1
|
Zhang Y, Zhao J, Zhao H, Lu X, Jia X, Zhao X, Xu G. Reference Intervals of Serum Metabolites and Lipids of a Healthy Chinese Population Determined by Liquid Chromatography-Mass Spectrometry. Metabolites 2025; 15:106. [PMID: 39997731 PMCID: PMC11857409 DOI: 10.3390/metabo15020106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Metabolomics serves as a very useful tool for elucidating disease mechanisms and identifying biomarkers. Establishing reference intervals (RIs) of metabolites in a healthy population is crucial to the application of metabolomics in life sciences and clinics. Methods: We enrolled 615 healthy Chinese adults aged between 21 and 85 years. Their health status was ascertained through clinical examinations, biochemical parameters, and medical history. Targeted metabolomics and lipidomics analyses were applied to quantify 705 metabolites and lipids in the serum, establishing RIs and investigating the effect of sex and age on the metabolome and lipidome. Results: This study is the first large-scale effort in China to establish RIs for metabolites in the apparently healthy population. We found that most of the sex-related metabolites, including amino acids, acyl-carnitines and triacylglycerols, had higher concentrations in males, while the other sex-related lipids showed higher concentrations in females. Most of the age-related metabolites increased with age, including those associated with protein synthesis, nitric oxide synthesis, energy metabolism, and lipid metabolism. Conclusions: This study gives the reference intervals of the healthy Chinese metabolome and lipidome and their relationship with sex and age, which facilitates life sciences and precision medicine, especially for disease research and biomarker discovery.
Collapse
Affiliation(s)
- Yuqing Zhang
- School of Chemistry, Dalian University of Technology, Dalian 116024, China;
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China; (J.Z.); (X.L.)
| | - Jinhui Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China; (J.Z.); (X.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Zhao
- Department of the Health Checkup Center, The Second Hospital of Dalian Medical University, Dalian 116023, China (X.J.)
| | - Xin Lu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China; (J.Z.); (X.L.)
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Xueni Jia
- Department of the Health Checkup Center, The Second Hospital of Dalian Medical University, Dalian 116023, China (X.J.)
| | - Xinjie Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China; (J.Z.); (X.L.)
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Guowang Xu
- School of Chemistry, Dalian University of Technology, Dalian 116024, China;
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China; (J.Z.); (X.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| |
Collapse
|
2
|
Wasilewski A, Wasilewska E, Serrafi A. Exploring Diagnostic Markers and Therapeutic Targets in Parkinson's Disease: A Comprehensive 1H-NMR Metabolomic Analysis - Systematic Review. Arch Immunol Ther Exp (Warsz) 2025; 73:aite-2025-0011. [PMID: 40214076 DOI: 10.2478/aite-2025-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/05/2025] [Indexed: 04/19/2025]
Abstract
Parkinson's disease (PD) affects millions of people globally. Accurate early diagnosis remains a challenge due to the lack of specific biomarkers. This systematic review explores the potential of 1H-NMR metabolomics in identifying diagnostic markers and therapeutic targets for PD. A comprehensive analysis was conducted across databases such as Scopus, Web of Science, PubMed, and Embase, focusing on studies that utilized 1H-NMR spectroscopy to profile metabolites associated with PD progression. The review identifies key metabolites-glutamate, taurine, myo-inositol, glutamine, and creatine-that play critical roles in the pathophysiology of PD. Glutamate, linked to excitotoxicity and neuronal degeneration, emerges as a prominent target for therapeutic intervention, while taurine is associated with oxidative stress. Myo-inositol, a key regulator of autophagy, underscores the biochemical dysregulation associated with PD, similar to glutamine and glutamate. Creatine's role in neuronal energy metabolism suggests potential avenues for treatment focused on energy supplementation. The reproducibility of metabolite findings varied, indicating the complexity of PD's metabolomic landscape. Despite challenges in consistency, these metabolites hold promise as biomarkers for diagnosing PD and tracking disease progression. The review underscores the need for further validation of these markers and their integration with other omics technologies to enhance PD management. By identifying key metabolic pathways, this study opens new directions for personalized medicine, offering potential therapeutic targets to slow disease progression and improve patient outcomes.
Collapse
Affiliation(s)
- Andrzej Wasilewski
- Student Scientific Association of Medical Chemistry and Immunochemistry, Wroclaw Medical University, Wroclaw, Poland
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Eliza Wasilewska
- Department of Allergology, Medical University of Gdansk, Gdansk, Poland
- Center of Rare Disease, Centre University Hospital, Gdansk, Poland
| | - Agata Serrafi
- Department of Immunochemistry and Chemistry, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
3
|
Labory J, Njomgue-Fotso E, Bottini S. Benchmarking feature selection and feature extraction methods to improve the performances of machine-learning algorithms for patient classification using metabolomics biomedical data. Comput Struct Biotechnol J 2024; 23:1274-1287. [PMID: 38560281 PMCID: PMC10979063 DOI: 10.1016/j.csbj.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Objective Classification tasks are an open challenge in the field of biomedicine. While several machine-learning techniques exist to accomplish this objective, several peculiarities associated with biomedical data, especially when it comes to omics measurements, prevent their use or good performance achievements. Omics approaches aim to understand a complex biological system through systematic analysis of its content at the molecular level. On the other hand, omics data are heterogeneous, sparse and affected by the classical "curse of dimensionality" problem, i.e. having much fewer observation, samples (n) than omics features (p). Furthermore, a major problem with multi-omics data is the imbalance either at the class or feature level. The objective of this work is to study whether feature extraction and/or feature selection techniques can improve the performances of classification machine-learning algorithms on omics measurements. Methods Among all omics, metabolomics has emerged as a powerful tool in cancer research, facilitating a deeper understanding of the complex metabolic landscape associated with tumorigenesis and tumor progression. Thus, we selected three publicly available metabolomics datasets, and we applied several feature extraction techniques both linear and non-linear, coupled or not with feature selection methods, and evaluated the performances regarding patient classification in the different configurations for the three datasets. Results We provide general workflow and guidelines on when to use those techniques depending on the characteristics of the data available. To further test the extension of our approach to other omics data, we have included a transcriptomics and a proteomics data. Overall, for all datasets, we showed that applying supervised feature selection improves the performances of feature extraction methods for classification purposes. Scripts used to perform all analyses are available at: https://github.com/Plant-Net/Metabolomic_project/.
Collapse
Affiliation(s)
- Justine Labory
- Université Côte d′Azur, Center of Modeling Simulation and Interactions, Nice, France
- INRAE, Université Côte d′Azur, CNRS, Institut Sophia Agrobiotech, Sophia-Antipolis, France
- Université Côte d′Azur, Inserm U1081, CNRS UMR 7284, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | | | - Silvia Bottini
- Université Côte d′Azur, Center of Modeling Simulation and Interactions, Nice, France
- INRAE, Université Côte d′Azur, CNRS, Institut Sophia Agrobiotech, Sophia-Antipolis, France
| |
Collapse
|
4
|
Chappel JR, Kirkwood-Donelson KI, Dodds JN, Fleming J, Reif DM, Baker ES. Streamlining Phenotype Classification and Highlighting Feature Candidates: A Screening Method for Non-Targeted Ion Mobility Spectrometry-Mass Spectrometry (IMS-MS) Data. Anal Chem 2024; 96:15970-15979. [PMID: 39292613 PMCID: PMC11480931 DOI: 10.1021/acs.analchem.4c03256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Nontargeted analysis (NTA) is increasingly utilized for its ability to identify key molecular features beyond known targets in complex samples. NTA is particularly advantageous in exploratory studies aimed at identifying phenotype-associated features or molecules able to classify various sample types. However, implementing NTA involves extensive data analyses and labor-intensive annotations. To address these limitations, we developed a rapid data screening capability compatible with NTA data collected on a liquid chromatography, ion mobility spectrometry, and mass spectrometry (LC-IMS-MS) platform that allows for sample classification while highlighting potential features of interest. Specifically, this method aggregates the thousands of IMS-MS spectra collected across the LC space for each sample and collapses the LC dimension, resulting in a single summed IMS-MS spectrum for screening. The summed IMS-MS spectra are then analyzed with a bootstrapped Lasso technique to identify key regions or coordinates for phenotype classification via support vector machines. Molecular annotations are then performed by examining the features present in the selected coordinates, highlighting potential molecular candidates. To demonstrate this summed IMS-MS screening approach, we applied it to clinical plasma lipidomic NTA data and exposomic NTA data from water sites with varying contaminant levels. Distinguishing coordinates were observed in both studies, enabling the evaluation of phenotypic molecular annotations and resulting in screening models capable of classifying samples with up to a 25% increase in accuracy compared to models using annotated data.
Collapse
Affiliation(s)
- Jessie R Chappel
- Bioinformatics Research Center, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Kaylie I Kirkwood-Donelson
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Durham, North Carolina 27709, United States
| | - James N Dodds
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Jonathon Fleming
- Bioinformatics Research Center, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - David M Reif
- Predictive Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Durham, North Carolina 27709, United States
| | - Erin S Baker
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| |
Collapse
|
5
|
Liu YA, Aboud O, Dahabiyeh LA, Bloch O, Fiehn O. Metabolomic characterization of human glioblastomas and patient plasma: a pilot study. F1000Res 2024; 13:98. [PMID: 39371551 PMCID: PMC11452765 DOI: 10.12688/f1000research.143642.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/08/2024] Open
Abstract
Background Glioblastoma (GBM) is a clinically challenging primary brain tumor with poor survival outcome despite surgical resection and intensive chemoradiation. The metabolic heterogeneity of GBM can become biomarkers for treatment response, resistance, and outcome prediction. The aim of the study is to investigate metabolic distinctions between primary and recurrent GBM tissue and patient plasma to establish feasibility for metabolic profiling. Methods A single-center cohort study analyzed tissue and blood samples from 15 patients with GBM using untargeted metabolomic/lipidomic assays. Metabolomic, lipidomic, and biogenic amine analyses were conducted on GBM tissue and patient plasma at diagnosis and recurrence using untargeted mass spectrometry. The study utilized a small but longitudinally collected cohort to evaluate alteration in metabolites, lipids, and biogenic amines between specimens at diagnosis and recurrence. Results Exploratory analysis revealed significant alteration in metabolites, lipids, and biogenic amines between diagnostic and recurrent states in both tumor and plasma specimens. Notable metabolites differed at recurrence, including N-alpha-methylhistamine, glycerol-3-phosphate, phosphocholine, and succinic acid in tissue, and indole-3-acetate, and urea in plasma. Principal component analysis revealed distinct metabolomic profiles between tumor tissue and patient plasma. Distinct metabolic profiles were observed in GBM tissue and patient plasma at recurrence, demonstrating the feasibility of using metabolomic methodologies for longitudinal studies. One patient exhibited a unique tumor resistance signature at diagnosis, possibly indicating a high-risk metabolomic phenotype. Conclusions In this small cohort, the findings suggest the potential of metabolomic signatures of GBM tissue and patient plasma for risk stratification, outcome prediction, and the development of novel adjuvant metabolic-targeting therapies. The findings suggest metabolic discrepancies at diagnosis and recurrence in tissue and plasma, highlighting potential implications for evaluation of clinical response. The identification of significant changes in metabolite abundance emphasizes the need for larger studies using targeted metabolomics to validate and further explore these profiles.
Collapse
Affiliation(s)
- Yin Allison Liu
- Department of Opthalmology, University of California Davis, Davis, California, USA
- Department of Neurology, University of California Davis, Davis, California, USA
- Department of Neurosurgery, University of California Davis, Davis, California, USA
| | - Orwa Aboud
- Department of Opthalmology, University of California Davis, Davis, California, USA
- Department of Neurology, University of California Davis, Davis, California, USA
- Department of Neurosurgery, University of California Davis, Davis, California, USA
- Comprehensive Cancer Center, University of California Davis, Davis, California, USA
| | - Lina A. Dahabiyeh
- West Coast Metabolomics Center, University of California Davis, Davis, California, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Jordan, Amman, Amman Governorate, Jordan
| | - Orin Bloch
- Department of Opthalmology, University of California Davis, Davis, California, USA
- Department of Neurosurgery, University of California Davis, Davis, California, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, California, USA
| |
Collapse
|
6
|
Ghosh N, Lejonberg C, Czuba T, Dekkers K, Robinson R, Ärnlöv J, Melander O, Smith ML, Evans AM, Gidlöf O, Gerszten RE, Lind L, Engström G, Fall T, Smith JG. Analysis of plasma metabolomes from 11 309 subjects in five population-based cohorts. Sci Rep 2024; 14:8933. [PMID: 38637659 PMCID: PMC11026396 DOI: 10.1038/s41598-024-59388-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Plasma metabolomics holds potential for precision medicine, but limited information is available to compare the performance of such methods across multiple cohorts. We compared plasma metabolite profiles after an overnight fast in 11,309 participants of five population-based Swedish cohorts (50-80 years, 52% women). Metabolite profiles were uniformly generated at a core laboratory (Metabolon Inc.) with untargeted liquid chromatography mass spectrometry and a comprehensive reference library. Analysis of a second sample obtained one year later was conducted in a subset. Of 1629 detected metabolites, 1074 (66%) were detected in all cohorts while only 10% were unique to one cohort, most of which were xenobiotics or uncharacterized. The major classes were lipids (28%), xenobiotics (22%), amino acids (14%), and uncharacterized (19%). The most abundant plasma metabolome components were the major dietary fatty acids and amino acids, glucose, lactate and creatinine. Most metabolites displayed a log-normal distribution. Temporal variability was generally similar to clinical chemistry analytes but more pronounced for xenobiotics. Extensive metabolite-metabolite correlations were observed but mainly restricted to within each class. Metabolites were broadly associated with clinical factors, particularly body mass index, sex and renal function. Collectively, our findings inform the conduct and interpretation of metabolite association and precision medicine studies.
