhibit hypoxia responsiveness, but which were absent in the label-free shotgun data. We established a "distance" score (|zHMPOS - zPOS|), and "sensitivity" score (|zHypoxia - zNormoxia) to quantitatively evaluate the proteome shifts exhibited by OS cells in response to hypoxia. Evaluation of the sensitivity scores for the proteome shifts observed and principal component analysis of the hypoxia-responsive proteins indicated that both cell types acquire a proteome that supports a Warburg phenotype with enhanced cell migration and proliferation characteristics. Cell migration and glucose uptake assays combined with protein function inhibitor studies provided further support that hypoxia-driven adaption of pathways associated with glycolytic metabolism, collagen biosynthesis and remodeling, redox regulation and immunomodulatory proteins typify a proteotype associated with an aggressive cancer cell phenotype. Our findings further suggest that proteins involved in collagen remodeling and immune editing may warrant further evaluation as potential targets for anti-metastatic treatment strategies in osteosarcoma.
Collapse
Affiliation(s)
- Zifeng Song
- Department of Chemistry, Oregon State University, Oregon, USA
| | - Martin C Pearce
- Department of Environmental & Molecular Toxicology, Oregon State University, Oregon, USA
| | - Yuan Jiang
- Department of Statistics, Oregon State University, Oregon, USA
| | - Liping Yang
- Department of Chemistry, Oregon State University, Oregon, USA
| | - Cheri Goodall
- College of Veterinary Medicine, Oregon State University, Oregon, USA
| | | | - Milan Milovancev
- College of Veterinary Medicine, Oregon State University, Oregon, USA
| | - Shay Bracha
- College of Veterinary Medicine, Oregon State University, Oregon, USA
| | - Siva K Kolluri
- Department of Environmental & Molecular Toxicology, Oregon State University, Oregon, USA
- Linus Pauling Institute, Oregon State University, Oregon, USA
| | - Claudia S Maier
- Department of Chemistry, Oregon State University, Oregon, USA.
- Linus Pauling Institute, Oregon State University, Oregon, USA.
| |
Collapse
|
158
|
Value of quantifying ABC transporters by mass spectrometry and impact on in vitro-to-in vivo prediction of transporter-mediated drug-drug interactions of rivaroxaban. Eur J Pharm Biopharm 2020; 148:27-37. [PMID: 31945490 DOI: 10.1016/j.ejpb.2020.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/04/2019] [Accepted: 01/10/2020] [Indexed: 11/20/2022]
Abstract
ABC transporters, such as P-gp and BCRP, are involved in rivaroxaban pharmacokinetics and can lead to drug-drug interactions (DDIs). Investigations of the victim role for rivaroxaban and transporter-mediated DDI are commonly performed using in vitro models. However, interpretation of rivaroxaban efflux transport and DDI studies in cell models may be influenced by P-gp and BCRP transporter abundance. This study aimed to develop an LC-MS/MS quantification method for assessing the relationship between transporter expression and functionality in Caco-2ATCC, Caco-2ECACC, MDCK-MDR1, MDCK-BCRP cell models. First, the relative and absolute quantities of the transporters were determined by LC-MS/MS. P-gp and BCRP expression was then confirmed by western blotting and immunofluorescence staining. Finally, P-gp and BCRP functional activities and half-inhibitory concentrations (IC50s) of two specific inhibitors (verapamil and ko143) were determined by bidirectional transport experiments. P-gp and BCRP protein expression was detected at the cell membrane and was greater in the respective transfected models. Efflux ratios were correlated with P-gp and BCRP quantities. The lowest IC50s were obtained in the MDCK-MDR1 and MDCK-BCRP models for verapamil and ko143, respectively. In conclusion, this study demonstrated that LC-MS/MS can accurately quantify P-gp and BCRP efflux transporters and thereby improve the interpretation of transport data and in vitro-in vivo correlations.
Collapse
|
159
|
Shao X, Taha IN, Clauser KR, Gao Y(T, Naba A. MatrisomeDB: the ECM-protein knowledge database. Nucleic Acids Res 2020; 48:D1136-D1144. [PMID: 31586405 PMCID: PMC6943062 DOI: 10.1093/nar/gkz849] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/13/2019] [Accepted: 09/30/2019] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix (ECM) is a complex and dynamic meshwork of cross-linked proteins that supports cell polarization and functions and tissue organization and homeostasis. Over the past few decades, mass-spectrometry-based proteomics has emerged as the method of choice to characterize the composition of the ECM of normal and diseased tissues. Here, we present a new release of MatrisomeDB, a searchable collection of curated proteomic data from 17 studies on the ECM of 15 different normal tissue types, six cancer types (different grades of breast cancers, colorectal cancer, melanoma, and insulinoma) and other diseases including vascular defects and lung and liver fibroses. MatrisomeDB (http://www.pepchem.org/matrisomedb) was built by retrieving raw mass spectrometry data files and reprocessing them using the same search parameters and criteria to allow for a more direct comparison between the different studies. The present release of MatrisomeDB includes 847 human and 791 mouse ECM proteoforms and over 350 000 human and 600 000 mouse ECM-derived peptide-to-spectrum matches. For each query, a hierarchically-clustered tissue distribution map, a peptide coverage map, and a list of post-translational modifications identified, are generated. MatrisomeDB is the most complete collection of ECM proteomic data to date and allows the building of a comprehensive ECM atlas.
Collapse
Affiliation(s)
- Xinhao Shao
- College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Isra N Taha
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | - Yu (Tom) Gao
- College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
- University of Illinois at Chicago Cancer Center, Chicago, IL 60612, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
- University of Illinois at Chicago Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
160
|
Mendes ML, Fougeras MR, Dittmar G. Analysis of ubiquitin signaling and chain topology cross-talk. J Proteomics 2020; 215:103634. [PMID: 31918034 DOI: 10.1016/j.jprot.2020.103634] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/13/2019] [Accepted: 01/05/2020] [Indexed: 12/12/2022]
Abstract
Protein ubiquitination is a powerful post-translational modification implicated in many cellular processes. Although ubiquitination is associated with protein degradation, depending on the topology of polyubiquitin chains, protein ubiquitination is connected to non-degradative events in DNA damage response, cell cycle control, immune response, trafficking, intracellular localization, and vesicle fusion events. It has been shown that a ubiquitin chain can contain two or more topologies at the same time. These branched chains add another level of complexity to ubiquitin signaling, increasing its versatility and specificity. Mass spectrometry-based proteomics has been playing an important role in the identification of all types of ubiquitin chains and linkages. This review aims to provide an overview of ubiquitin chain topology and associated signaling pathways and discusses the MS-based proteomic methodologies used to determine such topologies. SIGNIFICANCE: Ubiquitination plays important roles in many cellular processes. Proteins can be monoubiquitinated or polyubiquitinated forming non-branched or branched chains in a high number of possible combinations, each associated with different cellular processes. The detection and the topology of ubiquitin chains is thus of extreme importance in order to explain such processes. Advances in mass spectrometry based proteomics allowed for the discovery and topology mapping of many ubiquitin chains. This review revisits the state of the art in ubiquitin chain identification by mass spectrometry and gives an insight on the implication of such chains in many cellular processes.
Collapse
Affiliation(s)
- Marta L Mendes
- Proteomics of Cellular Signaling, Quantitative Biology Unit, Luxembourg Institute of Health, 1a Rue Thomas Edison, 1445 Strassen, Luxembourg
| | - Miriam R Fougeras
- Proteomics of Cellular Signaling, Quantitative Biology Unit, Luxembourg Institute of Health, 1a Rue Thomas Edison, 1445 Strassen, Luxembourg; Faculty of Science, Technology and Communication, University of Luxembourg, 2 avenue de l'Université, 4365, Esch-sur-Alzette, Luxembourg
| | - Gunnar Dittmar
- Proteomics of Cellular Signaling, Quantitative Biology Unit, Luxembourg Institute of Health, 1a Rue Thomas Edison, 1445 Strassen, Luxembourg; Faculty of Science, Technology and Communication, University of Luxembourg, 2 avenue de l'Université, 4365, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
161
|
Suárez-Fernández A, González-Antuña A, Rodríguez-González P, Alonso JIG. Determination of Cystatin C in human urine by isotope dilution tandem mass spectrometry. J Pharm Biomed Anal 2020; 177:112889. [PMID: 31568966 DOI: 10.1016/j.jpba.2019.112889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/13/2019] [Accepted: 09/19/2019] [Indexed: 11/29/2022]
Abstract
This work presents the development of a methodology for the accurate and precise quantification of the renal biomarker Cystatin C in human urine by Isotope Dilution Mass Spectrometry (IDMS). The procedure is based on the addition of a known quantity of the proteotypic peptide ALDFAVG*EYNK labelled with 13C2-glycine to the urine sample followed by protein hydrolysis using trypsin. Then, preconcentration and purification of the isotope diluted peptide was carried out by a selective monoclonal antibody bound to magnetic beads and final measurement was done after injection of the sample in a HPLC-MS/MS triple quadrupole instrument. The isotopic distribution of the isotope diluted proteotypic peptide was measured by low resolution selected reaction monitoring. Using this aquisition mode, the bandpass of the first quadrupole was widened (FWHM =13 u) so the whole isotopic clusters for both the natural abundance and the labelled peptides entered the collision cell. The proposed acquisition mode provided similar accuracy and precision than the regular SRM mode (FWHM =0.7 u) but a higher sensitivity was observed. The purification of the sample by antibody based enrichment of the target peptide was shown to remove interfering compounds more efficiently in comparison with a sample purification based on semipreparative liquid chromatography. Using 5 ng of the labelled peptide it was possible to quantify Cystatin C in human urine in patients with normal and impaired renal function. Recoveries from 100 to 104% were obtained in samples containing from 90 to 700 μg L-1 of Cystatin C with relative standard deviations from 0.5 to 6%. The stability of Cystatin C in urine samples was evaluated under different storage conditions showing that only when the urine samples were stored at room temperature during more than 10 days, a significant degradation of Cystatin C was observed.
Collapse
Affiliation(s)
- Amanda Suárez-Fernández
- Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, Julián Clavería 8, 33006 Oviedo, Spain
| | - Ana González-Antuña
- Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, Julián Clavería 8, 33006 Oviedo, Spain
| | - Pablo Rodríguez-González
- Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, Julián Clavería 8, 33006 Oviedo, Spain.
| | - J Ignacio García Alonso
- Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, Julián Clavería 8, 33006 Oviedo, Spain
| |
Collapse
|
162
|
Young JB, Keppel TR, Waas M, Salmon AE, Buchberger AR, Skumatz CM, Gundry RL, Kassem IS. Quantitative proteomic analysis of aqueous humor after rabbit lensectomy reveals differences in coagulation and immunomodulatory proteins. Mol Omics 2020; 16:126-137. [DOI: 10.1039/c9mo00169g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Proteomics of juvenile rabbit aqueous humor was evaluated before and after surgery to determine proteins responsible for postoperative outcomes.
