51
|
Gumpenberger T, Brezina S, Keski-Rahkonen P, Baierl A, Robinot N, Leeb G, Habermann N, Kok DEG, Scalbert A, Ueland PM, Ulrich CM, Gsur A. Untargeted Metabolomics Reveals Major Differences in the Plasma Metabolome between Colorectal Cancer and Colorectal Adenomas. Metabolites 2021; 11:119. [PMID: 33669644 PMCID: PMC7922413 DOI: 10.3390/metabo11020119] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/09/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Sporadic colorectal cancer is characterized by a multistep progression from normal epithelium to precancerous low-risk and high-risk adenomas to invasive cancer. Yet, the underlying molecular mechanisms of colorectal carcinogenesis are not completely understood. Within the "Metabolomic profiles throughout the continuum of colorectal cancer" (MetaboCCC) consortium we analyzed data generated by untargeted, mass spectrometry-based metabolomics using plasma from 88 colorectal cancer patients, 200 patients with high-risk adenomas and 200 patients with low-risk adenomas recruited within the "Colorectal Cancer Study of Austria" (CORSA). Univariate logistic regression models comparing colorectal cancer to adenomas resulted in 442 statistically significant molecular features. Metabolites discriminating colorectal cancer patients from those with adenomas in our dataset included acylcarnitines, caffeine, amino acids, glycerophospholipids, fatty acids, bilirubin, bile acids and bacterial metabolites of tryptophan. The data obtained discovers metabolite profiles reflecting metabolic differences between colorectal cancer and colorectal adenomas and delineates a potentially underlying biological interpretation.
Collapse
Affiliation(s)
- Tanja Gumpenberger
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (T.G.); (S.B.)
| | - Stefanie Brezina
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (T.G.); (S.B.)
| | - Pekka Keski-Rahkonen
- International Agency for Research on Cancer, 69372 Lyon, France; (P.K.-R.); (N.R.); (A.S.)
| | - Andreas Baierl
- Department of Statistics and Operations Research, University of Vienna, 1090 Vienna, Austria;
| | - Nivonirina Robinot
- International Agency for Research on Cancer, 69372 Lyon, France; (P.K.-R.); (N.R.); (A.S.)
| | - Gernot Leeb
- Department of Internal Medicine, Hospital Oberpullendorf, 7350 Oberpullendorf, Austria;
| | - Nina Habermann
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Genome Biology, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Dieuwertje E G Kok
- Division of Human Nutrition and Health, Wageningen University & Research, 6708 Wageningen, The Netherlands;
| | - Augustin Scalbert
- International Agency for Research on Cancer, 69372 Lyon, France; (P.K.-R.); (N.R.); (A.S.)
| | | | - Cornelia M Ulrich
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA;
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84108, USA
| | - Andrea Gsur
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (T.G.); (S.B.)
| |
Collapse
|
52
|
Wu Z, Huang Z, Wu Y, Jin Y, Wang Y, Zhao H, Chen S, Wu S, Gao X. Risk stratification for mortality in cardiovascular disease survivors: A survival conditional inference tree analysis. Nutr Metab Cardiovasc Dis 2021; 31:420-428. [PMID: 33223407 DOI: 10.1016/j.numecd.2020.09.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/20/2020] [Accepted: 09/23/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND AND AIMS Efficient analysis strategies for complex network with cardiovascular disease (CVD) risk stratification remain lacking. We sought to identify an optimized model to study CVD prognosis using survival conditional inference tree (SCTREE), a machine-learning method. METHODS AND RESULTS We identified 5379 new onset CVD from 2006 (baseline) to May, 2017 in the Kailuan I study including 101,510 participants (the training dataset). The second cohort composing 1,287 CVD survivors was used to validate the algorithm (the Kailuan II study, n = 57,511). All variables (e.g., age, sex, family history of CVD, metabolic risk factors, renal function indexes, heart rate, atrial fibrillation, and high sensitivity C-reactive protein) were measured at baseline and biennially during the follow-up period. Up to December 2017, we documented 1,104 deaths after CVD in the Kailuan I study and 170 deaths in the Kailuan II study. Older age, hyperglycemia and proteinuria were identified by the SCTREE as main predictors of post-CVD mortality. CVD survivors in the high risk group (presence of 2-3 of these top risk factors), had higher mortality risk in the training dataset (hazard ratio (HR): 5.41; 95% confidence Interval (CI): 4.49-6.52) and in the validation dataset (HR: 6.04; 95%CI: 3.59-10.2), than those in the lowest risk group (presence of 0-1 of these factors). CONCLUSION Older age, hyperglycemia and proteinuria were the main predictors of post-CVD mortality. TRIAL REGISTRATION ChiCTR-TNRC-11001489.
Collapse
Affiliation(s)
- Zhijun Wu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhe Huang
- Department of Cardiology, Kailuan Hospital, Tangshan, China
| | - Yuntao Wu
- Department of Cardiology, Kailuan Hospital, Tangshan, China
| | - Yao Jin
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanxiu Wang
- Department of Cardiology, Kailuan Hospital, Tangshan, China
| | - Haiyan Zhao
- Department of Cardiology, Kailuan Hospital, Tangshan, China
| | - Shuohua Chen
- Health Care Center, Kailuan Medical Group, Tangshan, China
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital, Tangshan, China.
| | - Xiang Gao
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, USA.
| |
Collapse
|
53
|
Abstract
Advances in high-throughput biotechnologies have facilitated omics profiling, a key component of precision phenotyping, in patients with pulmonary vascular disease. Omics provides comprehensive information pertaining to genes, transcripts, proteins, and metabolites. The resulting omics big datasets may be integrated for more robust results and are amenable to analysis using machine learning or newer analytical methodologies, such as network analysis. Results from fully integrated multi-omics datasets combined with clinical data are poised to provide novel insight into pulmonary vascular disease as well as diagnose the presence of disease and prognosticate outcomes.
