1
|
La Salvia A, Lens-Pardo A, López-López A, Carretero-Puche C, Capdevila J, Benavent M, Jiménez-Fonseca P, Castellano D, Alonso T, Teule A, Custodio A, Tafuto S, La Casta A, Spada F, Lopez-Gonzalvez A, Gil-Calderon B, Espinosa-Olarte P, Barbas C, Garcia-Carbonero R, Soldevilla B. Metabolomic profile of neuroendocrine tumors identifies methionine, porphyrin, and tryptophan metabolisms as key dysregulated pathways associated with patient survival. Eur J Endocrinol 2024; 190:62-74. [PMID: 38033321 DOI: 10.1093/ejendo/lvad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023]
Abstract
OBJECTIVE Metabolic profiling is a valuable tool to characterize tumor biology but remains largely unexplored in neuroendocrine tumors (NETs). Our aim was to comprehensively assess the metabolomic profile of NETs and identify novel prognostic biomarkers and dysregulated molecular pathways. DESIGN AND METHODS Multiplatform untargeted metabolomic profiling (GC-MS, CE-MS, and LC-MS) was performed in plasma from 77 patients with G1-2 extra-pancreatic NETs enrolled in the AXINET trial (NCT01744249) (study cohort) and from 68 non-cancer individuals (control). The prognostic value of each differential metabolite (n = 155) in NET patients (P < .05) was analyzed by univariate and multivariate analyses adjusted for multiple testing and other confounding factors. Related pathways were explored by Metabolite Set Enrichment Analysis (MSEA) and Metabolite Pathway Analysis (MPA). RESULTS Thirty-four metabolites were significantly associated with progression-free survival (PFS) (n = 16) and/or overall survival (OS) (n = 27). Thirteen metabolites remained significant independent prognostic factors in multivariate analysis, 3 of them with a significant impact on both PFS and OS. Unsupervised clustering of these 3 metabolites stratified patients in 3 distinct prognostic groups (1-year PFS of 71.1%, 47.7%, and 15.4% (P = .012); 5-year OS of 69.7%, 32.5%, and 27.7% (P = .003), respectively). The MSEA and MPA of the 13-metablolite signature identified methionine, porphyrin, and tryptophan metabolisms as the 3 most relevant dysregulated pathways associated with the prognosis of NETs. CONCLUSIONS We identified a metabolomic signature that improves prognostic stratification of NET patients beyond classical prognostic factors for clinical decisions. The enriched metabolic pathways identified reveal novel tumor vulnerabilities that may foster the development of new therapeutic strategies for these patients.
Collapse
Affiliation(s)
- Anna La Salvia
- Center of Experimental Oncology, Gastrointestinal and Neuroendrocrine Tumors Research Group, Research Institute Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Oncology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- National Center for Drug Research and Evaluation, National Institute of Health (ISS), 00161 Rome, Italy
| | - Alberto Lens-Pardo
- Center of Experimental Oncology, Gastrointestinal and Neuroendrocrine Tumors Research Group, Research Institute Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Angel López-López
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Centre for Metabolomics and Bioanalysis (CEMBIO), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28925 Madrid, Spain
| | - Carlos Carretero-Puche
- Center of Experimental Oncology, Gastrointestinal and Neuroendrocrine Tumors Research Group, Research Institute Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Jaume Capdevila
- Vall Hebron University Hospital and Vall Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Marta Benavent
- Medical Oncology Department, Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), 41013 Seville, Spain
| | - Paula Jiménez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, ISPA, 33011 Oviedo, Spain
| | - Daniel Castellano
- Oncology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Teresa Alonso
- Medical Oncology, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Alexandre Teule
- Institut Català d'Oncologia (ICO)-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908 L'Hospitalet del Llobregat, Barcelona, Spain
| | - Ana Custodio
- Department of Medical Oncology, Hospital Universitario La Paz, CIBERONC CB16/12/00398, 28046 Madrid, Spain
| | - Salvatore Tafuto
- Sarcomas and Rare Tumours Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Adelaida La Casta
- Department of Medical Oncology, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), 20014 San Sebastián, Spain
| | - Francesca Spada
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Angeles Lopez-Gonzalvez
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Centre for Metabolomics and Bioanalysis (CEMBIO), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28925 Madrid, Spain
| | - Beatriz Gil-Calderon
- Center of Experimental Oncology, Gastrointestinal and Neuroendrocrine Tumors Research Group, Research Institute Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Paula Espinosa-Olarte
- Center of Experimental Oncology, Gastrointestinal and Neuroendrocrine Tumors Research Group, Research Institute Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Oncology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Coral Barbas
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Centre for Metabolomics and Bioanalysis (CEMBIO), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28925 Madrid, Spain
| | - Rocio Garcia-Carbonero
- Center of Experimental Oncology, Gastrointestinal and Neuroendrocrine Tumors Research Group, Research Institute Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Oncology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
- Medicine Department, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Beatriz Soldevilla
- Center of Experimental Oncology, Gastrointestinal and Neuroendrocrine Tumors Research Group, Research Institute Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
- Genetics, Physiology and Microbiology Department, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| |
Collapse
|
2
|
Chen C, Ye L, Yi J, Liu T, Li Z. FN1 mediated activation of aspartate metabolism promotes the progression of triple-negative and luminal a breast cancer. Breast Cancer Res Treat 2023; 201:515-533. [PMID: 37458908 DOI: 10.1007/s10549-023-07032-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/28/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Breast cancer (BC) is regarded as one of the most common cancers diagnosed among the female population and has an extremely high mortality rate. It is known that Fibronectin 1 (FN1) drives the occurrence and development of a variety of cancers through metabolic reprogramming. Aspartic acid is considered to be an important substrate for nucleotide synthesis. However, the regulatory mechanism between FN1 and aspartate metabolism is currently unclear. METHODS We used RNA sequencing (RNA seq) and liquid chromatography-mass spectrometry to analyze the tumor tissues and paracancerous tissues of patients. MCF7 and MDA-MB-231 cells were used to explore the effects of FN1-regulated aspartic acid metabolism on cell survival, invasion, migration and tumor growth. We used PCR, Western blot, immunocytochemistry and immunofluorescence techniques to study it. RESULTS We found that FN1 was highly expressed in tumor tissues, especially in Lumina A and TNBC subtypes, and was associated with poor prognosis. In vivo and in vitro experiments showed that silencing FN1 inhibits the activation of the YAP1/Hippo pathway by enhancing YAP1 phosphorylation, down-regulates SLC1A3-mediated aspartate uptake and utilization by tumor cells, inhibits BC cell proliferation, invasion and migration, and promotes apoptosis. In addition, inhibition of FN1 combined with the YAP1 inhibitor or SLC1A3 inhibitor can effectively inhibit tumor growth, of which inhibition of FN1 combined with the YAP1 inhibitor is more effective. CONCLUSION Targeting the "FN1/YAP1/SLC1A3/Aspartate metabolism" regulatory axis provides a new target for BC diagnosis and treatment. This study also revealed that intratumoral metabolic heterogeneity plays an important role in the progression of different subtypes of breast cancer.
