Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Edison Dong is active.

Publication


Featured researches published by Edison Dong.


Nucleic Acids Research | 2012

HMDB 3.0—The Human Metabolome Database in 2013

David S. Wishart; Timothy Jewison; Anchi Guo; Michael Wilson; Craig Knox; Yifeng Liu; Yannick Djoumbou; Rupasri Mandal; Farid Aziat; Edison Dong; Souhaila Bouatra; Igor Sinelnikov; David Arndt; Jianguo Xia; Philip Liu; Faizath S. Yallou; Trent C. Bjorndahl; Rolando Perez-Pineiro; Roman Eisner; Felicity Allen; Vanessa Neveu; Russell Greiner; Augustin Scalbert

The Human Metabolome Database (HMDB) (www.hmdb.ca) is a resource dedicated to providing scientists with the most current and comprehensive coverage of the human metabolome. Since its first release in 2007, the HMDB has been used to facilitate research for nearly 1000 published studies in metabolomics, clinical biochemistry and systems biology. The most recent release of HMDB (version 3.0) has been significantly expanded and enhanced over the 2009 release (version 2.0). In particular, the number of annotated metabolite entries has grown from 6500 to more than 40 000 (a 600% increase). This enormous expansion is a result of the inclusion of both ‘detected’ metabolites (those with measured concentrations or experimental confirmation of their existence) and ‘expected’ metabolites (those for which biochemical pathways are known or human intake/exposure is frequent but the compound has yet to be detected in the body). The latest release also has greatly increased the number of metabolites with biofluid or tissue concentration data, the number of compounds with reference spectra and the number of data fields per entry. In addition to this expansion in data quantity, new database visualization tools and new data content have been added or enhanced. These include better spectral viewing tools, more powerful chemical substructure searches, an improved chemical taxonomy and better, more interactive pathway maps. This article describes these enhancements to the HMDB, which was previously featured in the 2009 NAR Database Issue. (Note to referees, HMDB 3.0 will go live on 18 September 2012.).


Nucleic Acids Research | 2009

HMDB: a knowledgebase for the human metabolome

David S. Wishart; Craig Knox; Anchi Guo; Roman Eisner; Nelson Young; Bijaya Gautam; David Hau; Nick Psychogios; Edison Dong; Souhaila Bouatra; Rupasri Mandal; Igor Sinelnikov; Jianguo Xia; Leslie Jia; Joseph A. Cruz; Emilia Lim; Constance A. Sobsey; Savita Shrivastava; Paul Huang; Philip Liu; Lydia Fang; Jun Peng; Ryan Fradette; Dean Cheng; Dan Tzur; Melisa Clements; Avalyn Lewis; Andrea De Souza; Azaret Zuniga; Margot Dawe

The Human Metabolome Database (HMDB, http://www.hmdb.ca) is a richly annotated resource that is designed to address the broad needs of biochemists, clinical chemists, physicians, medical geneticists, nutritionists and members of the metabolomics community. Since its first release in 2007, the HMDB has been used to facilitate the research for nearly 100 published studies in metabolomics, clinical biochemistry and systems biology. The most recent release of HMDB (version 2.0) has been significantly expanded and enhanced over the previous release (version 1.0). In particular, the number of fully annotated metabolite entries has grown from 2180 to more than 6800 (a 300% increase), while the number of metabolites with biofluid or tissue concentration data has grown by a factor of five (from 883 to 4413). Similarly, the number of purified compounds with reference to NMR, LC-MS and GC-MS spectra has more than doubled (from 380 to more than 790 compounds). In addition to this significant expansion in database size, many new database searching tools and new data content has been added or enhanced. These include better algorithms for spectral searching and matching, more powerful chemical substructure searches, faster text searching software, as well as dedicated pathway searching tools and customized, clickable metabolic maps. Changes to the user-interface have also been implemented to accommodate future expansion and to make database navigation much easier. These improvements should make the HMDB much more useful to a much wider community of users.


PLOS ONE | 2011

The Human Serum Metabolome

Nikolaos Psychogios; David Hau; Jun Peng; An Chi Guo; Rupasri Mandal; Souhaila Bouatra; Igor Sinelnikov; Ramanarayan Krishnamurthy; Roman Eisner; Bijaya Gautam; Nelson Young; Jianguo Xia; Craig Knox; Edison Dong; Paul Huang; Zsuzsanna Hollander; Theresa L. Pedersen; Steven R. Smith; Fiona Bamforth; Russell Greiner; Bruce M. McManus; John W. Newman; Theodore L. Goodfriend; David S. Wishart

