1
|
Qiu C, Liao J, Gheissari R, Li C, Kapai A, Conti DV, Jones DP, Bastain TM, Breton CV, Chen Z. Dysregulated maternal and newborn fatty acid, sugar and amino acid metabolism associated with high birth weight. Int J Obes (Lond) 2025:10.1038/s41366-025-01775-9. [PMID: 40247088 DOI: 10.1038/s41366-025-01775-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/19/2025]
Abstract
OBJECTIVE This study aims to find maternal and neonatal metabolomic signatures that contribute to the adverse birthweight outcomes including abnormally high and low birth weight. We also investigated the role of metabolomic signatures in the associations of maternal risk factors such as parity and gestational weight gain with adverse birthweight outcomes. METHODS Ninety-six pregnant women and their newborns from the MADRES prospective cohort were studied. Maternal serum at third trimester and newborn cord blood were assayed for untargeted metabolomics using mass-spectrometry. Metabolome-wide association analysis was conducted to assess maternal and newborn metabolomic features association with birth weight Z-score, followed by network analysis of maternal and newborn metabolomics. Lastly, the contribution of maternal and newborn metabolomics to associations between maternal risk factors and newborn birthweight was assessed. RESULTS Maternal gestational weight gain and parity were positively associated with newborn birthweight. Maternal glucose and branched-chain amino acid metabolism pathways and newborn's fatty acid, glucose metabolism and C21-steroid hormone biosynthesis were significantly enriched with high birth weight Z-score. Dysregulation in these pathways linked maternal factors such as gestational weight gain and parity with high birth weight Z-score. CONCLUSION Our findings indicate that altered maternal sugar and energy metabolism, newborn sugar and amino acid metabolism, and newborn C21-steroid hormone biosynthesis were associated with high birth weight. Dysregulated metabolism in pregnant women and newborn may contribute to the pathophysiological mechanisms linking maternal excessive gestational weight gain and multiparity with high birth weight.
Collapse
Affiliation(s)
- Chenyu Qiu
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Jiawen Liao
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Roya Gheissari
- Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Claire Li
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Anika Kapai
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - David V Conti
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Dean P Jones
- Department of Medicine, Emory University, Atlanta, GA, USA
| | - Theresa M Bastain
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Carrie V Breton
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| | - Zhanghua Chen
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Zhou S, Han X, Shi Z, Zhang Q, Cox J, Pak V. Aminoacetone is associated with sleep latency. Sleep Med 2025; 128:183-186. [PMID: 39954376 DOI: 10.1016/j.sleep.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/25/2025] [Accepted: 02/02/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION This study conducted a targeted analysis of metabolites involved in inflammatory, oxidative stress, and neuronal pathways that were previously implicated with sleepiness based on our prior work identifying metabolites in these pathways in persons with OSA. We examine associations between sleep latency (based on the Pittsburgh Sleep Quality Index) which is an important indicator of sleep quality and daytime sleepiness, and metabolic concentrations to identify potential mechanisms linking the two. METHODS This cross-sectional study included 88 participants newly diagnosed and untreated for OSA who underwent an overnight in-lab or at home sleep study recruited from the Emory Mechanisms of Sleepiness Symptoms Study (EMOSS). Fasting morning blood plasma samples were collected after the overnight sleep study. A multiple linear regression model was utilized to examine the association between metabolites of interest and sleep latency, controlling for baseline covariates of age, sex, body mass index (BMI), race, smoking status, and apnea hypopnea index (AHI). RESULTS Among the targeted metabolites, aminoacetone showed a significant association with sleep latency (adjusted mean (SE) = -0.39 (0.11); p = 0.00072). Participants with sleep onset latency >60 min had lower levels of aminoacetone than patients with sleep onset latency <15 min. CONCLUSION This is the first study to evaluate the relationship between aminoacetone and sleep latency in participants with newly diagnosed OSA. Our findings suggests that aminoacetone is associated with sleep latency in participants with OSA.
Collapse
Affiliation(s)
- Sirui Zhou
- Emory University-College of Arts and Sciences, Atlanta, GA, USA
| | - Xue Han
- School of Nursing, Emory University, 1520 Clifton Road, 243, Atlanta, GA, 30322, USA
| | - Zhenzhen Shi
- The Program for Experimental and Theoretical Modeling, Division of Hepatology, Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - John Cox
- School of Nursing, Emory University, 1520 Clifton Road, 243, Atlanta, GA, 30322, USA
| | - Victoria Pak
- School of Nursing, Emory University, 1520 Clifton Road, 243, Atlanta, GA, 30322, USA; Emory Rollins School of Public Health, Atlanta, GA, USA.
| |
Collapse
|
3
|
Wang P, Li Z, Song Y, Zhang B, Fan C. Resveratrol-driven macrophage polarization: unveiling mechanisms and therapeutic potential. Front Pharmacol 2025; 15:1516609. [PMID: 39872049 PMCID: PMC11770351 DOI: 10.3389/fphar.2024.1516609] [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: 10/24/2024] [Accepted: 12/23/2024] [Indexed: 01/29/2025] Open
Abstract
Resveratrol, a polyphenolic compound known for its diverse biological activities, has demonstrated multiple pharmacological effects, including anti-inflammatory, anti-aging, anti-diabetic, anti-cancer, and cardiovascular protective properties. Recent studies suggest that these effects are partly mediated through the regulation of macrophage polarization, wherein macrophages differentiate into pro-inflammatory M1 or anti-inflammatory M2 phenotypes. Our review highlights how resveratrol modulates macrophage polarization through various signaling pathways to achieve therapeutic effects. For example, resveratrol can activate the senescence-associated secretory phenotype (SASP) pathway and inhibit the signal transducer and activator of transcription (STAT3) and sphingosine-1-phosphate (S1P)-YAP signaling axes, promoting M1 polarization or suppressing M2 polarization, thereby inhibiting tumor growth. Conversely, it can promote M2 polarization or suppress M1 polarization by inhibiting the NF-κB signaling pathway or activating the PI3K/Akt and AMP-activated protein kinase (AMPK) pathways, thus alleviating inflammatory responses. Notably, the effect of resveratrol on macrophage polarization is concentration-dependent; moderate concentrations tend to promote M1 polarization, while higher concentrations may favor M2 polarization. This concentration dependence offers new perspectives for clinical treatment but also underscores the necessity for precise dosage control when using resveratrol. In summary, resveratrol exhibits significant potential in regulating macrophage polarization and treating related diseases.
Collapse
Affiliation(s)
- Panting Wang
- Department of Neurosurgery West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing Sichuan University, Chengdu, China
| | - Zixi Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yixuan Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bowei Zhang
- Southwest Institute of Technical Physics, Chengdu, China
| | - Chaofeng Fan
- Department of Neurosurgery West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Shao K, Zou R, Zhang Z, Mandemaker LDB, Timbie S, Smith RD, Durkin AM, Dusza HM, Meirer F, Weckhuysen BM, Alderete TL, Vermeulen R, Walker DI. Advancements in Assays for Micro- and Nanoplastic Detection: Paving the Way for Biomonitoring and Exposomics Studies. Annu Rev Pharmacol Toxicol 2025; 65:567-585. [PMID: 39270670 DOI: 10.1146/annurev-pharmtox-030424-112828] [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] [Indexed: 09/15/2024]
Abstract
Although plastic pollution and exposure to plastic-related compounds have received worldwide attention, health risks associated with micro- and nanoplastics (MNPs) are largely unknown. Emerging evidence suggests MNPs are present in human biofluids and tissue, including blood, breast milk, stool, lung tissue, and placenta; however, exposure assessment is limited and the extent of human exposure to MNPs is not well known. While there is a critical need to establish robust and scalable biomonitoring strategies to assess human exposure to MNPs and plastic-related chemicals, over 10,000 chemicals have been linked to plastic manufacturing with no existing standardized approaches to account for even a fraction of these exposures. This review provides an overview of the status of methods for measuring MNPs and associated plastic-related chemicals in humans, with a focus on approaches that could be adapted for population-wide biomonitoring and integration with biological response measures to develop hypotheses on potential health effects of plastic exposures. We also examine the exposure risks associated with the widespread use of chemical additives in plastics. Despite advancements in analytical techniques, there remains a pressing need for standardized measurement protocols and untargeted, high-throughput analysis methods to enable comprehensive MNP biomonitoring to identify key MNP exposures in human populations. This review aims to merge insights into the toxicological effects of MNPs and plastic additives with an evaluation of analytical challenges, advocating for enhanced research methods to fully assess, understand, and mitigate the public health implications of MNPs.