Collapse
Affiliation(s)
- Chen Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Leiguang Ye
- Department of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jinfeng Yi
- Department of Pathology, Harbin Medical University, Harbin, 150081, China
| | - Tang Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Zhigao Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
3
|
Johnson RK, Brunetti T, Quinn K, Doenges K, Campbell M, Arehart C, Taub MA, Mathias RA, Reisdorph N, Barnes KC, Daya M. Discovering metabolite quantitative trait loci in asthma using an isolated population. J Allergy Clin Immunol 2022; 149:1807-1811.e16. [PMID: 34780848 PMCID: PMC9081120 DOI: 10.1016/j.jaci.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Integration of metabolomics with genetics may advance understanding of disease pathogenesis but has been underused in asthma genetic studies. OBJECTIVE We sought to discover new genetic effects in asthma and to characterize the molecular consequences of asthma genetic risk through integration with the metabolome in a homogeneous population. METHODS From fasting serum samples collected on 348 Tangier Island residents, we quantified 2612 compounds using untargeted metabolomics. Genotyping was performed using Illumina's MEGA array imputed to the TOPMed reference panel. To prioritize metabolites for genome-wide association analysis, we performed a metabolome-wide association study with asthma, selecting asthma-associated metabolites with heritability q value less than 0.01 for genome-wide association analysis. We also tested the association between all metabolites and 8451 candidate asthma single nucleotide polymorphisms previously associated with asthma in the UK Biobank. We followed up significant associations by characterizing shared genetic signal for metabolites and asthma using colocalization analysis. For detailed Methods, please see this article's Online Repository at www.jacionline.org. RESULTS A total of 60 metabolites were associated with asthma (P < .01), including 40 heritable metabolites tested in genome-wide association analysis. We observed a strong association peak for the endocannabinoid linoleoyl ethanolamide on chromosome 6 in VNN1 (P < 2.7 × 10-9). We found strong evidence (colocalization posterior probability >75%) for a shared causal variant between 3 metabolites and asthma, including the polyamine acisoga and variants in LPP, and derivative leukotriene B4 and intergenic variants in chr10p14. CONCLUSIONS We identified novel metabolite quantitative trait loci with asthma associations. Identification and characterization of these genetically driven metabolites may provide insight into the functional consequences of genetic risk factors for asthma.
Collapse
Affiliation(s)
- Randi K Johnson
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo.
| | - Tonya Brunetti
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Kevin Quinn
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Katrina Doenges
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Monica Campbell
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Christopher Arehart
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Margaret A Taub
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md
| | - Rasika A Mathias
- Division of Allergy & Clinical Immunology, Department of Medicine, Johns Hopkins University, Baltimore, Md
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Kathleen C Barnes
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Michelle Daya
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| |
Collapse
|
4
|
McCord JP, Groff LC, Sobus JR. Quantitative non-targeted analysis: Bridging the gap between contaminant discovery and risk characterization. ENVIRONMENT INTERNATIONAL 2022; 158:107011. [PMID: 35386928 PMCID: PMC8979303 DOI: 10.1016/j.envint.2021.107011] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Chemical risk assessments follow a long-standing paradigm that integrates hazard, dose-response, and exposure information to facilitate quantitative risk characterization. Targeted analytical measurement data directly support risk assessment activities, as well as downstream risk management and compliance monitoring efforts. Yet, targeted methods have struggled to keep pace with the demands for data regarding the vast, and growing, number of known chemicals. Many contemporary monitoring studies therefore utilize non-targeted analysis (NTA) methods to screen for known chemicals with limited risk information. Qualitative NTA data has enabled identification of previously unknown compounds and characterization of data-poor compounds in support of hazard identification and exposure assessment efforts. In spite of this, NTA data have seen limited use in risk-based decision making due to uncertainties surrounding their quantitative interpretation. Significant efforts have been made in recent years to bridge this quantitative gap. Based on these advancements, quantitative NTA data, when coupled with other high-throughput data streams and predictive models, are poised to directly support 21st-century risk-based decisions. This article highlights components of the chemical risk assessment process that are influenced by NTA data, surveys the existing literature for approaches to derive quantitative estimates of chemicals from NTA measurements, and presents a conceptual framework for incorporating NTA data into contemporary risk assessment frameworks.
