1
|
Baskaran K, Moshkovich M, Hart L, Shah N, Chowdhury F, Shanmuganathan M, Britz-McKibbin P, Pai N. The role of urine metabolomics in the diagnosis and management of adult and pediatric Crohn's disease and ulcerative colitis. Biomarkers 2024:1-10. [PMID: 39642943 DOI: 10.1080/1354750x.2024.2438734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Urine metabolomics offers a non-invasive approach to diagnose and manage inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), by identifying distinct metabolic signatures. OBJECTIVES This narrative review summarizes current findings on urinary metabolites in IBD, evaluating their roles in disease differentiation, assessment of activity, and monitoring therapeutic response. METHODS A comprehensive literature search of PubMed and MEDLINE up to October 2023 was conducted using keywords, such as 'urine metabolomics', 'inflammatory bowel disease', 'Crohn's disease', 'ulcerative colitis', and 'urinary biomarkers'. Studies were included that described alterations to metabolic pathways, including those related to the urea cycle, central energy metabolism (Krebs cycle), amino acid metabolism, and neurotransmitters. RESULTS Specific urinary metabolites differentiate IBD patients from healthy controls and between CD and UC. Decreased urinary levels of hippurate, acetate, methanol, formate, and methylamine are observed in IBD, indicating altered gut microbiota. In CD patients, urea cycle alterations include reduced urinary urea and ornithine with increased arginine. Changes in Krebs cycle intermediates show decreased citrate and succinate in adults, but increased fumarate and isocitrate in pediatric patients, reflecting energy metabolism differences. Amino acid metabolism differs by age: Adults exhibit decreased urinary asparagine, lysine, and histidine, while pediatric patients show increased methionine, proline, aspartic acid, and isoleucine. Elevated urinary neurotransmitters like dopamine are noted in pediatric IBD patients. Urine metabolomics also can monitor treatment efficacy by distinguishing responders from non-responders to therapies and differentiating active disease from remission. CONCLUSION Urine metabolomics provides promising, non-invasive biomarkers to enhance IBD diagnostics by distinguishing CD from UC and offering insights into underlying metabolic disturbances, paving the way for more precise, accessible patient care.
Collapse
Affiliation(s)
- Kanish Baskaran
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michal Moshkovich
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lara Hart
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Nyah Shah
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Fariha Chowdhury
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Meera Shanmuganathan
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Nikhil Pai
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, McMaster Children's Hospital, Hamilton, Ontario, Canada
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Lambert A, Budinich M, Mahé M, Chaffron S, Eveillard D. Community metabolic modeling of host-microbiota interactions through multi-objective optimization. iScience 2024; 27:110092. [PMID: 38952683 PMCID: PMC11215293 DOI: 10.1016/j.isci.2024.110092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/12/2024] [Accepted: 05/21/2024] [Indexed: 07/03/2024] Open
Abstract
The human gut microbiota comprises various microorganisms engaged in intricate interactions among themselves and with the host, affecting its health. While advancements in omics technologies have led to the inference of clear associations between microbiome composition and health conditions, we usually lack a causal and mechanistic understanding of these associations. For modeling mechanisms driving the interactions, we simulated the organism's metabolism using in silico genome-scale metabolic models (GEMs). We used multi-objective optimization to predict and explain metabolic interactions among gut microbes and an intestinal epithelial cell. We developed a score integrating model simulation results to predict the type (competition, neutralism, mutualism) and quantify the interaction between several organisms. This framework uncovered a potential cross-feeding for choline, explaining the predicted mutualism between Lactobacillus rhamnosus GG and the epithelial cell. Finally, we analyzed a five-organism ecosystem, revealing that a minimal microbiota can favor the epithelial cell's maintenance.