Collapse
Affiliation(s)
- Nilanjana Ghosh
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and the Department of Cardiology, Sahlgrenska University Hospital, SE-413 45, Gothenburg, Sweden
| | - Carl Lejonberg
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Tomasz Czuba
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and the Department of Cardiology, Sahlgrenska University Hospital, SE-413 45, Gothenburg, Sweden
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Koen Dekkers
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Johan Ärnlöv
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Olle Melander
- Department of Internal Medicine, Clinical Sciences, Lund University, Malmö, Sweden
| | - Maya Landenhed Smith
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and the Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Olof Gidlöf
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gunnar Engström
- Cardiovascular Epidemiology, Clinical Sciences, Lund University, Malmö, Sweden
| | - Tove Fall
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - J Gustav Smith
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and the Department of Cardiology, Sahlgrenska University Hospital, SE-413 45, Gothenburg, Sweden.
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden.
- Department of Heart Failure and Valvular Disease, Skåne University Hospital, Lund, Sweden.
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden.
| |
Collapse
|
7
|
Calzadilla N, Jayawardena D, Qazi A, Sharma A, Mongan K, Comiskey S, Eathara A, Saksena S, Dudeja PK, Alrefai WA, Gill RK. Serotonin Transporter Deficiency Induces Metabolic Alterations in the Ileal Mucosa. Int J Mol Sci 2024; 25:4459. [PMID: 38674044 PMCID: PMC11049861 DOI: 10.3390/ijms25084459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Serotonin transporter (SERT) deficiency has been implicated in metabolic syndrome, intestinal inflammation, and microbial dysbiosis. Interestingly, changes in microbiome metabolic capacity and several alterations in host gene expression, including lipid metabolism, were previously observed in SERT-/- mice ileal mucosa. However, the precise host or microbial metabolites altered by SERT deficiency that may contribute to the pleiotropic phenotype of SERT KO mice are not yet understood. This study investigated the hypothesis that SERT deficiency impacts lipid and microbial metabolite abundances in the ileal mucosa, where SERT is highly expressed. Ileal mucosal metabolomics was performed by Metabolon on wild-type (WT) and homozygous SERT knockout (KO) mice. Fluorescent-activated cell sorting (FACS) was utilized to measure immune cell populations in ileal lamina propria to assess immunomodulatory effects caused by SERT deficiency. SERT KO mice exhibited a unique ileal mucosal metabolomic signature, with the most differentially altered metabolites being lipids. Such changes included increased diacylglycerols and decreased monoacylglycerols in the ileal mucosa of SERT KO mice compared to WT mice. Further, the ileal mucosa of SERT KO mice exhibited several changes in microbial-related metabolites known to play roles in intestinal inflammation and insulin resistance. SERT KO mice also had a significant reduction in the abundance of ileal group 3 innate lymphoid cells (ILC3). In conclusion, SERT deficiency induces complex alterations in the ileal mucosal environment, indicating potential links between serotonergic signaling, gut microbiota, mucosal immunity, intestinal inflammation, and metabolic syndrome.
Collapse
Affiliation(s)
- Nathan Calzadilla
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA;
| | - Dulari Jayawardena
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
| | - Aisha Qazi
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
| | - Anchal Sharma
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
| | - Kai Mongan
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
| | - Shane Comiskey
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
| | - Abhijith Eathara
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
| | - Seema Saksena
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Pradeep K. Dudeja
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Waddah A. Alrefai
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Ravinder K. Gill
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA; (D.J.); (A.Q.); (A.S.); (K.M.); (S.C.); (A.E.); (S.S.); (P.K.D.); (W.A.A.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
8
|
Yin G, Sun Z, Wang Z, Xia Y, Cheng L, Qin G, Aschalew ND, Liu H, Zhang X, Wu Q, Zhang W, Zhao W, Wang T, Zhen Y. Mechanistic insights into inositol-mediated rumen function promotion and metabolic alteration using in vitro and in vivo models. Front Vet Sci 2024; 11:1359234. [PMID: 38435365 PMCID: PMC10904589 DOI: 10.3389/fvets.2024.1359234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/01/2024] [Indexed: 03/05/2024] Open
Abstract
Inositol is a bioactive factor that is widely found in nature; however, there are few studies on its use in ruminant nutrition. This study investigated the effects of different inositol doses and fermentation times on rumen fermentation and microbial diversity, as well as the levels of rumen and blood metabolites in sheep. Rumen fermentation parameters, microbial diversity, and metabolites after different inositol doses were determined in vitro. According to the in vitro results, six small-tailed Han sheep fitted with permanent rumen fistulas were used in a 3 × 3 Latin square feeding experiment where inositol was injected into the rumen twice a day and rumen fluid and blood samples were collected. The in vitro results showed that inositol could increase in vitro dry matter digestibility, in vitro crude protein digestibility, NH3-N, acetic acid, propionic acid, and rumen microbial diversity and affect rumen metabolic pathways (p < 0.05). The feeding experiment results showed that inositol increased the blood concentration of high-density lipoprotein and IgG, IgM, and IL-4 levels. The rumen microbial composition was significantly affected (p < 0.05). Differential metabolites in the rumen were mainly involved in ABC transporters, biotin metabolism, and phenylalanine metabolism, whereas those in the blood were mainly involved in arginine biosynthesis and glutathione and tyrosine metabolism. In conclusion, inositol improves rumen function, affects rumen microorganisms and rumen and blood metabolites and may reduce inflammation, improving animal health.
Collapse
Affiliation(s)
- Guopei Yin
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Zhe Sun
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun, China
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science and Technology Co., Ltd., Changchun, China
| | - Zhanqing Wang
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yuanhong Xia
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Long Cheng
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Guixin Qin
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Natnael D. Aschalew
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- College of Agriculture and Environmental Science, Dilla University, Dila, Ethiopia
| | - Hongyun Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xuefeng Zhang
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Qilu Wu
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Weigang Zhang
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science and Technology Co., Ltd., Changchun, China
| | - Wei Zhao
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science and Technology Co., Ltd., Changchun, China
| | - Tao Wang
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science and Technology Co., Ltd., Changchun, China
| | - Yuguo Zhen
- Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, Key Laboratory of Animal Production Product Quality and Security Ministry of Education, JLAU-Borui Dairy Science and Technology R&D Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science and Technology Co., Ltd., Changchun, China
| |
Collapse
|
9
|
Pautova AK. Metabolic Profiling of Aromatic Compounds. Metabolites 2024; 14:107. [PMID: 38392999 PMCID: PMC10890443 DOI: 10.3390/metabo14020107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Metabolic profiling is a powerful modern tool in searching for novel biomarkers and indicators of normal or pathological processes in the body [...].
Collapse
Affiliation(s)
- Alisa K Pautova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25-2 Petrovka Str., 107031 Moscow, Russia
| |
Collapse
|
10
|
Gątarek P, Kałużna-Czaplińska J. Integrated metabolomics and proteomics analysis of plasma lipid metabolism in Parkinson's disease. Expert Rev Proteomics 2024; 21:13-25. [PMID: 38346207 DOI: 10.1080/14789450.2024.2315193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024]
Abstract
INTRODUCTION Metabolomics and proteomics are two growing fields of science which may shed light on the molecular mechanisms that contribute to neurodegenerative diseases. Studies focusing on these aspects can reveal specific metabolites and proteins that can halt or reverse the progressive neurodegenerative process leading to dopaminergic cell death in the brain. AREAS COVERED In this article, an overview of the current status of metabolomic and proteomic profiling in the neurodegenerative disease such as Parkinson's disease (PD) is presented. We discuss the importance of state-of-the-art metabolomics and proteomics using advanced analytical methodologies and their potential for discovering new biomarkers in PD. We critically review the research to date, highlighting how metabolomics and proteomics can have an important impact on early disease diagnosis, future therapy development and the identification of new biomarkers. Finally, we will discuss interactions between lipids and α-synuclein (SNCA) and also consider the role of SNCA in lipid metabolism. EXPERT OPINION Metabolomic and proteomic studies contribute to understanding the biological basis of PD pathogenesis, identifying potential biomarkers and introducing new therapeutic strategies. The complexity and multifactorial nature of this disease requires a comprehensive approach, which can be achieved by integrating just these two omic studies.
Collapse
Affiliation(s)
- Paulina Gątarek
- Institute Of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Lodz, Poland
- CONEM Poland Chemistry and Nutrition Research Group, Lodz University of Technology, Lodz, Poland
| | - Joanna Kałużna-Czaplińska
- Institute Of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Lodz, Poland
- CONEM Poland Chemistry and Nutrition Research Group, Lodz University of Technology, Lodz, Poland
| |
Collapse
|
11
|
Cai L, Vu HS, Gu W, Chen H, Franklin J, Haidar LA, Wu Z, Pan C, Cai F, Nguyen P, Ko B, Yang C, Zacharias LG, Sudderth J, Montgomery S, Uhles C, Fisher H, Hudnall J, Hornbuckle C, Quinn C, Michel D, Umaña L, Scheuerle A, McNutt MC, Gotway GK, Afroze B, Ni M, DeBerardinis R. An interactive web application for exploring human plasma and fibroblast metabolomics data from patients with inborn errors of metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.571124. [PMID: 38168314 PMCID: PMC10760037 DOI: 10.1101/2023.12.11.571124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Metabolomic profiling is instrumental in understanding the systemic and cellular impact of inborn errors of metabolism (IEMs), monogenic disorders caused by pathogenic genomic variants in genes involved in metabolism. This study encompasses untargeted metabolomics analysis of plasma from 474 individuals and fibroblasts from 67 subjects, incorporating healthy controls, patients with 65 different monogenic diseases, and numerous undiagnosed cases. We introduce a web application designed for the in-depth exploration of this extensive metabolomics database. The application offers a user-friendly interface for data review, download, and detailed analysis of metabolic deviations linked to IEMs at the level of individual patients or groups of patients with the same diagnosis. It also provides interactive tools for investigating metabolic relationships and offers comparative analyses of plasma and fibroblast profiles. This tool emphasizes the metabolic interplay within and across biological matrices, enriching our understanding of metabolic regulation in health and disease. As a resource, the application provides broad utility in research, offering novel insights into metabolic pathways and their alterations in various disorders.
Collapse
|
12
|
Zhao T, Yan Q, Wang C, Zeng J, Zhang R, Wang H, Pu L, Dai X, Liu H, Han L. Identification of Serum Biomarkers of Ischemic Stroke in a Hypertensive Population Based on Metabolomics and Lipidomics. Neuroscience 2023; 533:22-35. [PMID: 37806545 DOI: 10.1016/j.neuroscience.2023.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023]
Abstract
Hypertensive individuals are at a high risk of stroke, and thus, prevention of stroke in hypertensive patients is essential. Metabolomics and lipidomics can be used to identify diagnostic biomarkers and conduct early assessments of stroke risk in hypertensive populations. In this study, serum samples were collected from 30 hypertensive ischemic stroke (IS), 30 matched hypertensive and 30 matched healthy participants. Metabolomics and lipidomics analyses were conducted via liquid chromatography-tandem mass spectrometry, and the data were analyzed using multivariate and univariate statistical methods. A random forest algorithm and binary logistic regression were used to screen the biomarkers and establish diagnostic model. We detected 21 differential metabolites and 38 differential lipids between the hypertensive IS and healthy group. Moreover, we found 18 differential metabolites and 31 differential lipids between the hypertensive IS and hypertension group. In particular, the following seven metabolites or lipids distinguished the hypertensive IS from the healthy group: 4-hydroxyphenylpyruvic acid, cafestol, phosphatidylethanolamine (PE) (18:0p/18:2), PE (16:0e/20:4), (O-acyI)-1-hydroxy fatty acid (36:3), PE (16:0p/20:3) and PE (18:1p/18:2) (rep). The following seven biomarkers distinguished the hypertensive IS from the hypertension group: diglyceride (DG) (20:1/18:2), PE (18:0p/18:2), PE (16:0e/22:5), phosphatidylcholine (40:7), dimethylphosphatidylethanolamine (50:3), DG (18:1/18:2), and 4-hydroxyphenylpyruvic acid. The aforementioned panels had good diagnostic and predictive ability for hypertensive IS. Our study determines the metabolomic and lipidomic profiles of hypertensive IS patients and thereby identifies potential biomarkers of the presence of IS in hypertensive populations.