Collapse
Affiliation(s)
- James P. McCord
- Center for Environmental Measurement and Modeling, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Research Triangle Park, NC 27711, USA
- Corresponding author. (J.P. McCord)
| | - Louis C. Groff
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Research Triangle Park, NC 27711, USA
- Oak Ridge Institute for Science and Education (ORISE) Participant, 109 T.W. Alexander Drive, Research Triangle Park, NC 27711, USA
| | - Jon R. Sobus
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Research Triangle Park, NC 27711, USA
| |
Collapse
|
5
|
Place BJ, Ulrich EM, Challis JK, Chao A, Du B, Favela K, Feng YL, Fisher CM, Gardinali P, Hood A, Knolhoff AM, McEachran AD, Nason SL, Newton SR, Ng B, Nuñez J, Peter KT, Phillips AL, Quinete N, Renslow R, Sobus JR, Sussman EM, Warth B, Wickramasekara S, Williams AJ. An Introduction to the Benchmarking and Publications for Non-Targeted Analysis Working Group. Anal Chem 2021; 93:16289-16296. [PMID: 34842413 PMCID: PMC8848292 DOI: 10.1021/acs.analchem.1c02660] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Non-targeted analysis (NTA) encompasses a rapidly evolving set of mass spectrometry techniques aimed at characterizing the chemical composition of complex samples, identifying unknown compounds, and/or classifying samples, without prior knowledge regarding the chemical content of the samples. Recent advances in NTA are the result of improved and more accessible instrumentation for data generation and analysis tools for data evaluation and interpretation. As researchers continue to develop NTA approaches in various scientific fields, there is a growing need to identify, disseminate, and adopt community-wide method reporting guidelines. In 2018, NTA researchers formed the Benchmarking and Publications for Non-Targeted Analysis Working Group (BP4NTA) to address this need. Consisting of participants from around the world and representing fields ranging from environmental science and food chemistry to 'omics and toxicology, BP4NTA provides resources addressing a variety of challenges associated with NTA. Thus far, BP4NTA group members have aimed to establish a consensus on NTA-related terms and concepts and to create consistency in reporting practices by providing resources on a public Web site, including consensus definitions, reference content, and lists of available tools. Moving forward, BP4NTA will provide a setting for NTA researchers to continue discussing emerging challenges and contribute to additional harmonization efforts.
Collapse
Affiliation(s)
- Benjamin J. Place
- National Institute of Standards and Technology, Gaithersburg, MD, USA 20899,Corresponding author,
| | - Elin M. Ulrich
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Research Triangle Park, NC, USA 27711
| | | | - Alex Chao
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Research Triangle Park, NC, USA 27711
| | - Bowen Du
- Southern California Coastal Water Research Project Authority, Costa Mesa, CA, USA 92626
| | - Kristin Favela
- Southwest Research Institute, San Antonio, TX, USA 78238
| | - Yong-Lai Feng
- Exposure and Biomonitoring Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada, K1A 0K9
| | - Christine M. Fisher
- U.S. Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, MD, USA 20740
| | - Piero Gardinali
- Institute of Environment & Department of Chemistry and Biochemistry, Florida International University, North Miami, FL 33181
| | - Alan Hood
- U.S. Food and Drug Administration, Center for Devices and Radiological Health, Silver Spring, MD, USA 20993
| | - Ann M. Knolhoff
- U.S. Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, MD, USA 20740
| | | | - Sara L. Nason
- Connecticut Agricultural Experiment Station, New Haven, CT, USA 06511
| | - Seth R. Newton
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Research Triangle Park, NC, USA 27711
| | - Brian Ng
- Institute of Environment & Department of Chemistry and Biochemistry, Florida International University, North Miami, FL 33181
| | - Jamie Nuñez
- Pacific Northwest National Laboratory, Richland, WA, USA 99352
| | - Katherine T. Peter
- National Institute of Standards and Technology, Charleston, SC, USA 29412
| | - Allison L. Phillips
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA 27711
| | - Natalia Quinete
- Institute of Environment & Department of Chemistry and Biochemistry, Florida International University, North Miami, FL 33181
| | - Ryan Renslow
- Pacific Northwest National Laboratory, Richland, WA, USA 99352
| | - Jon R. Sobus
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Research Triangle Park, NC, USA 27711
| | - Eric M. Sussman
- U.S. Food and Drug Administration, Center for Devices and Radiological Health, Silver Spring, MD, USA 20993
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Samanthi Wickramasekara
- U.S. Food and Drug Administration, Center for Devices and Radiological Health, Silver Spring, MD, USA 20993
| | - Antony J. Williams
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Research Triangle Park, NC, USA 27711
| |
Collapse
|
6
|
Anderson BG, Raskind A, Habra H, Kennedy RT, Evans CR. Modifying Chromatography Conditions for Improved Unknown Feature Identification in Untargeted Metabolomics. Anal Chem 2021; 93:15840-15849. [PMID: 34794310 PMCID: PMC10634695 DOI: 10.1021/acs.analchem.1c02149] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Untargeted metabolomics is an essential component of systems biology research, but it is plagued by a high proportion of detectable features not identified with a chemical structure. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) experiments produce spectra that can be searched against databases to help identify or classify these unknowns, but many features do not generate spectra of sufficient quality to enable successful annotation. Here, we explore alterations to gradient length, mass loading, and rolling precursor ion exclusion parameters for reversed phase liquid chromatography (RPLC) and hydrophilic interaction liquid chromatography (HILIC) that improve compound identification performance for human plasma samples. A manual review of spectral matches from the HILIC data set was used to determine reasonable thresholds for search score and other metrics to enable semi-automated MS/MS data analysis. Compared to typical LC-MS/MS conditions, methods adapted for compound identification increased the total number of unique metabolites that could be matched to a spectral database from 214 to 2052. Following data alignment, 68.0% of newly identified features from the modified conditions could be detected and quantitated using a routine 20-min LC-MS run. Finally, a localized machine learning model was developed to classify the remaining unknowns and select a subset that shared spectral characteristics with successfully identified features. A total of 576 and 749 unidentified features in the HILIC and RPLC data sets were classified by the model as high-priority unknowns or higher-importance targets for follow-up analysis. Overall, our study presents a simple strategy to more deeply annotate untargeted metabolomics data for a modest additional investment of time and sample.