Collapse
Affiliation(s)
- Anna Lambert
- Nantes Université, École Centrale Nantes, CNRS, LS2N, UMR 6004, 44000 Nantes, France
| | - Marko Budinich
- Nantes Université, École Centrale Nantes, CNRS, LS2N, UMR 6004, 44000 Nantes, France
| | - Maxime Mahé
- Nantes Université, Inserm, TENS UMR1235, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Samuel Chaffron
- Nantes Université, École Centrale Nantes, CNRS, LS2N, UMR 6004, 44000 Nantes, France
| | - Damien Eveillard
- Nantes Université, École Centrale Nantes, CNRS, LS2N, UMR 6004, 44000 Nantes, France
| |
Collapse
|
3
|
Heinken A, El Kouche S, Guéant-Rodriguez RM, Guéant JL. Towards personalized genome-scale modeling of inborn errors of metabolism for systems medicine applications. Metabolism 2024; 150:155738. [PMID: 37981189 DOI: 10.1016/j.metabol.2023.155738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
Inborn errors of metabolism (IEMs) are a group of more than 1000 inherited diseases that are individually rare but have a cumulative global prevalence of 50 per 100,000 births. Recently, it has been recognized that like common diseases, patients with rare diseases can greatly vary in the manifestation and severity of symptoms. Here, we review omics-driven approaches that enable an integrated, holistic view of metabolic phenotypes in IEM patients. We focus on applications of Constraint-based Reconstruction and Analysis (COBRA), a widely used mechanistic systems biology approach, to model the effects of inherited diseases. Moreover, we review evidence that the gut microbiome is also altered in rare diseases. Finally, we outline an approach using personalized metabolic models of IEM patients for the prediction of biomarkers and tailored therapeutic or dietary interventions. Such applications could pave the way towards personalized medicine not just for common, but also for rare diseases.
Collapse
Affiliation(s)
- Almut Heinken
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France.
| | - Sandra El Kouche
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France
| | - Rosa-Maria Guéant-Rodriguez
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France; National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, Nancy F-54000, France
| | - Jean-Louis Guéant
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France; National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, Nancy F-54000, France
| |
Collapse
|
4
|
Sridhar S, Bhalla P, Kullu J, Veerapaneni S, Sahoo S, Bhatt N, Suraishkumar GK. A reactive species reactions module for integration into genome-scale metabolic models for improved insights: Application to cancer. Metab Eng 2023; 80:78-93. [PMID: 37689259 DOI: 10.1016/j.ymben.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/07/2023] [Accepted: 08/28/2023] [Indexed: 09/11/2023]
Abstract
Reactive species (RS) play significant roles in many disease contexts. Despite their crucial roles in diseases including cancer, the RS are not adequately modeled in the genome-scale metabolic (GSM) models, which are used to understand cell metabolism in disease contexts. We have developed a scalable RS reactions module that can be integrated with any Recon 3D-derived human metabolic model, or after fine-tuning, with any metabolic model. With RS-integration, the GSM models of three cancers (basal-like triple negative breast cancer (TNBC), high grade serous ovarian carcinoma (HGSOC) and colorectal cancer (CRC)) built from Recon 3D, precisely highlighted the increases/decreases in fluxes (dysregulation) occurring in important pathways of these cancers. These dysregulations were not prominent in the standard cancer models without the RS module. Further, the results from these RS-integrated cancer GSM models suggest the following decreasing order in the ease of ferroptosis-targeting to treat the cancers: TNBC > HGSOC > CRC.
Collapse
Affiliation(s)
- Subasree Sridhar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences Building - 1 and 2, Indian Institute of Technology Madras, Chennai, 600 036, India; Centre for Integrative Biology and Systems medicinE, Indian Institute of Technology Madras, Chennai, 600 036, India
| | - Prerna Bhalla
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences Building - 1 and 2, Indian Institute of Technology Madras, Chennai, 600 036, India
| | - Justin Kullu
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences Building - 1 and 2, Indian Institute of Technology Madras, Chennai, 600 036, India
| | - Sriya Veerapaneni
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences Building - 1 and 2, Indian Institute of Technology Madras, Chennai, 600 036, India
| | - Swagatika Sahoo
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, 600 036, India
| | - Nirav Bhatt
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences Building - 1 and 2, Indian Institute of Technology Madras, Chennai, 600 036, India; Centre for Integrative Biology and Systems medicinE, Indian Institute of Technology Madras, Chennai, 600 036, India; Robert Bosch Centre for Data Science and Artificial Intelligence, Indian Institute of Technology Madras, Chennai, 600 036, India.
| | - G K Suraishkumar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences Building - 1 and 2, Indian Institute of Technology Madras, Chennai, 600 036, India.
| |
Collapse
|
5
|
Sen P, Orešič M. Integrating Omics Data in Genome-Scale Metabolic Modeling: A Methodological Perspective for Precision Medicine. Metabolites 2023; 13:855. [PMID: 37512562 PMCID: PMC10383060 DOI: 10.3390/metabo13070855] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Recent advancements in omics technologies have generated a wealth of biological data. Integrating these data within mathematical models is essential to fully leverage their potential. Genome-scale metabolic models (GEMs) provide a robust framework for studying complex biological systems. GEMs have significantly contributed to our understanding of human metabolism, including the intrinsic relationship between the gut microbiome and the host metabolism. In this review, we highlight the contributions of GEMs and discuss the critical challenges that must be overcome to ensure their reproducibility and enhance their prediction accuracy, particularly in the context of precision medicine. We also explore the role of machine learning in addressing these challenges within GEMs. The integration of omics data with GEMs has the potential to lead to new insights, and to advance our understanding of molecular mechanisms in human health and disease.