Continuing improvements in analytical technology along with an increased interest in performing comprehensive, quantitative metabolic profiling, is leading to increased interest pressures within the metabolomics community to develop centralized metabolite reference resources for certain clinically important biofluids, such as cerebrospinal fluid, urine and blood. As part of an ongoing effort to systematically characterize the human metabolome through the Human Metabolome Project, we have undertaken the task of characterizing the human serum metabolome. In doing so, we have combined targeted and non-targeted NMR, GC-MS and LC-MS methods with computer-aided literature mining to identify and quantify a comprehensive, if not absolutely complete, set of metabolites commonly detected and quantified (with todays technology) in the human serum metabolome. Our use of multiple metabolomics platforms and technologies allowed us to substantially enhance the level of metabolome coverage while critically assessing the relative strengths and weaknesses of these platforms or technologies. Tables containing the complete set of 4229 confirmed and highly probable human serum compounds, their concentrations, related literature references and links to their known disease associations are freely available at http://www.serummetabolome.ca.


PLOS ONE | 2013

The Human Urine Metabolome

Souhaila Bouatra; Farid Aziat; Rupasri Mandal; An Chi Guo; Michael Wilson; Craig Knox; Trent C. Bjorndahl; Ramanarayan Krishnamurthy; Fozia Saleem; Philip Liu; Zerihun T. Dame; Jenna Poelzer; Jessica Huynh; Faizath Yallou; Nick Psychogios; Edison Dong; Ralf Bogumil; Cornelia Roehring; David S. Wishart

Urine has long been a “favored” biofluid among metabolomics researchers. It is sterile, easy-to-obtain in large volumes, largely free from interfering proteins or lipids and chemically complex. However, this chemical complexity has also made urine a particularly difficult substrate to fully understand. As a biological waste material, urine typically contains metabolic breakdown products from a wide range of foods, drinks, drugs, environmental contaminants, endogenous waste metabolites and bacterial by-products. Many of these compounds are poorly characterized and poorly understood. In an effort to improve our understanding of this biofluid we have undertaken a comprehensive, quantitative, metabolome-wide characterization of human urine. This involved both computer-aided literature mining and comprehensive, quantitative experimental assessment/validation. The experimental portion employed NMR spectroscopy, gas chromatography mass spectrometry (GC-MS), direct flow injection mass spectrometry (DFI/LC-MS/MS), inductively coupled plasma mass spectrometry (ICP-MS) and high performance liquid chromatography (HPLC) experiments performed on multiple human urine samples. This multi-platform metabolomic analysis allowed us to identify 445 and quantify 378 unique urine metabolites or metabolite species. The different analytical platforms were able to identify (quantify) a total of: 209 (209) by NMR, 179 (85) by GC-MS, 127 (127) by DFI/LC-MS/MS, 40 (40) by ICP-MS and 10 (10) by HPLC. Our use of multiple metabolomics platforms and technologies allowed us to identify several previously unknown urine metabolites and to substantially enhance the level of metabolome coverage. It also allowed us to critically assess the relative strengths and weaknesses of different platforms or technologies. The literature review led to the identification and annotation of another 2206 urinary compounds and was used to help guide the subsequent experimental studies. An online database containing the complete set of 2651 confirmed human urine metabolite species, their structures (3079 in total), concentrations, related literature references and links to their known disease associations are freely available at http://www.urinemetabolome.ca.


Journal of Maternal-fetal & Neonatal Medicine | 2012

Metabolomics and first-trimester prediction of early-onset preeclampsia.

Ray O. Bahado-Singh; Ranjit Akolekar; Rupasri Mandal; Edison Dong; Jianguo Xia; Michael Kruger; David S. Wishart; Kypros H. Nicolaides

Objective: To evaluate the use of metabolomics for the first-trimester detection of maternal metabolic dysfunction and prediction of subsequent development of early-onset preeclampsia (PE). Study design: This was a case-control study of maternal plasma samples collected at 11–13 weeks’ gestation from 30 women who had subsequently developed PE requiring delivery before 34 weeks and 60 unaffected controls. Nuclear magnetic Resonance (NMR) spectroscopy was used to identify and quantify metabolomic changes in cases versus controls. Both genetic computing and standard statistical analyses were performed to predict the development of PE from the metabolite concentrations alone as well as the combination of metabolite concentrations with maternal characteristics and first-trimester uterine artery Doppler pulsatility index (PI). Results: Significant differences between cases and controls were found for 20 metabolites. A combination of four of these metabolites (citrate, glycerol, hydroxyisovalerate, and methionine) appeared highly predictive of PE with an estimated detection rate of 75.9%, at a false-positive rate (FPR) of 4.9%. The predictive performance was improved by the addition of uterine artery Doppler PI and fetal crown-rump length (CRL) and with an estimated detection rate of 82.6%, at a FPR of 1.6%. Conclusion: A profound change in the first-trimester metabolite profile was noted in women who had subsequently developed early-onset PE. Preliminary algorithms appeared highly sensitive for first trimester prediction of early onset PE.