Collapse
Affiliation(s)
- Kuanliang Shao
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA;
| | - Runyu Zou
- Division of Toxicology, Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Zhuoyue Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA;
| | - Laurens D B Mandemaker
- Inorganic Chemistry and Catalysis Group, Institute for Sustainable and Circular Chemistry, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, The Netherlands
| | - Sarah Timbie
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA;
| | - Ronald D Smith
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA;
| | - Amanda M Durkin
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hanna M Dusza
- Division of Toxicology, Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Florian Meirer
- Inorganic Chemistry and Catalysis Group, Institute for Sustainable and Circular Chemistry, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, The Netherlands
| | - Bert M Weckhuysen
- Inorganic Chemistry and Catalysis Group, Institute for Sustainable and Circular Chemistry, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, The Netherlands
| | - Tanya L Alderete
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Roel Vermeulen
- Division of Toxicology, Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Douglas I Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA;
| |
Collapse
|
5
|
Huneault HE, Chen CY, Cohen CC, Liu X, Jarrell ZR, He Z, DeSantos KE, Welsh JA, Maner-Smith KM, Ortlund EA, Schwimmer JB, Vos MB. Lipidome Changes Associated with a Diet-Induced Reduction in Hepatic Fat among Adolescent Boys with Metabolic Dysfunction-Associated Steatotic Liver Disease. Metabolites 2024; 14:191. [PMID: 38668319 PMCID: PMC11052520 DOI: 10.3390/metabo14040191] [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: 02/10/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Little is known about lipid changes that occur in the setting of metabolic-dysfunction-associated steatotic liver disease (MASLD) regression. We previously reported improvements in hepatic steatosis, de novo lipogenesis (DNL), and metabolomic profiles associated with oxidative stress, inflammation, and selected lipid metabolism in 40 adolescent boys (11-16 y) with hepatic steatosis ≥5% (98% meeting the definition of MASLD). Participants were randomized to a low-free-sugar diet (LFSD) (n = 20) or usual diet (n = 20) for 8 weeks. Here, we employed untargeted/targeted lipidomics to examine lipid adaptations associated with the LFSD and improvement of hepatic steatosis. Our LC-MS/MS analysis revealed decreased triglycerides (TGs), diacylglycerols (DGs), cholesteryl esters (ChE), lysophosphatidylcholine (LPC), and phosphatidylcholine (PC) species with the diet intervention (p < 0.05). Network analysis demonstrated significantly lower levels of palmitate-enriched TG species post-intervention, mirroring the previously shown reduction in DNL in response to the LFSD. Targeted oxylipins analysis revealed a decrease in the abundance of 8-isoprostane and 14,15-DiHET and an increase in 8,9-DiHET (p < 0.05). Overall, we observed reductions in TGs, DGs, ChE, PC, and LPC species among participants in the LFSD group. These same lipids have been associated with MASLD progression; therefore, our findings may indicate normalization of key biological processes, including lipid metabolism, insulin resistance, and lipotoxicity. Additionally, our targeted oxylipins assay revealed novel changes in eicosanoids, suggesting improvements in oxidative stress. Future studies are needed to elucidate the mechanisms of these findings and prospects of these lipids as biomarkers of MASLD regression.
Collapse
Affiliation(s)
- Helaina E. Huneault
- Nutrition & Health Sciences Doctoral Program, Laney Graduate School, Emory University, Atlanta, GA 30322, USA; (J.A.W.); (M.B.V.)
| | - Chih-Yu Chen
- Department of Biochemistry, Emory School of Medicine, Emory University, Atlanta, GA 30329, USA; (C.-Y.C.); (X.L.); (E.A.O.)
| | - Catherine C. Cohen
- Section of Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (K.M.M.-S.)
| | - Xueyun Liu
- Department of Biochemistry, Emory School of Medicine, Emory University, Atlanta, GA 30329, USA; (C.-Y.C.); (X.L.); (E.A.O.)
| | - Zachery R. Jarrell
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Zhulin He
- Pediatric Biostatistics Core, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Karla E. DeSantos
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, GA 30322, USA;
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Jean A. Welsh
- Nutrition & Health Sciences Doctoral Program, Laney Graduate School, Emory University, Atlanta, GA 30322, USA; (J.A.W.); (M.B.V.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Kristal M. Maner-Smith
- Section of Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (K.M.M.-S.)
| | - Eric A. Ortlund
- Department of Biochemistry, Emory School of Medicine, Emory University, Atlanta, GA 30329, USA; (C.-Y.C.); (X.L.); (E.A.O.)
| | - Jeffrey B. Schwimmer
- Department of Gastroenterology, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA;
- Department of Pediatrics, School of Medicine, University of California, San Diego, CA 92093, USA
| | - Miriam B. Vos
- Nutrition & Health Sciences Doctoral Program, Laney Graduate School, Emory University, Atlanta, GA 30322, USA; (J.A.W.); (M.B.V.)
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, GA 30322, USA;
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
6
|
Huneault HE, Gent AE, Cohen CC, He Z, Jarrell ZR, Kamaleswaran R, Vos MB. Validation of a screening panel for pediatric metabolic dysfunction-associated steatotic liver disease using metabolomics. Hepatol Commun 2024; 8:e0375. [PMID: 38407264 PMCID: PMC10898657 DOI: 10.1097/hc9.0000000000000375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/19/2023] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as NAFLD, is the most common liver disease in children. Liver biopsy remains the gold standard for diagnosis, although more efficient screening methods are needed. We previously developed a novel NAFLD screening panel in youth using machine learning applied to high-resolution metabolomics and clinical phenotype data. Our objective was to validate this panel in a separate cohort, which consisted of a combined cross-sectional sample of 161 children with stored frozen samples (75% male, 12.8±2.6 years of age, body mass index 31.0±7.0 kg/m2, 81% with MASLD, 58% Hispanic race/ethnicity). METHODS Clinical data were collected from all children, and high-resolution metabolomics was performed using their fasting serum samples. MASLD was assessed by MRI-proton density fat fraction or liver biopsy and cardiometabolic factors. Our previously developed panel included waist circumference, triglycerides, whole-body insulin sensitivity index, 3 amino acids, 2 phospholipids, dihydrothymine, and 2 unknowns. To improve feasibility, a simplified version without the unknowns was utilized in the present study. Since the panel was modified, the data were split into training (67%) and test (33%) sets to assess the validity of the panel. RESULTS Our present highest-performing modified model, with 4 clinical variables and 8 metabolomics features, achieved an AUROC of 0.92, 95% sensitivity, and 80% specificity for detecting MASLD in the test set. CONCLUSIONS Therefore, this panel has promising potential for use as a screening tool for MASLD in youth.
Collapse
Affiliation(s)
- Helaina E. Huneault
- Nutrition & Health Sciences Program, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Alasdair E. Gent
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Catherine C. Cohen
- Section of Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Zhulin He
- Department of Pediatrics, Pediatric Biostatistics Core, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Zachery R. Jarrell
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA
| | - Rishikesan Kamaleswaran
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Miriam B. Vos
- Nutrition & Health Sciences Program, Laney Graduate School, Emory University, Atlanta, Georgia, USA
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Jian J, He D, Gao S, Tao X, Dong X. Pharmacokinetics in Pharmacometabolomics: Towards Personalized Medication. Pharmaceuticals (Basel) 2023; 16:1568. [PMID: 38004434 PMCID: PMC10675232 DOI: 10.3390/ph16111568] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Indiscriminate drug administration may lead to drug therapy results with varying effects on patients, and the proposal of personalized medication can help patients to receive effective drug therapy. Conventional ways of personalized medication, such as pharmacogenomics and therapeutic drug monitoring (TDM), can only be implemented from a single perspective. The development of pharmacometabolomics provides a research method for the realization of precise drug administration, which integrates the environmental and genetic factors, and applies metabolomics technology to study how to predict different drug therapeutic responses of organisms based on baseline metabolic levels. The published research on pharmacometabolomics has achieved satisfactory results in predicting the pharmacokinetics, pharmacodynamics, and the discovery of biomarkers of drugs. Among them, the pharmacokinetics related to pharmacometabolomics are used to explore individual variability in drug metabolism from the level of metabolism of the drugs in vivo and the level of endogenous metabolite changes. By searching for relevant literature with the keyword "pharmacometabolomics" on the two major literature retrieval websites, PubMed and Web of Science, from 2006 to 2023, we reviewed articles in the field of pharmacometabolomics that incorporated pharmacokinetics into their research. This review explains the therapeutic effects of drugs on the body from the perspective of endogenous metabolites and pharmacokinetic principles, and reports the latest advances in pharmacometabolomics related to pharmacokinetics to provide research ideas and methods for advancing the implementation of personalized medication.