Collapse
Affiliation(s)
- Partho Sen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 702 81 Örebro, Sweden
| | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 702 81 Örebro, Sweden
| |
Collapse
|
6
|
Lamichhane S, Sen P, Dickens AM, Alves MA, Härkönen T, Honkanen J, Vatanen T, Xavier RJ, Hyötyläinen T, Knip M, Orešič M. Dysregulation of secondary bile acid metabolism precedes islet autoimmunity and type 1 diabetes. Cell Rep Med 2022; 3:100762. [PMID: 36195095 PMCID: PMC9589006 DOI: 10.1016/j.xcrm.2022.100762] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/28/2022] [Accepted: 09/10/2022] [Indexed: 11/13/2022]
Abstract
The gut microbiota is crucial in the regulation of bile acid (BA) metabolism. However, not much is known about the regulation of BAs during progression to type 1 diabetes (T1D). Here, we analyzed serum and stool BAs in longitudinal samples collected at 3, 6, 12, 18, 24, and 36 months of age from children who developed a single islet autoantibody (AAb) (P1Ab; n = 23) or multiple islet AAbs (P2Ab; n = 13) and controls (CTRs; n = 38) who remained AAb negative. We also analyzed the stool microbiome in a subgroup of these children. Factor analysis showed that age had the strongest impact on both BA and microbiome profiles. We found that at an early age, systemic BAs and microbial secondary BA pathways were altered in the P2Ab group compared with the P1Ab and CTR groups. Our findings thus suggest that dysregulated BA metabolism in early life may contribute to the risk and pathogenesis of T1D.
Collapse
Affiliation(s)
- Santosh Lamichhane
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Partho Sen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland,School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden
| | - Alex M. Dickens
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland,Department of Chemistry, University of Turku, 20520 Turku, Finland
| | - Marina Amaral Alves
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland,Walter Mors Institute of Research on Natural Products, Federal University of Rio de Janeiro, 21941-599 Rio de Janeiro, Brazil
| | - Taina Härkönen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jarno Honkanen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tommi Vatanen
- The Liggins Institute, University of Auckland, Auckland, New Zealand,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland,Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
| | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland,School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden,Corresponding author
| |
Collapse
|
7
|
Thiele I, Fleming RM. Whole-body metabolic modelling predicts isoleucine dependency of SARS-CoV-2 replication. Comput Struct Biotechnol J 2022; 20:4098-4109. [PMID: 35874091 PMCID: PMC9296228 DOI: 10.1016/j.csbj.2022.07.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/10/2022] [Indexed: 11/21/2022] Open
Abstract
We aimed at investigating host-virus co-metabolism during SARS-CoV-2 infection. Therefore, we extended comprehensive sex-specific, whole-body organ resolved models of human metabolism with the necessary reactions to replicate SARS-CoV-2 in the lung as well as selected peripheral organs. Using this comprehensive host-virus model, we obtained the following key results: 1. The predicted maximal possible virus shedding rate was limited by isoleucine availability. 2. The supported initial viral load depended on the increase in CD4+ T-cells, consistent with the literature. 3. During viral infection, the whole-body metabolism changed including the blood metabolome, which agreed well with metabolomic studies from COVID-19 patients and healthy controls. 4. The virus shedding rate could be reduced by either inhibition of the guanylate kinase 1 or availability of amino acids, e.g., in the diet. 5. The virus variants differed in their maximal possible virus shedding rates, which could be inversely linked to isoleucine occurrences in the sequences. Taken together, this study presents the metabolic crosstalk between host and virus and emphasises the role of amino acid metabolism during SARS-CoV-2 infection, in particular of isoleucine. As such, it provides an example of how computational modelling can complement more canonical approaches to gain insight into host-virus crosstalk and to identify potential therapeutic strategies.