Collapse
Affiliation(s)
- Brady G. Anderson
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109
- Biomedical Research Core Facilities Metabolomics Core, University of Michigan, Ann Arbor MI 48109
| | - Alexander Raskind
- Biomedical Research Core Facilities Metabolomics Core, University of Michigan, Ann Arbor MI 48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Hani Habra
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Robert T. Kennedy
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109
- Biomedical Research Core Facilities Metabolomics Core, University of Michigan, Ann Arbor MI 48109
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109
| | - Charles R. Evans
- Biomedical Research Core Facilities Metabolomics Core, University of Michigan, Ann Arbor MI 48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
7
|
Pekov SI, Sorokin AA, Kuzin AA, Bocharov KV, Bormotov DS, Shivalin AS, Shurkhay VA, Potapov AA, Nikolaev EN, Popov IA. Analysis of Phosphatidylcholines Alterations in Human Glioblastomas Ex Vivo. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2021. [DOI: 10.1134/s1990750821030070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
8
|
Im S, Lee J, Kim S. Preliminary Comparison of Subcortical Structures in Elderly Subclinical Depression: Structural Analysis with 3T MRI. Exp Neurobiol 2021; 30:183-202. [PMID: 33972469 PMCID: PMC8118753 DOI: 10.5607/en20056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/19/2021] [Accepted: 02/17/2021] [Indexed: 01/23/2023] Open
Abstract
Depression in the elderly population has shown increased likelihood of neurological disorders due to structural changes in the subcortical area. However, further investigation into depression related subcortical changes is needed due to mismatches in structural analysis results between studies as well as scarcities in research regarding subcortical connectivity patterns of subclinical depression populations. This study aims to investigate structural differences in subcortical regions of aged participants with subclinical depression using 3Tesla MRI. In structural analysis, volumes of each subcortical region were measured to observe the volumetric difference and asymmetry between groups, but no significant difference was found. In addition, fractional anisotropy (FA) and apparent diffusion coefficient (ADC) did not show any significant differences between groups. Structural analysis using probabilistic tractography indicated that the connection strength between left nucleus accumbens-right hippocampus, and right thalamus-right caudate was higher in the control group than the subclinical depression group. The differences in subcortical connection strength of subclinical depression groups, have shown to correlate with emotional and cognitive disorders, such as anxiety and memory impairment. We believe that the analysis of structural differences and cross-regional network measures in subcortical structures can help identify neurophysiological changes occurring in subclinical depression.
Collapse
Affiliation(s)
- SangJin Im
- Lee Gil Ya Cancer & Diabetes Institute, Gachon University, Incheon 21999, Korea
| | - Jeonghwan Lee
- Department of Psychiatry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Siekyeong Kim
- Department of Psychiatry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| |
Collapse
|
9
|
Quality Assessment of Untargeted Analytical Data in a Large-Scale Metabolomic Study. J Clin Med 2021; 10:jcm10091826. [PMID: 33922230 PMCID: PMC8122759 DOI: 10.3390/jcm10091826] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Large-scale metabolomic studies have become common, and the reliability of the peak data produced by the various instruments is an important issue. However, less attention has been paid to the large number of uncharacterized peaks in untargeted metabolomics data. In this study, we tested various criteria to assess the reliability of 276 and 202 uncharacterized peaks that were detected in a gathered set of 30 plasma and urine quality control samples, respectively, using capillary electrophoresis-time-of-flight mass spectrometry (CE-TOFMS). The linear relationship between the amounts of pooled samples and the corresponding peak areas was one of the criteria used to select reliable peaks. We used samples from approximately 3000 participants in the Tsuruoka Metabolome Cohort Study to investigate patterns of the areas of these uncharacterized peaks among the samples and clustered the peaks by combining the patterns and differences in the migration times. Our assessment pipeline removed substantial numbers of unreliable or redundant peaks and detected 35 and 74 reliable uncharacterized peaks in plasma and urine, respectively, some of which may correspond to metabolites involved in important physiological processes such as disease progression. We propose that our assessment pipeline can be used to help establish large-scale untargeted clinical metabolomic studies.
Collapse
|
10
|
Pekov SI, Sorokin AA, Kuzin AA, Bocharov KV, Bormotov DS, Shivalin AS, Shurkhay VA, Potapov AA, Nikolaev EN, Popov IA. [Analysis of phosphatidylcholines alterations in human glioblastoma multiform tissues ex vivo]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:81-87. [PMID: 33645525 DOI: 10.18097/pbmc20216701081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Significant metabolism alteration is accompanying the cell malignization process. Energy metabolism disturbance leads to the activation of de novo synthesis and beta-oxidation processes of lipids and fatty acids in a cancer cell, which becomes an indicator of pathological processes inside the cell. The majority of studies dealing with lipid metabolism alterations in glial tumors are performed using the cell lines in vitro or animal models. However, such conditions do not entirely represent the physiological conditions of cell growth or possible cells natural variability. This work presents the results of the data obtained by applying ambient mass spectrometry to human glioblastoma multiform tissues. By analyzing a relatively large cohort of primary and secondary glioblastoma samples, we identify the alterations in cells lipid composition, which accompanied the development of grade IV brain tumors. We demonstrate that primary glioblastomas, as well as ones developed from astrocytomas, are enriched with mono- and diunsaturated phosphatidylcholines (PC 26:1, 30:2, 32:1, 32:2, 34:1, 34:2). Simultaneously, the saturated and polyunsaturated phosphatidylcholines and phosphatidylethanolamines decrease. These alterations are obviously linked to the availability of the polyunsaturated fatty acids and activation of the de novo lipid synthesis and beta-oxidation pathways under the anaerobic conditions in the tumor core.