Collapse
Affiliation(s)
- Jingai Jian
- School of Medicine, Shanghai University, Shanghai 200444, China; (J.J.); (D.H.)
| | - Donglin He
- School of Medicine, Shanghai University, Shanghai 200444, China; (J.J.); (D.H.)
| | - Songyan Gao
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China;
| | - Xia Tao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xin Dong
- School of Medicine, Shanghai University, Shanghai 200444, China; (J.J.); (D.H.)
| |
Collapse
|
8
|
He D, Yan Q, Uppal K, Walker DI, Jones DP, Ritz B, Heck JE. Metabolite Stability in Archived Neonatal Dried Blood Spots Used for Epidemiologic Research. Am J Epidemiol 2023; 192:1720-1730. [PMID: 37218607 PMCID: PMC11004922 DOI: 10.1093/aje/kwad122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 09/01/2022] [Accepted: 05/17/2023] [Indexed: 05/24/2023] Open
Abstract
Epidemiologic studies of low-frequency exposures or outcomes using metabolomics analyses of neonatal dried blood spots (DBS) often require assembly of samples with substantial differences in duration of storage. Independent assessment of stability of metabolites in archived DBS will enable improved design and interpretation of epidemiologic research utilizing DBS. Neonatal DBS routinely collected and stored as part of the California Genetic Disease Screening Program between 1983 and 2011 were used. The study population included 899 children without cancer before age 6 years, born in California. High-resolution metabolomics with liquid-chromatography mass spectrometry was performed, and the relative ion intensities of common metabolites and selected xenobiotic metabolites of nicotine (cotinine and hydroxycotinine) were evaluated. In total, we detected 26,235 mass spectral features across 2 separate chromatography methods (C18 hydrophobic reversed-phase chromatography and hydrophilic-interaction liquid chromatography). For most of the 39 metabolites related to nutrition and health status, we found no statistically significant annual trends across the years of storage. Nicotine metabolites were captured in the DBS with relatively stable intensities. This study supports the usefulness of DBS stored long-term for epidemiologic studies of the metabolome. -Omics-based information gained from DBS may also provide a valuable tool for assessing prenatal environmental exposures in child health research.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Julia E Heck
- Correspondence to Dr. Julia E. Heck, College of Health and Public Service, UNT 1155 Union Circle #311340, Denton, TX 76203-5017 (e-mail: )
| |
Collapse
|
9
|
Welsh JA, Pyo E, Huneault H, Gonzalez Ramirez L, Alazraki A, Alli R, Dunbar SB, Khanna G, Knight-Scott J, Pimentel A, Reed B, Rodney-Somersall C, Santoro N, Umpierrez G, Vos MB. Study protocol for a randomized, controlled trial using a novel, family-centered diet treatment to prevent nonalcoholic fatty liver disease in Hispanic children. Contemp Clin Trials 2023; 129:107170. [PMID: 37019180 PMCID: PMC10734403 DOI: 10.1016/j.cct.2023.107170] [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: 01/19/2023] [Revised: 03/20/2023] [Accepted: 03/25/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the leading liver disorder among U.S. children and is most prevalent among Hispanic children with obesity. Previous research has shown that reducing the consumption of free sugars (added sugars + naturally occurring sugars in fruit juice) can reverse liver steatosis in adolescents with NAFLD. This study aims to determine if a low-free sugar diet (LFSD) can prevent liver fat accumulation and NAFLD in high-risk children. METHODS In this randomized controlled trial, we will enroll 140 Hispanic children aged 6 to 9 years who are ≥50th percentile BMI and without a previous diagnosis of NAFLD. Participants will be randomly assigned to either an experimental (LFSD) or a control (usual diet + educational materials) group. The one-year intervention includes removal of foods high in free sugars from the home at baseline, provision of LFSD household groceries for the entire family (weeks 1-4, 12, 24, and 36), dietitian-guided family grocery shopping sessions (weeks 12, 24, and 36), and ongoing education and motivational interviewing to promote LFSD. Both groups complete assessment measures at baseline, 6, 12, 18, and 24 months. Primary study outcomes are percent hepatic fat at 12 months and incidence of clinically significant hepatic steatosis (>5%) + elevated liver enzymes at 24 months. Secondary outcomes include metabolic markers potentially mediating or moderating NAFLD pathogenesis. DISCUSSION This protocol describes the rationale, eligibility criteria, recruitment strategies, analysis plan as well as a novel dietary intervention design. Study results will inform future dietary guidelines for pediatric NAFLD prevention. TRIAL REGISTRATION ClinicalTrials.gov, NCT05292352.
Collapse
Affiliation(s)
- J A Welsh
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States; Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, United States
| | - E Pyo
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, United States
| | - H Huneault
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, United States
| | - L Gonzalez Ramirez
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, United States
| | - A Alazraki
- Department of Radiology, Emory University School of Medicine, Atlanta, GA, United States; Department of Radiology, Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - R Alli
- Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - S B Dunbar
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - G Khanna
- Department of Radiology, Emory University School of Medicine, Atlanta, GA, United States; Department of Radiology, Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Jack Knight-Scott
- Department of Radiology, Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - A Pimentel
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States; Grady Memorial Hospital, Atlanta, GA, United States
| | - B Reed
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States; Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - C Rodney-Somersall
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States; Grady Memorial Hospital, Atlanta, GA, United States
| | - N Santoro
- Department of Pediatrics, Kansas Medical Center, Kansas City, KS, United States; Department of Medicine and Health Sciences, "V.Tiberio" University of Molise, Campobasso, Italy; Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| | - G Umpierrez
- Grady Memorial Hospital, Atlanta, GA, United States; Division of Endocrinology, Metabolism, Emory University School of Medicine, Atlanta, GA, United States
| | - M B Vos
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States; Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, United States; Children's Healthcare of Atlanta, Atlanta, GA, United States.
| |
Collapse
|
10
|
Cohen CC, Huneault H, Accardi CJ, Jones DP, Liu K, Maner-Smith KM, Song M, Welsh JA, Ugalde-Nicalo PA, Schwimmer JB, Vos MB. Metabolome × Microbiome Changes Associated with a Diet-Induced Reduction in Hepatic Fat among Adolescent Boys. Metabolites 2023; 13:401. [PMID: 36984841 PMCID: PMC10053986 DOI: 10.3390/metabo13030401] [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: 02/07/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Dietary sugar reduction is one therapeutic strategy for improving nonalcoholic fatty liver disease (NAFLD), and the underlying mechanisms for this effect warrant further investigation. Here, we employed metabolomics and metagenomics to examine systemic biological adaptations associated with dietary sugar restriction and (subsequent) hepatic fat reductions in youth with NAFLD. Data/samples were from a randomized controlled trial in adolescent boys (11-16 years, mean ± SD: 13.0 ± 1.9 years) with biopsy-proven NAFLD who were either provided a low free-sugar diet (LFSD) (n = 20) or consumed their usual diet (n = 20) for 8 weeks. Plasma metabolomics was performed on samples from all 40 participants by coupling hydrophilic interaction liquid chromatography (HILIC) and C18 chromatography with mass spectrometry. In a sub-sample (n = 8 LFSD group and n = 10 usual diet group), 16S ribosomal RNA (rRNA) sequencing was performed on stool to examine changes in microbial composition/diversity. The diet treatment was associated with differential expression of 419 HILIC and 205 C18 metabolite features (p < 0.05), which were enriched in amino acid pathways, including methionine/cysteine and serine/glycine/alanine metabolism (p < 0.05), and lipid pathways, including omega-3 and linoleate metabolism (p < 0.05). Quantified metabolites that were differentially changed in the LFSD group, compared to usual diet group, and representative of these enriched metabolic pathways included increased serine (p = 0.001), glycine (p = 0.004), 2-aminobutyric acid (p = 0.012), and 3-hydroxybutyric acid (p = 0.005), and decreased linolenic acid (p = 0.006). Microbiome changes included an increase in richness at the phylum level and changes in a few genera within Firmicutes. In conclusion, the LFSD treatment, compared to usual diet, was associated with metabolome and microbiome changes that may reflect biological mechanisms linking dietary sugar restriction to a therapeutic decrease in hepatic fat. Studies are needed to validate our findings and test the utility of these "omics" changes as response biomarkers.
Collapse
Affiliation(s)
- Catherine C. Cohen
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Helaina Huneault
- Nutrition & Health Sciences Doctoral Program, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Carolyn J. Accardi
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Dean P. Jones
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ken Liu
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Kristal M. Maner-Smith
- Emory Integrated Lipidomics Core, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ming Song
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Jean A. Welsh
- Nutrition & Health Sciences Doctoral Program, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Patricia A. Ugalde-Nicalo
- Department of Gastroenterology, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA
- Department of Pediatrics, School of Medicine, University of California, San Diego, CA 92093, USA
| | - Jeffrey B. Schwimmer
- Department of Gastroenterology, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA
- Department of Pediatrics, School of Medicine, University of California, San Diego, CA 92093, USA
| | - Miriam B. Vos
- Nutrition & Health Sciences Doctoral Program, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
11
|
Untargeted, High-Resolution Metabolomics in Pediatric Eosinophilic Esophagitis. J Pediatr Gastroenterol Nutr 2023; 76:355-363. [PMID: 36728821 DOI: 10.1097/mpg.0000000000003693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND/OBJECTIVES Eosinophilic esophagitis (EoE) is an inflammatory disease of unclear etiology. The aim of this study was to use untargeted plasma metabolomics to identify metabolic pathway alterations associated with EoE to better understand the pathophysiology. METHODS This prospective, case-control study included 72 children, aged 1-17 years, undergoing clinically indicated upper endoscopy (14 diagnosed with EoE and 58 controls). Fasting plasma samples were analyzed for metabolomics by high-resolution dual-chromatography mass spectrometry. Analysis was performed on sex-matched groups at a 2:1 ratio. Significant differences among the plasma metabolite features between children with and without EoE were determined using multivariate regression analysis and were annotated with a network-based algorithm. Subsequent pathway enrichment analysis was performed. RESULTS Patients with EoE had a higher proportion of atopic disease (85.7% vs 50%, P = 0.019) and any allergies (100% vs 57.1%, P = 0.0005). Analysis of the dual chromatography features resulted in a total of 918 metabolites that differentiated EoE and controls. Glycerophospholipid metabolism was significantly enriched with the greatest number of differentiating metabolites and overall pathway enrichment ( P < 0.01). Multiple amino and fatty acid pathways including linoleic acid were also enriched, as well as pyridoxine metabolism ( P < 0.01). CONCLUSIONS In this pilot study, we found differences in metabolites involved in glycerophospholipid and inflammation pathways in pediatric patients with EoE using untargeted metabolomics, as well as overlap with amino acid metabolome alterations found in atopic disease.