Collapse
Affiliation(s)
- Ines Thiele
- School of Medicine, National University of Galway, Galway, Ireland
- Ryan Institute, National University of Galway, Galway, Ireland
- Division of Microbiology, National University of Galway, Galway, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Ronan M.T. Fleming
- School of Medicine, National University of Galway, Galway, Ireland
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
8
|
Mohammad FK, Palukuri MV, Shivakumar S, Rengaswamy R, Sahoo S. A Computational Framework for Studying Gut-Brain Axis in Autism Spectrum Disorder. Front Physiol 2022; 13:760753. [PMID: 35330929 PMCID: PMC8940246 DOI: 10.3389/fphys.2022.760753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/17/2022] [Indexed: 12/28/2022] Open
Abstract
Introduction The integrity of the intestinal epithelium is crucial for human health and is harmed in autism spectrum disorder (ASD). An aberrant gut microbial composition resulting in gut-derived metabolic toxins was found to damage the intestinal epithelium, jeopardizing tissue integrity. These toxins further reach the brain via the gut-brain axis, disrupting the normal function of the brain. A mechanistic understanding of metabolic disturbances in the brain and gut is essential to design effective therapeutics and early intervention to block disease progression. Herein, we present a novel computational framework integrating constraint based tissue specific metabolic (CBM) model and whole-body physiological pharmacokinetics (PBPK) modeling for ASD. Furthermore, the role of gut microbiota, diet, and oxidative stress is analyzed in ASD. Methods A representative gut model capturing host-bacteria and bacteria-bacteria interaction was developed using CBM techniques and patient data. Simultaneously, a PBPK model of toxin metabolism was assembled, incorporating multi-scale metabolic information. Furthermore, dynamic flux balance analysis was performed to integrate CBM and PBPK. The effectiveness of a probiotic and dietary intervention to improve autism symptoms was tested on the integrated model. Results The model accurately highlighted critical metabolic pathways of the gut and brain that are associated with ASD. These include central carbon, nucleotide, and vitamin metabolism in the host gut, and mitochondrial energy and amino acid metabolisms in the brain. The proposed dietary intervention revealed that a high-fiber diet is more effective than a western diet in reducing toxins produced inside the gut. The addition of probiotic bacteria Lactobacillus acidophilus, Bifidobacterium longum longum, Akkermansia muciniphila, and Prevotella ruminicola to the diet restores gut microbiota balance, thereby lowering oxidative stress in the gut and brain. Conclusion The proposed computational framework is novel in its applicability, as demonstrated by the determination of the whole-body distribution of ROS toxins and metabolic association in ASD. In addition, it emphasized the potential for developing novel therapeutic strategies to alleviate autism symptoms. Notably, the presented integrated model validates the importance of combining PBPK modeling with COBRA -specific tissue details for understanding disease pathogenesis.
Collapse
Affiliation(s)
- Faiz Khan Mohammad
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, India
| | - Meghana Venkata Palukuri
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, India.,Initiative for Biological Systems Engineering, Indian Institute of Technology Madras, Chennai, India
| | - Shruti Shivakumar
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, India.,Initiative for Biological Systems Engineering, Indian Institute of Technology Madras, Chennai, India
| | - Raghunathan Rengaswamy
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, India.,Initiative for Biological Systems Engineering, Indian Institute of Technology Madras, Chennai, India
| | - Swagatika Sahoo
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, India.,Initiative for Biological Systems Engineering, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
9
|
Metabolic modeling of host-microbe interactions for therapeutics in colorectal cancer. NPJ Syst Biol Appl 2022; 8:1. [PMID: 35046399 PMCID: PMC8770697 DOI: 10.1038/s41540-021-00210-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
The onset of colorectal cancer (CRC) is often attributed to gut bacterial dysbiosis, and thus gut microbiota are highly relevant in devising treatment strategies. Certain gut microbes, like Enterococcus spp., exhibit remarkable anti-neoplastic and probiotic properties, which can aid in silver nanoparticle (AgNPs) induced reactive oxygen species (ROS)-based CRC treatment. However, the effects of AgNPs on gut microbial metabolism have not been reported thus far. In this study, a detailed systems-level understanding of ROS metabolism in Enterococcus durans (E. durans), a representative gut microbe, was gained using constraint-based modeling, wherein, the critical association between ROS and folate metabolism was established. Experimental studies involving low AgNP concentration treatment of E. durans cultures confirmed these modeling predictions (an increased extracellular folate concentration by 52%, at the 9th h of microbial growth, was observed). Besides, the computational studies established various metabolic pathways involving amino acids, energy metabolites, nucleotides, and SCFAs as the key players in elevating folate levels on ROS exposure. The anti-cancer potential of E. durans was also studied through MTT analysis of HCT 116 cells treated with microbial culture (AgNP treated) supernatant. A decrease in cell viability by 19% implicated the role of microbial metabolites (primarily folate) in causing cell death. The genome-scale modeling approach was then extended to extensively model CRC metabolism, as well as CRC-E. durans interactions in the context of CRC treatment, using tissue-specific metabolic models of CRC and healthy colon. These findings on further validation can facilitate the development of robust and effective cancer therapy.