Collapse
Affiliation(s)
- S I Pekov
- Skolkovo Institute of Science and Technology, Moscow, Russia; Moscow Institute of Physics and Technology (National Research University), Moscow, Russia
| | - A A Sorokin
- Moscow Institute of Physics and Technology (National Research University), Moscow, Russia
| | - A A Kuzin
- Moscow Institute of Physics and Technology (National Research University), Moscow, Russia
| | - K V Bocharov
- Semenov Federal Center of Chemical Physic of RAS, Moscow, Russia
| | - D S Bormotov
- Moscow Institute of Physics and Technology (National Research University), Moscow, Russia
| | - A S Shivalin
- Moscow Institute of Physics and Technology (National Research University), Moscow, Russia
| | - V A Shurkhay
- Moscow Institute of Physics and Technology (National Research University), Moscow, Russia; N.N. Burdenko National Medical Research Center of Neurosurgery, Moscow, Russia
| | - A A Potapov
- N.N. Burdenko National Medical Research Center of Neurosurgery, Moscow, Russia
| | - E N Nikolaev
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - I A Popov
- Moscow Institute of Physics and Technology (National Research University), Moscow, Russia
| |
Collapse
|
11
|
Sands CJ, Gómez-Romero M, Correia G, Chekmeneva E, Camuzeaux S, Izzi-Engbeaya C, Dhillo WS, Takats Z, Lewis MR. Representing the Metabolome with High Fidelity: Range and Response as Quality Control Factors in LC-MS-Based Global Profiling. Anal Chem 2021; 93:1924-1933. [PMID: 33448796 DOI: 10.1021/acs.analchem.0c03848] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Liquid chromatography-mass spectrometry (LC-MS) is a powerful and widely used technique for measuring the abundance of chemical species in living systems. Its sensitivity, analytical specificity, and direct applicability to biofluids and tissue extracts impart great promise for the discovery and mechanistic characterization of biomarker panels for disease detection, health monitoring, patient stratification, and treatment personalization. Global metabolic profiling applications yield complex data sets consisting of multiple feature measurements for each chemical species observed. While this multiplicity can be useful in deriving enhanced analytical specificity and chemical identities from LC-MS data, data set inflation and quantitative imprecision among related features is problematic for statistical analyses and interpretation. This Perspective provides a critical evaluation of global profiling data fidelity with respect to measurement linearity and the quantitative response variation observed among components of the spectra. These elements of data quality are widely overlooked in untargeted metabolomics yet essential for the generation of data that accurately reflect the metabolome. Advanced feature filtering informed by linear range estimation and analyte response factor assessment is advocated as an attainable means of controlling LC-MS data quality in global profiling studies and exemplified herein at both the feature and data set level.
Collapse
Affiliation(s)
- Caroline J Sands
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom.,Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - María Gómez-Romero
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom.,Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Gonçalo Correia
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom.,Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Elena Chekmeneva
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom.,Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Stephane Camuzeaux
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom.,Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Chioma Izzi-Engbeaya
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0HS, United Kingdom
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0HS, United Kingdom
| | - Zoltan Takats
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom.,Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Matthew R Lewis
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom.,Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| |
Collapse
|
12
|
Luan H, Jiang X, Ji F, Lan Z, Cai Z, Zhang W. CPVA: a web-based metabolomic tool for chromatographic peak visualization and annotation. Bioinformatics 2020; 36:3913-3915. [PMID: 32186699 DOI: 10.1093/bioinformatics/btaa200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/02/2020] [Accepted: 03/17/2020] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Liquid chromatography-mass spectrometry-based non-targeted metabolomics is routinely performed to qualitatively and quantitatively analyze a tremendous amount of metabolite signals in complex biological samples. However, false-positive peaks in the datasets are commonly detected as metabolite signals by using many popular software, resulting in non-reliable measurement. RESULTS To reduce false-positive calling, we developed an interactive web tool, termed CPVA, for visualization and accurate annotation of the detected peaks in non-targeted metabolomics data. We used a chromatogram-centric strategy to unfold the characteristics of chromatographic peaks through visualization of peak morphology metrics, with additional functions to annotate adducts, isotopes and contaminants. CPVA is a free, user-friendly tool to help users to identify peak background noises and contaminants, resulting in decrease of false-positive or redundant peak calling, thereby improving the data quality of non-targeted metabolomics studies. AVAILABILITY AND IMPLEMENTATION The CPVA is freely available at http://cpva.eastus.cloudapp.azure.com. Source code and installation instructions are available on GitHub: https://github.com/13479776/cpva. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hemi Luan
- School of Medicine.,SUSTech Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, China
| | - Xingen Jiang
- Beijing University of Chinese Medicine, Shenzhen Hospital, Shenzhen, China
| | - Fenfen Ji
- Department of Chemistry, State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | | | - Zongwei Cai
- Department of Chemistry, State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | | |
Collapse
|
13
|