Collapse
Affiliation(s)
- Jonathon B. Young
- Cell Biology, Neurobiology, & Anatomy
- Medical College of Wisconsin
- The Eye Institute
- Milwaukee
- USA
| | - Theodore R. Keppel
- Center for Biomedical Mass Spectrometry Research
- Medical College of Wisconsin
- Milwaukee
- USA
| | - Matthew Waas
- Biochemistry, Medical College of Wisconsin
- Milwaukee
- USA
| | - Alexander E. Salmon
- Cell Biology, Neurobiology, & Anatomy
- Medical College of Wisconsin
- The Eye Institute
- Milwaukee
- USA
| | - Amanda Rae Buchberger
- Center for Biomedical Mass Spectrometry Research
- Medical College of Wisconsin
- Milwaukee
- USA
| | | | - Rebekah L. Gundry
- Center for Biomedical Mass Spectrometry Research
- Medical College of Wisconsin
- Milwaukee
- USA
- Biochemistry, Medical College of Wisconsin
| | - Iris S. Kassem
- Cell Biology, Neurobiology, & Anatomy
- Medical College of Wisconsin
- The Eye Institute
- Milwaukee
- USA
| |
Collapse
|
163
|
Captur G, Heywood WE, Coats C, Rosmini S, Patel V, Lopes LR, Collis R, Patel N, Syrris P, Bassett P, O'Brien B, Moon JC, Elliott PM, Mills K. Identification of a Multiplex Biomarker Panel for Hypertrophic Cardiomyopathy Using Quantitative Proteomics and Machine Learning. Mol Cell Proteomics 2020; 19:114-127. [PMID: 31243064 PMCID: PMC6944230 DOI: 10.1074/mcp.ra119.001586] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/24/2019] [Indexed: 12/22/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is defined by pathological left ventricular hypertrophy (LVH). It is the commonest inherited cardiac condition and a significant number of high risk cases still go undetected until a sudden cardiac death (SCD) event. Plasma biomarkers do not currently feature in the assessment of HCM disease progression, which is tracked by serial imaging, or in SCD risk stratification, which is based on imaging parameters and patient/family history. There is a need for new HCM plasma biomarkers to refine disease monitoring and improve patient risk stratification. To identify new plasma biomarkers for patients with HCM, we performed exploratory myocardial and plasma proteomics screens and subsequently developed a multiplexed targeted liquid chromatography-tandem/mass spectrometry-based assay to validate the 26 peptide biomarkers that were identified. The association of discovered biomarkers with clinical phenotypes was prospectively tested in plasma from 110 HCM patients with LVH (LVH+ HCM), 97 controls, and 16 HCM sarcomere gene mutation carriers before the development of LVH (subclinical HCM). Six peptides (aldolase fructose-bisphosphate A, complement C3, glutathione S-transferase omega 1, Ras suppressor protein 1, talin 1, and thrombospondin 1) were increased significantly in the plasma of LVH+ HCM compared with controls and correlated with imaging markers of phenotype severity: LV wall thickness, mass, and percentage myocardial scar on cardiovascular magnetic resonance imaging. Using supervised machine learning (ML), this six-biomarker panel differentiated between LVH+ HCM and controls, with an area under the curve of ≥ 0.87. Five of these peptides were also significantly increased in subclinical HCM compared with controls. In LVH+ HCM, the six-marker panel correlated with the presence of nonsustained ventricular tachycardia and the estimated five-year risk of sudden cardiac death. Using quantitative proteomic approaches, we have discovered six potentially useful circulating plasma biomarkers related to myocardial substrate changes in HCM, which correlate with the estimated sudden cardiac death risk.
Collapse
Affiliation(s)
- Gabriella Captur
- UCL MRC Unit for Lifelong Health and Ageing, 1-19 Torrington Place, Fitzrovia, London WC1E 7HB, UK; Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, St. Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, UK
| | - Wendy E Heywood
- Translational Mass Spectrometry Research Group, UCL Institute of Child Health and Great Ormond Street Hospital, 30 Guilford Street, London WC1N 1EH, UK; Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Caroline Coats
- Translational Mass Spectrometry Research Group, UCL Institute of Child Health and Great Ormond Street Hospital, 30 Guilford Street, London WC1N 1EH, UK; Institute of Cardiovascular Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Stefania Rosmini
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, St. Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, UK
| | - Vimal Patel
- Institute of Cardiovascular Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Luis R Lopes
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, St. Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, UK; Institute of Cardiovascular Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Richard Collis
- Institute of Cardiovascular Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Nina Patel
- Translational Mass Spectrometry Research Group, UCL Institute of Child Health and Great Ormond Street Hospital, 30 Guilford Street, London WC1N 1EH, UK; Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Petros Syrris
- Institute of Cardiovascular Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Paul Bassett
- Biostatistics Joint Research Office, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ben O'Brien
- Department of Perioperative Medicine, St. Bartholomew's Hospital and Barts Heart Center, West Smithfield, London, EC1A 7BE, UK; William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - James C Moon
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, St. Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, UK; Institute of Cardiovascular Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Perry M Elliott
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, St. Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, UK; Institute of Cardiovascular Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, UCL Institute of Child Health and Great Ormond Street Hospital, 30 Guilford Street, London WC1N 1EH, UK; Institute of Child Health, University College London, London, WC1N 1EH, UK.
| |
Collapse
|
164
|
Jiang W, Ma T, Zhang C, Tang X, Xu Q, Meng X, Ma T. Identification of urinary candidate biomarkers of cisplatin-induced nephrotoxicity in patients with carcinoma. J Proteomics 2020; 210:103533. [DOI: 10.1016/j.jprot.2019.103533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/23/2019] [Accepted: 09/19/2019] [Indexed: 10/25/2022]
|
165
|
Klein JA, Zaia J. A Perspective on the Confident Comparison of Glycoprotein Site-Specific Glycosylation in Sample Cohorts. Biochemistry 2019; 59:3089-3097. [PMID: 31833756 DOI: 10.1021/acs.biochem.9b00730] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein glycosylation, resulting from glycosyl transferase reactions under complex control in the secretory pathway, consists of a distribution of related glycoforms at each glycosylation site. Because the biosynthetic substrate concentration and transport rates depend on architecture and other aspects of cellular phenotypes, site-specific glycosylation cannot be predicted accurately from genomic, transcriptomic, or proteomic information. Rather, it is necessary to quantify glycosylation at each protein site and how this changes among a sample cohort to provide information about disease mechanisms. At present, mature mass spectrometry-based methods allow for qualitative assignment of the glycan composition and glycosylation site of singly glycosylated proteolytic peptides. To make such quantitative comparisons, it is necessary to sample the glycosylation distribution with sufficient coverage and accuracy for confident assessment of the glycosylation changes that occur in the biological cohort. In this Perspective, we discuss the unmet needs for mass spectrometry acquisition methods and bioinformatics for the confident comparison of protein site-specific glycosylation among sample cohorts.
Collapse
|
166
|
Kwon OK, Ha YS, Lee JN, Kim S, Lee H, Chun SY, Kwon TG, Lee S. Comparative Proteome Profiling and Mutant Protein Identification in Metastatic Prostate Cancer Cells by Quantitative Mass Spectrometry-based Proteogenomics. Cancer Genomics Proteomics 2019; 16:273-286. [PMID: 31243108 DOI: 10.21873/cgp.20132] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/AIM Prostate cancer (PCa) is the most frequent cancer found in males worldwide. The aim of this study was to identify new biomarkers using mutated peptides for the prognosis and prediction of advanced PCa, based on proteogenomics. MATERIALS AND METHODS The tryptic peptides were analyzed by tandem mass tag-based quantitative proteomics. Proteogenomics were used to identify mutant peptides as novel biomarkers in advanced PCa. RESULTS Using a human database, increased levels of INTS7 and decreased levels of SH3BGRL were found to be associated with the aggressiveness of PCa. Using proteogenomics and a cancer mutation database, 70 mutant peptides were identified in PCa cell lines. Using parallel reaction monitoring, the expression of seven mutant peptides was found to be altered in tumors, amongst which CAPN2 D22E was the most significantly up-regulated mutant peptide in PCa tissues. CONCLUSION Altered mutant peptides present in PCa tissue could be used as new biomarkers in advanced PCa.
Collapse
Affiliation(s)
- Oh Kwang Kwon
- BK21 Plus KNU Multi-Omics-based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Department of Urology, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jun Nyung Lee
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Department of Urology, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sunjoo Kim
- BK21 Plus Team for Creative Leader Program for Pharmacomics-based Future, Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hyesuk Lee
- BK21 Plus Team for Creative Leader Program for Pharmacomics-based Future, Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - So Young Chun
- Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Tae Gyun Kwon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea .,Department of Urology, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics-based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
167
|
Minikel EV, Kuhn E, Cocco AR, Vallabh SM, Hartigan CR, Reidenbach AG, Safar JG, Raymond GJ, McCarthy MD, O'Keefe R, Llorens F, Zerr I, Capellari S, Parchi P, Schreiber SL, Carr SA. Domain-specific Quantification of Prion Protein in Cerebrospinal Fluid by Targeted Mass Spectrometry. Mol Cell Proteomics 2019; 18:2388-2400. [PMID: 31558565 PMCID: PMC6885701 DOI: 10.1074/mcp.ra119.001702] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/16/2019] [Indexed: 01/11/2023] Open
Abstract
Therapies currently in preclinical development for prion disease seek to lower prion protein (PrP) expression in the brain. Trials of such therapies are likely to rely on quantification of PrP in cerebrospinal fluid (CSF) as a pharmacodynamic biomarker and possibly as a trial endpoint. Studies using PrP ELISA kits have shown that CSF PrP is lowered in the symptomatic phase of disease, a potential confounder for reading out the effect of PrP-lowering drugs in symptomatic patients. Because misfolding or proteolytic cleavage could potentially render PrP invisible to ELISA even if its concentration were constant or increasing in disease, we sought to establish an orthogonal method for CSF PrP quantification. We developed a multi-species targeted mass spectrometry method based on multiple reaction monitoring (MRM) of nine PrP tryptic peptides quantified relative to an isotopically labeled recombinant protein standard for human samples, or isotopically labeled synthetic peptides for nonhuman species. Analytical validation experiments showed process replicate coefficients of variation below 15%, good dilution linearity and recovery, and suitable performance for both CSF and brain homogenate and across humans as well as preclinical species of interest. In n = 55 CSF samples from individuals referred to prion surveillance centers with rapidly progressive dementia, all six human PrP peptides, spanning the N- and C-terminal domains of PrP, were uniformly reduced in prion disease cases compared with individuals with nonprion diagnoses. Thus, lowered CSF PrP concentration in prion disease is a genuine result of the disease process and not an artifact of ELISA-based measurement. As a result, dose-finding studies for PrP lowering drugs may need to be conducted in presymptomatic at-risk individuals rather than in symptomatic patients. We provide a targeted mass spectrometry-based method suitable for preclinical quantification of CSF PrP as a tool for drug development.