Collapse
|
12
|
Pak VM, Russell K, Shi Z, Zhang Q, Cox J, Uppal K, Yu T, Hertzberg V, Liu K, Ioachimescu OC, Collop N, Bliwise DL, Kutner NG, Rogers A, Dunbar SB. Sphinganine is associated with 24-h MAP in the non-sleepy with OSA. Metabolomics 2022; 18:23. [PMID: 35391564 DOI: 10.1007/s11306-021-01860-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 12/07/2021] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Excessive daytime sleepiness is a debilitating symptom of obstructive sleep apnea (OSA) linked to cardiovascular disease, and metabolomic mechanisms underlying this relationship remain unknown. We examine whether metabolites from inflammatory and oxidative stress-related pathways that were identified in our prior work could be involved in connecting the two phenomena. METHODS This study included 57 sleepy (Epworth Sleepiness Scale (ESS) ≥ 10) and 37 non-sleepy (ESS < 10) participants newly diagnosed and untreated for OSA that completed an overnight in-lab or at home sleep study who were recruited from the Emory Mechanisms of Sleepiness Symptoms Study (EMOSS). Differences in fasting blood samples of metabolites were explored in participants with sleepiness versus those without and multiple linear regression models were utilized to examine the association between metabolites and mean arterial pressure (MAP). RESULTS The 24-h MAP was higher in sleepy 92.8 mmHg (8.4) as compared to non-sleepy 88.8 mmHg (8.1) individuals (P = 0.03). Although targeted metabolites were not significantly associated with MAP, when we stratified by sleepiness group, we found that sphinganine is significantly associated with MAP (Estimate = 8.7, SE = 3.7, P = 0.045) in non-sleepy patients when controlling for age, BMI, smoking status, and apnea-hypopnea index (AHI). CONCLUSION This is the first study to evaluate the relationship of inflammation and oxidative stress related metabolites in sleepy versus non-sleepy participants with newly diagnosed OSA and their association with 24-h MAP. Our study suggests that Sphinganine is associated with 24 hour MAP in the non-sleepy participants with OSA.
Collapse
Affiliation(s)
- Victoria M Pak
- School of Nursing, Emory University, 1520 Clifton Road, 243, Atlanta, GA, 30322, USA.
- Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| | - Katherine Russell
- School of Nursing, Emory University, 1520 Clifton Road, 243, Atlanta, GA, 30322, USA
| | - Zhenzhen Shi
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Gangarosa Department of Environmental Health, Atlanta, GA, 30322, USA
| | - Qiang Zhang
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Gangarosa Department of Environmental Health, Atlanta, GA, 30322, USA
| | - John Cox
- School of Nursing, Emory University, 1520 Clifton Road, 243, Atlanta, GA, 30322, USA
| | - Karan Uppal
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Tianwei Yu
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Shenzhen Research Institute of Big Data, and School of Data Science, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Vicki Hertzberg
- School of Nursing, Emory University, 1520 Clifton Road, 243, Atlanta, GA, 30322, USA
| | - Ken Liu
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Octavian C Ioachimescu
- School of Medicine, Emory University, Atlanta, GA, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University - School of Medicine, Atlanta, GA, 30322, USA
- Sleep Medicine Section, Atlanta VA Healthcare System, Atlanta, GA, 30322, USA
| | - Nancy Collop
- School of Medicine, Emory University, Atlanta, GA, USA
| | | | - Nancy G Kutner
- Gangarosa Department of Environmental Health, Atlanta, GA, 30322, USA
| | - Ann Rogers
- School of Nursing, Emory University, 1520 Clifton Road, 243, Atlanta, GA, 30322, USA
| | - Sandra B Dunbar
- School of Nursing, Emory University, 1520 Clifton Road, 243, Atlanta, GA, 30322, USA
| |
Collapse
|
13
|
Gonzalez-Covarrubias V, Martínez-Martínez E, del Bosque-Plata L. The Potential of Metabolomics in Biomedical Applications. Metabolites 2022; 12:metabo12020194. [PMID: 35208267 PMCID: PMC8880031 DOI: 10.3390/metabo12020194] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 12/12/2022] Open
Abstract
The metabolome offers a dynamic, comprehensive, and precise picture of the phenotype. Current high-throughput technologies have allowed the discovery of relevant metabolites that characterize a wide variety of human phenotypes with respect to health, disease, drug monitoring, and even aging. Metabolomics, parallel to genomics, has led to the discovery of biomarkers and has aided in the understanding of a diversity of molecular mechanisms, highlighting its application in precision medicine. This review focuses on the metabolomics that can be applied to improve human health, as well as its trends and impacts in metabolic and neurodegenerative diseases, cancer, longevity, the exposome, liquid biopsy development, and pharmacometabolomics. The identification of distinct metabolomic profiles will help in the discovery and improvement of clinical strategies to treat human disease. In the years to come, metabolomics will become a tool routinely applied to diagnose and monitor health and disease, aging, or drug development. Biomedical applications of metabolomics can already be foreseen to monitor the progression of metabolic diseases, such as obesity and diabetes, using branched-chain amino acids, acylcarnitines, certain phospholipids, and genomics; these can assess disease severity and predict a potential treatment. Future endeavors should focus on determining the applicability and clinical utility of metabolomic-derived markers and their appropriate implementation in large-scale clinical settings.
Collapse
Affiliation(s)
| | - Eduardo Martínez-Martínez
- Laboratory of Cell Communication and Extracellular Vesicles, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Laura del Bosque-Plata
- Laboratory of Nutrigenetics and Nutrigenomics, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
- Correspondence: ; Tel.: +52-55-53-50-1974
| |
Collapse
|
14
|
Lai W, Du D, Chen L. Metabolomics Provides Novel Insights into Epilepsy Diagnosis and Treatment: A Review. Neurochem Res 2022; 47:844-859. [PMID: 35067830 DOI: 10.1007/s11064-021-03510-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/04/2021] [Accepted: 12/14/2021] [Indexed: 02/05/2023]
Abstract
Epilepsy is one of the most common diseases of the central nervous system. The diagnosis of epilepsy mainly depends on electroencephalograms and symptomatology, while diagnostic biofluid markers are still lacking. In addition, approximately 30% of patients with epilepsy (PWE) show a poor response to the currently available anti-seizure medicines. An increasing number of studies have reported alterations in the blood, brain tissue, cerebrospinal fluid and urine metabolome in PWE and animal models of epilepsy. The aim of this review was to identify potential metabolic biomarkers and pathways that might facilitate diagnostic, therapeutic and prognostic determination in PWE and the understanding of the pathogenesis of the disease. The PubMed and Embase databases were searched for metabolomic studies of PWE and epileptic models published before December 2020. The study objectives, types of models and reported differentially altered metabolites were examined and compared. Pathway analyses were performed using MetaboAnalyst 5.0 online software. Thirty-five studies were included in this review. Metabolites such as glutamate, lactate and citrate were disturbed in both PWE and epileptic models, which might be potential biomarkers of epilepsy. Metabolic pathways including alanine, aspartate and glutamate metabolism; glycine, serine and threonine metabolism; glycerophospholipid metabolism; glyoxylate and dicarboxylate metabolism; and arginine and proline metabolism were involved in epilepsy. These pathways might play important roles in the pathogenesis of the disease. This review summarizes metabolites and metabolic pathways related to epilepsy and provides a novel perspective for the identification of potential biomarkers and therapeutic targets for epilepsy.
Collapse
Affiliation(s)
- Wanlin Lai
- Department of Neurology, West China Hospital of Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Dan Du
- West China-Washington Mitochondria and Metabolism Center, Advanced Mass Spectrometry Centre, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Lei Chen
- Department of Neurology, West China Hospital of Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
15
|
Chung MK, Smith MR, Lin Y, Walker DI, Jones D, Patel CJ, Kong SW. Plasma metabolomics of autism spectrum disorder and influence of shared components in proband families. EXPOSOME 2021; 1:osab004. [PMID: 35028569 PMCID: PMC8739333 DOI: 10.1093/exposome/osab004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 11/25/2022]
Abstract
Prevalence of autism spectrum disorder (ASD) has been increasing in the United States in the past decades. The exact mechanisms remain enigmatic, and diagnosis of the disease still relies primarily on assessment of behavior. We first used a case-control design (75 idiopathic cases and 29 controls, enrolled at Boston Children's Hospital from 2007-2012) to identify plasma biomarkers of ASD through a metabolome-wide association study approach. Then we leveraged a family-based design (31 families) to investigate the influence of shared genetic and environmental components on the autism-associated features. Using untargeted high-resolution mass spectrometry metabolomics platforms, we detected 19 184 features. Of these, 191 were associated with ASD (false discovery rate < 0.05). We putatively annotated 30 features that had an odds ratio (OR) between <0.01 and 5.84. An identified endogenous metabolite, O-phosphotyrosine, was associated with an extremely low autism odds (OR 0.17; 95% confidence interval 0.06-0.39). We also found that glutathione metabolism was associated with ASD (P = 0.048). Correlations of the significant features between proband and parents were low (median = 0.09). Of the 30 annotated features, the median correlations within families (proband-parents) were -0.15 and 0.24 for the endogenous and exogenous metabolites, respectively. We hypothesize that, without feature identification, family-based correlation analysis of autism-associated features can be an alternative way to assist the prioritization of potentially diagnostic features. A panel of ASD diagnostic metabolic markers with high specificity could be derived upon further studies.