Zhao H, Shen J, Moore SC, Ye Y, Wu X, Esteva FJ, Tripathy D, Chow WH. Breast cancer risk in relation to plasma metabolites among Hispanic and African American women. Breast Cancer Res Treat 2019; 176:687-696. [PMID: 30771047 PMCID: PMC6588417 DOI: 10.1007/s10549-019-05165-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/09/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE The metabolic etiology of breast cancer has been explored in the past several years using metabolomics. However, most of these studies only included non-Hispanic White individuals. METHODS To fill this gap, we performed a two-step (discovery and validation) metabolomics profiling in plasma samples from 358 breast cancer patients and 138 healthy controls. All study subjects were either Hispanics or non-Hispanic African Americans. RESULTS A panel of 14 identified metabolites significantly differed between breast cancer cases and healthy controls in both the discovery and validation sets. Most of these identified metabolites were lipids. In the pathway analysis, citrate cycle (TCA cycle), arginine and proline metabolism, and linoleic acid metabolism pathways were observed, and they significantly differed between breast cancer cases and healthy controls in both sets. From those 14 metabolites, we selected 9 non-correlated metabolites to generate a metabolic risk score. Increased metabolites risk score was associated with a 1.87- and 1.63-fold increased risk of breast cancer in the discovery and validation sets, respectively (Odds ratio (OR) 1.87, 95% Confidence interval (CI) 1.50, 2.32; OR 1.63, 95% CI 1.36, 1.95). CONCLUSIONS In summary, our study identified metabolic profiles and pathways that significantly differed between breast cancer cases and healthy controls in Hispanic or non-Hispanic African American women. The results from our study might provide new insights on the metabolic etiology of breast cancer.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Jie Shen
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Steven C Moore
- Divisions of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Yuanqing Ye
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Francisco J Esteva
- Perlmutter Cancer Center at New York University Langone Health, New York, NY, 10016, USA
| | - Debasish Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, racial Houston, TX, 77030, USA
| | - Wong-Ho Chow
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
14
|
Uncovering mechanisms of global ocean change effects on the Dungeness crab (Cancer magister) through metabolomics analysis. Sci Rep 2019; 9:10717. [PMID: 31341175 PMCID: PMC6656712 DOI: 10.1038/s41598-019-46947-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 07/04/2019] [Indexed: 01/22/2023] Open
Abstract
The Dungeness crab is an economically and ecologically important species distributed along the North American Pacific coast. To predict how Dungeness crab may physiologically respond to future global ocean change on a molecular level, we performed untargeted metabolomic approaches on individual Dungeness crab juveniles reared in treatments that mimicked current and projected future pH and dissolved oxygen conditions. We found 94 metabolites and 127 lipids responded in a condition-specific manner, with a greater number of known compounds more strongly responding to low oxygen than low pH exposure. Pathway analysis of these compounds revealed that juveniles may respond to low oxygen through evolutionarily conserved processes including downregulating glutathione biosynthesis and upregulating glycogen storage, and may respond to low pH by increasing ATP production. Most interestingly, we found that the response of juveniles to combined low pH and low oxygen exposure was most similar to the low oxygen exposure response, indicating low oxygen may drive the physiology of juvenile crabs more than pH. Our study elucidates metabolic dynamics that expand our overall understanding of how the species might respond to future ocean conditions and provides a comprehensive dataset that could be used in future ocean acidification response studies.
Collapse
|
15
|
Shen J, Hernandez D, Ye Y, Wu X, Chow WH, Zhao H. Metabolic hormones and breast cancer risk among Mexican American Women in the Mano a Mano Cohort Study. Sci Rep 2019; 9:9989. [PMID: 31292496 PMCID: PMC6620309 DOI: 10.1038/s41598-019-46429-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/23/2019] [Indexed: 12/19/2022] Open
Abstract
C-peptide, insulin, leptin, and other metabolic hormones are assumed to play roles in breast cancer development; though, results are inconsistent. In this prospective case-control study nested within the Mano a Mano Cohort Study, we assessed the risk of breast cancer with regard to plasma levels of c-peptide, gastric inhibitory polypeptide, insulin, leptin, monocyte chemoattractant protein-1, pancreatic polypeptide, and peptide YY. Among women followed for a median of 8.5 years, 109 breast cancer cases were identified and frequency-matched to 327 controls at a ratio of 1:3. Overall, only c-peptide was observed significantly associated with breast cancer risk. High c-peptide levels (≥ the median level of controls) were significantly associated with increased breast cancer risk (odds ratio [OR] = 1.39, 95% confidence interval [CI]: 1.01, 2.44). In an analysis of participants stratified by age, the significant association between c-peptide levels and breast cancer risk was evident in only women age ≥51 years (OR = 1.53, 95% CI: 1.02, 3.27). Among women age <51 years, high leptin levels were significantly associated with decreased breast cancer risk (OR = 0.49, 95% CI: 0.24, 0.82). Our findings suggest that selected metabolic hormones are associated with breast cancer development in Mexican American women.
Collapse
Affiliation(s)
- Jie Shen
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daphne Hernandez
- Department of Health and Human Performance, University of Houston, Texas, USA
| | - Yuanqing Ye
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wong-Ho Chow
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hua Zhao
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
16
|
Beckers M, Jakobi AJ, Sachse C. Thresholding of cryo-EM density maps by false discovery rate control. IUCRJ 2019; 6:18-33. [PMID: 30713700 PMCID: PMC6327189 DOI: 10.1107/s2052252518014434] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/12/2018] [Indexed: 05/31/2023]
Abstract
Cryo-EM now commonly generates close-to-atomic resolution as well as intermediate resolution maps from macromolecules observed in isolation and in situ. Interpreting these maps remains a challenging task owing to poor signal in the highest resolution shells and the necessity to select a threshold for density analysis. In order to facilitate this process, a statistical framework for the generation of confidence maps by multiple hypothesis testing and false discovery rate (FDR) control has been developed. In this way, three-dimensional confidence maps contain signal separated from background noise in the form of local detection rates of EM density values. It is demonstrated that confidence maps and FDR-based thresholding can be used for the interpretation of near-atomic resolution single-particle structures as well as lower resolution maps determined by subtomogram averaging. Confidence maps represent a conservative way of interpreting molecular structures owing to minimized noise. At the same time they provide a detection error with respect to background noise, which is associated with the density and is particularly beneficial for the interpretation of weaker cryo-EM densities in cases of conformational flexibility and lower occupancy of bound molecules and ions in the structure.