Collapse
Affiliation(s)
- Jane A Leopold
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB0630K, Boston, MA 02115, USA.
| | - Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, T1218 Medical Center North, 1161 21st Avenue South, Nashville, TN 37232, USA
| |
Collapse
|
54
|
Pal S, Mondal S, Das G, Khatua S, Ghosh Z. Big data in biology: The hope and present-day challenges in it. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
55
|
Perakakis N, Stefanakis K, Mantzoros CS. The role of omics in the pathophysiology, diagnosis and treatment of non-alcoholic fatty liver disease. Metabolism 2020; 111S:154320. [PMID: 32712221 PMCID: PMC7377759 DOI: 10.1016/j.metabol.2020.154320] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifaceted metabolic disorder, whose spectrum covers clinical, histological and pathophysiological developments ranging from simple steatosis to non-alcoholic steatohepatitis (NASH) and liver fibrosis, potentially evolving into cirrhosis, hepatocellular carcinoma and liver failure. Liver biopsy remains the gold standard for diagnosing NAFLD, while there are no specific treatments. An ever-increasing number of high-throughput Omics investigations on the molecular pathobiology of NAFLD at the cellular, tissue and system levels produce comprehensive biochemical patient snapshots. In the clinical setting, these applications are considerably enhancing our efforts towards obtaining a holistic insight on NAFLD pathophysiology. Omics are also generating non-invasive diagnostic modalities for the distinct stages of NAFLD, that remain though to be validated in multiple, large, heterogenous and independent cohorts, both cross-sectionally as well as prospectively. Finally, they aid in developing novel therapies. By tracing the flow of information from genomics to epigenomics, transcriptomics, proteomics, metabolomics, lipidomics and glycomics, the chief contributions of these techniques in understanding, diagnosing and treating NAFLD are summarized herein.
Collapse
Affiliation(s)
- Nikolaos Perakakis
- Department of Internal Medicine, Boston VA Healthcare system and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA..
| | - Konstantinos Stefanakis
- Department of Internal Medicine, Boston VA Healthcare system and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christos S Mantzoros
- Department of Internal Medicine, Boston VA Healthcare system and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
56
|
Vila IK, Fretaud M, Vlachakis D, Laguette N, Langevin C. Animal Models for the Study of Nucleic Acid Immunity: Novel Tools and New Perspectives. J Mol Biol 2020; 432:5529-5543. [PMID: 32860771 PMCID: PMC7611023 DOI: 10.1016/j.jmb.2020.08.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 02/08/2023]
Abstract
Unresolved inflammation fosters and supports a wide range of human pathologies. There is growing evidence for a role played by cytosolic nucleic acids in initiating and supporting pathological chronic inflammation. In particular, the cGAS-STING pathway has emerged as central to the mounting of nucleic acid-dependent type I interferon responses, leading to the identification of small-molecule modulators of STING that have raised clinical interest. However, several new challenges have emerged, representing potential obstacles to efficient clinical translation. Indeed, the current literature underscores that nucleic acid-induced inflammatory responses are subjected to several layers of regulation, further suggesting complex coordination at the cell-type, tissue or organism level. Untangling the underlying processes is paramount to the identification of specific therapeutic strategies targeting deleterious inflammation. Herein, we present an overview of human pathologies presenting with deregulated interferon levels and with accumulation of cytosolic nucleic acids. We focus on the central role of the STING adaptor protein in these pathologies and discuss how in vivo models have forged our current understanding of nucleic acid immunity. We present our opinion on the advantages and limitations of zebrafish and mice models to highlight their complementarity for the study of inflammatory human pathologies and the development of therapeutics. Finally, we discuss high-throughput screening strategies that generate multi-parametric datasets that allow integrative analysis of heterogeneous information (imaging and omics approaches). These approaches are likely to structure the future of screening strategies for the treatment of human pathologies.
Collapse
Affiliation(s)
- Isabelle K Vila
- Institut de Génétique Humaine, CNRS, Université de Montpellier, Molecular Basis of Inflammation Laboratory, Montpellier, France.
| | - Maxence Fretaud
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France
| | - Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; University Research Institute of Maternal and Child Health & Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nadine Laguette
- Institut de Génétique Humaine, CNRS, Université de Montpellier, Molecular Basis of Inflammation Laboratory, Montpellier, France
| | | |
Collapse
|
57
|
Ma A, McDermaid A, Xu J, Chang Y, Ma Q. Integrative Methods and Practical Challenges for Single-Cell Multi-omics. Trends Biotechnol 2020; 38:1007-1022. [PMID: 32818441 PMCID: PMC7442857 DOI: 10.1016/j.tibtech.2020.02.013] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/19/2022]
Abstract
Fast-developing single-cell multimodal omics (scMulti-omics) technologies enable the measurement of multiple modalities, such as DNA methylation, chromatin accessibility, RNA expression, protein abundance, gene perturbation, and spatial information, from the same cell. scMulti-omics can comprehensively explore and identify cell characteristics, while also presenting challenges to the development of computational methods and tools for integrative analyses. Here, we review these integrative methods and summarize the existing tools for studying a variety of scMulti-omics data. The various functionalities and practical challenges in using the available tools in the public domain are explored through several case studies. Finally, we identify remaining challenges and future trends in scMulti-omics modeling and analyses.
Collapse
Affiliation(s)
- Anjun Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43235, USA
| | - Adam McDermaid
- Imagenetics, Sanford Health, Sioux Falls, SD 57104, USA; Department of Internal Medicine, University of South Dakota, Virmillion, SD 57069, USA
| | - Jennifer Xu
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43235, USA; Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yuzhou Chang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43235, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43235, USA.
| |
Collapse
|
58
|
Aleksandrova K, Egea Rodrigues C, Floegel A, Ahrens W. Omics Biomarkers in Obesity: Novel Etiological Insights and Targets for Precision Prevention. Curr Obes Rep 2020; 9:219-230. [PMID: 32594318 PMCID: PMC7447658 DOI: 10.1007/s13679-020-00393-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Omics-based technologies were suggested to provide an advanced understanding of obesity etiology and its metabolic consequences. This review highlights the recent developments in "omics"-based research aimed to identify obesity-related biomarkers. RECENT FINDINGS Recent advances in obesity and metabolism research increasingly rely on new technologies to identify mechanisms in the development of obesity using various "omics" platforms. Genetic and epigenetic biomarkers that translate into changes in transcriptome, proteome, and metabolome could serve as targets for obesity prevention. Despite a number of promising candidate biomarkers, there is an increased demand for larger prospective cohort studies to validate findings and determine biomarker reproducibility before they can find applications in primary care and public health. "Omics" biomarkers have advanced our knowledge on the etiology of obesity and its links with chronic diseases. They bring substantial promise in identifying effective public health strategies that pave the way towards patient stratification and precision prevention.