Collapse
Affiliation(s)
- Ming Kei Chung
- Department of Biomedical Informatics, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Matthew Ryan Smith
- Division of Pulmonary Medicine, Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Yufei Lin
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dean Jones
- Division of Pulmonary Medicine, Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Sek Won Kong
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, USA
| |
Collapse
|
16
|
Husebye ESN, Riedel B, Bjørke-Monsen AL, Spigset O, Daltveit AK, Gilhus NE, Bjørk MH. Vitamin B status and association with antiseizure medication in pregnant women with epilepsy. Epilepsia 2021; 62:2968-2980. [PMID: 34590314 DOI: 10.1111/epi.17076] [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: 06/09/2021] [Revised: 08/23/2021] [Accepted: 09/07/2021] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Antiseizure medication (ASM) use interacts with vitamin B status in nonpregnant epilepsy populations. We aimed to examine the association between ASM and vitamin B status in pregnant women with epilepsy. METHODS We performed a cross-sectional study of pregnancies in women with epilepsy enrolled in the Norwegian Mother, Father and Child Cohort Study from 1999 to 2008. Data on ASM and vitamin supplement use were collected from questionnaires. We analyzed maternal plasma concentrations of ASM and metabolites of folate, including unmetabolized folic acid (UMFA), riboflavin (vitamin B2), pyridoxine (vitamin B6), and niacin (vitamin B3) during gestational weeks 17-19. RESULTS We included 227 singleton pregnancies exposed to ASM with available plasma samples (median maternal age 29 years, range 18 to 41 years). From the preconception period to gestational week 20, any supplement of folic acid was reported in 208 of pregnancies (94%), riboflavin in 72 (33%), pyridoxine in 77 (35%), and niacin in 45 (20%). High ASM concentrations correlated with high concentrations of UMFA and inactive folate metabolites, and with low concentrations of riboflavin and metabolically active pyridoxine. There was no association between ASM and niacin status. SIGNIFICANCE ASM concentrations during pregnancy were associated with vitamin B status in pregnant women with epilepsy. Additional studies are needed to determine the clinical impact of these findings, and to define the optimal vitamin doses that should be recommended to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Elisabeth Synnøve Nilsen Husebye
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Bettina Riedel
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anne-Lise Bjørke-Monsen
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Olav Spigset
- Department of Clinical Pharmacology, St. Olav University Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Kjersti Daltveit
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway.,Department of Health Registries, Norwegian Institute of Public Health, Bergen, Norway
| | - Nils Erik Gilhus
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Marte Helene Bjørk
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
17
|
Schmidt JC, Dougherty BV, Beger RD, Jones DP, Schmidt MA, Mattes WB. Metabolomics as a Truly Translational Tool for Precision Medicine. Int J Toxicol 2021; 40:413-426. [PMID: 34514887 DOI: 10.1177/10915818211039436] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metabolomics is unique among omics technologies in being applicable to metabolism and toxicity studies broadly across organisms (e.g., humans, other mammals, model organisms, and even bacteria) and across biological materials (e.g., blood, urine, saliva, biopsy, and stool), including cultured cells and subcellular fractions. Metabolomics can be used to characterize biologic response patterns in humans as well as to support mechanistic studies in model systems and ex vivo studies. A broad range of resources are available, including publicly accessible data repositories (e.g., Metabolomics Workbench), tools for biostatistics and bioinformatics (e.g., MetaboAnalyst), metabolite identification (e.g., Metlin), and pathway analysis (e.g., Kyoto Encyclopedia of Genes and Genomes). Thus, metabolomics is more than a promise of the future; metabolomics is already available as a translational approach to facilitate precision medicine. This ACT Symposium review will contain an introduction to metabolomics in toxicity studies followed by sections on translational metabolic networks, translational metabolite biomarkers of acetaminophen-induced acute liver injury, translational framework using high-resolution metabolomics for integrated pharmacokinetics and pharmacodynamics, and precision medicine applications: extracting actionable targets from untargeted metabolomics data following one year in space.
Collapse
Affiliation(s)
| | - Bonnie V Dougherty
- Department of Biomedical Engineering, 2358University of Virginia, Charlottesville, VA, USA
| | - Richard D Beger
- Division of Systems Biology, 4136National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, 1371Emory University School of Medicine, Atlanta, GA, USA
| | - Michael A Schmidt
- 466810Sovaris Aerospace, Boulder, CO, USA.,Advanced Pattern Analysis & Countermeasures Group, Boulder, CO, USA
| | - William B Mattes
- Division of Systems Biology, 4136National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
18
|
Tetro N, Hamed R, Berman E, Eyal S. Effects of antiseizure medications on placental cells: Focus on heterodimeric placental carriers. Epilepsy Res 2021; 174:106664. [PMID: 34022522 DOI: 10.1016/j.eplepsyres.2021.106664] [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] [Received: 03/01/2021] [Revised: 04/24/2021] [Accepted: 05/06/2021] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Appropriate placental nutrient transfer is essential for optimal fetal development. We have previously shown that antiseizure medications (ASMs) can alter the expression of placental carriers for folate and thyroid hormones. Here we extended our analysis to heterodimeric carriers that mediate the placental uptake of amino acids and antioxidant precursors. We focused on the L-type amino acid transporter (LAT)2/SLC7A8, the cystine/glutamate antiporter xCT/SLC7A11, and their chaperone 4F2hc/SLC3A2. METHODS BeWo cells were exposed for two or five days to therapeutic concentrations of valproate, levetiracetam, carbamazepine, lamotrigine, or lacosamide. Transcript levels were measured by quantitative PCR. Levetiracetam effects on placental carriers were further explored using a tailored gene array. RESULTS At five days, 30 μg/mL levetiracetam (high therapeutic concentrations) significantly reduced the expression of all studied genes (p < 0.05). Carbamazepine treatment was associated with lower SLC7A8 (LAT2) expression (p < 0.05), whereas valproate increased the transcript levels of this transporter by up to 2.0-fold (p < 0.01). Some of these effects were already observed after two incubation days. Lamotrigine did not alter gene expression, and lacosamide slightly elevated SLC3A2 levels (p < 0.05). The array analysis confirmed the trends observed for levetiracetam and identified additional affected genes. SIGNIFICANCE Altered expression of placental heterodimeric transporters may represent a mechanism by which ASM affect fetal development. The placental effects are differential, with valproate, carbamazepine and levetiracetam as the more active compounds. The concentration-dependence of those ASM effects are in line with established dose-dependent teratogenicity implying that ASM doses should be adjusted during pregnancy with caution.
Collapse
Affiliation(s)
- Nino Tetro
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Roua Hamed
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Erez Berman
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sara Eyal
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
19
|
Li Y, Zhang S, Snyder MP, Meador KJ. Precision medicine in women with epilepsy: The challenge, systematic review, and future direction. Epilepsy Behav 2021; 118:107928. [PMID: 33774354 PMCID: PMC8653993 DOI: 10.1016/j.yebeh.2021.107928] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/01/2021] [Accepted: 03/07/2021] [Indexed: 11/29/2022]
Abstract
Epilepsy is one of the most prevalent neurologic conditions, affecting almost 70 million people worldwide. In the United States, 1.3 million women with epilepsy (WWE) are in their active reproductive years. Women with epilepsy (WWE) face gender-specific challenges such as pregnancy, seizure exacerbation with hormonal pattern fluctuations, contraception, fertility, and menopause. Precision medicine, which applies state-of-the art molecular profiling to diagnostic, prognostic, and therapeutic problems, has the potential to advance the care of WWE by precisely tailoring individualized management to each patient's needs. For example, antiseizure medications (ASMs) are among the most common teratogens prescribed to women of childbearing potential. Teratogens act in a dose-dependent manner on a susceptible genotype. However, the genotypes at risk for ASM-induced teratogenic deficits are unknown. Here we summarize current challenging issues for WWE, review the state-of-art tools for clinical precision medicine approaches, perform a systematic review of pharmacogenomic approaches in management for WWE, and discuss potential future directions in this field. We envision a future in which precision medicine enables a new practice style that puts focus on early detection, prediction, and targeted therapies for WWE.