American Journal of Obstetrics and Gynecology | 2013

First-trimester metabolomic detection of late-onset preeclampsia

Ray O. Bahado-Singh; Ranjit Akolekar; Rupasri Mandal; Edison Dong; Jianguo Xia; Michael Kruger; David S. Wishart; Kypros H. Nicolaides

OBJECTIVE We sought to identify first-trimester maternal serum biomarkers for the prediction of late-onset preeclampsia (PE) using metabolomic analysis. STUDY DESIGN In a case-control study, nuclear magnetic resonance-based metabolomic analysis was performed on first-trimester maternal serum between 11(+0)-13(+6) weeks of gestation. There were 30 cases of late-onset PE, i.e., requiring delivery ≥37 weeks, and 59 unaffected controls. The concentrations of 40 metabolites were compared between the 2 groups. We also compared 30 early-onset cases to the late-onset group. RESULTS A total of 14 metabolites were significantly elevated and 3 significantly reduced in first-trimester serum of late-onset PE patients. A complex model consisting of multiple metabolites and maternal demographic characteristics had a 76.6% sensitivity at 100% specificity for PE detection. A simplified model using fewer predictors yielded 60% sensitivity at 96.6% specificity. Strong separation of late- vs early-onset PE groups was achieved. CONCLUSION Significant differences in the first-trimester metabolites were noted in women who went on to developed late-onset PE and between early- and late-onset PE.


American Journal of Obstetrics and Gynecology | 2013

Metabolomic analysis for first-trimester Down syndrome prediction

Ray O. Bahado-Singh; Ranjit Akolekar; Rupasri Mandal; Edison Dong; Jianguo Xia; Michael Kruger; David S. Wishart; Kypros H. Nicolaides

OBJECTIVE The objective of the study was to perform first-trimester maternal serum metabolomic analysis and compare the results in aneuploid vs Down syndrome (DS) pregnancies. STUDY DESIGN This was a case-control study of pregnancies between 11+0 and 13+6 weeks. There were 30 DS cases and 60 controls in which first-trimester maternal serum was analyzed. Nuclear magnetic resonance-based metabolomic analysis was performed for DS prediction. RESULTS Concentrations of 11 metabolites were significantly different in the serum of DS pregnancies. The combination of 3-hydroxyisovalerate, 3-hydroxybuterate, and maternal age had a 51.9% sensitivity at 1.9% false-positive rate for DS detection. One multimarker algorithm had 70% sensitivity at 1.7% false-positive rate. Novel markers such as 3-hydroxybutyrate, involved in brain growth and myelination, and 2-hydroxybutyrate, involved in the defense against oxidative stress, were found to be abnormal. CONCLUSION The study reports novel metabolomic markers for the first-trimester prediction of fetal DS. Metabolomics provided insights into the cellular dysfunction in DS.


American Journal of Obstetrics and Gynecology | 2015

Validation of metabolomic models for prediction of early-onset preeclampsia.

Ray O. Bahado-Singh; Argyro Syngelaki; Ranjit Akolekar; Rupsari Mandal; Trent C. Bjondahl; Beomsoo Han; Edison Dong; Samuel T. Bauer; Zeynep Alpay-Savasan; Stewart F. Graham; Onur Turkoglu; David S. Wishart; Kypros H. Nicolaides

OBJECTIVE We sought to perform validation studies of previously published and newly derived first-trimester metabolomic algorithms for prediction of early preeclampsia (PE). STUDY DESIGN Nuclear magnetic resonance-based metabolomic analysis was performed on first-trimester serum in 50 women who subsequently developed early PE and in 108 first-trimester controls. Random stratification and allocation was used to divide cases into a discovery group (30 early PE and 65 controls) for generation of the biomarker model(s) and a validation group (20 early PE and 43 controls) to ensure an unbiased assessment of the predictive algorithms. Cross-validation testing on the different algorithms was performed to confirm their robustness before use. Metabolites, demographic features, clinical characteristics, and uterine Doppler pulsatility index data were evaluated. Area under the receiver operator characteristic curve (AUC), 95% confidence interval (CI), sensitivity, and specificity of the biomarker models were derived. RESULTS Validation testing found that the metabolite-only model had an AUC of 0.835 (95% CI, 0.769-0.941) with a 75% sensitivity and 74.4% specificity and for the metabolites plus uterine Doppler pulsatility index model it was 0.916 (95% CI, 0.836-0.996), 90%, and 88.4%, respectively. Predictive metabolites included arginine and 2-hydroxybutyrate, which are known to be involved in vascular dilation, and insulin resistance and impaired glucose regulation, respectively. CONCLUSION We found confirmatory evidence that first-trimester metabolomic biomarkers can predict future development of early PE.