Collapse
Affiliation(s)
- Krasimira Aleksandrova
- Nutrition, Immunity and Metabolism Senior Scientist Group, Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany.
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany.
| | - Caue Egea Rodrigues
- Nutrition, Immunity and Metabolism Senior Scientist Group, Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Anna Floegel
- Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
| | - Wolfgang Ahrens
- Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
- Faculty of Mathematics and Computer Science, University of Bremen, Bremen, Germany
| |
Collapse
|
59
|
Alfano R, Chadeau-Hyam M, Ghantous A, Keski-Rahkonen P, Chatzi L, Perez AE, Herceg Z, Kogevinas M, de Kok TM, Nawrot TS, Novoloaca A, Patel CJ, Pizzi C, Robinot N, Rusconi F, Scalbert A, Sunyer J, Vermeulen R, Vrijheid M, Vineis P, Robinson O, Plusquin M. A multi-omic analysis of birthweight in newborn cord blood reveals new underlying mechanisms related to cholesterol metabolism. Metabolism 2020; 110:154292. [PMID: 32553738 PMCID: PMC7450273 DOI: 10.1016/j.metabol.2020.154292] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Birthweight reflects in utero exposures and later health evolution. Despite existing studies employing high-dimensional molecular measurements, the understanding of underlying mechanisms of birthweight remains limited. METHODS To investigate the systems biology of birthweight, we cross-sectionally integrated the methylome, the transcriptome, the metabolome and a set of inflammatory proteins measured in cord blood samples, collected from four birth-cohorts (n = 489). We focused on two sets of 68 metabolites and 903 CpGs previously related to birthweight and investigated the correlation structures existing between these two sets and all other omic features via bipartite Pearson correlations. RESULTS This dataset revealed that the set of metabolome and methylome signatures of birthweight have seven signals in common, including three metabolites [PC(34:2), plasmalogen PC(36:4)/PC(O-36:5), and a compound with m/z of 781.0545], two CpGs (on the DHCR24 and SC4MOL gene), and two proteins (periostin and CCL22). CCL22, a macrophage-derived chemokine has not been previously identified in relation to birthweight. Since the results of the omics integration indicated the central role of cholesterol metabolism, we explored the association of cholesterol levels in cord blood with birthweight in the ENVIRONAGE cohort (n = 1097), finding that higher birthweight was associated with increased high-density lipoprotein cholesterol and that high-density lipoprotein cholesterol was lower in small versus large for gestational age newborns. CONCLUSIONS Our data suggests that an integration of different omic-layers in addition to single omics studies is a useful approach to generate new hypotheses regarding biological mechanisms. CCL22 and cholesterol metabolism in cord blood play a mechanistic role in birthweight.
Collapse
Affiliation(s)
- Rossella Alfano
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom; Medical Research Council-Health Protection Agency Centre for Environment and Health, Imperial College London, London, United Kingdom; Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Marc Chadeau-Hyam
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom; Medical Research Council-Health Protection Agency Centre for Environment and Health, Imperial College London, London, United Kingdom; Institute for Risk Assessment Sciences (IRAS), Division of Environmental Epidemiology, Utrecht University, Utrecht, the Netherlands
| | - Akram Ghantous
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008 Lyon, France
| | - Pekka Keski-Rahkonen
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008 Lyon, France
| | - Leda Chatzi
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90007, United States; Department of Social Medicine, University of Crete, Heraklion, Crete, Greece
| | - Almudena Espin Perez
- Department of Biomedical Informatics Research, Stanford University, CA, United States
| | - Zdenko Herceg
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008 Lyon, France
| | - Manolis Kogevinas
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain; ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain; Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Theo M de Kok
- Department of Toxicogenomics, Maastricht University, Maastricht, the Netherlands
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium; Environment & Health Unit, Leuven University, Leuven, Belgium
| | - Alexei Novoloaca
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008 Lyon, France
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, United States
| | - Costanza Pizzi
- Department of Medical Sciences, University of Turin and CPO-Piemonte, Torino, Italy
| | - Nivonirina Robinot
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008 Lyon, France
| | - Franca Rusconi
- Unit of Epidemiology, Anna Meyer Children's University Hospital, Florence, Italy
| | - Augustin Scalbert
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008 Lyon, France
| | - Jordi Sunyer
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain; Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Roel Vermeulen
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom; Institute for Risk Assessment Sciences (IRAS), Division of Environmental Epidemiology, Utrecht University, Utrecht, the Netherlands
| | - Martine Vrijheid
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain; ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Paolo Vineis
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom; Medical Research Council-Health Protection Agency Centre for Environment and Health, Imperial College London, London, United Kingdom; Human Genetic Foundation (HuGeF), Turin, Italy
| | - Oliver Robinson
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom
| | - Michelle Plusquin
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom; Medical Research Council-Health Protection Agency Centre for Environment and Health, Imperial College London, London, United Kingdom; Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
60
|
Patel SK, George B, Rai V. Artificial Intelligence to Decode Cancer Mechanism: Beyond Patient Stratification for Precision Oncology. Front Pharmacol 2020; 11:1177. [PMID: 32903628 PMCID: PMC7438594 DOI: 10.3389/fphar.2020.01177] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
The multitude of multi-omics data generated cost-effectively using advanced high-throughput technologies has imposed challenging domain for research in Artificial Intelligence (AI). Data curation poses a significant challenge as different parameters, instruments, and sample preparations approaches are employed for generating these big data sets. AI could reduce the fuzziness and randomness in data handling and build a platform for the data ecosystem, and thus serve as the primary choice for data mining and big data analysis to make informed decisions. However, AI implication remains intricate for researchers/clinicians lacking specific training in computational tools and informatics. Cancer is a major cause of death worldwide, accounting for an estimated 9.6 million deaths in 2018. Certain cancers, such as pancreatic and gastric cancers, are detected only after they have reached their advanced stages with frequent relapses. Cancer is one of the most complex diseases affecting a range of organs with diverse disease progression mechanisms and the effectors ranging from gene-epigenetics to a wide array of metabolites. Hence a comprehensive study, including genomics, epi-genomics, transcriptomics, proteomics, and metabolomics, along with the medical/mass-spectrometry imaging, patient clinical history, treatments provided, genetics, and disease endemicity, is essential. Cancer Moonshot℠ Research Initiatives by NIH National Cancer Institute aims to collect as much information as possible from different regions of the world and make a cancer data repository. AI could play an immense role in (a) analysis of complex and heterogeneous data sets (multi-omics and/or inter-omics), (b) data integration to provide a holistic disease molecular mechanism, (c) identification of diagnostic and prognostic markers, and (d) monitor patient's response to drugs/treatments and recovery. AI enables precision disease management well beyond the prevalent disease stratification patterns, such as differential expression and supervised classification. This review highlights critical advances and challenges in omics data analysis, dealing with data variability from lab-to-lab, and data integration. We also describe methods used in data mining and AI methods to obtain robust results for precision medicine from "big" data. In the future, AI could be expanded to achieve ground-breaking progress in disease management.