Collapse
|
10
|
Petersen AØ, Julienne H, Hyötyläinen T, Sen P, Fan Y, Pedersen HK, Jäntti S, Hansen TH, Nielsen T, Jørgensen T, Hansen T, Myers PN, Nielsen HB, Ehrlich SD, Orešič M, Pedersen O. Conjugated C-6 hydroxylated bile acids in serum relate to human metabolic health and gut Clostridia species. Sci Rep 2021; 11:13252. [PMID: 34168163 PMCID: PMC8225906 DOI: 10.1038/s41598-021-91482-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/24/2021] [Indexed: 12/13/2022] Open
Abstract
Knowledge about in vivo effects of human circulating C-6 hydroxylated bile acids (BAs), also called muricholic acids, is sparse. It is unsettled if the gut microbiome might contribute to their biosynthesis. Here, we measured a range of serum BAs and related them to markers of human metabolic health and the gut microbiome. We examined 283 non-obese and obese Danish adults from the MetaHit study. Fasting concentrations of serum BAs were quantified using ultra-performance liquid chromatography-tandem mass-spectrometry. The gut microbiome was characterized with shotgun metagenomic sequencing and genome-scale metabolic modeling. We find that tauro- and glycohyocholic acid correlated inversely with body mass index (P = 4.1e-03, P = 1.9e-05, respectively), waist circumference (P = 0.017, P = 1.1e-04, respectively), body fat percentage (P = 2.5e-03, P = 2.3e-06, respectively), insulin resistance (P = 0.051, P = 4.6e-4, respectively), fasting concentrations of triglycerides (P = 0.06, P = 9.2e-4, respectively) and leptin (P = 0.067, P = 9.2e-4). Tauro- and glycohyocholic acids, and tauro-a-muricholic acid were directly linked with a distinct gut microbial community primarily composed of Clostridia species (P = 0.037, P = 0.013, P = 0.027, respectively). We conclude that serum conjugated C-6-hydroxylated BAs associate with measures of human metabolic health and gut communities of Clostridia species. The findings merit preclinical interventions and human feasibility studies to explore the therapeutic potential of these BAs in obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Anders Ø Petersen
- Department of Health Technology, Technical University of Denmark, 2800, Lyngby, Denmark
| | - Hanna Julienne
- Department of Computational Biology-USR 3756 CNRS, Institut Pasteur, 75015, Paris, France
| | | | - Partho Sen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
| | - Yong Fan
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Helle Krogh Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
- Clinical Microbiomics A/S, Fruebjergvej 3 , 2100 , Copenhagen, Denmark
| | - Sirkku Jäntti
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014, Helsinki, Finland
| | - Tue H Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Trine Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Torben Jørgensen
- Department of Public Health, Faculty of Health and Medical Sciences , University of Copenhagen , Blegdamsvej 3B , 2200 , Copenhagen, Denmark
- Faculty of Medicine, Aalborg University, Niels Jernes Vej 10, 9200 , Aalborg East, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Pernille Neve Myers
- Department of Health Technology, Technical University of Denmark, 2800, Lyngby, Denmark
| | - H Bjørn Nielsen
- Clinical Microbiomics A/S, Fruebjergvej 3 , 2100 , Copenhagen, Denmark
| | - S Dusko Ehrlich
- Department of Computational Biology-USR 3756 CNRS, Institut Pasteur, 75015, Paris, France
| | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
- School of Medical Sciences, Örebro University, 702 81, Örebro, Sweden
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark.
| |
Collapse
|
11
|
Cheng Y, Schlosser P, Hertel J, Sekula P, Oefner PJ, Spiekerkoetter U, Mielke J, Freitag DF, Schmidts M, Kronenberg F, Eckardt KU, Thiele I, Li Y, Köttgen A. Rare genetic variants affecting urine metabolite levels link population variation to inborn errors of metabolism. Nat Commun 2021; 12:964. [PMID: 33574263 PMCID: PMC7878905 DOI: 10.1038/s41467-020-20877-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolite levels in urine may provide insights into genetic mechanisms shaping their related pathways. We therefore investigate the cumulative contribution of rare, exonic genetic variants on urine levels of 1487 metabolites and 53,714 metabolite ratios among 4864 GCKD study participants. Here we report the detection of 128 significant associations involving 30 unique genes, 16 of which are known to underlie inborn errors of metabolism. The 30 genes are strongly enriched for shared expression in liver and kidney (odds ratio = 65, p-FDR = 3e-7), with hepatocytes and proximal tubule cells as driving cell types. Use of UK Biobank whole-exome sequencing data links genes to diseases connected to the identified metabolites. In silico constraint-based modeling of gene knockouts in a virtual whole-body, organ-resolved metabolic human correctly predicts the observed direction of metabolite changes, highlighting the potential of linking population genetics to modeling. Our study implicates candidate variants and genes for inborn errors of metabolism.