Collapse
Affiliation(s)
- Tian Zhao
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No. 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, China.
| | - Qianqian Yan
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, China.
| | - Changyi Wang
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen 518000, China.
| | - Jingjing Zeng
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No. 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, China.
| | - Ruijie Zhang
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No. 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, China.
| | - Han Wang
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No. 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, China.
| | - Liyuan Pu
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No. 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, China.
| | - Xiaoyu Dai
- Department of Anus & Intestine Surgery, Ningbo No. 2 Hospital, Ningbo 315000, China.
| | - Huina Liu
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No. 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, China.
| | - Liyuan Han
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No. 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, China.
| |
Collapse
|
13
|
Jalota A, Hershberger CE, Patel MS, Mian A, Faruqi A, Khademi G, Rotroff DM, Hill BT, Gupta N. Host metabolome predicts the severity and onset of acute toxicities induced by CAR T-cell therapy. Blood Adv 2023; 7:4690-4700. [PMID: 36399526 PMCID: PMC10468366 DOI: 10.1182/bloodadvances.2022007456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/04/2022] [Accepted: 08/20/2022] [Indexed: 11/19/2022] Open
Abstract
Anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is a highly effective treatment option for patients with relapsed/refractory large B-cell lymphoma. However, widespread use is deterred by the development of clinically significant acute inflammatory toxicities, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), that induce significant morbidity and require close monitoring. Identification of host biochemical signatures that predict the severity and time-to-onset of CRS and ICANS may assist patient stratification to enable timely mitigation strategies. Here, we report pretreatment host metabolites that are associated with CRS and ICANS induced by axicabtagene ciloleucel or tisagenlecleucel therapy. Both untargeted metabolomics analysis and validation using targeted assays revealed a significant association between the abundance of specific pretreatment biochemical entities and an increased risk and/or onset of clinically significant CRS (q < .1) and ICANS (q < .25). Higher pretreatment levels of plasma glucose and lower levels of cholesterol and glutamate were associated with a faster onset of CRS. In contrast, low baseline levels of the amino acids proline and glycine and the secondary bile acid isoursodeoxycholate were significantly correlated with clinically significant CRS. Lower concentration of the amino acid hydroxyproline was associated with higher grade and faster onset of ICANS, whereas low glutamine was negatively correlated with faster development of ICANS. Overall, our data indicate that the pretreatment host metabolome has biomarker potential in determining the risk of clinically significant CRS and ICANS, and may be useful in risk stratification of patients before anti-CD19 CAR T-cell therapy.
Collapse
Affiliation(s)
- Akansha Jalota
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH
| | | | - Manishkumar S. Patel
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH
| | - Agrima Mian
- Department of Internal Medicine, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Aiman Faruqi
- Cleveland Clinic Lerner College of Medicine, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Gholamreza Khademi
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland, OH
| | - Daniel M. Rotroff
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland, OH
- Cleveland Clinic Lerner College of Medicine, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Brian T. Hill
- Cleveland Clinic Lerner College of Medicine, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Neetu Gupta
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH
- Cleveland Clinic Lerner College of Medicine, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
14
|
Shields PG. Role of untargeted omics biomarkers of exposure and effect for tobacco research. ADDICTION NEUROSCIENCE 2023; 7:100098. [PMID: 37396411 PMCID: PMC10310069 DOI: 10.1016/j.addicn.2023.100098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Tobacco research remains a clear priority to improve individual and population health, and has recently become more complex with emerging combustible and noncombustible tobacco products. The use of omics methods in prevention and cessation studies are intended to identify new biomarkers for risk, compared risks related to other products and never use, and compliance for cessation and reinitation. to assess the relative effects of tobacco products to each other. They are important for the prediction of reinitiation of tobacco use and relapse prevention. In the research setting, both technical and clinical validation is required, which presents a number of complexities in the omics methodologies from biospecimen collection and sample preparation to data collection and analysis. When the results identify differences in omics features, networks or pathways, it is unclear if the results are toxic effects, a healthy response to a toxic exposure or neither. The use of surrogate biospecimens (e.g., urine, blood, sputum or nasal) may or may not reflect target organs such as the lung or bladder. This review describes the approaches for the use of omics in tobacco research and provides examples of prior studies, along with the strengths and limitations of the various methods. To date, there is little consistency in results, likely due to small number of studies, limitations in study size, the variability in the analytic platforms and bioinformatic pipelines, differences in biospecimen collection and/or human subject study design. Given the demonstrated value for the use of omics in clinical medicine, it is anticipated that the use in tobacco research will be similarly productive.
Collapse
Affiliation(s)
- Peter G. Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH
| |
Collapse
|
15
|
Siracusano M, Arturi L, Riccioni A, Noto A, Mussap M, Mazzone L. Metabolomics: Perspectives on Clinical Employment in Autism Spectrum Disorder. Int J Mol Sci 2023; 24:13404. [PMID: 37686207 PMCID: PMC10487559 DOI: 10.3390/ijms241713404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/09/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Precision medicine is imminent, and metabolomics is one of the main actors on stage. We summarize and discuss the current literature on the clinical application of metabolomic techniques as a possible tool to improve early diagnosis of autism spectrum disorder (ASD), to define clinical phenotypes and to identify co-occurring medical conditions. A review of the current literature was carried out after PubMed, Medline and Google Scholar were consulted. A total of 37 articles published in the period 2010-2022 was included. Selected studies involve as a whole 2079 individuals diagnosed with ASD (1625 males, 394 females; mean age of 10, 9 years), 51 with other psychiatric comorbidities (developmental delays), 182 at-risk individuals (siblings, those with genetic conditions) and 1530 healthy controls (TD). Metabolomics, reflecting the interplay between genetics and environment, represents an innovative and promising technique to approach ASD. The metabotype may mirror the clinical heterogeneity of an autistic condition; several metabolites can be expressions of dysregulated metabolic pathways thus liable of leading to clinical profiles. However, the employment of metabolomic analyses in clinical practice is far from being introduced, which means there is a need for further studies for the full transition of metabolomics from clinical research to clinical diagnostic routine.
Collapse
Affiliation(s)
- Martina Siracusano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
| | - Lucrezia Arturi
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
| | - Assia Riccioni
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
| | - Antonio Noto
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, SS 554, Km 4.5, 09042 Monserrato, Italy
| | - Michele Mussap
- Department of Surgical Sciences, School of Medicine, University of Cagliari, Cittadella Universitaria, SS 554, Km 4.5, 09042 Monserrato, Italy
| | - Luigi Mazzone
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
- Systems Medicine Department, University of Rome Tor Vergata, Montpellier Street 1, 00133 Rome, Italy
| |
Collapse
|
16
|
Calzadilla N, Qazi A, Sharma A, Mongan K, Comiskey S, Manne J, Youkhana AG, Khanna S, Saksena S, Dudeja PK, Alrefai WA, Gill RK. Mucosal Metabolomic Signatures in Chronic Colitis: Novel Insights into the Pathophysiology of Inflammatory Bowel Disease. Metabolites 2023; 13:873. [PMID: 37512580 PMCID: PMC10386370 DOI: 10.3390/metabo13070873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammatory bowel diseases (IBD) involve complex interactions among genetic factors, aberrant immune activation, and gut microbial dysbiosis. While metabolomic studies have focused on feces and serum, fewer investigations have examined the intestinal mucosa despite its crucial role in metabolite absorption and transport. The goals of this study were twofold: to test the hypothesis that gut microbial dysbiosis from chronic intestinal inflammation leads to mucosal metabolic alterations suitable for therapeutic targeting, and to address gaps in metabolomic studies of intestinal inflammation that have overlooked the mucosal metabolome. The chronic DSS colitis was induced for five weeks in 7-9-week-old wild-type C57BL/6J male mice followed by microbial profiling with targeted 16srRNA sequencing service. Mucosal metabolite measurements were performed by Metabolon (Morrisville, NC). The data were analyzed using the bioinformatic tools Pathview, MetOrigin, and Metaboanalyst. The novel findings demonstrated increases in several host- and microbe-derived purine, pyrimidine, endocannabinoid, and ceramide metabolites in colitis. Origin analysis revealed that microbial-related tryptophan metabolites kynurenine, anthranilate, 5-hydroxyindoleacetate, and C-glycosyltryptophan were significantly increased in colon mucosa during chronic inflammation and strongly correlated with disease activity. These findings offer new insights into the pathophysiology of IBD and provide novel potential targets for microbial-based therapeutics.
Collapse
Affiliation(s)
- Nathan Calzadilla
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Aisha Qazi
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Anchal Sharma
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Kai Mongan
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Shane Comiskey
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jahnavi Manne
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Alvin G Youkhana
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Sonam Khanna
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Seema Saksena
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Pradeep K Dudeja
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Waddah A Alrefai
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Ravinder K Gill
- Division of Gastroenterology & Hepatology, University of Illinois Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
17
|
Chen Y, Xu W, Zhang W, Tong R, Yuan A, Li Z, Jiang H, Hu L, Huang L, Xu Y, Zhang Z, Sun M, Yan X, Chen AF, Qian K, Pu J. Plasma metabolic fingerprints for large-scale screening and personalized risk stratification of metabolic syndrome. Cell Rep Med 2023; 4:101109. [PMID: 37467725 PMCID: PMC10394172 DOI: 10.1016/j.xcrm.2023.101109] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/01/2023] [Accepted: 06/16/2023] [Indexed: 07/21/2023]
Abstract
Direct diagnosis and accurate assessment of metabolic syndrome (MetS) allow for prompt clinical interventions. However, traditional diagnostic strategies overlook the complex heterogeneity of MetS. Here, we perform metabolomic analysis in 13,554 participants from the natural cohort and identify 26 hub plasma metabolic fingerprints (PMFs) associated with MetS and its early identification (pre-MetS). By leveraging machine-learning algorithms, we develop robust diagnostic models for pre-MetS and MetS with convincing performance through independent validation. We utilize these PMFs to assess the relative contributions of the four major MetS risk factors in the general population, ranked as follows: hyperglycemia, hypertension, dyslipidemia, and obesity. Furthermore, we devise a personalized three-dimensional plasma metabolic risk (PMR) stratification, revealing three distinct risk patterns. In summary, our study offers effective screening tools for identifying pre-MetS and MetS patients in the general community, while defining the heterogeneous risk stratification of metabolic phenotypes in real-world settings.
Collapse
Affiliation(s)
- Yifan Chen
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Wei Xu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Wei Zhang
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Renyang Tong
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Ancai Yuan
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Zheng Li
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Huiru Jiang
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Liuhua Hu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Lin Huang
- Country Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yudian Xu
- School of Biomedical Engineering, Institute of Medical Robotics and Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ziyue Zhang
- School of Biomedical Engineering, Institute of Medical Robotics and Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Mingze Sun
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Kun Qian
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China; School of Biomedical Engineering, Institute of Medical Robotics and Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Jun Pu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China.
| |
Collapse
|
18
|
Vernardis SI, Demichev V, Lemke O, Grüning NM, Messner C, White M, Pietzner M, Peluso A, Collet TH, Henning E, Gille C, Campbell A, Hayward C, Porteous DJ, Marioni RE, Mülleder M, Zelezniak A, Wareham NJ, Langenberg C, Farooqi IS, Ralser M. The Impact of Acute Nutritional Interventions on the Plasma Proteome. J Clin Endocrinol Metab 2023; 108:2087-2098. [PMID: 36658456 PMCID: PMC10348471 DOI: 10.1210/clinem/dgad031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
CONTEXT Humans respond profoundly to changes in diet, while nutrition and environment have a great impact on population health. It is therefore important to deeply characterize the human nutritional responses. OBJECTIVE Endocrine parameters and the metabolome of human plasma are rapidly responding to acute nutritional interventions such as caloric restriction or a glucose challenge. It is less well understood whether the plasma proteome would be equally dynamic, and whether it could be a source of corresponding biomarkers. METHODS We used high-throughput mass spectrometry to determine changes in the plasma proteome of i) 10 healthy, young, male individuals in response to 2 days of acute caloric restriction followed by refeeding; ii) 200 individuals of the Ely epidemiological study before and after a glucose tolerance test at 4 time points (0, 30, 60, 120 minutes); and iii) 200 random individuals from the Generation Scotland study. We compared the proteomic changes detected with metabolome data and endocrine parameters. RESULTS Both caloric restriction and the glucose challenge substantially impacted the plasma proteome. Proteins responded across individuals or in an individual-specific manner. We identified nutrient-responsive plasma proteins that correlate with changes in the metabolome, as well as with endocrine parameters. In particular, our study highlights the role of apolipoprotein C1 (APOC1), a small, understudied apolipoprotein that was affected by caloric restriction and dominated the response to glucose consumption and differed in abundance between individuals with and without type 2 diabetes. CONCLUSION Our study identifies APOC1 as a dominant nutritional responder in humans and highlights the interdependency of acute nutritional response proteins and the endocrine system.
Collapse
Affiliation(s)
- Spyros I Vernardis
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
| | - Vadim Demichev
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Oliver Lemke
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Nana-Maria Grüning
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Christoph Messner
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
| | - Matt White
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, CB2 0SL, UK
- Computational Medicine, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Alina Peluso
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
| | - Tinh-Hai Collet
- Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
- Service of Endocrinology, Diabetology, Nutrition and Therapeutic Education, Department of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland
| | - Elana Henning
- Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Christoph Gille
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Michael Mülleder
- Core Facility High Throughput Mass Spectrometry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Aleksej Zelezniak
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius SE-412 96, Lithuania
- Randall Centre for Cell & Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, SE1 1UL London, UK
| | | | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, CB2 0SL, UK
- Computational Medicine, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, E1 1HH, UK
| | - I Sadaf Farooqi
- Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Markus Ralser
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
19
|
Sherlock L, Martin BR, Behsangar S, Mok KH. Application of novel AI-based algorithms to biobank data: uncovering of new features and linear relationships. Front Med (Lausanne) 2023; 10:1162808. [PMID: 37521348 PMCID: PMC10373878 DOI: 10.3389/fmed.2023.1162808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/16/2023] [Indexed: 08/01/2023] Open
Abstract
We independently analyzed two large public domain datasets that contain 1H-NMR spectral data from lung cancer and sex studies. The biobanks were sourced from the Karlsruhe Metabolomics and Nutrition (KarMeN) study and Bayesian Automated Metabolite Analyzer for NMR data (BATMAN) study. Our approach of applying novel artificial intelligence (AI)-based algorithms to NMR is an attempt to globalize metabolomics and demonstrate its clinical applications. The intention of this study was to analyze the resulting spectra in the biobanks via AI application to demonstrate its clinical applications. This technique enables metabolite mapping in areas of localized enrichment as a measure of true activity while also allowing for the accurate categorization of phenotypes.