Collapse
Affiliation(s)
- Yi Li
- Department of Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Sai Zhang
- Stanford Center for Genomics and Personalized Medicine, Department of Genetics, Stanford University School of Medicine, Stanford CA, 94305, USA
| | - Michael P. Snyder
- Stanford Center for Genomics and Personalized Medicine, Department of Genetics, Stanford University School of Medicine, Stanford CA, 94305, USA
| | - Kimford J. Meador
- Department of Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
20
|
Bradner JM, Kalia V, Lau FK, Sharma M, Bucher ML, Johnson M, Chen M, Walker DI, Jones DP, Miller GW. Genetic or Toxicant-Induced Disruption of Vesicular Monoamine Storage and Global Metabolic Profiling in Caenorhabditis elegans. Toxicol Sci 2021; 180:313-324. [PMID: 33538833 PMCID: PMC8041460 DOI: 10.1093/toxsci/kfab011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The proper storage and release of monoamines contributes to a wide range of neuronal activity. Here, we examine the effects of altered vesicular monoamine transport in the nematode Caenorhabditis elegans. The gene cat-1 is responsible for the encoding of the vesicular monoamine transporter (VMAT) in C. elegans and is analogous to the mammalian vesicular monoamine transporter 2 (VMAT2). Our laboratory has previously shown that reduced VMAT2 activity confers vulnerability on catecholamine neurons in mice. The purpose of this article was to determine whether this function is conserved and to determine the impact of reduced VMAT activity in C. elegans. Here we show that deletion of cat-1/VMAT increases sensitivity to the neurotoxicant 1-methyl-4-phenylpyridinium (MPP+) as measured by enhanced degeneration of dopamine neurons. Reduced cat-1/VMAT also induces changes in dopamine-mediated behaviors. High-resolution mass spectrometry-based metabolomics in the whole organism reveals changes in amino acid metabolism, including tyrosine metabolism in the cat-1/VMAT mutants. Treatment with MPP+ disrupted tryptophan metabolism. Both conditions altered glycerophospholipid metabolism, suggesting a convergent pathway of neuronal dysfunction. Our results demonstrate the evolutionarily conserved nature of monoamine function in C. elegans and further suggest that high-resolution mass spectrometry-based metabolomics can be used in this model to study environmental and genetic contributors to complex human disease.
Collapse
Affiliation(s)
- Joshua M Bradner
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Fion K Lau
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Monica Sharma
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Michelle Johnson
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, USA
| | - Merry Chen
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Dean P Jones
- Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia 30303, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| |
Collapse
|
21
|
|
22
|
Saigusa D, Matsukawa N, Hishinuma E, Koshiba S. Identification of biomarkers to diagnose diseases and find adverse drug reactions by metabolomics. Drug Metab Pharmacokinet 2020; 37:100373. [PMID: 33631535 DOI: 10.1016/j.dmpk.2020.11.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Metabolomics has been widely used for investigating the biological functions of disease expression and has the potential to discover biomarkers in circulating biofluids or tissue extracts that reflect in phenotypic changes. Metabolic profiling has advantages because of the use of unbiased techniques, including multivariate analysis, and has been applied in pharmacological studies to predict therapeutic and adverse reactions of drugs, which is called pharmacometabolomics (PMx). Nuclear magnetic resonance (NMR)- and mass spectrometry (MS)-based metabolomics has contributed to the discovery of recent disease biomarkers; however, the optimal strategy for the study purpose must be selected from many established protocols, methodologies and analytical platforms. Additionally, information on molecular localization in tissue is essential for further functional analyses related to therapeutic and adverse effects of drugs in the process of drug development. MS imaging (MSI) is a promising technology that can visualize molecules on tissue surfaces without labeling and thus provide localized information. This review summarizes recent uses of MS-based global and wide-targeted metabolomics technologies and the advantages of the MSI approach for PMx and highlights the PMx technique for the biomarker discovery of adverse drug effects.
Collapse
Affiliation(s)
- Daisuke Saigusa
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Naomi Matsukawa
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Eiji Hishinuma
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan.
| | - Seizo Koshiba
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan; Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan.
| |
Collapse
|
23
|
Donatti A, Canto AM, Godoi AB, da Rosa DC, Lopes-Cendes I. Circulating Metabolites as Potential Biomarkers for Neurological Disorders-Metabolites in Neurological Disorders. Metabolites 2020; 10:E389. [PMID: 33003305 PMCID: PMC7601919 DOI: 10.3390/metabo10100389] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
There are, still, limitations to predicting the occurrence and prognosis of neurological disorders. Biomarkers are molecules that can change in different conditions, a feature that makes them potential tools to improve the diagnosis of disease, establish a prognosis, and monitor treatments. Metabolites can be used as biomarkers, and are small molecules derived from the metabolic process found in different biological media, such as tissue samples, cells, or biofluids. They can be identified using various strategies, targeted or untargeted experiments, and by different techniques, such as high-performance liquid chromatography, mass spectrometry, or nuclear magnetic resonance. In this review, we aim to discuss the current knowledge about metabolites as biomarkers for neurological disorders. We will present recent developments that show the need and the feasibility of identifying such biomarkers in different neurological disorders, as well as discuss relevant research findings in the field of metabolomics that are helping to unravel the mechanisms underlying neurological disorders. Although several relevant results have been reported in metabolomic studies in patients with neurological diseases, there is still a long way to go for the clinical use of metabolites as potential biomarkers in these disorders, and more research in the field is needed.
Collapse
Affiliation(s)
- Amanda Donatti
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Tessália Vieira de Camargo, 126 Cidade Universitária “Zeferino Vaz”, Campinas SP 13083-887, Brazil; (A.D.); (A.M.C.); (A.B.G.); (D.C.d.R.)
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas SP 13083-887, Brazil
| | - Amanda M. Canto
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Tessália Vieira de Camargo, 126 Cidade Universitária “Zeferino Vaz”, Campinas SP 13083-887, Brazil; (A.D.); (A.M.C.); (A.B.G.); (D.C.d.R.)
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas SP 13083-887, Brazil
| | - Alexandre B. Godoi
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Tessália Vieira de Camargo, 126 Cidade Universitária “Zeferino Vaz”, Campinas SP 13083-887, Brazil; (A.D.); (A.M.C.); (A.B.G.); (D.C.d.R.)
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas SP 13083-887, Brazil
| | - Douglas C. da Rosa
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Tessália Vieira de Camargo, 126 Cidade Universitária “Zeferino Vaz”, Campinas SP 13083-887, Brazil; (A.D.); (A.M.C.); (A.B.G.); (D.C.d.R.)
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas SP 13083-887, Brazil
| | - Iscia Lopes-Cendes
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Tessália Vieira de Camargo, 126 Cidade Universitária “Zeferino Vaz”, Campinas SP 13083-887, Brazil; (A.D.); (A.M.C.); (A.B.G.); (D.C.d.R.)
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas SP 13083-887, Brazil
| |
Collapse
|
24
|
Cioffi CE, Narayan KMV, Liu K, Uppal K, Jones DP, Tran V, Yu T, Alvarez JA, Bellissimo MP, Maner-Smith KM, Pierpoint B, Caprio S, Santoro N, Vos MB. Hepatic fat is a stronger correlate of key clinical and molecular abnormalities than visceral and abdominal subcutaneous fat in youth. BMJ Open Diabetes Res Care 2020; 8:e001126. [PMID: 32699106 PMCID: PMC7380953 DOI: 10.1136/bmjdrc-2019-001126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/27/2020] [Accepted: 05/13/2020] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Body fat distribution is strongly associated with cardiometabolic disease (CMD), but the relative importance of hepatic fat as an underlying driver remains unclear. Here, we applied a systems biology approach to compare the clinical and molecular subnetworks that correlate with hepatic fat, visceral fat, and abdominal subcutaneous fat distribution. RESEARCH DESIGN AND METHODS This was a cross-sectional sub-study of 283 children/adolescents (7-19 years) from the Yale Pediatric NAFLD Cohort. Untargeted, high-resolution metabolomics (HRM) was performed on plasma and combined with existing clinical variables including hepatic and abdominal fat measured by MRI. Integrative network analysis was coupled with pathway enrichment analysis and multivariable linear regression (MLR) to examine which metabolites and clinical variables associated with each fat depot. RESULTS The data divided into four communities of correlated variables (|r|>0.15, p<0.05) after integrative network analysis. In the largest community, hepatic fat was associated with eight clinical biomarkers, including measures of insulin resistance and dyslipidemia, and 878 metabolite features that were enriched predominantly in amino acid (AA) and lipid pathways in pathway enrichment analysis (p<0.05). Key metabolites associated with hepatic fat included branched-chain AAs (valine and isoleucine/leucine), aromatic AAs (tyrosine and tryptophan), serine, glycine, alanine, and pyruvate, as well as several acylcarnitines and glycerophospholipids (all q<0.05 in MLR adjusted for covariates). The other communities detected in integrative network analysis consisted of abdominal visceral, superficial subcutaneous, and deep subcutaneous fats, but no clinical variables, fewer metabolite features (280, 312, and 74, respectively), and limited findings in pathway analysis. CONCLUSIONS These data-driven findings show a stronger association of hepatic fat with key CMD risk factors compared with abdominal fats. The molecular network identified using HRM that associated with hepatic fat provides insight into potential mechanisms underlying the hepatic fat-insulin resistance interface in youth.