Collapse
Affiliation(s)
- Sandip Kumar Patel
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Bhawana George
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vineeta Rai
- Department of Entomology & Plant Pathology, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
61
|
Gkouskou K, Vlastos I, Karkalousos P, Chaniotis D, Sanoudou D, Eliopoulos AG. The "Virtual Digital Twins" Concept in Precision Nutrition. Adv Nutr 2020; 11:1405-1413. [PMID: 32770212 PMCID: PMC7666894 DOI: 10.1093/advances/nmaa089] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/15/2020] [Accepted: 07/06/2020] [Indexed: 12/13/2022] Open
Abstract
Nutritional and lifestyle changes remain at the core of healthy aging and disease prevention. Accumulating evidence underscores the impact of genetic, metabolic, and host gut microbial factors on individual responses to nutrients, paving the way for the stratification of nutritional guidelines. However, technological advances that incorporate biological, nutritional, lifestyle, and health data at an unprecedented scale and depth conceptualize a future where preventative dietary interventions will exceed stratification and will be highly individualized. We herein discuss how genetic information combined with longitudinal metabolomic, immune, behavioral, and gut microbial parameters, and bioclinical variables could define a digital replica of oneself, a "virtual digital twin," which could serve to guide nutrition in a personalized manner. Such a model may revolutionize the management of obesity and its comorbidities, and provide a pillar for healthy aging.
Collapse
Affiliation(s)
| | - Ioannis Vlastos
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros Karkalousos
- Department of Biomedical Sciences, University of West Attica, Athens, Greece
| | - Dimitrios Chaniotis
- Department of Biomedical Sciences, University of West Attica, Athens, Greece
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece,Center for New Biotechnologies and Precision Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece,Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | |
Collapse
|
62
|
Ghaffari MH, Jahanbekam A, Post C, Sadri H, Schuh K, Koch C, Sauerwein H. Discovery of different metabotypes in overconditioned dairy cows by means of machine learning. J Dairy Sci 2020; 103:9604-9619. [PMID: 32747103 DOI: 10.3168/jds.2020-18661] [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: 04/06/2020] [Accepted: 05/22/2020] [Indexed: 01/13/2023]
Abstract
Using data from targeted metabolomics in serum in combination with machine learning (ML) approaches, we aimed at (1) identifying divergent metabotypes in overconditioned cows and at (2) exploring how metabotypes are associated with lactation performance, blood metabolites, and hormones. In a previously established animal model, 38 pregnant multiparous Holstein cows were assigned to 2 groups that were fed differently to reach either high (HBCS) or normal (NBCS) body condition score (BCS) and backfat thickness (BFT) until dryoff at -49 d before calving [NBCS: BCS < 3.5 (3.02 ± 0.24) and BFT < 1.2 cm (0.92 ± 0.21), mean ± SD; HBCS: BCS > 3.75 (3.82 ± 0.33) and BFT > 1.4 cm (2.36 ± 0.35)]. Cows were then fed the same diets during the dry period and the subsequent lactation, and maintained the differences in BFT and BCS throughout the study. Blood samples were collected weekly from 7 wk antepartum (ap) to 12 wk postpartum (pp) to assess serum concentrations of metabolites (by targeted metabolomics and by classical analyses) and metabolic hormones. Metabolic clustering by applying 4 supervised ML-based classifiers [sequential minimal optimization (SMO), random forest (RF), alternating decision tree (ADTree), and naïve Bayes-updatable (NB)] on the changes (d 21 pp minus d 49 ap) in concentrations of 170 serum metabolites resulted in 4 distinct metabolic clusters: HBCS predicted HBCS (HBCS-PH, n = 13), HBCS predicted NBCS (HBCS-PN, n = 6), NBCS predicted NBCS (NBCS-PN, n = 15), and NBCS predicted HBCS (NBCS-PH, n = 4). The accuracies of SMO, RF, ADTree, and NB classifiers were >70%. Because the number of NBCS-PH cows was low, we did not consider this group for further comparisons. Dry matter intake (kg/d and percentage of body weight) and energy intake were greater in HBCS-PN than in HBCS-PH in early lactation, and HBCS-PN also reached a positive energy balance earlier than did HBCS-PH. Milk yield was not different between groups, but milk protein percentage was greater in HBCS-PN than in HBCS-PH cows. The circulating concentrations of fatty acids (FA) increased during early lactation in both groups, but HBCS-PN cows had lower concentrations of β-hydroxybutyrate, indicating lower ketogenesis compared with HBCS-PH cows. The concentrations of insulin, insulin-like growth factor 1, leptin, adiponectin, haptoglobin, glucose, and revised quantitative insulin sensitivity check index did not differ between the groups, whereas serum concentrations of glycerophospholipids were lower before calving in HBCS-PH than in HBCS-PN cows. Glycine was the only amino acid that had higher concentration after calving in HBCS-PH than in HBCS-PN cows. The circulating concentrations of some short- (C2, C3, and C4) and long-chain (C12, C16:0, C18:0, and C18:1) acylcarnitines on d 21 pp were greater in HBCS-PH than in HBCS-PN cows, indicating incomplete FA oxidation. In conclusion, the use of ML approaches involving data from targeted metabolomics in serum is a promising method for differentiating divergent metabotypes from apparently similar BCS phenotypes. Further investigations, using larger numbers of cows and farms, are warranted for confirmation of this finding.