Collapse
Affiliation(s)
- Lee Sherlock
- Meta-Flux Ltd., Dublin, Ireland
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry and Immunology, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | | | | | - K. H. Mok
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry and Immunology, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| |
Collapse
|
20
|
Opoku R, DeCata J, Phillips CL, Schulz LC. Effect of Genetically Reduced Maternal Myostatin on Late Gestation Maternal, Fetal, and Placental Metabolomes in Mice. Metabolites 2023; 13:719. [PMID: 37367877 PMCID: PMC10302353 DOI: 10.3390/metabo13060719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
Myostatin (gene symbol: Mstn) is an autocrine and paracrine inhibitor of muscle growth. Pregnant mice with genetically reduced levels of myostatin give birth to offspring with greater adult muscle mass and bone biomechanical strength. However, maternal myostatin is not detectable in fetal circulations. Fetal growth is dependent on the maternal environment, and the provisioning of nutrients and growth factors by the placenta. Thus, this study examined the effect of reduced maternal myostatin on maternal and fetal serum metabolomes, as well as the placental metabolome. Fetal and maternal serum metabolomes were highly distinct, which is consistent with the role of the placenta in creating a specific fetal nutrient environment. There was no effect from myostatin on maternal glucose tolerance or fasting insulin. In comparisons between pregnant control and Mstn+/- mice, there were more significantly different metabolite concentrations in fetal serum, at 50, than in the mother's serum at 33, confirming the effect of maternal myostatin reduction on the fetal metabolic milieu. Polyamines, lysophospholipids, fatty acid oxidation, and vitamin C, in fetal serum, were all affected by maternal myostatin reduction.
Collapse
Affiliation(s)
- Ruth Opoku
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA; (R.O.); (J.D.)
| | - Jenna DeCata
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA; (R.O.); (J.D.)
| | | | - Laura C. Schulz
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
21
|
Dai HD, Qiu F, Jackson K, Fruttiger M, Rizzo WB. Untargeted Metabolomic Analysis of Sjögren-Larsson Syndrome Reveals a Distinctive Pattern of Multiple Disrupted Biochemical Pathways. Metabolites 2023; 13:682. [PMID: 37367841 DOI: 10.3390/metabo13060682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023] Open
Abstract
Sjögren-Larsson syndrome (SLS) is a rare inherited neurocutaneous disease characterized by ichthyosis, spastic diplegia or tetraplegia, intellectual disability and a distinctive retinopathy. SLS is caused by bi-allelic mutations in ALDH3A2, which codes for fatty aldehyde dehydrogenase (FALDH) and results in abnormal lipid metabolism. The biochemical abnormalities in SLS are not completely known, and the pathogenic mechanisms leading to symptoms are still unclear. To search for pathways that are perturbed in SLS, we performed untargeted metabolomic screening in 20 SLS subjects along with age- and sex-matched controls. Of 823 identified metabolites in plasma, 121 (14.7%) quantitatively differed in the overall SLS cohort from controls; 77 metabolites were decreased and 44 increased. Pathway analysis pointed to disrupted metabolism of sphingolipids, sterols, bile acids, glycogen, purines and certain amino acids such as tryptophan, aspartate and phenylalanine. Random forest analysis identified a unique metabolomic profile that had a predictive accuracy of 100% for discriminating SLS from controls. These results provide new insight into the abnormal biochemical pathways that likely contribute to disease in SLS and may constitute a biomarker panel for diagnosis and future therapeutic studies.
Collapse
Affiliation(s)
- Hongying Daisy Dai
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Fang Qiu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - William B Rizzo
- Department of Pediatrics and Child Health Research Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Children's Hospital & Medical Center, Omaha, NE 68114, USA
| |
Collapse
|
22
|
Lee WD, Liang L, AbuSalim J, Jankowski CS, Samarah LZ, Neinast MD, Rabinowitz JD. Impact of acute stress on murine metabolomics and metabolic flux. Proc Natl Acad Sci U S A 2023; 120:e2301215120. [PMID: 37186827 PMCID: PMC10214130 DOI: 10.1073/pnas.2301215120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Plasma metabolite concentrations and labeling enrichments are common measures of organismal metabolism. In mice, blood is often collected by tail snip sampling. Here, we systematically examined the effect of such sampling, relative to gold-standard sampling from an in-dwelling arterial catheter, on plasma metabolomics and stable isotope tracing. We find marked differences between the arterial and tail circulating metabolome, which arise from two major factors: handling stress and sampling site, whose effects were deconvoluted by taking a second arterial sample immediately after tail snip. Pyruvate and lactate were the most stress-sensitive plasma metabolites, rising ~14 and ~5-fold. Both acute handling stress and adrenergic agonists induce extensive, immediate production of lactate, and modest production of many other circulating metabolites, and we provide a reference set of mouse circulatory turnover fluxes with noninvasive arterial sampling to avoid such artifacts. Even in the absence of stress, lactate remains the highest flux circulating metabolite on a molar basis, and most glucose flux into the TCA cycle in fasted mice flows through circulating lactate. Thus, lactate is both a central player in unstressed mammalian metabolism and strongly produced in response to acute stress.
Collapse
Affiliation(s)
- Won Dong Lee
- Department of Chemistry, Princeton University, Princeton, NJ08544
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ08544
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ08544
| | - Lingfan Liang
- Department of Chemistry, Princeton University, Princeton, NJ08544
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ08544
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ08544
| | - Jenna AbuSalim
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ08544
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ08544
- Department of Molecular Biology, Princeton University, Princeton, NJ08544
| | - Connor S.R. Jankowski
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ08544
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ08544
- Department of Molecular Biology, Princeton University, Princeton, NJ08544
| | - Laith Z. Samarah
- Department of Chemistry, Princeton University, Princeton, NJ08544
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ08544
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ08544
| | - Michael D. Neinast
- Department of Chemistry, Princeton University, Princeton, NJ08544
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ08544
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ08544
| | - Joshua D. Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ08544
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ08544
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ08544
- Department of Molecular Biology, Princeton University, Princeton, NJ08544
| |
Collapse
|
23
|
Kolvatzis C, Tsakiridis I, Kalogiannidis IA, Tsakoumaki F, Kyrkou C, Dagklis T, Daniilidis A, Michaelidou AM, Athanasiadis A. Utilizing Amniotic Fluid Metabolomics to Monitor Fetal Well-Being: A Narrative Review of the Literature. Cureus 2023; 15:e36986. [PMID: 37139280 PMCID: PMC10150141 DOI: 10.7759/cureus.36986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2023] [Indexed: 04/03/2023] Open
Abstract
Fetal and perinatal periods are critical phases for long-term development. Early diagnosis of maternal complications is challenging due to the great complexity of these conditions. In recent years, amniotic fluid has risen in a prominent position in the latest efforts to describe and characterize prenatal development. Amniotic fluid may provide real-time information on fetal development and metabolism throughout pregnancy as substances from the placenta, fetal skin, lungs, gastric fluid, and urine are transferred between the mother and the fetus. Applying metabolomics to monitor fetal well-being, in such a context, could help in the understanding, diagnosis, and treatment of these conditions and is a promising area of research. This review shines a spotlight on recent amniotic fluid metabolomics studies and their methods as an interesting tool for the assessment of many conditions and the identification of biomarkers. Platforms in use, such as proton nuclear magnetic resonance (1H NMR) and ultra-high-performance liquid chromatography (UHPLC), have different merits, and a combinatorial approach could be valuable. Metabolomics may also be used in the quest for habitual diet-induced metabolic signals in amniotic fluid. Finally, analysis of amniotic fluid can provide information on exposure to exogenous substances by detecting the exact levels of metabolites carried to the fetus and associated metabolic effects.
Collapse
|
24
|
LeWitt PA, Li J, Wu KH, Lu M. Diagnostic metabolomic profiling of Parkinson's disease biospecimens. Neurobiol Dis 2023; 177:105962. [PMID: 36563791 DOI: 10.1016/j.nbd.2022.105962] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Reliable and sensitive biomarkers are needed for enhancing and predicting Parkinson's disease (PD) diagnosis. OBJECTIVE To investigate comprehensive metabolomic profiling of biochemicals in CSF and serum for determining diagnostic biomarkers of PD. METHODS Fifty subjects, symptomatic with PD for ≥5 years, were matched to 50 healthy controls (HCs). We used ultrahigh-performance liquid chromatography linked to tandem mass spectrometry (UHPLC-MS/MS) for measuring relative concentrations of ≤1.5 kDalton biochemicals. A reference library created from authentic standards facilitated chemical identifications. Analytes underwent univariate analysis for PD association, with false discovery rate-adjusted p-value (≤0.05) determinations. Multivariate analysis (for identifying a panel of biochemicals discriminating PD from HCs) used several biostatistical methods, including logistic LASSO regression. RESULTS Comparing PD and HCs, strong differentiation was achieved from CSF but not serum specimens. With univariate analysis, 21 CSF compounds exhibited significant differential concentrations. Logistic LASSO regression led to selection of 23 biochemicals (11 shared with those determined by the univariate analysis). The selected compounds, as a group, distinguished PD from HCs, with Area-Under-the-Receiver-Operating-Characteristic (ROC) curve of 0.897. With optimal cutoff, logistic LASSO achieved 100% sensitivity and 96% specificity (and positive and negative predictive values of 96% and 100%). Ten-fold cross-validation gave 84% sensitivity and 82% specificity (and 82% positive and 84% negative predictive values). From the logistic LASSO-chosen regression model, 2 polyamine metabolites (N-acetylcadaverine and N-acetylputrescine) were chosen and had the highest fold-changes in comparing PD to HCs. Another chosen biochemical, acisoga (N-(3-acetamidopropyl)pyrrolidine-2-one), also is a polyamine metabolism derivative. CONCLUSIONS UHPLC-MS/MS assays provided a metabolomic signature highly predictive of PD. These findings provide further evidence for involvement of polyamine pathways in the neurodegeneration of PD.
Collapse
Affiliation(s)
- Peter A LeWitt
- Departments of Neurology, Henry Ford Hospital, West Bloomfield, MI, USA; Wayne State University School of Medicine, West Bloomfield, MI, USA.
| | - Jia Li
- The Department of Public Health Science, Henry Ford Health System, Detroit, MI, USA
| | - Kuan-Han Wu
- The Department of Public Health Science, Henry Ford Health System, Detroit, MI, USA
| | - Mei Lu
- The Department of Public Health Science, Henry Ford Health System, Detroit, MI, USA
| |
Collapse
|
25
|
Proteomics: Application of next-generation proteomics in cancer research. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00016-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
26
|
Wang H, Jia H, Gao Y, Zhang H, Fan J, Zhang L, Ren F, Yin Y, Cai Y, Zhu J, Zhu ZJ. Serum metabolic traits reveal therapeutic toxicities and responses of neoadjuvant chemoradiotherapy in patients with rectal cancer. Nat Commun 2022; 13:7802. [PMID: 36528604 PMCID: PMC9759530 DOI: 10.1038/s41467-022-35511-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Neoadjuvant chemoradiotherapy (nCRT) has become the standard treatment for patients with locally advanced rectal cancer (LARC). Therapeutic efficacy of nCRT is significantly affected by treatment-induced diarrhea and hematologic toxicities. Metabolic alternations in cancer therapy are key determinants to therapeutic toxicities and responses, but exploration in large-scale clinical studies remains limited. Here, we analyze 743 serum samples from 165 LARC patients recruited in a phase III clinical study using untargeted metabolomics and identify responsive metabolic traits over the course of nCRT. Pre-therapeutic serum metabolites successfully predict the chances of diarrhea and hematologic toxicities during nCRT. Particularly, levels of acyl carnitines are linked to sex disparity in nCRT-induced diarrhea. Finally, we show that differences in phenylalanine metabolism and essential amino acid metabolism may underlie distinct therapeutic responses of nCRT. This study illustrates the metabolic dynamics over the course of nCRT and provides potential to guide personalized nCRT treatment using responsive metabolic traits.
Collapse
Affiliation(s)
- Hongmiao Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huixun Jia
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- Department of Biostatistics, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yang Gao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haosong Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jin Fan
- Department of Biostatistics, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Lijie Zhang
- Department of Biostatistics, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Fandong Ren
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yandong Yin
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yuping Cai
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Ji Zhu
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310005, China.
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, 310000, China.
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China.