Collapse
Affiliation(s)
- Yurong Cheng
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Johannes Hertel
- School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland
- University of Greifswald, University Medicine Greifswald, Department of Psychiatry and Psychotherapy, Greifswald, Germany
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Ute Spiekerkoetter
- Department of General Pediatrics and Adolescent Medicine, Medical Center and Faculty of Medicine - University of Freiburg, Freiburg, Germany
| | - Johanna Mielke
- Bayer AG, Division Pharmaceuticals, Open Innovation & Digital Technologies, Wuppertal, Germany
| | - Daniel F Freitag
- Bayer AG, Division Pharmaceuticals, Open Innovation & Digital Technologies, Wuppertal, Germany
| | - Miriam Schmidts
- Department of General Pediatrics and Adolescent Medicine, Medical Center and Faculty of Medicine - University of Freiburg, Freiburg, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, University of Erlangen-Nürnberg, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ines Thiele
- School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland
- Division of Microbiology, National University of Ireland, Galway, University Road, Galway, Ireland
- APC Microbiome Ireland, Galway, Ireland
| | - Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany.
- CIBSS - Centre for Integrative Biological Signalling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany.
| |
Collapse
|
12
|
Genome scale metabolic models and analysis for evaluating probiotic potentials. Biochem Soc Trans 2020; 48:1309-1321. [PMID: 32726414 DOI: 10.1042/bst20190668] [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] [Received: 01/27/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 11/17/2022]
Abstract
Probiotics are live beneficial microorganisms that can be consumed in the form of dairy and food products as well as dietary supplements to promote a healthy balance of gut bacteria in humans. Practically, the main challenge is to identify and select promising strains and formulate multi-strain probiotic blends with consistent efficacy which is highly dependent on individual dietary regimes, gut environments, and health conditions. Limitations of current in vivo and in vitro methods for testing probiotic strains can be overcome by in silico model guided systems biology approaches where genome scale metabolic models (GEMs) can be used to describe their cellular behaviors and metabolic states of probiotic strains under various gut environments. Here, we summarize currently available GEMs of microbial strains with probiotic potentials and propose a knowledge-based framework to evaluate metabolic capabilities on the basis of six probiotic criteria. They include metabolic characteristics, stability, safety, colonization, postbiotics, and interaction with the gut microbiome which can be assessed by in silico approaches. As such, the most suitable strains can be identified to design personalized multi-strain probiotics in the future.
Collapse
|
13
|
Frioux C, Singh D, Korcsmaros T, Hildebrand F. From bag-of-genes to bag-of-genomes: metabolic modelling of communities in the era of metagenome-assembled genomes. Comput Struct Biotechnol J 2020; 18:1722-1734. [PMID: 32670511 PMCID: PMC7347713 DOI: 10.1016/j.csbj.2020.06.028] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022] Open
Abstract
Metagenomic sequencing of complete microbial communities has greatly enhanced our understanding of the taxonomic composition of microbiotas. This has led to breakthrough developments in bioinformatic disciplines such as assembly, gene clustering, metagenomic binning of species genomes and the discovery of an incredible, so far undiscovered, taxonomic diversity. However, functional annotations and estimating metabolic processes from single species - or communities - is still challenging. Earlier approaches relied mostly on inferring the presence of key enzymes for metabolic pathways in the whole metagenome, ignoring the genomic context of such enzymes, resulting in the 'bag-of-genes' approach to estimate functional capacities of microbiotas. Here, we review recent developments in metagenomic bioinformatics, with a special focus on emerging technologies to simulate and estimate metabolic information, that can be derived from metagenomic assembled genomes. Genome-scale metabolic models can be used to model the emergent properties of microbial consortia and whole communities, and the progress in this area is reviewed. While this subfield of metagenomics is still in its infancy, it is becoming evident that there is a dire need for further bioinformatic tools to address the complex combinatorial problems in modelling the metabolism of large communities as a 'bag-of-genomes'.