Collapse
Affiliation(s)
- Morteza H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | | | - Christian Post
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - Hassan Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 516616471 Tabriz, Iran
| | - Katharina Schuh
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany; Department of Life Sciences and Engineering, Animal Nutrition and Hygiene Unit, University of Applied Sciences Bingen, 55411 Bingen am Rhein, Germany
| | - Christian Koch
- Educational and Research Centre for Animal Husbandry, Hofgut Neumühle, 67728 Münchweiler an der Alsenz, Germany
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| |
Collapse
|
63
|
Mahjoubin-Tehran M, De Vincentis A, Mikhailidis DP, Atkin SL, Mantzoros CS, Jamialahmadi T, Sahebkar A. Non-alcoholic fatty liver disease and steatohepatitis: State of the art on effective therapeutics based on the gold standard method for diagnosis. Mol Metab 2020; 50:101049. [PMID: 32673798 PMCID: PMC8324680 DOI: 10.1016/j.molmet.2020.101049] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/19/2020] [Accepted: 06/26/2020] [Indexed: 02/08/2023] Open
Abstract
Objective The prevalence of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis (NAFLD/NASH) is increasing. NAFLD/NASH may progress to cirrhosis and hepatocellular carcinoma. However, most patients with NAFLD/NASH will die from a vascular cause. There are no approved pharmacological treatments for NASH/NAFLD. Many clinical trials have been, or are being, undertaken; however, the challenge is the assessment of the clinical endpoint. The main objective of this narrative review was to evaluate the efficacy of drugs used in clinical trials for the treatment of NAFLD/NASH that included a liver biopsy as the gold standard. Methods A literature search was conducted using 3 databases (PubMed, Scopus, and Google Scholar) to identify the clinical trials that included liver biopsy assessment before and after treatment. Results Interventional clinical trials (n = 33) involving 18 different agents, alone and in combination, were identified. Pioglitazone is the only agent that has shown consistent benefit and efficacy in clinical trials. Pentoxifylline, rosiglitazone, and ursodeoxycholic acid had both positive and negative results from clinical trials. There is also evidence for vitamin E and metformin. Other drugs, including bicyclol, cysteamine bitartrate, l-carnitine, liraglutide, obeticholic acid, oligofructose, selonsertib, silymarin, and statins, each had a single clinical study. Conclusions In summary, the available molecules demonstrated a significant improvement in NASH and/or liver fibrosis in a minority of patients; thus, other drugs should be identified, possibly those acting on alternative pathophysiological pathways, and tested for their safety and efficacy. There are no currently approved pharmacological treatments for NASH/NAFLD. Confirmation of effective therapies for NAFLD/NASH is challenging due to the limitations of non-biopsy methods. We reviewed the efficacy of drugs used in NAFLD/NASH trials that included a liver biopsy as the gold standard.
Collapse
Affiliation(s)
- Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Antonio De Vincentis
- Clinical Medicine and Hepatology Unit, Campus Bio-Medico University of Rome, via Alvaro del Portillo, 200, 00128 Rome, Italy
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom
| | | | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA
| | - Tannaz Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.
| |
Collapse
|
64
|
Mudali D, Jeevanandam J, Danquah MK. Probing the characteristics and biofunctional effects of disease-affected cells and drug response via machine learning applications. Crit Rev Biotechnol 2020; 40:951-977. [PMID: 32633615 DOI: 10.1080/07388551.2020.1789062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Drug-induced transformations in disease characteristics at the cellular and molecular level offers the opportunity to predict and evaluate the efficacy of pharmaceutical ingredients whilst enabling the optimal design of new and improved drugs with enhanced pharmacokinetics and pharmacodynamics. Machine learning is a promising in-silico tool used to simulate cells with specific disease properties and to determine their response toward drug uptake. Differences in the properties of normal and infected cells, including biophysical, biochemical and physiological characteristics, plays a key role in developing fundamental cellular probing platforms for machine learning applications. Cellular features can be extracted periodically from both the drug treated, infected, and normal cells via image segmentations in order to probe dynamic differences in cell behavior. Cellular segmentation can be evaluated to reflect the levels of drug effect on a distinct cell or group of cells via probability scoring. This article provides an account for the use of machine learning methods to probe differences in the biophysical, biochemical and physiological characteristics of infected cells in response to pharmacokinetics uptake of drug ingredients for application in cancer, diabetes and neurodegenerative disease therapies.
Collapse
Affiliation(s)
- Deborah Mudali
- Department of Computer Science, University of Tennessee, Chattanooga, TN, USA
| | - Jaison Jeevanandam
- Department of Chemical Engineering, Faculty of Engineering and Science, Curtin University, Miri, Malaysia
| | - Michael K Danquah
- Chemical Engineering Department, University of Tennessee, Chattanooga, TN, USA
| |
Collapse
|
65
|
Castillo R, Scher JU. Not your average joint: Towards precision medicine in psoriatic arthritis. Clin Immunol 2020; 217:108470. [PMID: 32473975 DOI: 10.1016/j.clim.2020.108470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/31/2022]
Abstract
Precision medicine, propelled by advances in multi-omics methods and analytics, aims to revolutionize patient care by using clinically-actionable molecular markers to guide diagnostic and therapeutic decisions. We describe the applications of precision medicine in risk stratification, drug selection, and treatment response prediction in psoriatic arthritis, for which targeted, personalized approaches are steadily emerging.
Collapse
Affiliation(s)
- Rochelle Castillo
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, NY, United States of America
| | - Jose U Scher
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, NY, United States of America; Psoriatic Arthritis Center, New York University School of Medicine, New York, NY, United States of America.
| |
Collapse
|
66
|
Yamada R, Okada D, Wang J, Basak T, Koyama S. Interpretation of omics data analyses. J Hum Genet 2020; 66:93-102. [PMID: 32385339 PMCID: PMC7728595 DOI: 10.1038/s10038-020-0763-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/25/2020] [Accepted: 03/28/2020] [Indexed: 11/22/2022]
Abstract
Omics studies attempt to extract meaningful messages from large-scale and high-dimensional data sets by treating the data sets as a whole. The concept of treating data sets as a whole is important in every step of the data-handling procedures: the pre-processing step of data records, the step of statistical analyses and machine learning, translation of the outputs into human natural perceptions, and acceptance of the messages with uncertainty. In the pre-processing, the method by which to control the data quality and batch effects are discussed. For the main analyses, the approaches are divided into two types and their basic concepts are discussed. The first type is the evaluation of many items individually, followed by interpretation of individual items in the context of multiple testing and combination. The second type is the extraction of fewer important aspects from the whole data records. The outputs of the main analyses are translated into natural languages with techniques, such as annotation and ontology. The other technique for making the outputs perceptible is visualization. At the end of this review, one of the most important issues in the interpretation of omics data analyses is discussed. Omics studies have a large amount of information in their data sets, and every approach reveals only a very restricted aspect of the whole data sets. The understandable messages from these studies have unavoidable uncertainty.