Collapse
Affiliation(s)
- Maximilian Beckers
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
- Faculty of Biosciences, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Arjen J. Jakobi
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
- Hamburg Unit c/o DESY, Notkestrasse 85, 22607 Hamburg, Germany
- The Hamburg Centre for Ultrafast Imaging (CUI), Luruper Chaussee 149, 22761 Hamburg, Germany
| | - Carsten Sachse
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
- Ernst Ruska-Centre for Microscopy and Spectroscopy with Electrons (ER-C-3/Structural Biology), Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
17
|
Rattray NJW, Deziel NC, Wallach JD, Khan SA, Vasiliou V, Ioannidis JPA, Johnson CH. Beyond genomics: understanding exposotypes through metabolomics. Hum Genomics 2018; 12:4. [PMID: 29373992 PMCID: PMC5787293 DOI: 10.1186/s40246-018-0134-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/11/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Over the past 20 years, advances in genomic technology have enabled unparalleled access to the information contained within the human genome. However, the multiple genetic variants associated with various diseases typically account for only a small fraction of the disease risk. This may be due to the multifactorial nature of disease mechanisms, the strong impact of the environment, and the complexity of gene-environment interactions. Metabolomics is the quantification of small molecules produced by metabolic processes within a biological sample. Metabolomics datasets contain a wealth of information that reflect the disease state and are consequent to both genetic variation and environment. Thus, metabolomics is being widely adopted for epidemiologic research to identify disease risk traits. In this review, we discuss the evolution and challenges of metabolomics in epidemiologic research, particularly for assessing environmental exposures and providing insights into gene-environment interactions, and mechanism of biological impact. MAIN TEXT Metabolomics can be used to measure the complex global modulating effect that an exposure event has on an individual phenotype. Combining information derived from all levels of protein synthesis and subsequent enzymatic action on metabolite production can reveal the individual exposotype. We discuss some of the methodological and statistical challenges in dealing with this type of high-dimensional data, such as the impact of study design, analytical biases, and biological variance. We show examples of disease risk inference from metabolic traits using metabolome-wide association studies. We also evaluate how these studies may drive precision medicine approaches, and pharmacogenomics, which have up to now been inefficient. Finally, we discuss how to promote transparency and open science to improve reproducibility and credibility in metabolomics. CONCLUSIONS Comparison of exposotypes at the human population level may help understanding how environmental exposures affect biology at the systems level to determine cause, effect, and susceptibilities. Juxtaposition and integration of genomics and metabolomics information may offer additional insights. Clinical utility of this information for single individuals and populations has yet to be routinely demonstrated, but hopefully, recent advances to improve the robustness of large-scale metabolomics will facilitate clinical translation.
Collapse
Affiliation(s)
- Nicholas J. W. Rattray
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT USA
| | - Nicole C. Deziel
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT USA
| | - Joshua D. Wallach
- Collaboration for Research Integrity and Transparency (CRIT), Yale Law School, New Haven, CT USA
- Center for Outcomes Research and Evaluation (CORE), Yale-New Haven Health System, New Haven, CT USA
| | - Sajid A. Khan
- Department of Surgery, Section of Surgical Oncology, Yale University School of Medicine, New Haven, CT USA
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT USA
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT USA
| | - John P. A. Ioannidis
- Stanford Prevention Research Center, Department of Medicine, Stanford University, Stanford, CA USA
- Department of Health Research and Policy, Stanford University, Stanford, CA USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA USA
- Department of Statistics, Stanford University, Stanford, CA USA
- Meta-Research Innovation Center at Stanford, Stanford University, Stanford, CA USA
| | - Caroline H. Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT USA
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
18
|
Kiani-Pouya A, Roessner U, Jayasinghe NS, Lutz A, Rupasinghe T, Bazihizina N, Bohm J, Alharbi S, Hedrich R, Shabala S. Epidermal bladder cells confer salinity stress tolerance in the halophyte quinoa and Atriplex species. PLANT, CELL & ENVIRONMENT 2017; 40:1900-1915. [PMID: 28558173 DOI: 10.1111/pce.12995] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/21/2017] [Indexed: 05/02/2023]
Abstract
Epidermal bladder cells (EBCs) have been postulated to assist halophytes in coping with saline environments. However, little direct supporting evidence is available. Here, Chenopodium quinoa plants were grown under saline conditions for 5 weeks. One day prior to salinity treatment, EBCs from all leaves and petioles were gently removed by using a soft cosmetic brush and physiological, ionic and metabolic changes in brushed and non-brushed leaves were compared. Gentle removal of EBC neither initiated wound metabolism nor affected the physiology and biochemistry of control-grown plants but did have a pronounced effect on salt-grown plants, resulting in a salt-sensitive phenotype. Of 91 detected metabolites, more than half were significantly affected by salinity. Removal of EBC dramatically modified these metabolic changes, with the biggest differences reported for gamma-aminobutyric acid (GABA), proline, sucrose and inositol, affecting ion transport across cellular membranes (as shown in electrophysiological experiments). This work provides the first direct evidence for a role of EBC in salt tolerance in halophytes and attributes this to (1) a key role of EBC as a salt dump for external sequestration of sodium; (2) improved K+ retention in leaf mesophyll and (3) EBC as a storage space for several metabolites known to modulate plant ionic relations.