Collapse
Affiliation(s)
- Eric Vallabh Minikel
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115; Prion Alliance, Cambridge, MA 02139; Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142.
| | - Eric Kuhn
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115
| | - Alexandra R Cocco
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Sonia M Vallabh
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115; Prion Alliance, Cambridge, MA 02139
| | | | - Andrew G Reidenbach
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Jiri G Safar
- Departments of Pathology and Neurology Case Western Reserve University, Cleveland, OH 44106
| | - Gregory J Raymond
- Laboratory of Persistent Viral Diseases, NIAID Rocky Mountain Labs, Hamilton, MT 59840
| | - Michael D McCarthy
- Environmental Health and Safety, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Rhonda O'Keefe
- Environmental Health and Safety, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Franc Llorens
- National Reference Center for TSE, Georg-August University, Göttingen, 37073, Germany; Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Inga Zerr
- National Reference Center for TSE, Georg-August University, Göttingen, 37073, Germany
| | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, 40139, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, 40123, Italy
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, 40139, Italy; Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, 40138, Italy
| | - Stuart L Schreiber
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Department of Chemistry & Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Steven A Carr
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142.
| |
Collapse
|
168
|
Hober A, Edfors F, Ryaboshapkina M, Malmqvist J, Rosengren L, Percy AJ, Lind L, Forsström B, Uhlén M, Oscarsson J, Miliotis T. Absolute Quantification of Apolipoproteins Following Treatment with Omega-3 Carboxylic Acids and Fenofibrate Using a High Precision Stable Isotope-labeled Recombinant Protein Fragments Based SRM Assay. Mol Cell Proteomics 2019; 18:2433-2446. [PMID: 31591263 PMCID: PMC6885709 DOI: 10.1074/mcp.ra119.001765] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Indexed: 11/20/2022] Open
Abstract
Stable isotope-labeled standard (SIS) peptides are used as internal standards in targeted proteomics to provide robust protein quantification, which is required in clinical settings. However, SIS peptides are typically added post trypsin digestion and, as the digestion efficiency can vary significantly between peptides within a protein, the accuracy and precision of the assay may be compromised. These drawbacks can be remedied by a new class of internal standards introduced by the Human Protein Atlas project, which are based on SIS recombinant protein fragments called SIS PrESTs. SIS PrESTs are added initially to the sample and SIS peptides are released on trypsin digestion. The SIS PrEST technology is promising for absolute quantification of protein biomarkers but has not previously been evaluated in a clinical setting. An automated and scalable solid phase extraction workflow for desalting and enrichment of plasma digests was established enabling simultaneous preparation of up to 96 samples. Robust high-precision quantification of 13 apolipoproteins was achieved using a novel multiplex SIS PrEST-based LC-SRM/MS Tier 2 assay in non-depleted human plasma. The assay exhibited inter-day coefficients of variation between 1.5% and 14.5% (median = 3.5%) and was subsequently used to investigate the effects of omega-3 carboxylic acids (OM3-CA) and fenofibrate on these 13 apolipoproteins in human plasma samples from a randomized placebo-controlled trial, EFFECT I (NCT02354976). No significant changes were observed in the OM3-CA arm, whereas treatment with fenofibrate significantly increased apoAII and reduced apoB, apoCI, apoE and apoCIV levels. The reduction in apoCIV following fenofibrate treatment is a novel finding. The study demonstrates that SIS PrESTs can facilitate the generation of robust multiplexed biomarker Tier 2 assays for absolute quantification of proteins in clinical studies.
Collapse
Affiliation(s)
- Andreas Hober
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden; Department of Protein Science, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden; Department of Protein Science, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Maria Ryaboshapkina
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Jonas Malmqvist
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Louise Rosengren
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Andrew J Percy
- Department of Applications Development, Cambridge Isotope Laboratories, Inc., Tewksbury, MA 01876
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Björn Forsström
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden; Department of Protein Science, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden; Department of Protein Science, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Oscarsson
- Global Medicines Development, Cardiovascular, Renal and Metabolism, AstraZeneca, Gothenburg, Sweden
| | - Tasso Miliotis
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
169
|
van Huizen NA, van Rosmalen J, Dekker LJM, Coebergh van den Braak RRJ, Verhoef C, IJzermans JNM, Luider TM. Identification of a Collagen Marker in Urine Improves the Detection of Colorectal Liver Metastases. J Proteome Res 2019; 19:153-160. [PMID: 31721589 DOI: 10.1021/acs.jproteome.9b00474] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Previously, we reported a combination of an urine collagen alpha-1(I) natural occurring peptide (NOP) AGPP(-OH)GEAGKP(-OH)GEQGVP(-OH)GDLGAP(-OH)GP (AGP) and serum carcinoembryonic antigen (CEA) to have the potential to detect colorectal liver metastasis (CRLM). The combined method requires further adaption for better sensitivity and specificity prior to clinical implementation. This mass spectrometry study aimed to identify additional collagen NOPs in urine and determine the most discriminating NOP panel. We improved the combined method on the basis of analysis of urine samples from 100 healthy controls and 100 CRLM patients. Two additional NOPs were identified: GPPGEAGK(-OH)P(-OH)GEQGVP(-OH)GDLGAP(-OH)GP (GPP), collagen alpha-1(I), and GNDGARGSDGQPGPP(-OH)GP(-OH)P(-OH)GTAGFP(-OH)GSP(-OH)GAK(-OH)GEVGP (GND), collagen alpha-1(III). A molecular model combining NOPs (AGP, GPP, and GND) and CEA was generated. Molecules that did not contribute significantly were removed, resulting in a model consisting of GND and CEA. With this model, 88% sensitivity and 88% specificity were reached in the discovery set and 75% sensitivity and 100% specificity in the validation set (control, n = 12; CRLM, n = 10). The AUC of the ROC curve is significantly higher than the current model based on AGP and CEA (p = 3.3 × 10-4). The new model performs better than the currently used techniques in the clinic that have a 57-70% sensitivity and a 90-96% specificity.
Collapse
|
170
|
Abstract
Abstract
Precision oncology aims to tailor clinical decisions specifically to patients with the objective of improving treatment outcomes. This can be achieved by leveraging omics information for accurate molecular characterization of tumors. Tumor tissue biopsies are currently the main source of information for molecular profiling. However, biopsies are invasive and limited in resolving spatiotemporal heterogeneity in tumor tissues. Alternative non-invasive liquid biopsies can exploit patient’s body fluids to access multiple layers of tumor-specific biological information (genomes, epigenomes, transcriptomes, proteomes, metabolomes, circulating tumor cells, and exosomes). Analysis and integration of these large and diverse datasets using statistical and machine learning approaches can yield important insights into tumor biology and lead to discovery of new diagnostic, predictive, and prognostic biomarkers. Translation of these new diagnostic tools into standard clinical practice could transform oncology, as demonstrated by a number of liquid biopsy assays already entering clinical use. In this review, we highlight successes and challenges facing the rapidly evolving field of cancer biomarker research.
Lay Summary
Precision oncology aims to tailor clinical decisions specifically to patients with the objective of improving treatment outcomes. The discovery of biomarkers for precision oncology has been accelerated by high-throughput experimental and computational methods, which can inform fine-grained characterization of tumors for clinical decision-making. Moreover, advances in the liquid biopsy field allow non-invasive sampling of patient’s body fluids with the aim of analyzing circulating biomarkers, obviating the need for invasive tumor tissue biopsies. In this review, we highlight successes and challenges facing the rapidly evolving field of liquid biopsy cancer biomarker research.
Collapse
|
171
|
Park J, Oh HJ, Han D, Wang JI, Park IA, Ryu HS, Kim Y. Parallel Reaction Monitoring-Mass Spectrometry (PRM-MS)-Based Targeted Proteomic Surrogates for Intrinsic Subtypes in Breast Cancer: Comparative Analysis with Immunohistochemical Phenotypes. J Proteome Res 2019; 19:2643-2653. [DOI: 10.1021/acs.jproteome.9b00490] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Joonho Park
- Department of Biomedical Engineering, Seoul National University College of Medicine, 103 Daehak-ro, Seoul 03080, Korea
| | - Hyeon Jeong Oh
- Department of Pathology, Seoul National University Hospital, 101 Daehak-ro, Seoul 03080, Korea
| | - Dohyun Han
- Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Seoul 03080, Korea
| | - Joseph I. Wang
- Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Seoul 03080, Korea
| | - In Ae Park
- Department of Pathology, Seoul National University Hospital, 101 Daehak-ro, Seoul 03080, Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, 101 Daehak-ro, Seoul 03080, Korea
| | - Youngsoo Kim
- Department of Biomedical Engineering, Seoul National University College of Medicine, 103 Daehak-ro, Seoul 03080, Korea
| |
Collapse
|
172
|
Silva ARM, Toyoshima MTK, Passarelli M, Di Mascio P, Ronsein GE. Comparing Data-Independent Acquisition and Parallel Reaction Monitoring in Their Abilities To Differentiate High-Density Lipoprotein Subclasses. J Proteome Res 2019; 19:248-259. [PMID: 31697504 DOI: 10.1021/acs.jproteome.9b00511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
High-density lipoprotein (HDL) is a diverse group of particles with multiple cardioprotective functions. HDL proteome follows HDL particle complexity. Many proteins were described in HDL, but consistent quantification of HDL protein cargo is still a challenge. To address this issue, the aim of this work was to compare data-independent acquisition (DIA) and parallel reaction monitoring (PRM) methodologies in their abilities to differentiate HDL subclasses through their proteomes. To this end, we first evaluated the analytical performances of DIA and PRM using labeled peptides in pooled digested HDL as a biological matrix. Next, we compared the quantification capabilities of the two methodologies for 24 proteins found in HDL2 and HDL3 from 19 apparently healthy subjects. DIA and PRM exhibited comparable linearity, accuracy, and precision. Moreover, both methodologies worked equally well, differentiating HDL subclasses' proteomes with high precision. Our findings may help to understand HDL functional diversity.