Collapse
Affiliation(s)
- Ryo Yamada
- Unit of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Nanbusogo-Kenkyu-To-1, 5F, 53 Syogoin-Kawaramachi, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Daigo Okada
- Unit of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Nanbusogo-Kenkyu-To-1, 5F, 53 Syogoin-Kawaramachi, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Juan Wang
- Unit of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Nanbusogo-Kenkyu-To-1, 5F, 53 Syogoin-Kawaramachi, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tapati Basak
- Unit of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Nanbusogo-Kenkyu-To-1, 5F, 53 Syogoin-Kawaramachi, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Satoshi Koyama
- Unit of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Nanbusogo-Kenkyu-To-1, 5F, 53 Syogoin-Kawaramachi, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
67
|
Sahli-Costabal F, Seo K, Ashley E, Kuhl E. Classifying Drugs by their Arrhythmogenic Risk Using Machine Learning. Biophys J 2020; 118:1165-1176. [PMID: 32023435 DOI: 10.1101/545863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/27/2019] [Accepted: 01/13/2020] [Indexed: 05/25/2023] Open
Abstract
All medications have adverse effects. Among the most serious of these are cardiac arrhythmias. Current paradigms for drug safety evaluation are costly, lengthy, conservative, and impede efficient drug development. Here, we combine multiscale experiment and simulation, high-performance computing, and machine learning to create a risk estimator to stratify new and existing drugs according to their proarrhythmic potential. We capitalize on recent developments in machine learning and integrate information across 10 orders of magnitude in space and time to provide a holistic picture of the effects of drugs, either individually or in combination with other drugs. We show, both experimentally and computationally, that drug-induced arrhythmias are dominated by the interplay between two currents with opposing effects: the rapid delayed rectifier potassium current and the L-type calcium current. Using Gaussian process classification, we create a classifier that stratifies drugs into safe and arrhythmic domains for any combinations of these two currents. We demonstrate that our classifier correctly identifies the risk categories of 22 common drugs exclusively on the basis of their concentrations at 50% current block. Our new risk assessment tool explains under which conditions blocking the L-type calcium current can delay or even entirely suppress arrhythmogenic events. Using machine learning in drug safety evaluation can provide a more accurate and comprehensive mechanistic assessment of the proarrhythmic potential of new drugs. Our study paves the way toward establishing science-based criteria to accelerate drug development, design safer drugs, and reduce heart rhythm disorders.
Collapse
Affiliation(s)
| | - Kinya Seo
- Department of Medicine, Stanford University, Stanford, California
| | - Euan Ashley
- Department of Medicine, Stanford University, Stanford, California; Department of Pathology, Stanford University, Stanford, California
| | - Ellen Kuhl
- Department of Mechanical Engineering, Stanford University, Stanford, California; Department of Bioengineering, Stanford University, Stanford, California.
| |
Collapse
|
68
|
Sahli-Costabal F, Seo K, Ashley E, Kuhl E. Classifying Drugs by their Arrhythmogenic Risk Using Machine Learning. Biophys J 2020; 118:1165-1176. [PMID: 32023435 PMCID: PMC7063479 DOI: 10.1016/j.bpj.2020.01.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/27/2019] [Accepted: 01/13/2020] [Indexed: 12/17/2022] Open
Abstract
All medications have adverse effects. Among the most serious of these are cardiac arrhythmias. Current paradigms for drug safety evaluation are costly, lengthy, conservative, and impede efficient drug development. Here, we combine multiscale experiment and simulation, high-performance computing, and machine learning to create a risk estimator to stratify new and existing drugs according to their proarrhythmic potential. We capitalize on recent developments in machine learning and integrate information across 10 orders of magnitude in space and time to provide a holistic picture of the effects of drugs, either individually or in combination with other drugs. We show, both experimentally and computationally, that drug-induced arrhythmias are dominated by the interplay between two currents with opposing effects: the rapid delayed rectifier potassium current and the L-type calcium current. Using Gaussian process classification, we create a classifier that stratifies drugs into safe and arrhythmic domains for any combinations of these two currents. We demonstrate that our classifier correctly identifies the risk categories of 22 common drugs exclusively on the basis of their concentrations at 50% current block. Our new risk assessment tool explains under which conditions blocking the L-type calcium current can delay or even entirely suppress arrhythmogenic events. Using machine learning in drug safety evaluation can provide a more accurate and comprehensive mechanistic assessment of the proarrhythmic potential of new drugs. Our study paves the way toward establishing science-based criteria to accelerate drug development, design safer drugs, and reduce heart rhythm disorders.
Collapse
Affiliation(s)
| | - Kinya Seo
- Department of Medicine, Stanford University, Stanford, California
| | - Euan Ashley
- Department of Medicine, Stanford University, Stanford, California; Department of Pathology, Stanford University, Stanford, California
| | - Ellen Kuhl
- Department of Mechanical Engineering, Stanford University, Stanford, California; Department of Bioengineering, Stanford University, Stanford, California.
| |
Collapse
|
69
|
Banerjee S. Empowering Clinical Diagnostics with Mass Spectrometry. ACS OMEGA 2020; 5:2041-2048. [PMID: 32064364 PMCID: PMC7016904 DOI: 10.1021/acsomega.9b03764] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/21/2020] [Indexed: 05/20/2023]
Abstract
The unmet need for highly accurate methods of disease diagnosis poses new challenges for developments in laboratory medicine. Advances in mass spectrometry (MS)-based disease biomarker discoveries are continuously expanding the clinical diagnostic landscape. Although a number of MS-based in vitro diagnostics are already adopted in routine clinical practices, more are expected to undergo transition from bench to bedside in the near future. The ultrahigh sensitivity, specificity, and low turnaround time in molecular detection by MS make this technology highly powerful in disease detection and therapy monitoring. This mini-review highlights how MS has created a new paradigm in clinical diagnosis, which is growing in importance for public health.