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200031, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, 201210, China.
| |
Collapse
|
27
|
Mahajan UM, Oehrle B, Sirtl S, Alnatsha A, Goni E, Regel I, Beyer G, Vornhülz M, Vielhauer J, Chromik A, Bahra M, Klein F, Uhl W, Fahlbusch T, Distler M, Weitz J, Grützmann R, Pilarsky C, Weiss FU, Adam MG, Neoptolemos JP, Kalthoff H, Rad R, Christiansen N, Bethan B, Kamlage B, Lerch MM, Mayerle J. Independent Validation and Assay Standardization of Improved Metabolic Biomarker Signature to Differentiate Pancreatic Ductal Adenocarcinoma From Chronic Pancreatitis. Gastroenterology 2022; 163:1407-1422. [PMID: 35870514 DOI: 10.1053/j.gastro.2022.07.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/28/2022] [Accepted: 07/14/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma cancer (PDAC) is a highly lethal malignancy requiring efficient detection when the primary tumor is still resectable. We previously developed the MxPancreasScore comprising 9 analytes and serum carbohydrate antigen 19-9 (CA19-9), achieving an accuracy of 90.6%. The necessity for 5 different analytical platforms and multiple analytical runs, however, hindered clinical applicability. We therefore aimed to develop a simpler single-analytical run, single-platform diagnostic signature. METHODS We evaluated 941 patients (PDAC, 356; chronic pancreatitis [CP], 304; nonpancreatic disease, 281) in 3 multicenter independent tests, and identification (ID) and validation cohort 1 (VD1) and 2 (VD2) were evaluated. Targeted quantitative plasma metabolite analysis was performed on a liquid chromatography-tandem mass spectrometry platform. A machine learning-aided algorithm identified an improved (i-Metabolic) and minimalistic metabolic (m-Metabolic) signatures, and compared them for performance. RESULTS The i-Metabolic Signature, (12 analytes plus CA19-9) distinguished PDAC from CP with area under the curve (95% confidence interval) of 97.2% (97.1%-97.3%), 93.5% (93.4%-93.7%), and 92.2% (92.1%-92.3%) in the ID, VD1, and VD2 cohorts, respectively. In the VD2 cohort, the m-Metabolic signature (4 analytes plus CA19-9) discriminated PDAC from CP with a sensitivity of 77.3% and specificity of 89.6%, with an overall accuracy of 82.4%. For the subset of 45 patients with PDAC with resectable stages IA-IIB tumors, the sensitivity, specificity, and accuracy were 73.2%, 89.6%, and 82.7%, respectively; for those with detectable CA19-9 >2 U/mL, 81.6%, 88.7%, and 84.5%, respectively; and for those with CA19-9 <37 U/mL, 39.7%, 94.1%, and 76.3%, respectively. CONCLUSIONS The single-platform, single-run, m-Metabolic signature of just 4 metabolites used in combination with serum CA19-9 levels is an innovative accurate diagnostic tool for PDAC at the time of clinical presentation, warranting further large-scale evaluation.
Collapse
Affiliation(s)
- Ujjwal M Mahajan
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany
| | - Bettina Oehrle
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany
| | - Simon Sirtl
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany
| | - Ahmed Alnatsha
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany
| | - Elisabetta Goni
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany
| | - Ivonne Regel
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany
| | - Georg Beyer
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany
| | - Marlies Vornhülz
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany
| | - Jakob Vielhauer
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany
| | - Ansgar Chromik
- Department of General and Visceral Surgery, Asklepios Klinikum Hamburg, Hamburg, Germany
| | - Markus Bahra
- Zentrum für Onkologische Oberbauchchirurgie und Robotik, Krankenhaus Waldfriede, Berlin, Germany
| | - Fritz Klein
- Department of General, Visceral and Transplantation Surgery, Charité, Campus Virchow Klinikum, Berlin, Germany
| | - Waldemar Uhl
- Department of General and Visceral Surgery, Katholisches Klinikum Bochum, Bochum, Germany
| | - Tim Fahlbusch
- Department of General and Visceral Surgery, Katholisches Klinikum Bochum, Bochum, Germany
| | - Marius Distler
- Department for Visceral, Thoracic and Vascular Surgery, University Hospital, Technical University Dresden, Dresden, Germany
| | - Jürgen Weitz
- Department for Visceral, Thoracic and Vascular Surgery, University Hospital, Technical University Dresden, Dresden, Germany
| | - Robert Grützmann
- Department of Surgery, Erlangen University Hospital, Erlangen, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Erlangen, Germany
| | - Christian Pilarsky
- Department of Surgery, Erlangen University Hospital, Erlangen, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Erlangen, Germany
| | - Frank Ulrich Weiss
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - M Gordian Adam
- Metanomics Health GmbH, Berlin, Germany; biocrates life sciences ag, Innsbruck, Austria
| | - John P Neoptolemos
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Holger Kalthoff
- Section for Molecular Oncology, Institut for Experimental Cancer Research (IET), Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Roland Rad
- Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany; Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine and Center for Translational Cancer Research (TranslaTUM), Technische Universität München, Munich, Germany
| | - Nicole Christiansen
- Metanomics Health GmbH, Berlin, Germany; TrinamiX GmbH, Ludwigshafen am Rhein, Rheinland-Pfalz, Germany
| | | | | | - Markus M Lerch
- Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany; Department of Medicine A, University Medicine Greifswald, Greifswald, Germany; Ludwig Maximilian University Klinikum, Munich, Germany
| | - Julia Mayerle
- Department of Medicine II, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany; Bavarian Centre for Cancer Research (Bayerisches Zentrum für Krebsforschung), Munich, Germany.
| |
Collapse
|
28
|
Role of Intestinal Microbes in Chronic Liver Diseases. Int J Mol Sci 2022; 23:ijms232012661. [PMID: 36293518 PMCID: PMC9603943 DOI: 10.3390/ijms232012661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/15/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022] Open
Abstract
With the recent availability and upgrading of many emerging intestinal microbes sequencing technologies, our research on intestinal microbes is changing rapidly. A variety of investigations have found that intestinal microbes are essential for immune system regulation and energy metabolism homeostasis, which impacts many critical organs. The liver is the first organ to be traversed by the intestinal portal vein, and there is a strong bidirectional link between the liver and intestine. Many intestinal factors, such as intestinal microbes, bacterial composition, and intestinal bacterial metabolites, are deeply involved in liver homeostasis. Intestinal microbial dysbiosis and increased intestinal permeability are associated with the pathogenesis of many chronic liver diseases, such as alcoholic fatty liver disease (AFLD), non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), chronic hepatitis B (CHB), chronic hepatitis C (CHC), autoimmune liver disease (AIH) and the development of hepatocellular carcinoma (HCC). Intestinal permeability and dysbacteriosis often lead to Lipopolysaccharide (LPS) and metabolites entering in serum. Then, Toll-like receptors activation in the liver induces the exposure of the intestine and liver to many small molecules with pro-inflammatory properties. And all of these eventually result in various liver diseases. In this paper, we have discussed the current evidence on the role of various intestinal microbes in different chronic liver diseases. As well as potential new therapeutic approaches are proposed in this review, such as antibiotics, probiotics, and prebiotics, which may have an improvement in liver diseases.
Collapse
|
29
|
Xiang H, Chen H, Liu Y, Dodd D, Pao AC. Role of insulin resistance and the gut microbiome on urine oxalate excretion in ob/ob mice. Physiol Rep 2022; 10:e15357. [PMID: 35851836 PMCID: PMC9294392 DOI: 10.14814/phy2.15357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023] Open
Abstract
Ob/ob mice have recently emerged as a model for obesity-related hyperoxaluria as they are obese and excrete more urine oxalate compared to wild type mice. Ob/ob mice are deficient of leptin and develop obesity with hyperphagia and hyperinsulinemia. We hypothesized that insulin resistance and the gut microbiome contribute to hyperoxaluria in ob/ob mice. We developed a new liquid chromatography-mass spectrometry assay for urine oxalate and first compared urine oxalate excretion in ob/ob mice before and after ablation of intestinal bacteria with a standard antibiotic cocktail. We then compared urine oxalate excretion in ob/ob mice before and after leptin replacement or pioglitazone treatment, two maneuvers that reduce insulin resistance in ob/ob mice. Ob/ob mice excreted more oxalate into the urine in a 24-h period compared to wild type mice, but antibiotic, leptin, or pioglitazone treatment did not change urine oxalate excretion in ob/ob mice. Unexpectedly, we found that when food intake was carefully matched between ob/ob and wild type mice, the amount of 24-h urine oxalate excretion did not differ between the two mouse strains, suggesting that ob/ob mice excrete more urine oxalate because of hyperphagia. Since the level of urine oxalate excretion in wild type mice in our study was higher than those reported in prior studies, future work will be needed to standardize the measurement of urine oxalate and to define the range of urine oxalate excretion in wild type mice so that accurate and valid comparisons can be made between wild type mice and ob/ob mice or other mouse models.
Collapse
Affiliation(s)
- Hong Xiang
- Division of Nephrology, Department of MedicineStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Haoqing Chen
- Department of PathologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Yuanyuan Liu
- Department of PathologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Dylan Dodd
- Department of PathologyStanford University School of MedicineStanfordCaliforniaUSA
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Alan C. Pao
- Division of Nephrology, Department of MedicineStanford University School of MedicinePalo AltoCaliforniaUSA
- Department of UrologyStanford University School of MedicinePalo AltoCaliforniaUSA
- Veterans Affairs Palo Alto Health Care SystemPalo AltoCaliforniaUSA
| |
Collapse
|
30
|
Di Minno A, Gelzo M, Caterino M, Costanzo M, Ruoppolo M, Castaldo G. Challenges in Metabolomics-Based Tests, Biomarkers Revealed by Metabolomic Analysis, and the Promise of the Application of Metabolomics in Precision Medicine. Int J Mol Sci 2022; 23:5213. [PMID: 35563604 PMCID: PMC9103094 DOI: 10.3390/ijms23095213] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
Metabolomics helps identify metabolites to characterize/refine perturbations of biological pathways in living organisms. Pre-analytical, analytical, and post-analytical limitations that have hampered a wide implementation of metabolomics have been addressed. Several potential biomarkers originating from current targeted metabolomics-based approaches have been discovered. Precision medicine argues for algorithms to classify individuals based on susceptibility to disease, and/or by response to specific treatments. It also argues for a prevention-based health system. Because of its ability to explore gene-environment interactions, metabolomics is expected to be critical to personalize diagnosis and treatment. Stringent guidelines have been applied from the very beginning to design studies to acquire the information currently employed in precision medicine and precision prevention approaches. Large, prospective, expensive and time-consuming studies are now mandatory to validate old, and discover new, metabolomics-based biomarkers with high chances of translation into precision medicine. Metabolites from studies on saliva, sweat, breath, semen, feces, amniotic, cerebrospinal, and broncho-alveolar fluid are predicted to be needed to refine information from plasma and serum metabolome. In addition, a multi-omics data analysis system is predicted to be needed for omics-based precision medicine approaches. Omics-based approaches for the progress of precision medicine and prevention are expected to raise ethical issues.
Collapse
Affiliation(s)
- Alessandro Di Minno
- Dipartimento di Farmacia, University of Naples Federico II, 80131 Naples, Italy
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
| | - Monica Gelzo
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Marianna Caterino
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Michele Costanzo
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Margherita Ruoppolo
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
31
|
Wang Y, Juan L, Peng J, Wang T, Zang T, Wang Y. Explore potential disease related metabolites based on latent factor model. BMC Genomics 2022; 23:269. [PMID: 35387615 PMCID: PMC8985251 DOI: 10.1186/s12864-022-08504-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/17/2022] Open
Abstract
Background In biological systems, metabolomics can not only contribute to the discovery of metabolic signatures for disease diagnosis, but is very helpful to illustrate the underlying molecular disease-causing mechanism. Therefore, identification of disease-related metabolites is of great significance for comprehensively understanding the pathogenesis of diseases and improving clinical medicine. Results In the paper, we propose a disease and literature driven metabolism prediction model (DLMPM) to identify the potential associations between metabolites and diseases based on latent factor model. We build the disease glossary with disease terms from different databases and an association matrix based on the mapping between diseases and metabolites. The similarity of diseases and metabolites is used to complete the association matrix. Finally, we predict potential associations between metabolites and diseases based on the matrix decomposition method. In total, 1,406 direct associations between diseases and metabolites are found. There are 119,206 unknown associations between diseases and metabolites predicted with a coverage rate of 80.88%. Subsequently, we extract training sets and testing sets based on data increment from the database of disease-related metabolites and assess the performance of DLMPM on 19 diseases. As a result, DLMPM is proven to be successful in predicting potential metabolic signatures for human diseases with an average AUC value of 82.33%. Conclusion In this paper, a computational model is proposed for exploring metabolite-disease pairs and has good performance in predicting potential metabolites related to diseases through adequate validation. The results show that DLMPM has a better performance in prioritizing candidate diseases-related metabolites compared with the previous methods and would be helpful for researchers to reveal more information about human diseases.
Collapse
Affiliation(s)
- Yongtian Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China. .,Key Laboratory of Big Data Storage and Management Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, China.
| | - Liran Juan
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China.,Key Laboratory of Big Data Storage and Management Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, China
| | - Tao Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China.,Key Laboratory of Big Data Storage and Management Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, China
| | - Tianyi Zang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China.