Collapse
Affiliation(s)
- Catherine E Cioffi
- Nutrition and Health Sciences, Emory University Laney Graduate School, Atlanta, Georgia, USA
| | - K M Venkat Narayan
- Hubert Department of Global Health, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Ken Liu
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Karan Uppal
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dean P Jones
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - ViLinh Tran
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Tianwei Yu
- Department of Biostatistics, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Jessica A Alvarez
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Moriah P Bellissimo
- Nutrition and Health Sciences, Emory University Laney Graduate School, Atlanta, Georgia, USA
| | - Kristal M Maner-Smith
- Emory Integrated Lipidomics Core, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bridget Pierpoint
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sonia Caprio
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nicola Santoro
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Molise, Italy
| | - Miriam B Vos
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
25
|
Ritz B, Yan Q, Uppal K, Liew Z, Cui X, Ling C, Inoue K, von Ehrenstein O, Walker DI, Jones DP. Untargeted Metabolomics Screen of Mid-pregnancy Maternal Serum and Autism in Offspring. Autism Res 2020; 13:1258-1269. [PMID: 32496662 DOI: 10.1002/aur.2311] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 03/24/2020] [Accepted: 04/15/2020] [Indexed: 12/15/2022]
Abstract
Discovering pathophysiologic networks in a blood-based approach may help to generate valuable tools for early treatment or preventive measures in autism. To date targeted or untargeted metabolomics approaches to identify metabolic features and pathways affecting fetal neurodevelopment have rarely been applied to pregnancy samples, that is, an early period potentially relevant for the development of autism spectrum disorders (ASD). We conducted a population-based study relying on autism diagnoses retrieved from California Department of Developmental Services record. After linking cases to and sampling controls from birth certificates, we retrieved stored maternal mid-pregnancy serum samples collected as part of the California Prenatal Screening Program from the California Biobank for children born 2004 to 2010 in the central valley of California. We retrieved serum for 52 mothers whose children developed autism and 62 population controls originally selected from all eligible children matched by birth year and child's sex. Also, we required that these mothers were relatively low or unexposed to air pollution and select pesticides during early pregnancy. We identified differences in metabolite levels in several metabolic pathways, including glycosphingolipid biosynthesis and metabolism, N-glycan and pyrimidine metabolism, bile acid pathways and, importantly, C21-steroid hormone biosynthesis and metabolism. Disturbances in these pathways have been shown to be relevant for neurodevelopment in rare genetic syndromes or implicated in previous studies of autism. This study provides new insight into maternal mid-pregnancy metabolic features possibly related to the development of autism and an incentive to explore whether these pathways and metabolites are useful for early diagnosis, treatment, or prevention. LAY SUMMARY: This study found that in mid-pregnancy the blood of mothers who give birth to a child that develops autism has some characteristic features that are different from those of blood samples taken from control mothers. These features are related to biologic mechanisms that can affect fetal brain development. In the future, these insights may help identify biomarkers for early autism diagnosis and treatment or preventive measures. Autism Res 2020, 13: 1258-1269. © 2020 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA.,Department of Neurology, UCLA School of Medicine, Los Angeles, California, USA
| | - Qi Yan
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Zeyan Liew
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA.,Yale Center for Perinatal, Pediatric, and Environmental Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Xin Cui
- Perinatal Epidemiology and Health Outcomes Research Unit, Division of Neonatology, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, California, USA.,California Perinatal Quality Care Collaborative, Palo Alto, California, USA
| | - Chenxiao Ling
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Kosuke Inoue
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Ondine von Ehrenstein
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
26
|
Segers K, Zhang W, Aourz N, Bongaerts J, Declerck S, Mangelings D, Hankemeier T, De Bundel D, Vander Heyden Y, Smolders I, Ramautar R, Van Eeckhaut A. CE-MS metabolic profiling of volume-restricted plasma samples from an acute mouse model for epileptic seizures to discover potentially involved metabolomic features. Talanta 2020; 217:121107. [PMID: 32498853 DOI: 10.1016/j.talanta.2020.121107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 01/07/2023]
Abstract
Currently, a high variety of analytical techniques to perform metabolomics is available. One of these techniques is capillary electrophoresis coupled to mass spectrometry (CE-MS), which has emerged as a rather strong analytical technique for profiling polar and charged compounds. This work aims to discover with CE-MS potential metabolic consequences of evoked seizures in plasma by using a 6Hz acute corneal seizure mouse model. CE-MS is an appealing technique because of its capability to handle very small sample volumes, such as the 10 μL plasma samples obtained using capillary microsampling in this study. After liquid-liquid extraction, the samples were analyzed with CE-MS using low-pH separation conditions, followed by data analysis and biomarker identification. Both electrically induced seizures showed decreased values of methionine, lysine, glycine, phenylalanine, citrulline, 3-methyladenine and histidine in mice plasma. However, a second provoked seizure, 13 days later, showed a less pronounced decrease of the mean concentrations of these plasma metabolites, demonstrated by higher fold change ratios. Other obtained markers that can be related to seizure activities based on literature data, are isoleucine, serine, proline, tryptophan, alanine, arginine, valine and asparagine. Most amino acids showed relatively stable plasma concentrations between the basal levels (Time point 1) and after the 13-day wash-out period (Time point 3), which suggests its effectiveness. Overall, this work clearly demonstrated the possibility of profiling metabolite consequences related to seizure activities of an intrinsically low amount of body fluid using CE-MS. It would be useful to investigate and validate, in the future, the known and unknown metabolites in different animal models as well as in humans.
Collapse
Affiliation(s)
- Karen Segers
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Wei Zhang
- Biomedical Microscale Analytics, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333, CC Leiden, the Netherlands.
| | - Najat Aourz
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Jana Bongaerts
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Sven Declerck
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Debby Mangelings
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Thomas Hankemeier
- Biomedical Microscale Analytics, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333, CC Leiden, the Netherlands.
| | - Dimitri De Bundel
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Yvan Vander Heyden
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Rawi Ramautar
- Biomedical Microscale Analytics, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333, CC Leiden, the Netherlands.
| | - Ann Van Eeckhaut
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| |
Collapse
|
27
|
Mussap M, Loddo C, Fanni C, Fanos V. Metabolomics in pharmacology - a delve into the novel field of pharmacometabolomics. Expert Rev Clin Pharmacol 2020; 13:115-134. [PMID: 31958027 DOI: 10.1080/17512433.2020.1713750] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Pharmacometabolomics is an emerging science pursuing the application of precision medicine. Combining both genetic and environmental factors, the so-called pharmacometabolomic approach guides patient selection and stratification in clinical trials and optimizes personalized drug dosage, improving efficacy and safety.Areas covered: This review illustrates the progressive introduction of pharmacometabolomics as an innovative solution for enhancing the discovery of novel drugs and improving research and development (R&D) productivity of the pharmaceutical industry. An extended analysis on published pharmacometabolomics studies both in animal models and humans includes results obtained in several areas such as hepatology, gastroenterology, nephrology, neuropsychiatry, oncology, drug addiction, embryonic cells, neonatology, and microbiomics.Expert opinion: a tailored, individualized therapy based on the optimization of pharmacokinetics and pharmacodynamics, the improvement of drug efficacy, and the abolition of drug toxicity and adverse drug reactions is a key issue in precision medicine. Genetics alone has become insufficient for deciphring intra- and inter-individual variations in drug-response, since they originate both from genetic and environmental factors, including human microbiota composition. The association between pharmacogenomics and pharmacometabolomics may be considered the new strategy for an in-deep knowledge on changes and alterations in human and microbial metabolic pathways due to the action of a drug.
Collapse
Affiliation(s)
- Michele Mussap
- Laboratory Unit, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | | | - Claudia Fanni
- Division of Pediatrics, Rovigo Hospital, Rovigo, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
28
|
Yan Q, Liew Z, Uppal K, Cui X, Ling C, Heck JE, von Ehrenstein OS, Wu J, Walker DI, Jones DP, Ritz B. Maternal serum metabolome and traffic-related air pollution exposure in pregnancy. ENVIRONMENT INTERNATIONAL 2019; 130:104872. [PMID: 31228787 PMCID: PMC7017857 DOI: 10.1016/j.envint.2019.05.066] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 05/19/2023]
Abstract
BACKGROUND Maternal exposure to traffic-related air pollution during pregnancy has been shown to increase the risk of adverse birth outcomes and neurodevelopmental disorders. By utilizing high-resolution metabolomics (HRM), we investigated perturbations of the maternal serum metabolome in response to traffic-related air pollution to identify biological mechanisms. METHODS We retrieved stored mid-pregnancy serum samples from 160 mothers who lived in the Central Valley of California known for high air particulate levels. We estimated prenatal traffic-related air pollution exposure (carbon monoxide, nitric oxides, and particulate matter <2.5 μm) during first-trimester using the California Line Source Dispersion Model, version 4 (CALINE4) based on residential addresses recorded at birth. We used liquid chromatography-high resolution mass spectrometry to obtain untargeted metabolic profiles and partial least squares discriminant analysis (PLS-DA) to select metabolic features associated with air pollution exposure. Pathway analyses were employed to identify biologic pathways related to air pollution exposure. As potential confounders we included maternal age, maternal race/ethnicity, and maternal education. RESULTS In total we extracted 4038 and 4957 metabolic features from maternal serum samples in hydrophilic interaction (HILIC) chromatography (positive ion mode) and C18 (negative ion mode) columns, respectively. After controlling for confounding factors, PLS-DA (Variable Importance in Projection (VIP) ≥2) yielded 181 and 251 metabolic features (HILIC and C18, respectively) that discriminated between the high (n = 98) and low exposed (n = 62). Pathway enrichment analysis for discriminatory features associated with air pollution indicated that in maternal serum oxidative stress and inflammation related pathways were altered, including linoleate, leukotriene, and prostaglandin pathways. CONCLUSION The metabolomic features and pathways we found to be associated with air pollution exposure suggest that maternal exposure during pregnancy induces oxidative stress and inflammation pathways previously implicated in pregnancy complications and adverse outcomes.