Collapse
|
70
|
Boutari C, Bouzoni E, Joshi A, Stefanakis K, Farr OM, Mantzoros CS. Metabolism updates: new directions, techniques, and exciting research that is broadening the horizons. Metabolism 2020; 102:154009. [PMID: 31715175 DOI: 10.1016/j.metabol.2019.154009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Chrysoula Boutari
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Eirini Bouzoni
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Aditya Joshi
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Konstantinos Stefanakis
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Olivia M Farr
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA 02130, USA.
| |
Collapse
|
71
|
Perakakis N, Polyzos SA, Yazdani A, Sala-Vila A, Kountouras J, Anastasilakis AD, Mantzoros CS. Non-invasive diagnosis of non-alcoholic steatohepatitis and fibrosis with the use of omics and supervised learning: A proof of concept study. Metabolism 2019; 101:154005. [PMID: 31711876 DOI: 10.1016/j.metabol.2019.154005] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/23/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) affects 25-30% of the general population and is characterized by the presence of non-alcoholic fatty liver (NAFL) that can progress to non-alcoholic steatohepatitis (NASH), liver fibrosis and cirrhosis leading to hepatocellular carcinoma. To date, liver biopsy is the gold standard for the diagnosis of NASH and for staging liver fibrosis. This study aimed to train models for the non-invasive diagnosis of NASH and liver fibrosis based on measurements of lipids, glycans and biochemical parameters in peripheral blood and with the use of different machine learning methods. METHODS We performed a lipidomic, glycomic and free fatty acid analysis in serum samples of 49 healthy subjects and 31 patients with biopsy-proven NAFLD (15 with NAFL and 16 with NASH). The data from the above measurements combined with measurements of 4 hormonal parameters were analyzed with two different platforms and five different machine learning tools. RESULTS 365 lipids, 61 glycans and 23 fatty acids were identified with mass-spectrometry and liquid chromatography. Robust differences in the concentrations of specific lipid species were observed between healthy, NAFL and NASH subjects. One-vs-Rest (OvR) support vector machine (SVM) models with recursive feature elimination (RFE) including 29 lipids or combining lipids with glycans and/or hormones (20 or 10 variables total) could differentiate with very high accuracy (up to 90%) between the three conditions. In an exploratory analysis, a model consisting of 10 lipid species could robustly discriminate between the presence of liver fibrosis or not (98% accuracy). CONCLUSION We propose novel models utilizing lipids, hormones and glycans that can diagnose with high accuracy the presence of NASH, NAFL or healthy status. Additionally, we report a combination of lipids that can diagnose the presence of liver fibrosis. Both models should be further trained prospectively and validated in large independent cohorts.
Collapse
Affiliation(s)
- Nikolaos Perakakis
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Stergios A Polyzos
- First Department of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alireza Yazdani
- Division of Applied Mathematics, Brown University, Providence, RI 02906, USA
| | - Aleix Sala-Vila
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic of Barcelona, Villarroel 170, Barcelona 08036, Spain
| | - Jannis Kountouras
- Second Medical Clinic, Faculty of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | | | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
72
|
Anh NH, Long NP, Kim SJ, Min JE, Yoon SJ, Kim HM, Yang E, Hwang ES, Park JH, Hong SS, Kwon SW. Steroidomics for the Prevention, Assessment, and Management of Cancers: A Systematic Review and Functional Analysis. Metabolites 2019; 9:E199. [PMID: 31546652 PMCID: PMC6835899 DOI: 10.3390/metabo9100199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/09/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023] Open
Abstract
Steroidomics, an analytical technique for steroid biomarker mining, has received much attention in recent years. This systematic review and functional analysis, following the PRISMA statement, aims to provide a comprehensive review and an appraisal of the developments and fundamental issues in steroid high-throughput analysis, with a focus on cancer research. We also discuss potential pitfalls and proposed recommendations for steroidomics-based clinical research. Forty-five studies met our inclusion criteria, with a focus on 12 types of cancer. Most studies focused on cancer risk prediction, followed by diagnosis, prognosis, and therapy monitoring. Prostate cancer was the most frequently studied cancer. Estradiol, dehydroepiandrosterone, and cortisol were mostly reported and altered in at least four types of cancer. Estrogen and estrogen metabolites were highly reported to associate with women-related cancers. Pathway enrichment analysis revealed that steroidogenesis; androgen and estrogen metabolism; and androstenedione metabolism were significantly altered in cancers. Our findings indicated that estradiol, dehydroepiandrosterone, cortisol, and estrogen metabolites, among others, could be considered oncosteroids. Despite noble achievements, significant shortcomings among the investigated studies were small sample sizes, cross-sectional designs, potential confounding factors, and problematic statistical approaches. More efforts are required to establish standardized procedures regarding study design, analytical procedures, and statistical inference.
Collapse
Affiliation(s)
- Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | | | - Sun Jo Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Jung Eun Min
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Sang Jun Yoon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Hyung Min Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Eugine Yang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea.
| | - Eun Sook Hwang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea.
| | - Jeong Hill Park
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea.
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
73
|
Farr OM, Pilitsi E, Mantzoros CS. Of mice and men: incretin actions in the central nervous system. Metabolism 2019; 98:121-135. [PMID: 31173757 DOI: 10.1016/j.metabol.2019.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023]
Abstract
Incretins have risen to the forefront of therapies for obesity and related metabolic complications, primarily because of their efficacy and relatively few side effects. Importantly, their efficacy in altering energy balance and decreasing body weight is apparently through actions in the central nervous system (CNS); the latter may have implications beyond obesity per se, i.e. in other disease states associated with obesity including CNS-related disorders. Here, we first describe the role of the CNS in energy homeostasis and then the current state of knowledge in terms of incretin physiology, pathophysiology and efficacy in preclinical and clinical studies. In the future, more clinical studies are needed to fully map mechanistic pathways underlying incretin actions and outcomes in the human CNS. Additionally, future research will likely lead to the discovery of additional novel incretins and/or more efficacious medications with less side effects through the improvement of current compounds with properties that would allow them to have more favorable pharmacokinetic and pharmacodynamic profiles and/or by combining known and novel incretins into safe and more efficacious combination therapies leading ultimately to more tangible benefits for our patients.