Collapse
Affiliation(s)
- Clémence Frioux
- Inria, CNRS, INRAE Bordeaux, France
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, Norfolk, UK
| | - Dipali Singh
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, Norfolk, UK
| | - Tamas Korcsmaros
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, Norfolk, UK
- Digital Biology, Earlham Institute, Norwich, Norfolk, UK
| | - Falk Hildebrand
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, Norfolk, UK
- Digital Biology, Earlham Institute, Norwich, Norfolk, UK
| |
Collapse
|
14
|
Toroghi MK, Cluett WR, Mahadevan R. A Personalized Multiscale Modeling Framework for Dose Selection in Precision Medicine. Ind Eng Chem Res 2020. [DOI: 10.1021/acs.iecr.0c01070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Masood Khaksar Toroghi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada, M5S 3E5
| | - William R. Cluett
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada, M5S 3E5
| | - Radhakrishnan Mahadevan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada, M5S 3E5
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada, M5S 3E5
| |
Collapse
|
15
|
Thiele I, Sahoo S, Heinken A, Hertel J, Heirendt L, Aurich MK, Fleming RMT. Personalized whole-body models integrate metabolism, physiology, and the gut microbiome. Mol Syst Biol 2020; 16:e8982. [PMID: 32463598 PMCID: PMC7285886 DOI: 10.15252/msb.20198982] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022] Open
Abstract
Comprehensive molecular-level models of human metabolism have been generated on a cellular level. However, models of whole-body metabolism have not been established as they require new methodological approaches to integrate molecular and physiological data. We developed a new metabolic network reconstruction approach that used organ-specific information from literature and omics data to generate two sex-specific whole-body metabolic (WBM) reconstructions. These reconstructions capture the metabolism of 26 organs and six blood cell types. Each WBM reconstruction represents whole-body organ-resolved metabolism with over 80,000 biochemical reactions in an anatomically and physiologically consistent manner. We parameterized the WBM reconstructions with physiological, dietary, and metabolomic data. The resulting WBM models could recapitulate known inter-organ metabolic cycles and energy use. We also illustrate that the WBM models can predict known biomarkers of inherited metabolic diseases in different biofluids. Predictions of basal metabolic rates, by WBM models personalized with physiological data, outperformed current phenomenological models. Finally, integrating microbiome data allowed the exploration of host-microbiome co-metabolism. Overall, the WBM reconstructions, and their derived computational models, represent an important step toward virtual physiological humans.
Collapse
Affiliation(s)
- Ines Thiele
- School of MedicineNational University of IrelandGalwayIreland
- Discipline of MicrobiologySchool of Natural SciencesNational University of IrelandGalwayIreland
- APC MicrobiomeCorkIreland
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Swagatika Sahoo
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
- Present address:
Department of Chemical Engineering, and Initiative for Biological Systems Engineering (IBSE)Indian Institute of TechnologyChennaiIndia
| | - Almut Heinken
- School of MedicineNational University of IrelandGalwayIreland
| | - Johannes Hertel
- School of MedicineNational University of IrelandGalwayIreland
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldGermany
| | - Laurent Heirendt
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Maike K Aurich
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Ronan MT Fleming
- School of MedicineNational University of IrelandGalwayIreland
- Division of Analytical BiosciencesLeiden Academic Centre for Drug ResearchFaculty of ScienceUniversity of LeidenLeidenThe Netherlands
| |
Collapse
|
16
|
Blanco-Míguez A, Fdez-Riverola F, Sánchez B, Lourenço A. Resources and tools for the high-throughput, multi-omic study of intestinal microbiota. Brief Bioinform 2020; 20:1032-1056. [PMID: 29186315 DOI: 10.1093/bib/bbx156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/23/2017] [Indexed: 12/18/2022] Open
Abstract
The human gut microbiome impacts several aspects of human health and disease, including digestion, drug metabolism and the propensity to develop various inflammatory, autoimmune and metabolic diseases. Many of the molecular processes that play a role in the activity and dynamics of the microbiota go beyond species and genic composition and thus, their understanding requires advanced bioinformatics support. This article aims to provide an up-to-date view of the resources and software tools that are being developed and used in human gut microbiome research, in particular data integration and systems-level analysis efforts. These efforts demonstrate the power of standardized and reproducible computational workflows for integrating and analysing varied omics data and gaining deeper insights into microbe community structure and function as well as host-microbe interactions.