Collapse
Affiliation(s)
- Amanda R M Silva
- Departamento de Bioquímica , Instituto de Química, Universidade de São Paulo , São Paulo 05513970 , Brazil
| | - Marcos T K Toyoshima
- Laboratório de Lípides (LIM-10) , Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo , São Paulo 01246903 , Brazil.,Serviço de Onco-Endocrinologia, Instituto do Câncer do Estado de São Paulo Octávio Frias de Oliveira , Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo , São Paulo 01246000 , Brazil
| | - Marisa Passarelli
- Laboratório de Lípides (LIM-10) , Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo , São Paulo 01246903 , Brazil.,Programa de Pós-Graduação da Universidade Nove de Julho , São Paulo 01504001 , Brazil
| | - Paolo Di Mascio
- Departamento de Bioquímica , Instituto de Química, Universidade de São Paulo , São Paulo 05513970 , Brazil
| | - Graziella E Ronsein
- Departamento de Bioquímica , Instituto de Química, Universidade de São Paulo , São Paulo 05513970 , Brazil
| |
Collapse
|
173
|
Quantitative mass spectrometry-based proteomics in the era of model-informed drug development: Applications in translational pharmacology and recommendations for best practice. Pharmacol Ther 2019; 203:107397. [DOI: 10.1016/j.pharmthera.2019.107397] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/29/2019] [Indexed: 02/08/2023]
|
174
|
Liu X, Miao W, Huang M, Li L, Dai X, Wang Y. Elevated Hexokinase II Expression Confers Acquired Resistance to 4-Hydroxytamoxifen in Breast Cancer Cells. Mol Cell Proteomics 2019; 18:2273-2284. [PMID: 31519767 PMCID: PMC6823848 DOI: 10.1074/mcp.ra119.001576] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/26/2019] [Indexed: 12/11/2022] Open
Abstract
Tamoxifen has been clinically used in treating estrogen receptor (ER)-positive breast cancer for over 30 years. The most challenging aspect associated with tamoxifen therapy is the development of resistance in initially responsive breast tumors. We applied a parallel-reaction monitoring (PRM)-based quantitative proteomic method to examine the differential expression of kinase proteins in MCF-7 and the isogenic tamoxifen-resistant (TamR) cells. We were able to quantify the relative protein expression levels of 315 kinases, among which hexokinase 2 (HK2) and mTOR were up-regulated in TamR MCF-7 cells. We also observed that the TamR MCF-7 cells exhibited elevated rate of glycolysis than the parental MCF-7 cells. In addition, we found that phosphorylation of S6K - a target of mTOR - was much lower in TamR MCF-7 cells, and this phosphorylation level could be restored upon genetic depletion or pharmacological inhibition of HK2. Reciprocally, the level of S6K phosphorylation was diminished upon overexpression of HK2 in MCF-7 cells. Moreover, we observed that HK2 interacts with mTOR, and this interaction inhibits mTOR activity. Lower mTOR activity led to augmented autophagy, which conferred resistance of MCF-7 cells toward tamoxifen. Together, our study demonstrates that elevated expression of HK2 promotes autophagy through inhibiting the mTOR-S6K signaling pathway and results in resistance of MCF-7 breast cancer cells toward tamoxifen; thus, our results uncovered, for the first time, HK2 as a potential therapeutic target for overcoming tamoxifen resistance.
Collapse
Affiliation(s)
- Xiaochuan Liu
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Weili Miao
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Ming Huang
- Environmental Toxicology Graduate Program, University of California, Riverside, Riverside, CA 92521
| | - Lin Li
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Xiaoxia Dai
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521; Environmental Toxicology Graduate Program, University of California, Riverside, Riverside, CA 92521.
| |
Collapse
|
175
|
Radko SP, Poverennaya EV, Kurbatov LK, Ponomarenko EA, Lisitsa AV, Archakov AI. The "Missing" Proteome: Undetected Proteins, Not-Translated Transcripts, and Untranscribed Genes. J Proteome Res 2019; 18:4273-4276. [PMID: 31621326 DOI: 10.1021/acs.jproteome.9b00383] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The Chromosome-centric Human Proteome Project aims at characterizing the expression of proteins encoded in each chromosome at the tissue, cell, and subcellular levels. The proteomic profiling of a particular tissue or cell line commonly results in a substantial portion of proteins that are not observed (the "missing" proteome). The concurrent transcriptome profiling of the analyzed tissue/cells samples may help define the set of untranscribed genes in a given type of tissue or cell, thus narrowing the size of the "missing" proteome and allowing us to focus on defining the reasons behind undetected proteins, namely, whether they are technical (insufficient sensitivity of protein detection) or biological (correspond to not-translated transcripts). We believe that the quantitative polymerase chain reaction (qPCR) can provide an efficient approach to studying low-abundant transcripts related to undetected proteins due to its high sensitivity and the possibility of ensuring the specificity of detection via the simple Sanger sequencing of PCR products. Here we illustrated the feasibility of such an approach on a set of low-abundant transcripts. Although inapplicable to the analysis of whole transcriptome, qPCR can successfully be utilized to profile a limited cohort of transcripts encoded on a particular chromosome, as we previously demonstrated for human chromosome 18.
Collapse
Affiliation(s)
- Sergey P Radko
- Institute of Biomedical Chemistry , 119121 Moscow , Russia
| | | | | | | | | | | |
Collapse
|
176
|
Ignjatovic V, Geyer PE, Palaniappan KK, Chaaban JE, Omenn GS, Baker MS, Deutsch EW, Schwenk JM. Mass Spectrometry-Based Plasma Proteomics: Considerations from Sample Collection to Achieving Translational Data. J Proteome Res 2019; 18:4085-4097. [PMID: 31573204 DOI: 10.1021/acs.jproteome.9b00503] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The proteomic analysis of human blood and blood-derived products (e.g., plasma) offers an attractive avenue to translate research progress from the laboratory into the clinic. However, due to its unique protein composition, performing proteomics assays with plasma is challenging. Plasma proteomics has regained interest due to recent technological advances, but challenges imposed by both complications inherent to studying human biology (e.g., interindividual variability) and analysis of biospecimens (e.g., sample variability), as well as technological limitations remain. As part of the Human Proteome Project (HPP), the Human Plasma Proteome Project (HPPP) brings together key aspects of the plasma proteomics pipeline. Here, we provide considerations and recommendations concerning study design, plasma collection, quality metrics, plasma processing workflows, mass spectrometry (MS) data acquisition, data processing, and bioinformatic analysis. With exciting opportunities in studying human health and disease though this plasma proteomics pipeline, a more informed analysis of human plasma will accelerate interest while enhancing possibilities for the incorporation of proteomics-scaled assays into clinical practice.
Collapse
Affiliation(s)
- Vera Ignjatovic
- Haematology Research , Murdoch Children's Research Institute , Parkville , VIC 3052 , Australia.,Department of Paediatrics , The University of Melbourne , Parkville , VIC 3052 , Australia
| | - Philipp E Geyer
- NNF Center for Protein Research, Faculty of Health Sciences , University of Copenhagen , 2200 Copenhagen , Denmark.,Department of Proteomics and Signal Transduction , Max Planck Institute of Biochemistry , 82152 Martinsried , Germany
| | - Krishnan K Palaniappan
- Freenome , 259 East Grand Avenue , South San Francisco , California 94080 , United States
| | - Jessica E Chaaban
- Haematology Research , Murdoch Children's Research Institute , Parkville , VIC 3052 , Australia
| | - Gilbert S Omenn
- Departments of Computational Medicine & Bioinformatics, Human Genetics, and Internal Medicine and School of Public Health , University of Michigan , 100 Washtenaw Avenue , Ann Arbor , Michigan 48109-2218 , United States
| | - Mark S Baker
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences , Macquarie University , 75 Talavera Road , North Ryde , NSW 2109 , Australia
| | - Eric W Deutsch
- Institute for Systems Biology , 401 Terry Avenue North , Seattle , Washington 98109 , United States
| | - Jochen M Schwenk
- Affinity Proteomics, SciLifeLab , KTH Royal Institute of Technology , 171 65 Stockholm , Sweden
| |
Collapse
|
177
|
Vandenbrouck Y, Christiany D, Combes F, Loux V, Brun V. Bioinformatics Tools and Workflow to Select Blood Biomarkers for Early Cancer Diagnosis: An Application to Pancreatic Cancer. Proteomics 2019; 19:e1800489. [DOI: 10.1002/pmic.201800489] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/11/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Yves Vandenbrouck
- University of Grenoble Alpes, INSERM, CEA, IRIG‐BGE, U1038 Grenoble 38000 France
| | - David Christiany
- University of Grenoble Alpes, INSERM, CEA, IRIG‐BGE, U1038 Grenoble 38000 France
- MaIAGE, INRA, Université Paris‐Saclay Jouy‐en‐Josas 78350 France
| | - Florence Combes
- University of Grenoble Alpes, INSERM, CEA, IRIG‐BGE, U1038 Grenoble 38000 France
| | - Valentin Loux
- MaIAGE, INRA, Université Paris‐Saclay Jouy‐en‐Josas 78350 France
| | - Virginie Brun
- University of Grenoble Alpes, INSERM, CEA, IRIG‐BGE, U1038 Grenoble 38000 France
| |
Collapse
|
178
|
Fernández-Puente P, González-Rodríguez L, Calamia V, Picchi F, Lourido L, Camacho-Encina M, Oreiro N, Rocha B, Paz-González R, Marina A, García C, Blanco FJ, Ruiz-Romero C. Analysis of Endogenous Peptides Released from Osteoarthritic Cartilage Unravels Novel Pathogenic Markers. Mol Cell Proteomics 2019; 18:2018-2028. [PMID: 31352363 PMCID: PMC6773562 DOI: 10.1074/mcp.ra119.001554] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/08/2019] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is a pathology characterized by the loss of articular cartilage. In this study, we performed a peptidomic strategy to identify endogenous peptides (neopeptides) that are released from human osteoarthritic tissue, which may serve as disease markers. With this aim, secretomes of osteoarthritic and healthy articular cartilages obtained from knee and hip were analyzed by shotgun peptidomics. This discovery step led to the identification of 1175 different peptides, corresponding to 101 proteins, as products of the physiological or pathological turnover of cartilage extracellular matrix. Then, a targeted multiple reaction monitoring-mass spectrometry method was developed to quantify the panel of best marker candidates on a larger set of samples (n = 62). Statistical analyses were performed to evaluate the significance of the observed differences and the ability of the neopeptides to classify the tissue. Eight of them were differentially abundant in the media from wounded zones of OA cartilage compared with the healthy tissue (p < 0.05). Three neopeptides belonging to Clusterin and one from Cartilage Oligomeric Matrix Protein showed a disease-dependent decrease specifically in hip OA, whereas two from Prolargin (PRELP) and one from Cartilage Intermediate Layer Protein 1 were significantly increased in samples from knee OA. The release of one peptide from PRELP showed the best metrics for tissue classification (AUC = 0.834). The present study reveals specific neopeptides that are differentially released from knee or hip human osteoarthritic cartilage compared with healthy tissue. This evidences the intervention of characteristic pathogenic pathways in OA and provides a novel panel of peptidic candidates for biomarker development.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Biomarkers/metabolism
- Cartilage, Articular/cytology
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Case-Control Studies
- Cells, Cultured
- Chromatography, Liquid
- Culture Media, Conditioned/chemistry
- Extracellular Matrix/metabolism
- Female
- Humans
- Male
- Organ Specificity
- Osteoarthritis, Hip/metabolism
- Osteoarthritis, Hip/pathology
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/pathology
- Peptides/metabolism
- Proteomics/methods
- Tandem Mass Spectrometry
Collapse
Affiliation(s)
- Patricia Fernández-Puente
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain.; Agrupación Estratégica CICA - INIBIC, Universidade da Coruña, 15071 A Coruña, Spain
| | - Lucía González-Rodríguez
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Valentina Calamia
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Florencia Picchi
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Lucía Lourido
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - María Camacho-Encina
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Natividad Oreiro
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Beatriz Rocha
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Rocío Paz-González
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain
| | - Anabel Marina
- Centro de Biología Molecular Severo Ochoa, CSIC. Nicolás Cabrera, 1, 28049 Madrid, Spain
| | - Carlos García
- Centro de Biología Molecular Severo Ochoa, CSIC. Nicolás Cabrera, 1, 28049 Madrid, Spain
| | - Francisco J Blanco
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain.; Departamento de Medicina, Fisioterapia y Ciencias Biomédicas. Universidade da Coruña, 15006 A Coruña, Spain.; RIER-RED de Inflamación y Enfermedades Reumáticas, INIBIC-CHUAC, As Xubias 84, 15006 A Coruña, Spain.