Collapse
Affiliation(s)
- Olivia M Farr
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, United States of America.
| | - Eleni Pilitsi
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, United States of America
| | - Christos S Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, United States of America; Section of Endocrinology, VA Boston Healthcare System, Boston, MA 02130, United States of America
| |
Collapse
|
74
|
Katsiki N, Perakakis N, Mantzoros C. Effects of sodium-glucose co-transporter-2 (SGLT2) inhibitors on non-alcoholic fatty liver disease/non-alcoholic steatohepatitis: Ex quo et quo vadimus? Metabolism 2019; 98:iii-ix. [PMID: 31301336 DOI: 10.1016/j.metabol.2019.07.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Niki Katsiki
- Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Nikolaos Perakakis
- Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Christos Mantzoros
- Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
75
|
Wong YKE, Lam KW, Ho KY, Yu CSA, Cho CSW, Tsang HF, Chu MKM, Ng PWL, Tai CSW, Chan LWC, Wong EYL, Wong SCC. The applications of big data in molecular diagnostics. Expert Rev Mol Diagn 2019; 19:905-917. [PMID: 31422710 DOI: 10.1080/14737159.2019.1657834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yin Kwan Evelyn Wong
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| | - Ka Wai Lam
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| | - Ka Yi Ho
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| | | | - Chi Shing William Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region
| | - Hin Fung Tsang
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| | - Man Kee Maggie Chu
- Department of Life Science, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region
| | - Po Wah Lawrence Ng
- Department of Pathology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region
| | - Chi Shing William Tai
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| | - Lawrence Wing Chi Chan
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| | - Elaine Yue Ling Wong
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| | - Sze Chuen Cesar Wong
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| |
Collapse
|
76
|
Boutari C, Polyzos SA, Mantzoros CS. Of mice and men: Why progress in the pharmacological management of obesity is slower than anticipated and what could be done about it? Metabolism 2019; 96:vi-xi. [PMID: 30910448 DOI: 10.1016/j.metabol.2019.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/16/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Chrysoula Boutari
- Second Propedeutic Department of Internal Medicine, Faculty of Medicine, Aristotle University, Hippokration Hospital, Thessaloniki, Greece; Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Stergios A Polyzos
- First Department of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Section of Endocrinology, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
77
|
Papathanasiou AE, Nolen-Doerr E, Farr OM, Mantzoros CS. GEOFFREY HARRIS PRIZE LECTURE 2018: Novel pathways regulating neuroendocrine function, energy homeostasis and metabolism in humans. Eur J Endocrinol 2019; 180:R59-R71. [PMID: 30475221 PMCID: PMC6378110 DOI: 10.1530/eje-18-0847] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022]
Abstract
The discovery of leptin, an adipocyte-secreted hormone, set the stage for unraveling the mechanisms dictating energy homeostasis, revealing adipose tissue as an endocrine system that regulates appetite and body weight. Fluctuating leptin levels provide molecular signals to the brain regarding available energy reserves modulating energy homeostasis and neuroendocrine response in states of leptin deficiency and to a lesser extent in hyperleptinemic states. While leptin replacement therapy fails to provide substantial benefit in common obesity, it is an effective treatment for congenital leptin deficiency and states of acquired leptin deficiency such as lipodystrophy. Current evidence suggests that regulation of eating behavior in humans is not limited to homeostatic mechanisms and that the reward, attention, memory and emotion systems are involved, participating in a complex central nervous system network. It is critical to study these systems for the treatment of typical obesity. Although progress has been made, further studies are required to unravel the physiology, pathophysiology and neurobehavioral mechanisms underlying potential treatments for weight-related problems in humans.
Collapse
Affiliation(s)
| | - Eric Nolen-Doerr
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215
| | - Olivia M. Farr
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215
| | - Christos S. Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215
| |
Collapse
|
78
|
Kras KA, Langlais PR, Hoffman N, Roust LR, Benjamin TR, De Filippis EA, Dinu V, Katsanos CS. Obesity modifies the stoichiometry of mitochondrial proteins in a way that is distinct to the subcellular localization of the mitochondria in skeletal muscle. Metabolism 2018; 89:18-26. [PMID: 30253140 PMCID: PMC6221946 DOI: 10.1016/j.metabol.2018.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/01/2018] [Accepted: 09/19/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Skeletal muscle mitochondrial content and function appear to be altered in obesity. Mitochondria in muscle are found in well-defined regions within cells, and they are arranged in a way that form distinct subpopulations of subsarcolemmal (SS) and intermyofibrillar (IMF) mitochondria. We sought to investigate differences in the proteomes of SS and IMF mitochondria between lean subjects and subjects with obesity. METHODS We performed comparative proteomic analyses on SS and IMF mitochondria isolated from muscle samples obtained from lean subjects and subjects with obesity. Mitochondria were isolated using differential centrifugation, and proteins were subjected to label-free quantitative tandem mass spectrometry analyses. Collected data were evaluated for abundance of mitochondrial proteins using spectral counting. The Reactome pathway database was used to determine metabolic pathways that are altered in obesity. RESULTS Among proteins, 73 and 41 proteins showed different (mostly lower) expression in subjects with obesity in the SS and IMF mitochondria, respectively (false discovery rate-adjusted P ≤ 0.05). We specifically found an increase in proteins forming the tricarboxylic acid cycle and electron transport chain (ETC) complex II, but a decrease in proteins forming protein complexes I and III of the ETC and adenosine triphosphate (ATP) synthase in subjects with obesity in the IMF, but not SS, mitochondria. Obesity was associated with differential effects on metabolic pathways linked to protein translation in the SS mitochondria and ATP formation in the IMF mitochondria. CONCLUSIONS Obesity alters the expression of mitochondrial proteins regulating key metabolic processes in skeletal muscle, and these effects are distinct to mitochondrial subpopulations located in different regions of the muscle fibers. TRIAL REGISTRATION ClinicalTrials.gov (NCT01824173).
Collapse
Affiliation(s)
- Katon A Kras
- Center for Metabolic and Vascular Biology, Arizona State University, Scottsdale, AZ 85259, United States of America
| | - Paul R Langlais
- College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America
| | - Nyssa Hoffman
- Center for Metabolic and Vascular Biology, Arizona State University, Scottsdale, AZ 85259, United States of America
| | - Lori R Roust
- College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America
| | - Tonya R Benjamin
- College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America
| | - Elena A De Filippis
- College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America
| | - Valentin Dinu
- Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ 85259, United States of America
| | - Christos S Katsanos
- Center for Metabolic and Vascular Biology, Arizona State University, Scottsdale, AZ 85259, United States of America; College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America.
| |
Collapse
|