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
32
|
Wang Y, Shu W, Lin S, Wu J, Jiang M, Li S, Liu C, Li R, Pei C, Ding Y, Wan J, Di W. Hollow Cobalt Oxide/Carbon Hybrids Aid Metabolic Encoding for Active Systemic Lupus Erythematosus during Pregnancy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106412. [PMID: 35064740 DOI: 10.1002/smll.202106412] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/16/2021] [Indexed: 06/14/2023]
Abstract
A noninvasive, easy operation, and accurate diagnostic protocol is highly demanded to assess systemic lupus erythematosus (SLE) activity during pregnancy, promising real-time activity monitoring during the whole gestational period to reduce adverse pregnancy outcomes. Here, machine learning of serum metabolic fingerprints (SMFs) is developed to assess the SLE activity for pregnant women. The SMFs are directly extracted through a hollow-cobalt oxide/carbon (Co3 O4 /C)-composite-assisted laser desorption/ionization mass spectrometer (LDI MS) platform. The Co3 O4 /C composite owns enhanced light absorption, size-selective trapping, and better charge-hole separation, enabling improved ionization efficiency and selectivity for LDI MS detection toward small molecules. Metabolic fingerprints are collected from ≈0.1 µL serum within 1 s without enrichment and encoded by the optimized elastic net algorithm. The averaged area under the curve (AUC) value in the differentiation of active SLE from inactive SLE and healthy controls reaches 0.985 and 0.990, respectively. Further, a simplified panel based on four identified metabolites is built to distinguish SLE flares in pregnant women with the highest AUC value of 0.875 for the blind test. This work sets an accurate and practical protocol for SLE activity assessment during pregnancy, promoting precision diagnosis of disease status transitions in clinics.
Collapse
Affiliation(s)
- You Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
| | - Weikang Shu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Sihan Lin
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
| | - Jiayue Wu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
| | - Meng Jiang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
| | - Shumin Li
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Chao Liu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Rongxin Li
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Congcong Pei
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Yajie Ding
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Jingjing Wan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Wen Di
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, P. R. China
| |
Collapse
|
33
|
Durán AM, Beeson WL, Firek A, Cordero-MacIntyre Z, De León M. Dietary Omega-3 Polyunsaturated Fatty-Acid Supplementation Upregulates Protective Cellular Pathways in Patients with Type 2 Diabetes Exhibiting Improvement in Painful Diabetic Neuropathy. Nutrients 2022; 14:nu14040761. [PMID: 35215418 PMCID: PMC8876723 DOI: 10.3390/nu14040761] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Omega-3 polyunsaturated fatty acids (PUFAs) have been proposed to improve chronic neuroinflammatory diseases in peripheral and central nervous systems. For instance, docosahexaenoic acid (DHA) protects nerve cells from noxious stimuli in vitro and in vivo. Recent reports link PUFA supplementation to improving painful diabetic neuropathy (pDN) symptoms, but cellular mechanisms responsible for this therapeutic effect are not well understood. The objective of this study is to identify distinct cellular pathways elicited by dietary omega-3 PUFA supplementation in patients with type 2 diabetes mellitus (T2DM) affected by pDN. Methods: Forty volunteers diagnosed with type 2 diabetes were enrolled in the “En Balance-PLUS” diabetes education study. The volunteers participated in weekly lifestyle/nutrition education and daily supplementation with 1000 mg DHA and 200 mg eicosapentaenoic acid. The Short-Form McGill Pain Questionnaire validated clinical determination of baseline and post-intervention pain complaints. Laboratory and untargeted metabolomics analyses were conducted using blood plasma collected at baseline and after three months of participation in the dietary regimen. The metabolomics data were analyzed using random forest, hierarchical clustering, ingenuity pathway analysis, and metabolic pathway mapping. Results: The data show that metabolites involved in oxidative stress and glutathione production shifted significantly to a more anti-inflammatory state post supplementation. Example of these metabolites include cystathionine (+90%), S-methylmethionine (+9%), glycine cysteine-glutathione disulfide (+157%) cysteinylglycine (+19%), glutamate (−11%), glycine (+11%), and arginine (+13.4%). In addition, the levels of phospholipids associated with improved membrane fluidity such as linoleoyl-docosahexaenoyl-glycerol (18:2/22:6) (+253%) were significantly increased. Ingenuity pathway analysis suggested several key bio functions associated with omega-3 PUFA supplementation such as formation of reactive oxygen species (p = 4.38 × 10−4, z-score = −1.96), peroxidation of lipids (p = 2.24 × 10−5, z-score = −1.944), Ca2+ transport (p = 1.55 × 10−4, z-score = −1.969), excitation of neurons (p = 1.07 ×10−4, z-score = −1.091), and concentration of glutathione (p = 3.06 × 10−4, z-score = 1.974). Conclusion: The reduction of pro-inflammatory and oxidative stress pathways following dietary omega-3 PUFA supplementation is consistent with the promising role of these fatty acids in reducing adverse symptoms associated with neuroinflammatory diseases and painful neuropathy.
Collapse
Affiliation(s)
- Alfonso M. Durán
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
| | - W. Lawrence Beeson
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA 92350, USA
| | - Anthony Firek
- Comparative Effectiveness and Clinical Outcomes Research Center, Riverside University Health System Medical Center, Moreno Valley, CA 92555, USA;
| | - Zaida Cordero-MacIntyre
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA 92350, USA
| | - Marino De León
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
- Correspondence: ; Tel.: +1-909-558-9474
| |
Collapse
|
34
|
Brunmair J, Gotsmy M, Niederstaetter L, Neuditschko B, Bileck A, Slany A, Feuerstein ML, Langbauer C, Janker L, Zanghellini J, Meier-Menches SM, Gerner C. Finger sweat analysis enables short interval metabolic biomonitoring in humans. Nat Commun 2021; 12:5993. [PMID: 34645808 PMCID: PMC8514494 DOI: 10.1038/s41467-021-26245-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 09/22/2021] [Indexed: 01/28/2023] Open
Abstract
Metabolic biomonitoring in humans is typically based on the sampling of blood, plasma or urine. Although established in the clinical routine, these sampling procedures are often associated with a variety of compliance issues, which are impeding time-course studies. Here, we show that the metabolic profiling of the minute amounts of sweat sampled from fingertips addresses this challenge. Sweat sampling from fingertips is non-invasive, robust and can be accomplished repeatedly by untrained personnel. The sweat matrix represents a rich source for metabolic phenotyping. We confirm the feasibility of short interval sampling of sweat from the fingertips in time-course studies involving the consumption of coffee or the ingestion of a caffeine capsule after a fasting interval, in which we successfully monitor all known caffeine metabolites as well as endogenous metabolic responses. Fluctuations in the rate of sweat production are accounted for by mathematical modelling to reveal individual rates of caffeine uptake, metabolism and clearance. To conclude, metabotyping using sweat from fingertips combined with mathematical network modelling shows promise for broad applications in precision medicine by enabling the assessment of dynamic metabolic patterns, which may overcome the limitations of purely compositional biomarkers.
Collapse
Affiliation(s)
- Julia Brunmair
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Mathias Gotsmy
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Laura Niederstaetter
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Benjamin Neuditschko
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University and Medical University of Vienna, Vienna, Austria
| | - Astrid Slany
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Max Lennart Feuerstein
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Clemens Langbauer
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University and Medical University of Vienna, Vienna, Austria
| | - Jürgen Zanghellini
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Samuel M Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University and Medical University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria.
- Joint Metabolome Facility, University and Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
35
|
Santiago-Rodriguez TM, Hollister EB. Multi 'omic data integration: A review of concepts, considerations, and approaches. Semin Perinatol 2021; 45:151456. [PMID: 34256961 DOI: 10.1016/j.semperi.2021.151456] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The application of 'omic techniques including, but not limited to genomics/metagenomics, transcriptomics/meta-transcriptomics, proteomics/meta-proteomics, and metabolomics to generate multiple datasets from a single sample have facilitated hypothesis generation leading to the identification of biological, molecular and ecological functions and mechanisms, as well as associations and correlations. Despite their power and promise, a variety of challenges must be considered in the successful design and execution of a multi-omics study. In this review, various 'omic technologies applicable to single- and meta-organisms (i.e., host + microbiome) are described, and considerations for sample collection, storage and processing prior to data generation and analysis, as well as approaches to data storage, dissemination and analysis are discussed. Finally, case studies are included as examples of multi-omic applications providing novel insights and a more holistic understanding of biological processes.
Collapse
Affiliation(s)
| | - Emily B Hollister
- Diversigen, Inc, 3 Greenway Plaza, Suite 1575, Houston, TX 77046, USA.
| |
Collapse
|
36
|
Gao Y, Hou L, Gao J, Li D, Tian Z, Fan B, Wang F, Li S. Metabolomics Approaches for the Comprehensive Evaluation of Fermented Foods: A Review. Foods 2021; 10:2294. [PMID: 34681343 PMCID: PMC8534989 DOI: 10.3390/foods10102294] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Fermentation is an important process that can provide new flavors and nutritional and functional foods, to deal with changing consumer preferences. Fermented foods have complex chemical components that can modulate unique qualitative properties. Consequently, monitoring the small molecular metabolites in fermented food is critical to clarify its qualitative properties and help deliver personalized nutrition. In recent years, the application of metabolomics to nutrition research of fermented foods has expanded. In this review, we examine the application of metabolomics technologies in food, with a primary focus on the different analytical approaches suitable for food metabolomics and discuss the advantages and disadvantages of these approaches. In addition, we summarize emerging studies applying metabolomics in the comprehensive analysis of the flavor, nutrition, function, and safety of fermented foods, as well as emphasize the applicability of metabolomics in characterizing the qualitative properties of fermented foods.
Collapse
Affiliation(s)
- Yaxin Gao
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China; (Y.G.); (L.H.); (J.G.); (D.L.); (Z.T.); (B.F.)
| | - Lizhen Hou
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China; (Y.G.); (L.H.); (J.G.); (D.L.); (Z.T.); (B.F.)
| | - Jie Gao
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China; (Y.G.); (L.H.); (J.G.); (D.L.); (Z.T.); (B.F.)
| | - Danfeng Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China; (Y.G.); (L.H.); (J.G.); (D.L.); (Z.T.); (B.F.)
| | - Zhiliang Tian
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China; (Y.G.); (L.H.); (J.G.); (D.L.); (Z.T.); (B.F.)
| | - Bei Fan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China; (Y.G.); (L.H.); (J.G.); (D.L.); (Z.T.); (B.F.)
- Key Laboratory of Agro-Products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Fengzhong Wang
- Key Laboratory of Agro-Products Quality and Safety Control in Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shuying Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China; (Y.G.); (L.H.); (J.G.); (D.L.); (Z.T.); (B.F.)
| |
Collapse
|
37
|
Effects of dexmedetomidine, propofol, sevoflurane and S-ketamine on the human metabolome: A randomised trial using nuclear magnetic resonance spectroscopy. Eur J Anaesthesiol 2021; 39:521-532. [PMID: 34534172 DOI: 10.1097/eja.0000000000001591] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Pharmacometabolomics uses large-scale data capturing methods to uncover drug-induced shifts in the metabolic profile. The specific effects of anaesthetics on the human metabolome are largely unknown. OBJECTIVE We aimed to discover whether exposure to routinely used anaesthetics have an acute effect on the human metabolic profile. DESIGN Randomised, open-label, controlled, parallel group, phase IV clinical drug trial. SETTING The study was conducted at Turku PET Centre, University of Turku, Finland, 2016 to 2017. PARTICIPANTS One hundred and sixty healthy male volunteers were recruited. The metabolomic data of 159 were evaluable. INTERVENTIONS Volunteers were randomised to receive a 1-h exposure to equipotent doses (EC50 for verbal command) of dexmedetomidine (1.5 ng ml-1; n = 40), propofol (1.7 μg ml-1; n = 40), sevoflurane (0.9% end-tidal; n = 39), S-ketamine (0.75 μg ml-1; n = 20) or placebo (n = 20). MAIN OUTCOME MEASURES Metabolite subgroups of apolipoproteins and lipoproteins, cholesterol, glycerides and phospholipids, fatty acids, glycolysis, amino acids, ketone bodies, creatinine and albumin and the inflammatory marker GlycA, were analysed with nuclear magnetic resonance spectroscopy from arterial blood samples collected at baseline, after anaesthetic administration and 70 min postanaesthesia. RESULTS All metabolite subgroups were affected. Statistically significant changes vs. placebo were observed in 11.0, 41.3, 0.65 and 3.9% of the 155 analytes in the dexmedetomidine, propofol, sevoflurane and S-ketamine groups, respectively. Dexmedetomidine increased glucose, decreased ketone bodies and affected lipoproteins and apolipoproteins. Propofol altered lipoproteins, fatty acids, glycerides and phospholipids and slightly increased inflammatory marker glycoprotein acetylation. Sevoflurane was relatively inert. S-ketamine increased glucose and lactate, whereas branched chain amino acids and tyrosine decreased. CONCLUSION A 1-h exposure to moderate doses of routinely used anaesthetics led to significant and characteristic alterations in the metabolic profile. Dexmedetomidine-induced alterations mirror α2-adrenoceptor agonism. Propofol emulsion altered the lipid profile. The inertness of sevoflurane might prove useful in vulnerable patients. S-ketamine induced amino acid alterations might be linked to its suggested antidepressive properties. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02624401. URL: https://clinicaltrials.gov/ct2/show/NCT02624401.