Collapse
Affiliation(s)
- Ali Kiani-Pouya
- School of Land and Food, University of Tasmania, 7001, Hobart, Tasmania, Australia
| | - Ute Roessner
- School of BioSciences, The University of Melbourne, 3010, Parkville, Victoria, Australia
- Metabolomics Australia, School of BioSciences, The University of Melbourne, 3010, Parkville, Victoria, Australia
| | - Nirupama S Jayasinghe
- Metabolomics Australia, School of BioSciences, The University of Melbourne, 3010, Parkville, Victoria, Australia
| | - Adrian Lutz
- Metabolomics Australia, School of BioSciences, The University of Melbourne, 3010, Parkville, Victoria, Australia
| | - Thusitha Rupasinghe
- Metabolomics Australia, School of BioSciences, The University of Melbourne, 3010, Parkville, Victoria, Australia
| | - Nadia Bazihizina
- School of Land and Food, University of Tasmania, 7001, Hobart, Tasmania, Australia
- Deptartment of Agrifood Production and Environmental Science, University of Florence, I-50019, Florence, Italy
| | - Jennifer Bohm
- School of Land and Food, University of Tasmania, 7001, Hobart, Tasmania, Australia
- Institute for Molecular Plant Physiology and Biophysics, Biocenter, Würzburg University, 97082, Wurzburg, Germany
| | - Sulaiman Alharbi
- Zoology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Rainer Hedrich
- Institute for Molecular Plant Physiology and Biophysics, Biocenter, Würzburg University, 97082, Wurzburg, Germany
| | - Sergey Shabala
- School of Land and Food, University of Tasmania, 7001, Hobart, Tasmania, Australia
| |
Collapse
|
19
|
Dudzik D, Barbas-Bernardos C, García A, Barbas C. Quality assurance procedures for mass spectrometry untargeted metabolomics. a review. J Pharm Biomed Anal 2017; 147:149-173. [PMID: 28823764 DOI: 10.1016/j.jpba.2017.07.044] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/28/2017] [Accepted: 07/29/2017] [Indexed: 12/16/2022]
Abstract
Untargeted metabolomics, as a global approach, has already proven its great potential and capabilities for the investigation of health and disease, as well as the wide applicability for other research areas. Although great progress has been made on the feasibility of metabolomics experiments, there are still some challenges that should be faced and that includes all sources of fluctuations and bias affecting every step involved in multiplatform untargeted metabolomics studies. The identification and reduction of the main sources of unwanted variation regarding the pre-analytical, analytical and post-analytical phase of metabolomics experiments is essential to ensure high data quality. Nowadays, there is still a lack of information regarding harmonized guidelines for quality assurance as those available for targeted analysis. In this review, sources of variations to be considered and minimized along with methodologies and strategies for monitoring and improvement the quality of the results are discussed. The given information is based on evidences from different groups among our own experiences and recommendations for each stage of the metabolomics workflow. The comprehensive overview with tools presented here might serve other researchers interested in monitoring, controlling and improving the reliability of their findings by implementation of good experimental quality practices in the untargeted metabolomics study.
Collapse
Affiliation(s)
- Danuta Dudzik
- Center for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, San Pablo CEU University, Boadilla del Monte, ES-28668, Madrid, Spain.
| | - Cecilia Barbas-Bernardos
- Center for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, San Pablo CEU University, Boadilla del Monte, ES-28668, Madrid, Spain.
| | - Antonia García
- Center for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, San Pablo CEU University, Boadilla del Monte, ES-28668, Madrid, Spain.
| | - Coral Barbas
- Center for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, San Pablo CEU University, Boadilla del Monte, ES-28668, Madrid, Spain.
| |
Collapse
|
20
|
Aksenov AA, da Silva R, Knight R, Lopes NP, Dorrestein PC. Global chemical analysis of biology by mass spectrometry. Nat Rev Chem 2017. [DOI: 10.1038/s41570-017-0054] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
21
|
Park S, Lee J, Kim YH, Park J, Shin JW, Nam S. Clinical Relevance and Molecular Phenotypes in Gastric Cancer, of TP53 Mutations and Gene Expressions, in Combination With Other Gene Mutations. Sci Rep 2016; 6:34822. [PMID: 27708434 PMCID: PMC5052597 DOI: 10.1038/srep34822] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 09/21/2016] [Indexed: 01/19/2023] Open
Abstract
While altered TP53 is the most frequent mutation in gastric cancer (GC), its association with molecular or clinical phenotypes (e.g., overall survival, disease-free survival) remains little known. To that end, we can use genome-wide approaches to identify altered genes significantly related to mutated TP53. Here, we identified significant differences in clinical outcomes, as well as in molecular phenotypes, across specific GC tumor subpopulations, when combining TP53 with other signaling networks, including WNT and its related genes NRXN1, CTNNB1, SLITRK5, NCOR2, RYR1, GPR112, MLL3, MTUS2, and MYH6. Moreover, specific GC subpopulations indicated by dual mutation of NRXN1 and TP53 suggest different drug responses, according to the Connectivity Map, a pharmacological drug-gene association tool. Overall, TP53 mutation status in GC is significantly relevant to clinical or molecular categories. Thus, our approach can potentially provide a patient stratification strategy by dissecting previously unknown multiple TP53-mutated patient groups.
Collapse
Affiliation(s)
- Sungjin Park
- New Experimental Therapeutics Branch, National Cancer Center, Goyang-si Gyeonggi-do, 10408, Korea.,Department of Biomedical Engineering, Inje University, Gimhae, Gyeongnam, 50834, Korea.,Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Korea.,College of Medicine, Gachon University, Incheon, 21936, Korea
| | - Jinhyuk Lee
- Korean Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Korea.,Department of Nanobiotechnology and Bioinformatics, University of Science and Technology, Daejeon, 34113, Korea
| | - Yon Hui Kim
- New Experimental Therapeutics Branch, National Cancer Center, Goyang-si Gyeonggi-do, 10408, Korea.,CrystalGenomics, Inc., Seongnam-si, Gyeonggi-do, 13488, Korea
| | - Jaheun Park
- Digital Information Computing Center, Inje University, Gimhae, Gyeongnam, 50834, Korea
| | - Jung-Woog Shin
- Department of Biomedical Engineering, Inje University, Gimhae, Gyeongnam, 50834, Korea
| | - Seungyoon Nam
- New Experimental Therapeutics Branch, National Cancer Center, Goyang-si Gyeonggi-do, 10408, Korea.,Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Korea.,College of Medicine, Gachon University, Incheon, 21936, Korea
| |
Collapse
|