| | - Cristina Ruiz-Romero
- Proteomics Unit-PBR2-ProteoRed/ISCIII, Grupo de Investigación de Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS. As Xubias, 84, 15006 A Coruña, Spain.; CIBER-BBN Instituto de Salud Carlos III INIBIC-CHUAC As Xubias 84, 15006 A Coruna, Spain.
| |
Collapse
|
179
|
Newhardt MF, Batushansky A, Matsuzaki S, Young ZT, West M, Chin NC, Szweda LI, Kinter M, Humphries KM. Enhancing cardiac glycolysis causes an increase in PDK4 content in response to short-term high-fat diet. J Biol Chem 2019; 294:16831-16845. [PMID: 31562244 DOI: 10.1074/jbc.ra119.010371] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/18/2019] [Indexed: 12/17/2022] Open
Abstract
The healthy heart has a dynamic capacity to respond and adapt to changes in nutrient availability. Metabolic inflexibility, such as occurs with diabetes, increases cardiac reliance on fatty acids to meet energetic demands, and this results in deleterious effects, including mitochondrial dysfunction, that contribute to pathophysiology. Enhancing glucose usage may mitigate metabolic inflexibility and be advantageous under such conditions. Here, we sought to identify how mitochondrial function and cardiac metabolism are affected in a transgenic mouse model of enhanced cardiac glycolysis (GlycoHi) basally and following a short-term (7-day) high-fat diet (HFD). GlycoHi mice constitutively express an active form of phosphofructokinase-2, resulting in elevated levels of the PFK-1 allosteric activator fructose 2,6-bisphosphate. We report that basally GlycoHi mitochondria exhibit augmented pyruvate-supported respiration relative to fatty acids. Nevertheless, both WT and GlycoHi mitochondria had a similar shift toward increased rates of fatty acid-supported respiration following HFD. Metabolic profiling by GC-MS revealed distinct features based on both genotype and diet, with a unique increase in branched-chain amino acids in the GlycoHi HFD group. Targeted quantitative proteomics analysis also supported both genotype- and diet-dependent changes in protein expression and uncovered an enhanced expression of pyruvate dehydrogenase kinase 4 (PDK4) in the GlycoHi HFD group. These results support a newly identified mechanism whereby the levels of fructose 2,6-bisphosphate promote mitochondrial PDK4 levels and identify a secondary adaptive response that prevents excessive mitochondrial pyruvate oxidation when glycolysis is sustained after a high-fat dietary challenge.
Collapse
Affiliation(s)
- Maria F Newhardt
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104.,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Satoshi Matsuzaki
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Zachary T Young
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Melinda West
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Ngun Cer Chin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Luke I Szweda
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8573
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Kenneth M Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104 .,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| |
Collapse
|
180
|
Paulo JA, Gygi SP. mTMT: An Alternative, Nonisobaric, Tandem Mass Tag Allowing for Precursor-Based Quantification. Anal Chem 2019; 91:12167-12172. [PMID: 31490667 DOI: 10.1021/acs.analchem.9b03162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Stable isotope labeling of peptides is the basis for numerous mass-spectrometry-based quantification strategies. Isobaric tagging and metabolic labeling, namely, tandem mass tagging (TMT) and SILAC, are among the most widely used techniques for relative protein quantification. Here we report an alternative, precursor-based quantification method using nonisobaric TMT variants: TMTzero (TMT0) and superheavy TMT (shTMT). We term this strategy mass difference tandem mass tagging (mTMT). These TMT variants differ by 11 mass units; however, peptides labeled with these reagents coelute, analogous to SILAC-labeled peptide pairs. As a proof-of-concept, we profiled the proteomes of two cell lines that are frequently used in neuroscience studies, SH-SY5Y and SVGp12, using mTMT and standard SILAC-labeling approaches. We show similar quantified proteins and peptides for each method, with highly correlated fold-changes between workflows. We conclude that mTMT is a suitable alternative for precursor-based protein quantification.
Collapse
Affiliation(s)
- Joao A Paulo
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Steven P Gygi
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| |
Collapse
|
181
|
Fert-Bober J, Murray CI, Parker SJ, Van Eyk JE. Precision Profiling of the Cardiovascular Post-Translationally Modified Proteome: Where There Is a Will, There Is a Way. Circ Res 2019; 122:1221-1237. [PMID: 29700069 DOI: 10.1161/circresaha.118.310966] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There is an exponential increase in biological complexity as initial gene transcripts are spliced, translated into amino acid sequence, and post-translationally modified. Each protein can exist as multiple chemical or sequence-specific proteoforms, and each has the potential to be a critical mediator of a physiological or pathophysiological signaling cascade. Here, we provide an overview of how different proteoforms come about in biological systems and how they are most commonly measured using mass spectrometry-based proteomics and bioinformatics. Our goal is to present this information at a level accessible to every scientist interested in mass spectrometry and its application to proteome profiling. We will specifically discuss recent data linking various protein post-translational modifications to cardiovascular disease and conclude with a discussion for enablement and democratization of proteomics across the cardiovascular and scientific community. The aim is to inform and inspire the readership to explore a larger breadth of proteoform, particularity post-translational modifications, related to their particular areas of expertise in cardiovascular physiology.
Collapse
Affiliation(s)
- Justyna Fert-Bober
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Christopher I Murray
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Sarah J Parker
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA.
| | - Jennifer E Van Eyk
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
182
|
Drabovich AP, Saraon P, Drabovich M, Karakosta TD, Dimitromanolakis A, Hyndman ME, Jarvi K, Diamandis EP. Multi-omics Biomarker Pipeline Reveals Elevated Levels of Protein-glutamine Gamma-glutamyltransferase 4 in Seminal Plasma of Prostate Cancer Patients. Mol Cell Proteomics 2019; 18:1807-1823. [PMID: 31249104 PMCID: PMC6731075 DOI: 10.1074/mcp.ra119.001612] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Indexed: 11/06/2022] Open
Abstract
Seminal plasma, because of its proximity to prostate, is a promising fluid for biomarker discovery and noninvasive diagnostics. In this study, we investigated if seminal plasma proteins could increase diagnostic specificity of detecting primary prostate cancer and discriminate between high- and low-grade cancers. To select 147 most promising biomarker candidates, we combined proteins identified through five independent experimental or data mining approaches: tissue transcriptomics, seminal plasma proteomics, cell line secretomics, tissue specificity, and androgen regulation. A rigorous biomarker development pipeline based on selected reaction monitoring assays was designed to evaluate the most promising candidates. As a result, we qualified 76, and verified 19 proteins in seminal plasma of 67 negative biopsy and 152 prostate cancer patients. Verification revealed a prostate-specific, secreted and androgen-regulated protein-glutamine gamma-glutamyltransferase 4 (TGM4), which predicted prostate cancer on biopsy and outperformed age and serum Prostate-Specific Antigen (PSA). A machine-learning approach for data analysis provided improved multi-marker combinations for diagnosis and prognosis. In the independent verification set measured by an in-house immunoassay, TGM4 protein was upregulated 3.7-fold (p = 0.006) and revealed AUC = 0.66 for detecting prostate cancer on biopsy for patients with serum PSA ≥4 ng/ml and age ≥50. Very low levels of TGM4 (120 pg/ml) were detected in blood serum. Collectively, our study demonstrated rigorous evaluation of one of the remaining and not well-explored prostate-specific proteins within the medium-abundance proteome of seminal plasma. Performance of TGM4 warrants its further investigation within the distinct genomic subtypes and evaluation for the inclusion into emerging multi-biomarker panels.
Collapse
Affiliation(s)
- Andrei P Drabovich
- ‡Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5T 3L9 Canada; §Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, M5T 3L9 Canada; ¶Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, M5T 3L9 Canada.
| | - Punit Saraon
- ‡Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5T 3L9 Canada
| | | | - Theano D Karakosta
- §Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, M5T 3L9 Canada
| | | | - M Eric Hyndman
- **Department of Surgery, Division of Urology, Southern Alberta Institute of Urology, University of Calgary, Calgary, AB T2V 1P9, Canada
| | - Keith Jarvi
- ‡‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5T 3L9 Canada; §§Department of Surgery, Division of Urology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, M5T 3L9 Canada.
| | - Eleftherios P Diamandis
- ‡Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5T 3L9 Canada; §Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, M5T 3L9 Canada; ¶Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, M5T 3L9 Canada; ‡‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5T 3L9 Canada.
| |
Collapse
|
183
|
Borja T, Karim N, Goecker Z, Salemi M, Phinney B, Naeem M, Rice R, Parker G. Proteomic genotyping of fingermark donors with genetically variant peptides. Forensic Sci Int Genet 2019; 42:21-30. [DOI: 10.1016/j.fsigen.2019.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/09/2019] [Accepted: 05/26/2019] [Indexed: 01/31/2023]
|
184
|
Prasad B, Achour B, Artursson P, Hop CECA, Lai Y, Smith PC, Barber J, Wisniewski JR, Spellman D, Uchida Y, Zientek M, Unadkat JD, Rostami-Hodjegan A. Toward a Consensus on Applying Quantitative Liquid Chromatography-Tandem Mass Spectrometry Proteomics in Translational Pharmacology Research: A White Paper. Clin Pharmacol Ther 2019; 106:525-543. [PMID: 31175671 PMCID: PMC6692196 DOI: 10.1002/cpt.1537] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/22/2019] [Indexed: 12/18/2022]
Abstract
Quantitative translation of information on drug absorption, disposition, receptor engagement, and drug-drug interactions from bench to bedside requires models informed by physiological parameters that link in vitro studies to in vivo outcomes. To predict in vivo outcomes, biochemical data from experimental systems are routinely scaled using protein quantity in these systems and relevant tissues. Although several laboratories have generated useful quantitative proteomic data using state-of-the-art mass spectrometry, no harmonized guidelines exit for sample analysis and data integration to in vivo translation practices. To address this gap, a workshop was held on September 27 and 28, 2018, in Cambridge, MA, with 100 experts attending from academia, the pharmaceutical industry, and regulators. Various aspects of quantitative proteomics and its applications in translational pharmacology were debated. A summary of discussions and best practices identified by this expert panel are presented in this "White Paper" alongside unresolved issues that were outlined for future debates.