Journal of Maternal-fetal & Neonatal Medicine | 2017

Metabolomic determination of pathogenesis of late-onset preeclampsia

Ray O. Bahado-Singh; Argyro Syngelaki; Rupsari Mandal; Stewart F. Graham; Ranjit Akolekar; Beomsoo Han; Trent C. Bjondahl; Edison Dong; Samuel T. Bauer; Zeynep Alpay-Savasan; Onur Turkoglu; Dotun Ogunyemi; Liona Poon; David S. Wishart; Kypros H. Nicolaides

Abstract Objective: Our primary objective was to apply metabolomic pathway analysis of first trimester maternal serum to provide an insight into the pathogenesis of late-onset preeclampsia (late-PE) and thereby identify plausible therapeutic targets for PE. Methods: NMR-based metabolomics analysis was performed on 29 cases of late-PE and 55 unaffected controls. In order to achieve sufficient statistical power to perform the pathway analysis, these cases were combined with a group of previously analyzed specimens, 30 late-PE cases and 60 unaffected controls. Specimens from both groups of cases and controls were collected in the same clinical centers during the same time period. In addition, NMR analyses were performed in the same lab and using the same techniques. Results: We identified abnormalities in branch chain amino acids (valine, leucine and isoleucine) and propanoate, glycolysis, gluconeogenesis and ketone body metabolic pathways. The results suggest insulin resistance and metabolic syndrome, mitochondrial dysfunction and disturbance of energy metabolism, oxidative stress and lipid dysfunction in the pathogenesis of late PE and suggest a potential role for agents that reduce insulin resistance in PE. Conclusions: Branched chain amino acids are known markers of insulin resistance and strongly predict future diabetes development. The analysis provides independent evidence linking insulin resistance and late-PE and suggests a potentially important therapeutic role for pharmacologic agents that reduce insulin resistance for late-PE.


Metabolites | 2018

Pasture Feeding Changes the Bovine Rumen and Milk Metabolome

Tom F. O’Callaghan; Rosa Vázquez-Fresno; Arnau Serra-Cayuela; Edison Dong; Rupasri Mandal; D. Hennessy; Stephen McAuliffe; P. Dillon; David S. Wishart; Catherine Stanton; R.P. Ross

The purpose of this study was to examine the effects of two pasture feeding systems—perennial ryegrass (GRS) and perennial ryegrass and white clover (CLV)—and an indoor total mixed ration (TMR) system on the (a) rumen microbiome; (b) rumen fluid and milk metabolome; and (c) to assess the potential to distinguish milk from different feeding systems by their respective metabolomes. Rumen fluid was collected from nine rumen cannulated cows under the different feeding systems in early, mid and late lactation, and raw milk samples were collected from ten non-cannulated cows in mid-lactation from each of the feeding systems. The microbiota present in rumen liquid and solid portions were analysed using 16S rRNA gene sequencing, while 1H-NMR untargeted metabolomic analysis was performed on rumen fluid and raw milk samples. The rumen microbiota composition was not found to be significantly altered by any feeding system in this study, likely as a result of a shortened adaptation period (two weeks’ exposure time). In contrast, feeding system had a significant effect on both the rumen and milk metabolome. Increased concentrations of volatile fatty acids including acetic acid, an important source of energy for the cow, were detected in the rumen of TMR and CLV-fed cows. Pasture feeding resulted in significantly higher concentrations of isoacids in the rumen. The ruminal fluids of both CLV and GRS-fed cows were found to have increased concentrations of p-cresol, a product of microbiome metabolism. CLV feeding resulted in increased rumen concentrations of formate, a substrate compound for methanogenesis. The TMR feeding resulted in significantly higher rumen choline content, which contributes to animal health and milk production, and succinate, a product of carbohydrate metabolism. Milk and rumen-fluids were shown to have varying levels of dimethyl sulfone in each feeding system, which was found to be an important compound for distinguishing between the diets. CLV feeding resulted in increased concentrations of milk urea. Milk from pasture-based feeding systems was shown to have significantly higher concentrations of hippuric acid, a potential biomarker of pasture-derived milk. This study has demonstrated that 1H-NMR metabolomics coupled with multivariate analysis is capable of distinguishing both rumen-fluid and milk derived from cows on different feeding systems, specifically between indoor TMR and pasture-based diets used in this study.

Collaboration


Dive into the Edison Dong's collaboration.

Top Co-Authors

Avatar

David S. Wishart

International Agency for Research on Cancer

View shared research outputs
Top Co-Authors

Avatar

Rupasri Mandal

Polytechnic University of Catalonia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianguo Xia

International Agency for Research on Cancer

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge