Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 8th International Conference and Exhibition on Metabolomics & Systems Biology Singapore .

Day 1 :

Keynote Forum

Scott J. Tebbutt

University of British Columbia & PROOF Centre of Excellence, Canada

Keynote: Integration of metabolomic and other OMIC datasets to gain insights in human health and disease
Biography:

Scott J Tebbutt is an Associate Professor in the Department of Medicine, University of British Columbia and Principal Investigator at the Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, Canada. His research program is focused on molecular signatures of complex respiratory disease, including the early and late reactions in atopic asthma and allergic rhinitis. He is also the Chief Scientific Officer of the Prevention of Organ Failure (PROOF) Centre of Excellence. His responsibilities include evaluating new, high-performance technologies to improve biomarker discovery, as well as computational biology approaches to better deal with cell type heterogeneity and integration of OMIC datasets. He holds BA in Biochemistry from the University of Oxford and PhD in Molecular Genetics from the University of East Anglia, Cambridge Laboratory at John Innes Centre.

Abstract:

Systems biology combines information from different molecular layers to provide a holistic view of a biological system and unravel its complexities. Integration of data across these layers requires methods that include multivariate approaches, Bayesian methods and network analyses. Although metabolomic analysis can provide a valuable ‘snapshot’ of biochemical processes at a high level, it is the functional relationships among various additional elements including genes, proteins and cells that will provide a deeper understanding of the biology. Indeed, any single OMICs approach is unlikely to suffice to characterize the complexity of biological processes in health and disease. We are developing innovative OMIC integration via functional bioinformatics and data-driven statistical approaches and computational modeling to identify signals in complex OMICs datasets regarding mechanisms that drive biological processes in a variety of human health and disease conditions.

Biography:

Kenneth K Wu has received his MD from National Taiwan University, Taipei, Taiwan, MS from Yale University New Haven, CT, USA and PhD in Pharmacology from University of London, UK. He was the Huffington Chair and Director of Hematology Division at University of Texas Health Science Center at Houston and Distinguished Investigator and Former President of National Health Research Institutes (NHRI), Taiwan. He is currently a Distinguished Professor and Director of Metabolomic Medicine Research Center at CMU, Taiwan. He holds Ho Jin Dui Chair at NTHU and Distinguished Chair at National Taiwan University, Taiwan. His research interests have centered on thrombosis, vascular biology and prostaglandin synthesis and regulation. He is a Member of several distinguished academic societies and has received international recognitions and awards.

Abstract:

Statement of the Problem: Overexpression of cyclooxygenase-2 (COX-2) and pro-inflammatory (PI) cytokines is a major contributor to human systemic inflammatory disorders and cancer metastasis. Expression of COX-2 and PI cytokines are regulated by transcriptional mechanisms, but it remained unclear whether it is endogenously controlled by small molecules.

Conceptual & Experimental Orientation: We suspected that human cells such as fibroblasts release soluble factors into the extracellular milieu which act in a paracrine manner to control COX-2 and PI cytokine expression. Soluble factors isolated from normal cells inhibit COX-2 overexpression in inflammatory and cancer cells. NMR analysis suggests that the soluble factors contain indole moiety. We named the factors cytoguardins. We subsequently found that cancer cells do not release cytoguardins. Comparative metabolomics analysis coupled with molecular genetics and biochemical studies identify 5-methoxytryptophan (5-MTP) as a cytoguardin. Endothelial cells (ECs) produce abundant 5-MTP which accounts for a high human serum level of 5-MTP. 5-MTP is a powerful innate factor defending against systemic inflammation and cancer cell metastasis. Compromised 5-MTP production due to insulting agents such as LPS results in COX-2 and cytokine storm, organ failure and death in animal models. Administration of 5-MTP attenuates cytokine storm and prevents organ failure and death. 5-MTP inhibits cancer cell migration and EMT in vitro and cancer metastasis in murine xenograft tumor model. 5-MTP exerts its biological actions in part by blocking p38 MAPK, p300 HAT and NF-kB activation.

Conclusion & Significance: Human cells such as fibroblasts and endothelial cells produce and release 5-MTP to maintain inflammatory homeostasis and control cancer metastasis by inhibiting the expression of COX-2 and PI genes. It represents a new class of protective molecules and a valuable lead compound for developing new drugs against systemic inflammatory disorders and cancer metastasis. 

  • Clinical Metabolomics & Lipidomics | Cancer Therapeutic Approaches | Metabolic Syndrome
Biography:

Takeshi Kimura is a Board Member and Corporate Vice President for Ajinomoto Co., Inc. and is currently In-Charge of Research and Development, Intellectual Property, Quality Assurance and Regulatory Affairs. He has studied Cell and Molecular Biology at University of London, Kings College and obtained his PhD in Biochemistry from University of London in 1984. He was a Visiting Fellow and Visiting Associate at the National Institutes of Health in the USA before joining Ajinomoto in 1989. He has worked in research, regulatory affairs and quality assurance since then, helping to establish the basis of AminoIndex Technology while in research. He became Corporate Executive Officer in 2009 and Board Member in 2013. He is also a Member of the Board of Trustees for International Life Sciences Institute and Research Foundation, International Advisory Council Member for Monell Chemical Senses Center and Japanese Private Sector Member for APEC Policy Partnership for Food Security. 

Abstract:

Early detection remains the most valuable tool in the fight against chronic diseases. Especially, the need for an early cancer screening method is evident as prognosis depends on how early treatment measures can be taken. Although various analytical platforms exist, currently there are only a few screening tests that have both high sensitivity and the ability for early detection, and also these tests must be taken for each individual cancer. Metabolomics have profound capacity in the search for such candidate markers in that we can obtain comprehensive knowledge and data of metabolisms under specific physiological states. From early observations indicating that amino acids were a convenient subset of the metabolome to investigate changes in metabolism associated with various physiological states, we have developed a technological package (AminoIndex technology) to generate biomarkers for various disease and physiological states using plasma free amino acid concentration data. Risk screening for various cancers with AminoIndex technology have been offered commercially in Japan since 2011. More recently, we have developed novel LC-MS platform that enables high-throughput metabolite profiling including over 40 amino acids, amines and their derivatives. Furthermore, we have constructed a large scale plasma biospecimen bank as well as database containing health and medical information of more than 50,000 Japanese in total concerning multiple diseases (e.g. cancers, metabolic syndrome etc.) and their metabolomic profiles. By variable selection and regression analysis, so far we have developed multivariate indices that enable the early detection of multiple cancers and predictive risk assessment for diabetes and inflammatory bowel disease from a single blood draw. In order to achieve commercialization, various issues ranging from sample handling, throughput to standardization have to be overcome and some of these issues, which may be relevant to other biomarker commercialization, will also be addressed.

Biography:

Yukio Fujiki and his colleagues investigate cellular homeostasis involving subcellular organelles such as peroxisome. His lab tackles the problems involving membrane assembly, matrix protein import, morphogenesis and homeostasis of peroxisomes. His group successfully identified and isolated more than a dozen PEX genes including the first Zellweger gene PEX2. He and his colleagues unveil the roles of peroxins in peroxisome biogenesis and the pathogenesis of PBDs from the viewpoint of dysregulation of peroxisome homeostasis.

Abstract:

Cellular homeostasis is regulated by orchestrating the functions of organelles in response to the extracellular stimuli and/or intracellular signals. To elucidate the highly organized functions of intracellular organelles, peroxisome, a single membrane-bounded essential organelle has been used as a model compartment in mammalian cells. Peroxisomes are present in a wide variety of eukaryotic cells and they function in various metabolic pathways, including β-oxidation of very long chain fatty acids and the synthesis of ether-lipids such as plasmalogens. The functional consequence of human peroxisomes is highlighted by fatal genetic peroxisome biogenesis disorders (PBD) such as Zellweger syndrome (ZS). We successfully isolated a dozen Chinese Hamster Ovary (CHO) cell mutants defective in peroxisome biogenesis and identified PEX genes encoding peroxisome biogenesis factors termed peroxins, including PEX2, PEX6, PEX12, PEX26, by means of the genetic phenotype-complementation of CHO cell mutants. We also unveiled the roles of peroxins in peroxisomal membrane assembly including targeting mechanism of nascent C-tailed anchored proteins, matrix protein import and division. We are now focusing on the peroxisomal membrane targeting mechanism of nascent C-tailed anchored proteins. Physiological consequence of plasmalogens is highlighted by PBDs. Ablation of plasmalogen homeostasis is reported in several neurological diseases including Alzheimer’s disease. In cells from PBD patents, plasmenylethanolamine is remarkably reduced and phosphatidylethanolamine is increased. We have shown that plasmalogen biosynthesis is regulated by modulating stability of fatty acyl-CoA reductase (Far1) and that plasmalogen homeostasis plays an important role in cholesterol synthesis. Moreover, plasmalogens located in the inner leaflet of plasma membrane are sensed for monitoring cellular plasmalogen level.

Biography:

Ng Sean Pin is the Deputy Director of Singapore Phenome Center. His interest is in the application of high-throughput technologies in advancing clinical and biological sciences. 

Abstract:

Phenomics is defined as the acquisition of high-dimensional phenotypic data on an organism-wide scale. In other words, phenomics is the study of the phenotypes of an organism and the response of the phenotypes to genetic and environmental changes.​ In recent years, -omics studies have become increasingly important due to their potential to increase our understanding of how environmental factors and diseases affect human health. This leads to improvement in the treatment or therapeutic strategies, ultimately resulting in improved healthcare and a higher standard of living. As such, the main research areas of the Singapore Phenome Centre (SPC) are in the clinical, biological and environmental sciences. These research studies are centered on the profiling of critical biomolecules such as metabolites, lipids and proteins through the use of the state-of-the-art ultra-performance liquid chromatography mass spectrometry (UPLC-MS), imaging mass spectrometry and NMR spectroscopy technologies available at the Singapore Phenome Centre at NTU. SPC aims to deliver a world-class competency in metabolic phenotyping research in association with local and international research institutions, hospitals and industry. The Centre, officially launched in 2015, is a member of the International Phenome Centre Network (IPCN) headed by Imperial College London’s (ICL) very own National Phenome Centre (NPC). This allows SPC to harmonize its research methods and technologies with centers that are part of the IPCN, which opens the path to building a global infrastructure around phenotyping. Currently the center houses 8 quadrupole time-of-flight hybrid tandem mass spectrometers, two triple quadrupole mass spectrometers and one 600 MHz nuclear magnetic resonance spectroscope. SPC has since been involved in collaborations with PIs from National Cancer Centre Singapore (SGH), IMB and IMCB (A*STAR), National Neuroscience Institute on Brain Cancer Imaging, National Institute of Education, and different schools in NTU (i.e. LKC Medicine, School of Biological Sciences, Singapore Centre on Environmental Life Sciences Engineering).

Biography:

Andrew M Chan is currently a Professor and Chief of the Cancer Biology & Experimental Therapeutics Thematic Research Program at the School of Biomedical Sciences of the Chinese University of Hong Kong. He has obtained his PhD degree from the Chester Beatty Laboratory of the Institute of Cancer Research in London. He was a Fogarty International Fellow at the US National Cancer Institute in the Laboratory of Cellular & Molecular Biology. He was a Faculty Member at the Mount Sinai School of Medicine and the Medical College of Wisconsin. His research interest is in the area of cancer cell signaling and mouse models of human cancers with focuses on the PI3-K and Ras-mediated signaling mechanisms in brain, breast and lung cancers.

Abstract:

Metastatic diseases in lung adenocarcinoma are often associated with poor prognosis. We have previously characterized a brain metastasis cell line of lung cancer origin, A1115, which possessed heightened glycolytic activity when compared with other lung adenocarcinoma cell lines such as, A549, derived from a primary site. A1115 has greater lactate production than A549. Conversely, A549 has a greater oxygen uptake rate that could be effectively blocked by oligomycin. Indeed, the proliferation of A549 was hypersensitive to oligomycin treatment while A1115 was greatly inhibited by glucose deprivation. Western blotting analysis revealed both hexokinase I (HK1) and phosphoenolpyruvate carboxykinase 1 (PCK1) were preferentially expressed in A1115. For energy sensing signaling, AMPKα, which senses cellular AMP level, was phosphorylated in A549 but not in A1115. Indeed, the proliferative capacity of A1115 was drastically reduced by an AMPK activator, AICAR, while A549 was not affected. Metabolite profiles under reduced 1 g/L revealed a 5- to 24-fold increase in metabolites that are linked to purine metabolism (e.g. hypoxanthine, GMP, and adenosine). Treatment with a XOD inhibitor, allopurinol under reduced glucose condition increased cell viability in A1115 at low doses while toxic at high doses. This data suggests that heightened hypoxanthine may be a prosurvival adaptive response in A1115 under nutrient stress conditions. We further selected A1115 sublines that could survive at a low glucose concentration (0.15 g/L). Western blotting analysis revealed a drastic overexpression of PCK1 and PKC2 when compared to parental cells. Transcriptomic analysis was also performed to reveal genes that could play a role in the survival of brain metastatic lung cancer cells under nutrient stresses. Taken together, these results identified multiple vulnerable points in metabolic signaling that could be therapeutic targets for treating metastatic lung cancers.

Paul M. Yen

Duke-NUS Medical School, Singapore

Title: Thyroid hormone regulation of autophagy and metabolism
Biography:

Paul M Yen is a Professor at Duke-NUS Graduate Medical School in Singapore and Head of the Laboratory of Hormonal Regulation in the Cardiovascular and Metabolic Disorders Program. He has obtained his MD from Johns Hopkins, completed his Residency in Internal Medicine at University of Chicago and his Endocrinology Fellowship at NIH. Prior to Duke-NUS, he has served on the Faculty at Johns Hopkins, Harvard and as a Section Chief at NIDDK, NIH. He has served on the Editorial Boards of Endocrinology, Molecular Endocrinology and Thyroid. His current research interests are hormonal regulation of hepatic autophagy and lipid metabolism in non-alcoholic fatty liver disease as well as epigenetic regulation of metabolic genes by thyroid hormone

Abstract:

Statement of the Problem: Thyroid hormone (TH) plays a key role of metabolism and its dysfunction may be involved in non-alcoholic fatty liver disease (NAFLD). We previously showed that TH induces autophagy in the liver to promote the degradation and hydrolysis of triglycerides stored in fat droplets into free fatty acids (Lipophagy). Metabolomics analysis showed that TH also stimulated fatty acid β-oxidation.

Findings: TH increased mitophagy in hepatic cells in vivo that was associated with increased oxidative phosphorylation. This was evident from co-localization of autophagy/autolysosomal markers and mitochondria using confocal and electron microscopy. Furthermore, we found that T3 induced a concomitant increase in ROS that was crucial for TH-induced mitophagy through the AMPK/ULK1 pathway. Additionally, we showed that T3 is a potent inducer of mitochondrial biosynthesis and utilizes that activation of another nuclear hormone receptor ERRa to mediate many of its actions.

Conclusion & Significance: Our results describe a novel co-ordinated mechanism of TH-induced fatty acid β-oxidation that is dependent upon mitochondrial turnover. These findings suggest that low dose Levothyroxine or thyroid hormone analogs may be beneficial for patients with hepatosteatosis related to NAFLD and obesity.

Eun Jin Yang

Korea Institute of Oriental Medicine, Republic of Korea

Title: The effect of bee venom on neuroinflammation in a vascular dementia animal model
Biography:

Eun Jin Yang has completed her PhD from Yonsei University and Postdoctoral Studies from the University of Pennsylvania, USA. She is a Principal Researcher at Korea Institute of Oriental Medicine, South Korea. She has published more than 25 papers in reputed journals and has been serving as an Editorial Board Member.

Abstract:

Vascular dementia (VaD) is caused by the reduction of blood supply by vessel occlusion and is characterized by progressive cognitive decline. VaD incidence has been growing due to the aging population, placing greater strain on social and economic resources. However, the pathological mechanisms underlying VaD remain unclear. Many studies have used the bilateral common carotid artery occlusion (BCCAO) animal model to investigate potential therapeutics for VaD. In this study, we investigated whether bee venom (BV) improves cognitive function and reduces neuroinflammation in the hippocampus of BCCAO animals. Animals were randomly divided into three groups: a sham group (n=15), BCCAO control group (n=15), and BV-treated BCCAO group (n=15). BCCAO animals were treated with 0.1 μg/g BV at ST36 (BJoksamli^ acupoint) four times every other day. In order to investigate the effect of BV treatment on cognitive function, we performed a Y-maze test. In order to uncover any potential relationship between these results and neuroinflammation, we also performed Western blotting in the BCCAO group. Animals that had been treated with BV, showed an improved cognitive function and a reduced expression of neuroinflammatory proteins in the hippocampus, including Iba-1, TLR4, CD14, and TNF-α. Furthermore, we demonstrated that BV treatment increased pERK and BDNF in the hippocampus. The present study thus underlines the neuroprotective effect of BV treatment against BCCAO induced cognitive impairment and neuroinflammation. Our findings suggest that BV may be an effective complementary treatment for VaD, as it may improve cognitive function and attenuate neuroinflammation associated with dementia.

Biography:

Mohamed A Elrayess has completed his PhD at University College London (UCL) in Cardiovascular Genetics in 2002. He has then studied the therapeutic utilization of hematopoietic stem cells in cardiovascular disease at the Department of Medicine in UCL. He has spent over 7 years working as a Stem Cell Scientist in Eisai Ltd, a major international pharmaceutical company, leading projects focusing on stem cell therapy in various neurodegenerative diseases. He currently occupies a Senior Scientist position at Anti-Doping Lab Qatar, where he leads projects focusing on the role of stem cells in diabetes and genetics and metabolomics of elite athletes.

Abstract:

Background & Aim: The elite performance of professional athletes is associated with alterations in their systemic metabolic profiling. The objectives of this study were to compare the metabolic profiling between low and high power and endurance elite athletes and to highlight the underlying metabolic pathways.

Methods: Sera from one 191 elite athletes who passed anti-doping laboratories’ tests were profiled using non-targeted metabolomics-based mass spectroscopy combined with ultrahigh-performance liquid and gas. Differences in metabolic signatures were compared between low and high power and endurance groups by OPLS-DA and regression models.

Results & Conclusions: Data reveal that high performance athletes show a distinct metabolic profile that reflects steroid biosynthesis, fatty acid metabolism, oxidative stress and energy-related metabolites. Differences in performance-related metabolic profiles could shed light on the biochemical processes associated with their elite performance and potentially be utilized as biomarkers for endurance or power trainability in athletic candidates.

  • Special Session
Biography:

Maximilian Zucker has studied Physics at the Technical University of Munich, Germany, Business Administration at the University of Hagen, Germany and received his PhD in Physics in 2000 at the University of Bonn, Germany. Prior to joining numares HEALTH AG, he was the Managing Director (COO) of Roche Multiplate and Management Consultant at various consulting companies.

Abstract:

Statement of the Problem: In recent years, metabolomics has become a powerful tool for discovering biomarkers for diagnostic purposes. Although numerous promising biomarker patterns have been reported for a broad range of disorders, only few have entered clinical routine. Our presentation aims at analyzing this paradox.

Theory & Practice: The detection of disease-specific biomarker patterns and their transfer from theory into clinical practice as diagnostic tools requires robust, accurate, reproducible and cost/time-efficient methods. Thus, numares has developed the AXINON System encompassing the Magnetic Group Signaling (MGS) technology allowing quantitative analyses of metabolites in body fluids by nuclear magnetic resonance (NMR) spectroscopy. This standardized NMR-analysis is combined with internal quality control algorithms and innovative biostatistics.

Clinical Application: By meeting all these above requirements, we present the in vitro discovery, implementation and validation of novel biomarkers in the fields of urological malignancies, atherosclerosis and kidney disease as application examples. The general workflow included NMR analysis of several hundred patient samples in three distinct cohorts per use-case. Spectra of a first use-case specific cohort (training sets) were evaluated to identify spectral regions discriminating patients from controls. Subsequently, metabolites within these regions were identified, validated and quantified by fitting algorithms. In order to increase diagnostic sensitivity and specificity, single marker candidates were combined into multi-marker patterns, which were selected by using the second cohorts (optimization cohort). The final patterns were validated in the third independent test cohorts.

Conclusion & Significance: In order to implement metabolomics-based diagnostic test into clinical routine, several technical and algorithmic hurdles have to be tackled. It will only be possible to overcome the above mentioned paradox if new skills and technologies are added to what is otherwise required in more classical diagnostic approaches.