Collapse
Affiliation(s)
- Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, WA
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | | | | | - Philip C Smith
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Jacek R Wisniewski
- Biochemical Proteomics Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Daniel Spellman
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., West Point, PA
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | | | | | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
- Certara UK Ltd. (Simcyp Division), 1 Concourse Way, Sheffield, UK
| |
Collapse
|
185
|
Nielson CM, Jacobs JM, Orwoll ES. Proteomic studies of bone and skeletal health outcomes. Bone 2019; 126:18-26. [PMID: 30954730 PMCID: PMC7302501 DOI: 10.1016/j.bone.2019.03.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022]
Abstract
Proteins are an essential part of essentially all biological processes, and there is enormous variation in protein forms and concentrations that is not reflected in DNA or RNA. Recently there have been rapid advances in the ability to measure protein sequence, modification and concentration, particularly with methods based in mass spectrometry. Global measures of proteins in tissues or in the circulation provide a broad assessment of the proteome that can be extremely useful for discovery, and targeted proteomic measures can yield specific and sensitive assessments of specific peptides and proteins. While most proteomic measures are directed at the detection of consensus peptide sequences, mass spectrometry based proteomic methods also allow a detailed examination of the peptide sequence differences that result from genetic variants and that may have important effects on protein function. In evaluating proteomic data, a number of analytical considerations are important, including an understanding of missing data, the challenge of multiple testing and replication, and the use of rapidly evolving methods in systems biology. While proteomics has not yet had a major impact in skeletal research, interesting recent research has used these approaches in the study of bone cell biology and the discovery of biomarkers of skeletal disorders. Proteomics can be expected to have an increasing influence in the study of bone biology and pathophysiology.
Collapse
Affiliation(s)
| | - Jon M Jacobs
- Pacific Northwest National Laboratory, Richland, WA, USA
| | | |
Collapse
|
186
|
Exploring the extracellular matrix in health and disease using proteomics. Essays Biochem 2019; 63:417-432. [DOI: 10.1042/ebc20190001] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Abstract
The extracellular matrix (ECM) is a complex assembly of hundreds of proteins that constitutes the scaffold of multicellular organisms. In addition to providing architectural and mechanical support to the surrounding cells, it conveys biochemical signals that regulate cellular processes including proliferation and survival, fate determination, and cell migration. Defects in ECM protein assembly, decreased ECM protein production or, on the contrary, excessive ECM accumulation, have been linked to many pathologies including cardiovascular and skeletal diseases, cancers, and fibrosis. The ECM thus represents a potential reservoir of prognostic biomarkers and therapeutic targets. However, our understanding of the global protein composition of the ECM and how it changes during pathological processes has remained limited until recently.
In this mini-review, we provide an overview of the latest methodological advances in sample preparation and mass spectrometry-based proteomics that have permitted the profiling of the ECM of now dozens of normal and diseased tissues, including tumors and fibrotic lesions.
Collapse
|
187
|
Chen Y, Guenther JM, Gin JW, Chan LJG, Costello Z, Ogorzalek TL, Tran HM, Blake-Hedges JM, Keasling JD, Adams PD, García Martín H, Hillson NJ, Petzold CJ. Automated “Cells-To-Peptides” Sample Preparation Workflow for High-Throughput, Quantitative Proteomic Assays of Microbes. J Proteome Res 2019; 18:3752-3761. [DOI: 10.1021/acs.jproteome.9b00455] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
| | - Joel M. Guenther
- Sandia National Laboratories (NTESS), Livermore, California 94551, United States
| | | | | | | | | | - Huu M. Tran
- Sandia National Laboratories (NTESS), Livermore, California 94551, United States
| | | | - Jay D. Keasling
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720-1460, United States
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
- Center for Synthetic Biochemistry, Synthetic Biology Institute, Shenzhen Institutes for Advanced Technologies, Shenzhen 518000, China
| | | | | | | | | |
Collapse
|
188
|
Nishimura T, Nakamura H, Végvári Á, Marko-Varga G, Furuya N, Saji H. Current status of clinical proteogenomics in lung cancer. Expert Rev Proteomics 2019; 16:761-772. [PMID: 31402712 DOI: 10.1080/14789450.2019.1654861] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Lung cancer is the leading cause of cancer death worldwide. Proteogenomics, a way to integrate genomics, transcriptomics, and proteomics, have emerged as a way to understand molecular causes in cancer tumorigenesis. This understanding will help identify therapeutic targets that are urgently needed to improve individual patient outcomes. Areas covered: To explore underlying molecular mechanisms of lung cancer subtypes, several efforts have used proteogenomic approaches that integrate next generation sequencing (NGS) and mass spectrometry (MS)-based technologies. Expert opinion: A large-scale, MS-based, proteomic analysis, together with both NGS-based genomic data and clinicopathological information, will facilitate establishing extensive databases for lung cancer subtypes that can be used for further proteogenomic analyzes. Proteogenomic strategies will further be understanding of how major driver mutations affect downstream molecular networks, resulting in lung cancer progression and malignancy, and how therapy-resistant cancers resistant are molecularly structured. These strategies require advanced bioinformatics based on a dynamic theory of network systems, rather than statistics, to accurately identify mutant proteins and their affected key networks.
Collapse
Affiliation(s)
- Toshihide Nishimura
- Department of Translational Medicine Informatics, St. Marianna University School of Medicine , Kawasaki, Kanagawa , Japan
| | - Haruhiko Nakamura
- Department of Translational Medicine Informatics, St. Marianna University School of Medicine , Kawasaki, Kanagawa , Japan.,Department of Chest Surgery, St. Marianna University School of Medicine , Kawasaki, Kanagawa , Japan
| | - Ákos Végvári
- Proteomics Biomedicum, Division of Physiological Chemistry I, Department of Medical Biochemistry & Biophysics (MBB), Karolinska Institutet , Solna , Sweden
| | - György Marko-Varga
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University , Lund , Sweden.,Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö , Malmö , Sweden
| | - Naoki Furuya
- Department of Internal Medicine, Division of Respiratory Medicine, St. Marianna University School of Medicine , Kawasaki , Kanagawa , Japan
| | - Hisashi Saji
- Department of Chest Surgery, St. Marianna University School of Medicine , Kawasaki, Kanagawa , Japan
| |
Collapse
|
189
|
Bradshaw RA, Hondermarck H, Rodriguez H. Cancer Proteomics and the Elusive Diagnostic Biomarkers. Proteomics 2019; 19:e1800445. [PMID: 31172665 DOI: 10.1002/pmic.201800445] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/02/2019] [Indexed: 12/11/2022]
Abstract
Despite progress in genomic and proteomic technology and applications, the validation of cancer biomarkers of use as clinical early detection diagnostics has remained elusive. As described in this brief viewpoint, there are now recognized to be many types of clinical biomarkers and proteomic analyses, particularly when combined with other 'omic analyses, have been effective in many such biomarker identifications. However, in the area of early diagnosis of cancers, the problems associated with the conversion from identification to diagnostic have largely not been overcome. Notably, the Clinical Proteomic Tumor Analysis Consortium (CPTAC) of the National Cancer Institute (NCI), has been particularly successful in refining the analytical steps needed to tackle this challenging issue and has provided positive insight into how to solve many of the underlying problems. The potential for developing clinical diagnostics for early detection of highly lethal cancers and possible new therapeutic strategies through proteomic analyses, as seen through these CPTAC successes, is more promising than ever.
Collapse
Affiliation(s)
- Ralph A Bradshaw
- Department of Physiology and Biophysics, University of California, Irvine, CA, 92697, USA.,Department of Pharmacology, University of California, San Diego, CA, 92093, USA
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
190
|
Huang KL, Wu Y, Primeau T, Wang YT, Gao Y, McMichael JF, Scott AD, Cao S, Wendl MC, Johnson KJ, Ruggles K, Held J, Payne SH, Davies S, Dar A, Kinsinger CR, Mesri M, Rodriguez H, Ellis MJ, Townsend RR, Chen F, Fenyö D, Li S, Liu T, Carr SA, Ding L. Regulated Phosphosignaling Associated with Breast Cancer Subtypes and Druggability. Mol Cell Proteomics 2019; 18:1630-1650. [PMID: 31196969 PMCID: PMC6682998 DOI: 10.1074/mcp.ra118.001243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/02/2019] [Indexed: 12/25/2022] Open
Abstract
Aberrant phospho-signaling is a hallmark of cancer. We investigated kinase-substrate regulation of 33,239 phosphorylation sites (phosphosites) in 77 breast tumors and 24 breast cancer xenografts. Our search discovered 2134 quantitatively correlated kinase-phosphosite pairs, enriching for and extending experimental or binding-motif predictions. Among the 91 kinases with auto-phosphorylation, elevated EGFR, ERBB2, PRKG1, and WNK1 phosphosignaling were enriched in basal, HER2-E, Luminal A, and Luminal B breast cancers, respectively, revealing subtype-specific regulation. CDKs, MAPKs, and ataxia-telangiectasia proteins were dominant, master regulators of substrate-phosphorylation, whose activities are not captured by genomic evidence. We unveiled phospho-signaling and druggable targets from 113 kinase-substrate pairs and cascades downstream of kinases, including AKT1, BRAF and EGFR. We further identified kinase-substrate-pairs associated with clinical or immune signatures and experimentally validated activated phosphosites of ERBB2, EIF4EBP1, and EGFR. Overall, kinase-substrate regulation revealed by the largest unbiased global phosphorylation data to date connects driver events to their signaling effects.