Collapse
|
38
|
Aladelokun O, Hanley M, Mu J, Giardina JC, Rosenberg DW, Giardina C. Fatty acid metabolism and colon cancer protection by dietary methyl donor restriction. Metabolomics 2021; 17:80. [PMID: 34480220 PMCID: PMC8416812 DOI: 10.1007/s11306-021-01831-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/19/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION A methyl donor depleted (MDD) diet dramatically suppresses intestinal tumor development in Apc-mutant mice, but the mechanism of this prevention is not entirely clear. OBJECTIVES We sought to gain insight into the mechanisms of cancer suppression by the MDD diet and to identify biomarkers of cancer risk reduction. METHODS A plasma metabolomic analysis was performed on ApcΔ14/+ mice maintained on either a methyl donor sufficient (MDS) diet or the protective MDD diet. A group of MDS animals was also pair-fed with the MDD mice to normalize caloric intake, and another group was shifted from an MDD to MDS diet to determine the durability of the metabolic changes. RESULTS In addition to the anticipated changes in folate one-carbon metabolites, plasma metabolites related to fatty acid metabolism were generally decreased by the MDD diet, including carnitine, acylcarnitines, and fatty acids. Some fatty acid selectivity was observed; the levels of cancer-promoting arachidonic acid and 2-hydroxyglutarate were decreased by the MDD diet, whereas eicosapentaenoic acid (EPA) levels were increased. Machine-learning elastic net analysis revealed a positive association between the fatty acid-related compounds azelate and 7-hydroxycholesterol and tumor development, and a negative correlation with succinate and β-sitosterol. CONCLUSION Methyl donor restriction causes dramatic changes in systemic fatty acid metabolism. Regulating fatty acid metabolism through methyl donor restriction favorably effects fatty acid profiles to achieve cancer protection.
Collapse
Affiliation(s)
- Oladimeji Aladelokun
- Center for Molecular Oncology, University of Connecticut Health Center, The University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT, 06030-3101, USA.
| | - Matthew Hanley
- Center for Molecular Oncology, University of Connecticut Health Center, The University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT, 06030-3101, USA
| | - Jinjian Mu
- Statistical Consulting Services, University of Connecticut, Storrs, CT, USA
| | - John C Giardina
- Center for Health Decision Science, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Daniel W Rosenberg
- Center for Molecular Oncology, University of Connecticut Health Center, The University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT, 06030-3101, USA
| | - Charles Giardina
- Department of Molecular and Cellular Biology, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
39
|
Ganguly S, Finkelstein D, Shaw TI, Michalek RD, Zorn KM, Ekins S, Yasuda K, Fukuda Y, Schuetz JD, Mukherjee K, Schuetz EG. Metabolomic and transcriptomic analysis reveals endogenous substrates and metabolic adaptation in rats lacking Abcg2 and Abcb1a transporters. PLoS One 2021; 16:e0253852. [PMID: 34255797 PMCID: PMC8277073 DOI: 10.1371/journal.pone.0253852] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/14/2021] [Indexed: 12/21/2022] Open
Abstract
Abcg2/Bcrp and Abcb1a/Pgp are xenobiotic efflux transporters limiting substrate permeability in the gastrointestinal system and brain, and increasing renal and hepatic drug clearance. The systemic impact of Bcrp and Pgp ablation on metabolic homeostasis of endogenous substrates is incompletely understood. We performed untargeted metabolomics of cerebrospinal fluid (CSF) and plasma, transcriptomics of brain, liver and kidney from male Sprague Dawley rats (WT) and Bcrp/Pgp double knock-out (dKO) rats, and integrated metabolomic/transcriptomic analysis to identify putative substrates and perturbations in canonical metabolic pathways. A predictive Bayesian machine learning model was used to predict in silico those metabolites with greater substrate-like features for either transporters. The CSF and plasma levels of 169 metabolites, nutrients, signaling molecules, antioxidants and lipids were significantly altered in dKO rats, compared to WT rats. These metabolite changes suggested alterations in histidine, branched chain amino acid, purine and pyrimidine metabolism in the dKO rats. Levels of methylated and sulfated metabolites and some primary bile acids were increased in dKO CSF or plasma. Elevated uric acid levels appeared to be a primary driver of changes in purine and pyrimidine biosynthesis. Alterations in Bcrp/Pgp dKO CSF levels of antioxidants, precursors of neurotransmitters, and uric acid suggests the transporters may contribute to the regulation of a healthy central nervous system in rats. Microbiome-generated metabolites were found to be elevated in dKO rat plasma and CSF. The altered dKO metabolome appeared to cause compensatory transcriptional change in urate biosynthesis and response to lipopolysaccharide in brain, oxidation-reduction processes and response to oxidative stress and porphyrin biosynthesis in kidney, and circadian rhythm genes in liver. These findings present insight into endogenous functions of Bcrp and Pgp, the impact that transporter substrates, inhibitors or polymorphisms may have on metabolism, how transporter inhibition could rewire drug sensitivity indirectly through metabolic changes, and identify functional Bcrp biomarkers.
Collapse
Affiliation(s)
- Samit Ganguly
- Cancer & Developmental Biology Track, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Timothy I. Shaw
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | | | - Kimberly M. Zorn
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina, United States of America
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina, United States of America
| | - Kazuto Yasuda
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Yu Fukuda
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - John D. Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Kamalika Mukherjee
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Erin G. Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
40
|
Rivera-Velez SM, Navas J, Villarino NF. Applying metabolomics to veterinary pharmacology and therapeutics. J Vet Pharmacol Ther 2021; 44:855-869. [PMID: 33719079 DOI: 10.1111/jvp.12961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Metabolomics is the large-scale study of low-molecular-weight substances in a biological system in a given physiological state at a given time point. Metabolomics can be applied to identify predictors of inter-individual variability in drug response, provide clinicians with data useful for decision-making processes in drug selection, and inform about the pharmacokinetics and pharmacodynamics of a drug. It is, therefore, an exceptional approach for gaining new understanding effects in the field of comparative veterinary pharmacology. However, the incorporation of metabolomics into veterinary pharmacology and toxicology is not yet widespread, and this is probably, at least in part, a result of its highly multidisciplinary nature. This article reviews the potential applications of metabolomics in veterinary pharmacology and therapeutics. It integrates key concepts for designing metabolomics studies and analyzing and interpreting metabolomics data, providing solid foundations for applying metabolomics to the study of drugs in all veterinary species.
Collapse
Affiliation(s)
- Sol M Rivera-Velez
- Molecular Determinants Core, Johns Hopkins All Children's Hospital, Saint Petersburg, Florida, USA
| | - Jinna Navas
- Program in Individualized Medicine, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Nicolas F Villarino
- Program in Individualized Medicine, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
41
|
Li GB, Hu HR, Pan WF, Li B, Ou ZY, Liang HY, Li C. Plasma Metabolic Profiling of Pediatric Sepsis in a Chinese Cohort. Front Cell Dev Biol 2021; 9:643979. [PMID: 33659257 PMCID: PMC7917179 DOI: 10.3389/fcell.2021.643979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 01/15/2021] [Indexed: 11/13/2022] Open
Abstract
Sepsis represents one of the most pressing problems in pediatrics, characterized by pathogenic bacteria invading the blood, growing and multiplying in the blood circulation, and ultimately causing severe infections. Most children with sepsis have a rapid disease onset and frequently exhibit sudden high fever or first chills. Here we performed comprehensive metabolomic profiling of plasma samples collected from pediatric sepsis patients to identify specific metabolic alterations associated with these patients (n = 84, designated as case subjects) as compared to healthy cohorts (n = 59, designated as control subjects). Diagnostic models were constructed using MetaboAnalyst, R packages, and multiple statistical methods, such as orthogonal partial least squares-discriminant analysis, principal component analysis, volcano plotting, and one-way ANOVA. Our study revealed a panel of metabolites responsible for the discrimination between case and control subjects with a high predictive value of prognosis. Moreover, significantly altered metabolites in sepsis survivors versus deceased patients (non-survivors) were identified as those involved in amino acids, fatty acids, and carbohydrates metabolism. Nine metabolites including organic acids and fatty acids were also identified with significantly higher abundance in sepsis patients with related microbes, implicating greater potentials to distinguish bacterial species using metabolomic analysis than blood culture. Pathway enrichment analysis further revealed that fatty acid metabolism might play an important role in the pathogenesis of sepsis.
Collapse
Affiliation(s)
- Guo-Bang Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hong-Rong Hu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wen-Feng Pan
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bo Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Ying Ou
- Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hui-Ying Liang
- Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Cong Li
- Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Central Laboratory, Affiliated Dongguan People's Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
42
|
Metabolomic analysis of uremic pruritus in patients on hemodialysis. PLoS One 2021; 16:e0246765. [PMID: 33577623 PMCID: PMC7880487 DOI: 10.1371/journal.pone.0246765] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Pruritus is a common debilitating symptom experienced by hemodialysis patients. Treatment is difficult because the cause of uremic pruritus is not known. This study addressed the hypothesis that pruritus is caused by solutes that accumulate in the plasma when the kidneys fail. We sought to identify solutes responsible for uremic pruritus using metabolomic analysis to compare the plasma of hemodialysis patients with severe pruritus versus mild/no pruritus. Pruritus severity in hemodialysis patients was assessed using a 100-mm visual analogue scale (VAS), with severe pruritus defined as >70 mm and mild/no pruritus defined as <10 mm. Twelve patients with severe pruritus (Itch) and 24 patients with mild/no pruritus (No Itch) were included. Pre-treatment plasma and plasma ultrafiltrate were analyzed using an established metabolomic platform (Metabolon, Inc.). To identify solutes associated with pruritus, we compared the average peak area of each solute in the Itch patients to that of the No Itch patients using the false discovery rate (q value) and principal component analysis. Dialysis vintage, Kt/Vurea, and serum levels of calcium, phosphorus, PTH, albumin, ferritin, and hemoglobin were similar in the Itch and No Itch patients. Metabolomic analysis identified 1,548 solutes of which 609 were classified as uremic. No difference in the plasma or plasma ultrafiltrate levels of any solute or group of solutes was found between the Itch and No Itch patients. Metabolomic analysis of hemodialysis patients did not reveal any solutes associated with pruritus. A limitation of metabolomic analysis is that the solute of interest may not be included in the metabolomic platform’s chemical library. A role for uremic solutes in pruritus remains to be established.
Collapse
|
43
|
Abstract
Neurological disorders, including neurodegenerative diseases, have a significant negative impact on both patients and society at large. Since the prevalence of most of these disorders increases with age, the consequences for our aging population are only going to grow. It is now acknowledged that neurological disorders are multi-factorial involving disruptions in multiple cellular systems. While each disorder has specific initiating mechanisms and pathologies, certain common pathways appear to be involved in most, if not all, neurological disorders. Thus, it is becoming increasingly important to identify compounds that can modulate the multiple pathways that contribute to disease development or progression. One of these compounds is the flavonol fisetin. Fisetin has now been shown in preclinical models to be effective at preventing the development and/or progression of multiple neurological disorders including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, stroke (both ischemic and hemorrhagic) and traumatic brain injury as well as to reduce age-associated changes in the brain. These beneficial effects stem from its actions on multiple pathways associated with the different neurological disorders. These actions include its well characterized anti-inflammatory and anti-oxidant effects as well as more recently described effects on the regulated cell death oxytosis/ferroptosis pathway, the gut microbiome and its senolytic activity. Therefore, the growing body of pre-clinical data, along with fisetin’s ability to modulate a large number of pathways associated with brain dysfunction, strongly suggest that it would be worthwhile to pursue its therapeutic effects in humans.
Collapse
Affiliation(s)
- Pamela Maher
- Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA
| |
Collapse
|
44
|
Barnes S. Overview of Experimental Methods and Study Design in Metabolomics, and Statistical and Pathway Considerations. Methods Mol Biol 2021; 2104:1-10. [PMID: 31953809 DOI: 10.1007/978-1-0716-0239-3_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Metabolomics has become a powerful tool in biological and clinical investigations. This chapter reviews the technological basis of metabolomics and the considerations in answering biomedical questions. The workflow of metabolomics is explained in the sequence of data processing, quality control, metabolite annotation, statistical analysis, pathway analysis, and multi-omics integration. Reproducibility in both sample analysis and data analysis is key to the scientific progress, and the recommendation is made on reporting standards in publications. This chapter explains the technical aspects of metabolomics in the context of systems biology and applications to human health.
Collapse
Affiliation(s)
- Stephen Barnes
- Department of Pharmacology & Toxicology and Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
45
|
Li Y, Yin W, Zhan Y, Jia Y, Cui D, Zhang W, Chang Y. Comparative metabolome analysis provides new insights into increased larval mortality under seawater acidification in the sea urchin Strongylocentrotus intermedius. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 747:141206. [PMID: 32777501 DOI: 10.1016/j.scitotenv.2020.141206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 06/11/2023]
Abstract
Mortality and metabolic responses of four-armed larvae of Strongylocentrotus intermedius under CO2-induced seawater acidification were investigated. Gametes of S. intermedius were fertilized and developed to the four-armed larval stage in either current natural seawater pH levels (as Control; pH = 7.99 ± 0.01) or laboratory-controlled acidified conditions (OA1: ΔpH = -0.3 units; OA2: ΔpH = -0.4 units; OA3: ΔpH = -0.5 units) according to the predictions of the Intergovernmental Panel on Climate Change (IPCC). The degrees of spicule exposure and asymmetry and mortality of four-armed larvae of S. intermedius were observed; each had a significant linearly increasing trend as the seawater pH level decreased. Comparative metabolome analysis identified a total of 87 significantly differentially expressed metabolites (SDMs, UP: 57, DOWN: 30) in OA-treated groups compared with the control group. Twenty-three SDMs, including carnitine, lysophosphatidylcholine (LPC) 18:3, lysophosphatidyl ethanolamine (LPE) 16:1, glutathione (GSH) and L-ascorbate, exhibited a linear increasing trend with decreasing seawater pH. Nine SDMs exhibited a linear decreasing trend as the seawater pH declined, including hypoxanthine, guanine and thymidine. Among all SDMs, we further mined 48 potential metabolite biomarkers responding to seawater acidification in four-armed larvae of S. intermedius. These potential metabolite biomarkers were mainly enriched in five pathways: glycerophospholipid metabolism, glutathione metabolism, purine metabolism, pyrimidine metabolism and the tricarboxylic acid cycle (TCA cycle). Our results will enrich our knowledge of the molecular mechanisms employed by sea urchins in response to CO2-induced seawater acidification.
Collapse
Affiliation(s)
- Yingying Li
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning 116023, PR China
| | - Wenlu Yin
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning 116023, PR China
| | - Yaoyao Zhan
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning 116023, PR China.
| | - Yujie Jia
- College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Dongyao Cui
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning 116023, PR China
| | - Weijie Zhang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning 116023, PR China
| | - Yaqing Chang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian, Liaoning 116023, PR China.
| |
Collapse
|
46
|
A rapid GC method coupled with quadrupole or time of flight mass spectrometry for metabolomics analysis. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1160:122355. [PMID: 32920480 DOI: 10.1016/j.jchromb.2020.122355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 01/01/2023]
Abstract
Gas chromatography-mass spectrometry (GC-MS) is an ideal tool for analyzing the intermediates of tricarboxylic acid cycle and glycolysis, sugars, organic acids and amino acids, etc. High-throughput metabolomics methods are required by large-scale clinical researches, and time of flight mass spectrometry (TOF MS) having fast scanning rate is preferable for rapid GC. Quadrupole MS (qMS) instruments have 95% market share, and their potential in rapid metabolomics is worth being studied. In this work, a within 15-min GC program was established and matched by qMS scanning for plasma metabolome analysis after N-methyl-N-(trimethylsilyl)-trifluoroacetamide derivatization. Compared to the longer-time program GC-qMS method, the rapid GC-qMS method had nearly no metabolome information loss, and it had excellent profile performance in repeatability, intra-day and inter-day precision, sampling range, linearity and extraction recovery. Compared to TOF MS, qMS achieved similar results in investigating lung cancer serum metabolic disruptions. Partial least squares-discriminant analysis revealed that the two datasets acquired by qMS and TOF MS had very similar model parameters, and most of top ranked differential metabolites were the same. This study provides a rapid and economical GC-qMS metabolomics method for researchers. Still, MS having faster scanning rate and higher sensitivity are recommended, if possible, to detect more small peaks and some co-eluted peaks.
Collapse
|
47
|
Sorensen MJ, Kennedy RT. Capillary ultrahigh-pressure liquid chromatography-mass spectrometry for fast and high resolution metabolomics separations. J Chromatogr A 2020; 1635:461706. [PMID: 33229007 DOI: 10.1016/j.chroma.2020.461706] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022]
Abstract
LC-MS is an important tool for metabolomics due its high sensitivity and broad metabolite coverage. The goal of improving resolution and decreasing analysis time in HPLC has led to the use of 5 - 15 cm long columns packed with 1.7 - 1.9 µm particles requiring pressures of 8 - 12 kpsi. We report on the potential for capillary LC-MS based metabolomics utilizing porous C18 particles down to 1.1 µm diameter and columns up to 50 cm long with an operating pressure of 35 kpsi. Our experiments show that it is possible to pack columns with 1.1 µm porous particles to provide predicted improvements in separation time and efficiency. Using kinetic plots to guide the choice of column length and particle size, we packed 50 cm long columns with 1.7 µm particles and 20 cm long columns with 1.1 µm particles, which should produce equivalent performance in shorter times. Columns were tested by performing isocratic and gradient LC-MS analyses of small molecule metabolites and extracts from plasma. These columns provided approximately 100,000 theoretical plates for metabolite standards and peak capacities over 500 in 100 min for a complex plasma extract with robust interfacing to MS. To generate a given peak capacity, the 1.1 µm particles in 20 cm columns required roughly 75% of the time as 1.7 µm particles in 50 cm columns with both operated at 35 kpsi. The 1.1 µm particle packed columns generated a given peak capacity nearly 3 times faster than 1.7 µm particles in 15 cm columns operated at ~10 kpsi. This latter condition represents commercial state of the art for capillary LC. To consider practical benefits for metabolomics, the effect of different LC-MS variables on mass spectral feature detection was evaluated. Lower flow rates (down to 700 nL/min) and larger injection volumes (up to 1 µL) increased the features detected with modest loss in separation performance. The results demonstrate the potential for fast and high resolution separations for metabolomics using 1.1 µm particles operated at 35 kpsi for capillary LC-MS.
Collapse
Affiliation(s)
- Matthew J Sorensen
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Robert T Kennedy
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
48
|
Xu W, Lin J, Gao M, Chen Y, Cao J, Pu J, Huang L, Zhao J, Qian K. Rapid Computer-Aided Diagnosis of Stroke by Serum Metabolic Fingerprint Based Multi-Modal Recognition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002021. [PMID: 33173737 PMCID: PMC7610260 DOI: 10.1002/advs.202002021] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/30/2020] [Indexed: 05/07/2023]
Abstract
Stroke is a leading cause of mortality and disability worldwide, expected to result in 61 million disability-adjusted life-years in 2020. Rapid diagnostics is the core of stroke management for early prevention and medical treatment. Serum metabolic fingerprints (SMFs) reflect underlying disease progression, predictive of patient phenotypes. Deep learning (DL) encoding SMFs with clinical indexes outperforms single biomarkers, while posing challenges with poor prediction to interpret by feature selection. Herein, rapid computer-aided diagnosis of stroke is performed using SMF based multi-modal recognition by DL, to combine adaptive machine learning with a novel feature selection approach. SMFs are extracted by nano-assisted laser desorption/ionization mass spectrometry (LDI MS), consuming 100 nL of serum in seconds. A multi-modal recognition is constructed by integrating SMFs and clinical indexes with an enhanced area under curve (AUC) up to 0.845 for stroke screening, compared to single-modal diagnosis by only SMFs or clinical indexes. The prediction of DL is addressed by selecting 20 key metabolite features with differential regulation through a saliency map approach, shedding light on the molecular mechanisms in stroke. The approach highlights the emerging role of DL in precision medicine and suggests an expanding utility for computational analysis of SMFs in stroke screening.
Collapse
Affiliation(s)
- Wei Xu
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
- State Key Laboratory for Oncogenes and Related GenesSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Jixian Lin
- Department of NeurologyMinhang HospitalFudan University170 Xinsong RoadShanghai201199P. R. China
| | - Ming Gao
- School of Management Science and EngineeringDongbei University of Finance and EconomicsDalian116025P. R. China
- Center for Post‐doctoral Studies of Computer ScienceNortheastern UniversityShenyang110819P. R. China
| | - Yuhan Chen
- School of Management Science and EngineeringDongbei University of Finance and EconomicsDalian116025P. R. China
- Center for Post‐doctoral Studies of Computer ScienceNortheastern UniversityShenyang110819P. R. China
| | - Jing Cao
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
- State Key Laboratory for Oncogenes and Related GenesSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
- State Key Laboratory for Oncogenes and Related GenesSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Lin Huang
- Stem Cell Research CenterRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
| | - Jing Zhao
- Department of NeurologyMinhang HospitalFudan University170 Xinsong RoadShanghai201199P. R. China
| | - Kun Qian
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
- State Key Laboratory for Oncogenes and Related GenesSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| |
Collapse
|
49
|
Distinct Fecal and Plasma Metabolites in Children with Autism Spectrum Disorders and Their Modulation after Microbiota Transfer Therapy. mSphere 2020; 5:5/5/e00314-20. [PMID: 33087514 PMCID: PMC7580952 DOI: 10.1128/msphere.00314-20] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Despite the prevalence of autism and its extensive impact on our society, no U.S. Food and Drug Administration-approved treatment is available for this complex neurobiological disorder. Based on mounting evidences that support a link between autism and the gut microbiome, we previously performed a pioneering open-label clinical trial using intensive fecal microbiota transplant. The therapy significantly improved gastrointestinal and behavioral symptoms. Comprehensive metabolomic measurements in this study showed that children with autism spectrum disorder (ASD) had different levels of many plasma metabolites at baseline compared to those in typically developing children. Microbiota transfer therapy (MTT) had a systemic effect, resulting in substantial changes in plasma metabolites, driving a number of metabolites to be more similar to those from typically developing children. Our results provide evidence that changes in metabolites are one mechanism of the gut-brain connection mediated by the gut microbiota and offer plausible clinical evidence for a promising autism treatment and biomarkers. Accumulating evidence has strengthened a link between dysbiotic gut microbiota and autism. Fecal microbiota transplant (FMT) is a promising therapy to repair dysbiotic gut microbiota. We previously performed intensive FMT called microbiota transfer therapy (MTT) for children with autism spectrum disorders (ASD) and observed a substantial improvement of gastrointestinal and behavioral symptoms. We also reported modulation of the gut microbiome toward a healthy one. In this study, we report comprehensive metabolite profiles from plasma and fecal samples of the children who participated in the MTT trial. With 619 plasma metabolites detected, we found that the autism group had distinctive metabolic profiles at baseline. Eight metabolites (nicotinamide riboside, IMP, iminodiacetate, methylsuccinate, galactonate, valylglycine, sarcosine, and leucylglycine) were significantly lower in the ASD group at baseline, while caprylate and heptanoate were significantly higher in the ASD group. MTT drove global shifts in plasma profiles across various metabolic features, including nicotinate/nicotinamide and purine metabolism. In contrast, for 669 fecal metabolites detected, when correcting for multiple hypotheses, no metabolite was significantly different at baseline. Although not statistically significant, p-cresol sulfate was relatively higher in the ASD group at baseline, and after MTT, the levels decreased and were similar to levels in typically developing (TD) controls. p-Cresol sulfate levels were inversely correlated with Desulfovibrio, suggesting a potential role of Desulfovibrio on p-cresol sulfate modulation. Further studies of metabolites in a larger ASD cohort, before and after MTT, are warranted, as well as clinical trials of other therapies to address the metabolic changes which MTT was not able to correct. IMPORTANCE Despite the prevalence of autism and its extensive impact on our society, no U.S. Food and Drug Administration-approved treatment is available for this complex neurobiological disorder. Based on mounting evidences that support a link between autism and the gut microbiome, we previously performed a pioneering open-label clinical trial using intensive fecal microbiota transplant. The therapy significantly improved gastrointestinal and behavioral symptoms. Comprehensive metabolomic measurements in this study showed that children with autism spectrum disorder (ASD) had different levels of many plasma metabolites at baseline compared to those in typically developing children. Microbiota transfer therapy (MTT) had a systemic effect, resulting in substantial changes in plasma metabolites, driving a number of metabolites to be more similar to those from typically developing children. Our results provide evidence that changes in metabolites are one mechanism of the gut-brain connection mediated by the gut microbiota and offer plausible clinical evidence for a promising autism treatment and biomarkers.
Collapse
|
50
|
Practicing precision medicine with intelligently integrative clinical and multi-omics data analysis. Hum Genomics 2020; 14:35. [PMID: 33008459 PMCID: PMC7530549 DOI: 10.1186/s40246-020-00287-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
Precision medicine aims to empower clinicians to predict the most appropriate course of action for patients with complex diseases like cancer, diabetes, cardiomyopathy, and COVID-19. With a progressive interpretation of the clinical, molecular, and genomic factors at play in diseases, more effective and personalized medical treatments are anticipated for many disorders. Understanding patient’s metabolomics and genetic make-up in conjunction with clinical data will significantly lead to determining predisposition, diagnostic, prognostic, and predictive biomarkers and paths ultimately providing optimal and personalized care for diverse, and targeted chronic and acute diseases. In clinical settings, we need to timely model clinical and multi-omics data to find statistical patterns across millions of features to identify underlying biologic pathways, modifiable risk factors, and actionable information that support early detection and prevention of complex disorders, and development of new therapies for better patient care. It is important to calculate quantitative phenotype measurements, evaluate variants in unique genes and interpret using ACMG guidelines, find frequency of pathogenic and likely pathogenic variants without disease indicators, and observe autosomal recessive carriers with a phenotype manifestation in metabolome. Next, ensuring security to reconcile noise, we need to build and train machine-learning prognostic models to meaningfully process multisource heterogeneous data to identify high-risk rare variants and make medically relevant predictions. The goal, today, is to facilitate implementation of mainstream precision medicine to improve the traditional symptom-driven practice of medicine, and allow earlier interventions using predictive diagnostics and tailoring better-personalized treatments. We strongly recommend automated implementation of cutting-edge technologies, utilizing machine learning (ML) and artificial intelligence (AI) approaches for the multimodal data aggregation, multifactor examination, development of knowledgebase of clinical predictors for decision support, and best strategies for dealing with relevant ethical issues.
Collapse
|