Collapse
Affiliation(s)
- Qi Yan
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Zeyan Liew
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA; Yale Center for Perinatal, Pediatric, and Environmental Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Xin Cui
- Perinatal Epidemiology and Health Outcomes Research Unit, Division of Neonatology, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA, USA; California Perinatal Quality Care Collaborative, Palo Alto, CA, USA
| | - Chenxiao Ling
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Julia E Heck
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | | | - Jun Wu
- Program in Public Health, UCI Susan and Henry Samueli College of Health Sciences, Irvine, CA, USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Department of Medicine, Emory University, Atlanta, GA, USA
| | - Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Neurology, UCLA School of Medicine, CA, USA.
| |
Collapse
|
29
|
Khusial RD, Cioffi CE, Caltharp SA, Krasinskas AM, Alazraki A, Knight-Scott J, Cleeton R, Castillo-Leon E, Jones DP, Pierpont B, Caprio S, Santoro N, Akil A, Vos MB. Development of a Plasma Screening Panel for Pediatric Nonalcoholic Fatty Liver Disease Using Metabolomics. Hepatol Commun 2019; 3:1311-1321. [PMID: 31592078 PMCID: PMC6771165 DOI: 10.1002/hep4.1417] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in children, but diagnosis is challenging due to limited availability of noninvasive biomarkers. Machine learning applied to high-resolution metabolomics and clinical phenotype data offers a novel framework for developing a NAFLD screening panel in youth. Here, untargeted metabolomics by liquid chromatography-mass spectrometry was performed on plasma samples from a combined cross-sectional sample of children and adolescents ages 2-25 years old with NAFLD (n = 222) and without NAFLD (n = 337), confirmed by liver biopsy or magnetic resonance imaging. Anthropometrics, blood lipids, liver enzymes, and glucose and insulin metabolism were also assessed. A machine learning approach was applied to the metabolomics and clinical phenotype data sets, which were split into training and test sets, and included dimension reduction, feature selection, and classification model development. The selected metabolite features were the amino acids serine, leucine/isoleucine, and tryptophan; three putatively annotated compounds (dihydrothymine and two phospholipids); and two unknowns. The selected clinical phenotype variables were waist circumference, whole-body insulin sensitivity index (WBISI) based on the oral glucose tolerance test, and blood triglycerides. The highest performing classification model was random forest, which had an area under the receiver operating characteristic curve (AUROC) of 0.94, sensitivity of 73%, and specificity of 97% for detecting NAFLD cases. A second classification model was developed using the homeostasis model assessment of insulin resistance substituted for the WBISI. Similarly, the highest performing classification model was random forest, which had an AUROC of 0.92, sensitivity of 73%, and specificity of 94%. Conclusion: The identified screening panel consisting of both metabolomics and clinical features has promising potential for screening for NAFLD in youth. Further development of this panel and independent validation testing in other cohorts are warranted.
Collapse
Affiliation(s)
- Richard D Khusial
- Department of Pharmaceutical Sciences, College of Pharmacy Mercer University Atlanta GA
| | - Catherine E Cioffi
- Nutrition and Health Sciences, Laney Graduate School Emory University Atlanta GA
| | - Shelley A Caltharp
- Children's Healthcare of Atlanta Atlanta GA.,Department of Pathology and Laboratory Medicine Emory University School of Medicine Atlanta GA
| | - Alyssa M Krasinskas
- Department of Pathology and Laboratory Medicine Emory University School of Medicine Atlanta GA
| | - Adina Alazraki
- Children's Healthcare of Atlanta Atlanta GA.,Department of Radiology Emory University School of Medicine Atlanta GA
| | | | - Rebecca Cleeton
- Department of Pediatrics Emory University School of Medicine Atlanta GA
| | | | - Dean P Jones
- Department of Medicine Emory University School of Medicine Atlanta GA
| | | | - Sonia Caprio
- Department of Pediatrics Yale School of Medicine New Haven CT
| | - Nicola Santoro
- Department of Pediatrics Yale School of Medicine New Haven CT
| | - Ayman Akil
- Department of Pharmaceutical Sciences, College of Pharmacy Mercer University Atlanta GA
| | - Miriam B Vos
- Nutrition and Health Sciences, Laney Graduate School Emory University Atlanta GA.,Children's Healthcare of Atlanta Atlanta GA.,Department of Pediatrics Emory University School of Medicine Atlanta GA
| |
Collapse
|
30
|
Walker DI, Marder ME, Yano Y, Terrell M, Liang Y, Barr DB, Miller GW, Jones DP, Marcus M, Pennell KD. Multigenerational metabolic profiling in the Michigan PBB registry. ENVIRONMENTAL RESEARCH 2019; 172:182-193. [PMID: 30782538 PMCID: PMC6534816 DOI: 10.1016/j.envres.2019.02.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/12/2019] [Accepted: 02/12/2019] [Indexed: 05/17/2023]
Abstract
Although polychlorinated biphenyls and polybrominated biphenyls are no longer manufactured the United States, biomonitoring in human populations show that exposure to these pollutants persist in human tissues. The objective of this study was to identify metabolic variations associated with exposure to 2,2'4,4',5,5'-hexabromobiphenyl (PBB-153) and 2,2'4,4',5,5'-hexachlorobiphenyl (PCB-153) in two generations of participants enrolled in the Michigan PBB Registry (http://pbbregistry.emory.edu/). Untargeted, high-resolution metabolomic profiling of plasma collected from 156 individuals was completed using liquid chromatography with high-resolution mass spectrometry. PBB-153 and PCB-153 levels were measured in the same individuals using targeted gas chromatography-tandem mass spectrometry and tested for dose-dependent correlation with the metabolome. Biological response to these exposures were evaluated using identified endogenous metabolites and pathway enrichment. When compared to lipid-adjusted concentrations for adults in the National Health and Nutrition Examination Survey (NHANES) for years 2003-2004, PCB-153 levels were consistent with similarly aged individuals, whereas PBB-153 concentrations were elevated (p<0.0001) in participants enrolled in the Michigan PBB Registry. Metabolic alterations were correlated with PBB-153 and PCB-153 in both generations of participants, and included changes in pathways related to catecholamine metabolism, cellular respiration, essential fatty acids, lipids and polyamine metabolism. These pathways were consistent with pathophysiological changes observed in neurodegenerative disease and included previously identified metabolomic markers of Parkinson's disease. To determine if the metabolic alterations detected in this study are replicated other cohorts, we evaluated correlation of PBB-153 and PCB-153 with plasma fatty acids measured in NHANES. Both pollutants showed similar associations with fatty acids previously linked to PCB exposure. Thus, the results from this study show metabolic alterations correlated with PBB-153 and PCB-153 exposure can be detected in human populations and are consistent with health outcomes previously reported in epidemiological and mechanistic studies.
Collapse
Affiliation(s)
- Douglas I Walker
- Department of Civil and Environmental Engineering, Tufts University, 200 College Ave, Medford MA 02155, United States; Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, 615 Michael St, Atlanta GA 30322, United States.
| | - M Elizabeth Marder
- Department of Environmental Health, Rollins School of Public Health, Emory University, 1518 Clifton Rd, Atlanta GA 30322, United States.
| | - Yukiko Yano
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, 50 University Ave Hall #7360, Berkeley CA 94720, United States.
| | - Metrecia Terrell
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd, Atlanta GA 30322, United States.
| | - Yongliang Liang
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, 615 Michael St, Atlanta GA 30322, United States.
| | - Dana Boyd Barr
- Department of Environmental Health, Rollins School of Public Health, Emory University, 1518 Clifton Rd, Atlanta GA 30322, United States.
| | - Gary W Miller
- Department of Environmental Health, Rollins School of Public Health, Emory University, 1518 Clifton Rd, Atlanta GA 30322, United States.
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, 615 Michael St, Atlanta GA 30322, United States.
| | - Michele Marcus
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd, Atlanta GA 30322, United States.
| | - Kurt D Pennell
- Department of Civil and Environmental Engineering, Tufts University, 200 College Ave, Medford MA 02155, United States.
| |
Collapse
|
31
|
|
32
|
Walker DI, Valvi D, Rothman N, Lan Q, Miller GW, Jones DP. The metabolome: A key measure for exposome research in epidemiology. CURR EPIDEMIOL REP 2019; 6:93-103. [PMID: 31828002 PMCID: PMC6905435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
PURPOSE OF REVIEW Application of omics to study human health has created a new era of opportunities for epidemiology research. However, approaches to characterize exogenous health triggers have largely not leveraged advances in analytical platforms and big data. In this review, we highlight the exposome, which is defined as the cumulative measure of exposure and biological responses across a lifetime as a cornerstone for new epidemiology approaches to study complex and preventable human diseases. RECENT FINDINGS While no universal approach exists to measure the entirety of the exposome, use of high-resolution mass spectrometry methods provide distinct advantages over traditional biomonitoring and have provided key advances necessary for exposome research. Application to different study designs and recommendations for combining exposome data with novel data analytic frameworks to study complex interactions of multiple stressors are also discussed. SUMMARY Even though challenges still need to be addressed, advances in methods to characterize the exposome provide exciting new opportunities for epidemiology to support fundamental discoveries to improve public health.
Collapse
Affiliation(s)
- Douglas I. Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Damaskini Valvi
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston MA, United States
| | - Nathaniel Rothman
- Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Qing Lan
- Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Gary W. Miller
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York NY
| | - Dean P. Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|