Collapse
Affiliation(s)
- Kuan-Lin Huang
- ‡Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029; §Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029; ¶Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029.
| | - Yige Wu
- **Department of Medicine, Washington University in St. Louis, MO 63108; ‡‡McDonnell Genome Institute, Washington University in St. Louis, MO 63108
| | - Tina Primeau
- ‡‡McDonnell Genome Institute, Washington University in St. Louis, MO 63108
| | - Yi-Ting Wang
- §§§Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352
| | - Yuqian Gao
- §§§Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352
| | | | - Adam D Scott
- **Department of Medicine, Washington University in St. Louis, MO 63108; ‡‡McDonnell Genome Institute, Washington University in St. Louis, MO 63108
| | - Song Cao
- **Department of Medicine, Washington University in St. Louis, MO 63108; ‡‡McDonnell Genome Institute, Washington University in St. Louis, MO 63108
| | - Michael C Wendl
- ‡‡McDonnell Genome Institute, Washington University in St. Louis, MO 63108; §§Department of Genetics, Washington University in St. Louis, MO 63108; ¶¶Department of Mathematics, Washington University in St. Louis, MO 63108.
| | - Kimberly J Johnson
- ‖‖Siteman Cancer Center, Washington University in St. Louis, MO 63108; ‡‡‡Brown School of Social Work, Washington University in St. Louis, MO 63108
| | - Kelly Ruggles
- ¶¶¶Center for Health Informatics and Bioinformatics, New York University School of Medicine, New York, NY 10016
| | - Jason Held
- **Department of Medicine, Washington University in St. Louis, MO 63108; ‖‖Siteman Cancer Center, Washington University in St. Louis, MO 63108
| | - Samuel H Payne
- §§§Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352
| | - Sherri Davies
- **Department of Medicine, Washington University in St. Louis, MO 63108; ‖‖Siteman Cancer Center, Washington University in St. Louis, MO 63108
| | - Arvin Dar
- ‖Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | - Mehdi Mesri
- ‖‖‖National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Henry Rodriguez
- ‖‖‖National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Matthew J Ellis
- ‡‡‡‡Dan L. Duncan Cancer Center & Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - R Reid Townsend
- **Department of Medicine, Washington University in St. Louis, MO 63108; ‖‖Siteman Cancer Center, Washington University in St. Louis, MO 63108
| | - Feng Chen
- **Department of Medicine, Washington University in St. Louis, MO 63108; ‖‖Siteman Cancer Center, Washington University in St. Louis, MO 63108
| | - David Fenyö
- ¶¶Department of Mathematics, Washington University in St. Louis, MO 63108
| | - Shunqiang Li
- **Department of Medicine, Washington University in St. Louis, MO 63108; ‖‖Siteman Cancer Center, Washington University in St. Louis, MO 63108
| | - Tao Liu
- ‡‡McDonnell Genome Institute, Washington University in St. Louis, MO 63108
| | - Steven A Carr
- §§§§The Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Li Ding
- **Department of Medicine, Washington University in St. Louis, MO 63108; §§Department of Genetics, Washington University in St. Louis, MO 63108; ‖‖Siteman Cancer Center, Washington University in St. Louis, MO 63108
| |
Collapse
|
191
|
Saleh S, Staes A, Deborggraeve S, Gevaert K. Targeted Proteomics for Studying Pathogenic Bacteria. Proteomics 2019; 19:e1800435. [DOI: 10.1002/pmic.201800435] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/04/2019] [Indexed: 02/04/2023]
Affiliation(s)
- Sara Saleh
- Department of Biomedical SciencesInstitute of Tropical Medicine B‐2000 Antwerp Belgium
- VIB Center for Medical Biotechnology B‐9000 Ghent Belgium
- Department of Biomolecular MedicineGhent University B‐9000 Ghent Belgium
| | - An Staes
- VIB Center for Medical Biotechnology B‐9000 Ghent Belgium
- Department of Biomolecular MedicineGhent University B‐9000 Ghent Belgium
| | - Stijn Deborggraeve
- Department of Biomedical SciencesInstitute of Tropical Medicine B‐2000 Antwerp Belgium
| | - Kris Gevaert
- VIB Center for Medical Biotechnology B‐9000 Ghent Belgium
- Department of Biomolecular MedicineGhent University B‐9000 Ghent Belgium
| |
Collapse
|
192
|
Croote D, Braslavsky I, Quake SR. Addressing Complex Matrix Interference Improves Multiplex Food Allergen Detection by Targeted LC-MS/MS. Anal Chem 2019; 91:9760-9769. [PMID: 31339301 DOI: 10.1021/acs.analchem.9b01388] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The frequent use of precautionary food allergen labeling (PAL) such as "may contain" frustrates allergic individuals who rely on such labeling to determine whether a food is safe to consume. One technique to study whether foods contain allergens is targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) employing scheduled multiple reaction monitoring (MRM). However, the applicability of a single MRM method to many commercial foods is unknown as complex and heterogeneous interferences derived from the unique composition of each food matrix can hinder quantification of trace amounts of allergen contamination. We developed a freely available, open source software package MAtrix-Dependent Interference Correction (MADIC) to identify interference and applied it with a method targeting 14 allergens. Among 84 unique food products, we found patterns of allergen contamination such as wheat in grains, milk in chocolate-containing products, and soy in breads and corn flours. We also found additional instances of contamination in products with and without PAL as well as highly variable soy content in foods containing only soybean oil and/or soy lecithin. These results demonstrate the feasibility of applying LC-MS/MS to a variety of food products with sensitive detection of multiple allergens in spite of variable matrix interference.
Collapse
Affiliation(s)
- Derek Croote
- Department of Bioengineering , Stanford University , Stanford , California 94305 , United States
| | - Ido Braslavsky
- Department of Bioengineering , Stanford University , Stanford , California 94305 , United States.,Robert H. Smith Faculty of Agriculture, Food, and Environment , The Hebrew University of Jerusalem , Rehovot 7610001 , Israel
| | - Stephen R Quake
- Department of Bioengineering , Stanford University , Stanford , California 94305 , United States.,Department of Applied Physics , Stanford University , Stanford , California 94305 , United States.,Chan Zuckerberg Biohub , San Francisco , California 94158 , United States
| |
Collapse
|
193
|
Schaller TH, Foster MW, Thompson JW, Spasojevic I, Normantaite D, Moseley MA, Sanchez-Perez L, Sampson JH. Pharmacokinetic Analysis of a Novel Human EGFRvIII:CD3 Bispecific Antibody in Plasma and Whole Blood Using a High-Resolution Targeted Mass Spectrometry Approach. J Proteome Res 2019; 18:3032-3041. [PMID: 31267741 DOI: 10.1021/acs.jproteome.9b00145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Bispecific single chain antibody fragments (bi-scFv) represent an emerging class of biotherapeutics. We recently developed a fully human bi-scFv (EGFRvIII:CD3 bi-scFv) with the goal of redirecting CD3-expressing T cells to recognize and destroy malignant, EGFRvIII-expressing glioma. In mice, we showed that EGFRvIII:CD3 bi-scFv effectively treats orthotopic patient-derived malignant glioma and syngeneic glioblastoma. Here, we developed a targeted assay for pharmacokinetic (PK) analysis of EGFRvIII:CD3 bi-scFv, a necessary step in the drug development process. Using microflow liquid chromatography coupled to a high resolution parallel reaction monitoring mass spectrometry, and data analysis in Skyline, we developed a bottom-up proteomic assay for quantification of EGFRvIII:CD3 bi-scFv in both plasma and whole blood. Importantly, a protein calibrator, along with stable isotope-labeled EGFRvIII:CD3 bi-scFv protein, were used for absolute quantification. A PK analysis in a CD3 humanized mouse revealed that EGFRvIII:CD3 bi-scFv in plasma and whole blood has an initial half-life of ∼8 min and a terminal half-life of ∼2.5 h. Our results establish a sensitive, high-throughput assay for direct quantification of EGFRvIII:CD3 bi-scFv without the need for immunoaffinity enrichment. Moreover, these pharmacokinetic parameters will guide drug optimization and dosing regimens in future IND-enabling and phase I studies of EGFRvIII:CD3 bi-scFv.
Collapse
Affiliation(s)
- Teilo H Schaller
- Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , North Carolina , United States.,Department of Neurosurgery , Duke University Medical Center , Durham , North Carolina , United States.,Department of Pathology , Duke University Medical Center , Durham , North Carolina , United States
| | - Matthew W Foster
- Duke Proteomics and Metabolomics Shared Resource, Duke Center for Genomic and Computational Biology , Duke University , Durham , North Carolina , United States
| | - J Will Thompson
- Duke Proteomics and Metabolomics Shared Resource, Duke Center for Genomic and Computational Biology , Duke University , Durham , North Carolina , United States
| | - Ivan Spasojevic
- Duke Cancer Institute PK/PD Core Laboratory , Durham , North Carolina , United States.,Department of Medicine , Duke University School of Medicine , Durham , North Carolina , United States
| | - Deimante Normantaite
- Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , North Carolina , United States
| | - M Arthur Moseley
- Duke Proteomics and Metabolomics Shared Resource, Duke Center for Genomic and Computational Biology , Duke University , Durham , North Carolina , United States
| | - Luis Sanchez-Perez
- Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , North Carolina , United States.,Department of Neurosurgery , Duke University Medical Center , Durham , North Carolina , United States
| | - John H Sampson
- Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , North Carolina , United States.,Department of Neurosurgery , Duke University Medical Center , Durham , North Carolina , United States.,Department of Pathology , Duke University Medical Center , Durham , North Carolina , United States
| |
Collapse
|
194
|
Erozenci LA, Böttger F, Bijnsdorp IV, Jimenez CR. Urinary exosomal proteins as (pan‐)cancer biomarkers: insights from the proteome. FEBS Lett 2019; 593:1580-1597. [DOI: 10.1002/1873-3468.13487] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/31/2019] [Accepted: 06/05/2019] [Indexed: 01/17/2023]
Affiliation(s)
- Leyla Ayse Erozenci
- Department of Medical Oncology Cancer Center Amsterdam Amsterdam UMC Vrije Universiteit Amsterdam The Netherlands
- OncoProteomics Laboratory Cancer Center Amsterdam Amsterdam UMC Vrije Universiteit Amsterdam The Netherlands
| | - Franziska Böttger
- Department of Medical Oncology Cancer Center Amsterdam Amsterdam UMC Vrije Universiteit Amsterdam The Netherlands
- OncoProteomics Laboratory Cancer Center Amsterdam Amsterdam UMC Vrije Universiteit Amsterdam The Netherlands
| | - Irene V. Bijnsdorp
- OncoProteomics Laboratory Cancer Center Amsterdam Amsterdam UMC Vrije Universiteit Amsterdam The Netherlands
- Department of Urology Amsterdam UMC Vrije Universiteit Amsterdam The Netherlands
| | - Connie R. Jimenez
- Department of Medical Oncology Cancer Center Amsterdam Amsterdam UMC Vrije Universiteit Amsterdam The Netherlands
- OncoProteomics Laboratory Cancer Center Amsterdam Amsterdam UMC Vrije Universiteit Amsterdam The Netherlands
| |
Collapse
|
195
|
Zhang G, Li J, Zhang J, Liang X, Zhang X, Wang T, Yin S. Integrated Analysis of Transcriptomic, miRNA and Proteomic Changes of a Novel Hybrid Yellow Catfish Uncovers Key Roles for miRNAs in Heterosis. Mol Cell Proteomics 2019; 18:1437-1453. [PMID: 31092672 PMCID: PMC6601203 DOI: 10.1074/mcp.ra118.001297] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 04/09/2019] [Indexed: 01/14/2023] Open
Abstract
Heterosis is a complex biological phenomenon in which hybridization produces offspring that exhibit superior phenotypic characteristics compared with the parents. Heterosis is widely utilized in agriculture, for example in fish farming; however, its underlying molecular basis remains elusive. To gain a comprehensive and unbiased molecular understanding of fish heterosis, we analyzed the mRNA, miRNA, and proteomes of the livers of three catfish species, Pelteobagrus fulvidraco, P. vachelli, and their hybrid, the hybrid yellow catfish "Huangyou-1" (P. fulvidraco ♀ × P. vachelli ♂). Using next-generation sequencing and mass spectrometry, we show that the nonadditive, homoeolog expression bias and expression level dominance pattern were readily identified at the transcriptional, post-transcriptional, or protein levels, providing the evidence for the widespread presence of dominant models during hybridization. A number of predicted miRNA-mRNA-protein pairs were found and validated by qRT-PCR and PRM assays. Furthermore, several diverse key pathways were identified, including immune defense, metabolism, digestion and absorption, and cell proliferation and development, suggesting the vital mechanisms involved in the generation of the heterosis phenotype in progenies. We propose that the high parental expression of genes/proteins (growth, nutrition, feeding, and disease resistance) coupled with low parental miRNAs of the offspring, are inherited from the mother or father, thus indicating that the offspring were enriched with the advantages of the father or mother. We provide new and important information about the molecular mechanisms of heterosis, which represents a significant step toward a more complete elucidation of this phenomenon.
Collapse
Affiliation(s)
- Guosong Zhang
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; §Key Laboratory for Physiology Biochemistry and Application, School of Agriculture and Bioengineering, Heze University, Heze, Shandong 274015, China
| | - Jie Li
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China
| | - Jiajia Zhang
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China
| | - Xia Liang
- §Key Laboratory for Physiology Biochemistry and Application, School of Agriculture and Bioengineering, Heze University, Heze, Shandong 274015, China
| | - Xinyu Zhang
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China
| | - Tao Wang
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China
| | - Shaowu Yin
- From the ‡College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China;; ¶Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China.
| |
Collapse
|
196
|
Method Validation by CPTAC Guidelines for Multi-protein Marker Assays Using Multiple Reaction Monitoring-mass Spectrometry. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-018-0454-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
197
|
Kontostathi G, Makridakis M, Zoidakis J, Vlahou A. Applications of multiple reaction monitoring targeted proteomics assays in human plasma. Expert Rev Mol Diagn 2019; 19:499-515. [PMID: 31057016 DOI: 10.1080/14737159.2019.1615448] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Multiple (or selected) reaction monitoring-mass spectrometry (MRM/SRM) is a targeted proteomic method that can be used for relative and absolute quantification. Multiple reports exist supporting the potential of the approach in proteomic biomarker validation. Areas covered: To get an overview of the applications of MRM in protein quantification in plasma, a search in MedLine/PubMed was performed using the keywords: 'MRM/SRM plasma proteomic/proteomics/proteome'. The retrieved studies were further filtered to focus on disease biomarkers and the main results are summarized. Expert opinion: MRM is increasingly employed for the quantification of both well-established but also newly discovered putative biomarkers and occasionally their post-translationally modified forms in plasma. Fractionation is regularly required for the detection of low abundance proteins. Standardized procedures to facilitate assay establishment and marker quantification have been proposed and, in few cases, implemented. Nevertheless, in most cases, absolute quantification is not performed. To advance, multiple technical issues including the regular use of standard labeled peptides and appropriate quality controls to monitor assay performance should be considered. Additionally, clinical aspects involving careful study design to address biomarker clinical use should also be considered.
Collapse
Affiliation(s)
- Georgia Kontostathi
- a Biotechnology Division , Biomedical Research Foundation, Academy of Athens (BRFAA) , Athens , Greece
| | - Manousos Makridakis
- a Biotechnology Division , Biomedical Research Foundation, Academy of Athens (BRFAA) , Athens , Greece
| | - Jerome Zoidakis
- a Biotechnology Division , Biomedical Research Foundation, Academy of Athens (BRFAA) , Athens , Greece
| | - Antonia Vlahou
- a Biotechnology Division , Biomedical Research Foundation, Academy of Athens (BRFAA) , Athens , Greece
| |
Collapse
|
198
|
Validation of a multiplexed LC-MS/MS clinical assay to quantify insulin-like growth factor-binding proteins in human serum and its application in a clinical study. Toxicol Appl Pharmacol 2019; 371:74-83. [PMID: 30926377 DOI: 10.1016/j.taap.2019.03.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/14/2019] [Accepted: 03/25/2019] [Indexed: 11/18/2022]
Abstract
Circulating insulin-like growth factor-binding proteins (IGFBPs) continue to gain attention as biomarkers of drug activities on insulin like growth factor (IGF)/IGF receptor signaling pathways. A multiplexed LC-MS/MS method was validated for the absolute quantitation of IGFBPs in human serum. The method was used to measure screening concentrations of IGFBPs in spinal and bulbar muscular atrophy (SBMA) patients in a phase 2 clinical trial. Concentrations of IGFBP 1, 2, 3, and 5 were simultaneously determined based on representative signature peptides derived from an optimized trypsin digestion procedure. Signature peptide levels were absolutely quantitated using a sensitive/specific targeted LC-MS/MS method. Corresponding mass-shifted, stable isotope-labeled peptides were employed as internal standards. A true blank matrix for the quantitation of IGFBPs was not available since they are endogenous proteins in human serum. In this method, calibration standards/curves were prepared using authentic synthetic peptides spiked into a surrogate matrix. The surrogate matrix was generated from human serum treated in the same way as the study samples, but using iodoacetic acid instead of iodoacetamide as the alkylation reagent. This surrogate matrix approach allowed for the direct and sensitive/specific quantification of IGFBP 1, 2, 3, and 5 due to the lack of any endogenous background. Equivalent matrix effect and recovery of analytes was achieved for the authentic and surrogate matrices. The fully validated LC-MS/MS assay will allow further evaluation of the utility of IGFBP biomarkers in clinical trials.
Collapse
|
199
|
Zhang B, Whiteaker JR, Hoofnagle AN, Baird GS, Rodland KD, Paulovich AG. Clinical potential of mass spectrometry-based proteogenomics. Nat Rev Clin Oncol 2019; 16:256-268. [PMID: 30487530 PMCID: PMC6448780 DOI: 10.1038/s41571-018-0135-7] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer genomics research aims to advance personalized oncology by finding and targeting specific genetic alterations associated with cancers. In genome-driven oncology, treatments are selected for individual patients on the basis of the findings of tumour genome sequencing. This personalized approach has prolonged the survival of subsets of patients with cancer. However, many patients do not respond to the predicted therapies based on the genomic profiles of their tumours. Furthermore, studies pairing genomic and proteomic analyses of samples from the same tumours have shown that the proteome contains novel information that cannot be discerned through genomic analysis alone. This observation has led to the concept of proteogenomics, in which both types of data are leveraged for a more complete view of tumour biology that might enable patients to be more successfully matched to effective treatments than they would using genomics alone. In this Perspective, we discuss the added value of proteogenomics over the current genome-driven approach to the clinical characterization of cancers and summarize current efforts to incorporate targeted proteomic measurements based on selected/multiple reaction monitoring (SRM/MRM) mass spectrometry into the clinical laboratory to facilitate clinical proteogenomics.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Molecular and Human Genetics, Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey R Whiteaker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Andrew N Hoofnagle
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Geoffrey S Baird
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Karin D Rodland
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
- Department of Cell, Development and Cancer Biology, Oregon Health & Sciences University, Portland, OR, USA
| | - Amanda G Paulovich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Division of Medical Oncology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
200
|
Mnatsakanyan R, Shema G, Basik M, Batist G, Borchers CH, Sickmann A, Zahedi RP. Detecting post-translational modification signatures as potential biomarkers in clinical mass spectrometry. Expert Rev Proteomics 2019; 15:515-535. [PMID: 29893147 DOI: 10.1080/14789450.2018.1483340] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Numerous diseases are caused by changes in post-translational modifications (PTMs). Therefore, the number of clinical proteomics studies that include the analysis of PTMs is increasing. Combining complementary information-for example changes in protein abundance, PTM levels, with the genome and transcriptome (proteogenomics)-holds great promise for discovering important drivers and markers of disease, as variations in copy number, expression levels, or mutations without spatial/functional/isoform information is often insufficient or even misleading. Areas covered: We discuss general considerations, requirements, pitfalls, and future perspectives in applying PTM-centric proteomics to clinical samples. This includes samples obtained from a human subject, for instance (i) bodily fluids such as plasma, urine, or cerebrospinal fluid, (ii) primary cells such as reproductive cells, blood cells, and (iii) tissue samples/biopsies. Expert commentary: PTM-centric discovery proteomics can substantially contribute to the understanding of disease mechanisms by identifying signatures with potential diagnostic or even therapeutic relevance but may require coordinated efforts of interdisciplinary and eventually multi-national consortia, such as initiated in the cancer moonshot program. Additionally, robust and standardized mass spectrometry (MS) assays-particularly targeted MS, MALDI imaging, and immuno-MALDI-may be transferred to the clinic to improve patient stratification for precision medicine, and guide therapies.
Collapse
Affiliation(s)
- Ruzanna Mnatsakanyan
- a Protein Dynamics , Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V , Dortmund , 44227 , Germany
| | - Gerta Shema
- a Protein Dynamics , Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V , Dortmund , 44227 , Germany
| | - Mark Basik
- b Gerald Bronfman Department of Oncology , Jewish General Hospital, McGill University , Montreal , Quebec H4A 3T2 , Canada
| | - Gerald Batist
- b Gerald Bronfman Department of Oncology , Jewish General Hospital, McGill University , Montreal , Quebec H4A 3T2 , Canada
| | - Christoph H Borchers
- b Gerald Bronfman Department of Oncology , Jewish General Hospital, McGill University , Montreal , Quebec H4A 3T2 , Canada.,c University of Victoria-Genome British Columbia Proteomics Centre, University of Victoria , Victoria , British Columbia V8Z 7X8 , Canada.,d Department of Biochemistry and Microbiology , University of Victoria , Victoria , British Columbia , V8P 5C2 , Canada.,e Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University , Montreal , Quebec H3T 1E2 , Canada
| | - Albert Sickmann
- a Protein Dynamics , Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V , Dortmund , 44227 , Germany.,f Medizinische Fakultät, Medizinische Proteom-Center (MPC), Ruhr-Universität Bochum , 44801 Bochum , Germany.,g Department of Chemistry , College of Physical Sciences, University of Aberdeen , Aberdeen AB24 3FX , Scotland , United Kingdom
| | - René P Zahedi
- a Protein Dynamics , Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V , Dortmund , 44227 , Germany.,b Gerald Bronfman Department of Oncology , Jewish General Hospital, McGill University , Montreal , Quebec H4A 3T2 , Canada.,e Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University , Montreal , Quebec H3T 1E2 , Canada
| |
Collapse
|