Collapse
Affiliation(s)
| | | | | | - Anália Lourenço
- Dpto. de Informática - Universidade de Vigo, ESEI - Escuela Superior de Ingeniería Informática, Edificio politécnico, Campus Universitario As Lagoas s/n, 32004 Ourense, Spain
| |
Collapse
|
17
|
Wegrzyn AB, Herzog K, Gerding A, Kwiatkowski M, Wolters JC, Dolga AM, van Lint AEM, Wanders RJA, Waterham HR, Bakker BM. Fibroblast-specific genome-scale modelling predicts an imbalance in amino acid metabolism in Refsum disease. FEBS J 2020; 287:5096-5113. [PMID: 32160399 PMCID: PMC7754141 DOI: 10.1111/febs.15292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/25/2020] [Accepted: 03/10/2020] [Indexed: 12/14/2022]
Abstract
Refsum disease (RD) is an inborn error of metabolism that is characterised by a defect in peroxisomal α‐oxidation of the branched‐chain fatty acid phytanic acid. The disorder presents with late‐onset progressive retinitis pigmentosa and polyneuropathy and can be diagnosed biochemically by elevated levels of phytanate in plasma and tissues of patients. To date, no cure exists for RD, but phytanate levels in patients can be reduced by plasmapheresis and a strict diet. In this study, we reconstructed a fibroblast‐specific genome‐scale model based on the recently published, FAD‐curated model, based on Recon3D reconstruction. We used transcriptomics (available via GEO database with identifier GSE138379), metabolomics and proteomics (available via ProteomeXchange with identifier PXD015518) data, which we obtained from healthy controls and RD patient fibroblasts incubated with phytol, a precursor of phytanic acid. Our model correctly represents the metabolism of phytanate and displays fibroblast‐specific metabolic functions. Using this model, we investigated the metabolic phenotype of RD at the genome scale, and we studied the effect of phytanate on cell metabolism. We identified 53 metabolites that were predicted to discriminate between healthy and RD patients, several of which with a link to amino acid metabolism. Ultimately, these insights in metabolic changes may provide leads for pathophysiology and therapy. Databases Transcriptomics data are available via GEO database with identifier GSE138379, and proteomics data are available via ProteomeXchange with identifier PXD015518.
Collapse
Affiliation(s)
- Agnieszka B Wegrzyn
- Systems Medicine of Metabolism and Signalling, Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, The Netherlands.,Analytical Biosciences and Metabolomics, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| | - Katharina Herzog
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Location AMC, University of Amsterdam, The Netherlands.,Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Sweden
| | - Albert Gerding
- Systems Medicine of Metabolism and Signalling, Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, The Netherlands.,Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Marcel Kwiatkowski
- Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, The Netherlands.,Mass Spectrometric Proteomics and Metabolomics, Institute of Biochemistry, University of Innsbruck, Austria
| | - Justina C Wolters
- Laboratory of Paediatrics, University Medical Centre Groningen, University of Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Alida E M van Lint
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Location AMC, University of Amsterdam, The Netherlands
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Location AMC, University of Amsterdam, The Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Location AMC, University of Amsterdam, The Netherlands
| | - Barbara M Bakker
- Systems Medicine of Metabolism and Signalling, Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, The Netherlands
| |
Collapse
|
18
|
Zhu C, Laghi L, Zhang Z, He Y, Wu D, Zhang H, Huang Y, Li C, Zou L. First Steps toward the Giant Panda Metabolome Database: Untargeted Metabolomics of Feces, Urine, Serum, and Saliva by 1H NMR. J Proteome Res 2020; 19:1052-1059. [PMID: 31994893 DOI: 10.1021/acs.jproteome.9b00564] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Differences in the concentration of metabolites in the biofluids of animals closely reflect their physiological diversities. In order to set the basis for a metabolomic atlas for giant panda (Ailuropoda melanoleuca), we characterized the metabolome of healthy giant panda feces (23), urine (16), serum (6), and saliva (4) samples by means of 1H NMR. A total of 107 metabolites and a core metabolome of 12 metabolites was quantified across the four biological matrices. Through univariate analysis followed by robust principal component analysis, we were able to describe how the molecular profile observed in giant panda urine and feces was affected by gender and age. Among the molecules modified by age in feces, fucose plays a peculiar role because it is related to the digestion of bamboo's hemicellulose, which is considered as the main source of energy for giant panda. A metagenomic investigation directed toward this molecule showed that its concentration was indeed positively related to the two-component system pathway and negatively related to the amino sugar and nucleotide sugar metabolism pathway. Such work is meant to provide a robust framework for further -omics research studies on giant panda to accelerate our understanding of the interaction of giant panda with its natural environment.
Collapse
Affiliation(s)
- Chenglin Zhu
- Centre of Food-omics, Department of Agro-Food Science and Technology, University of Bologna, Cesena 47521, Italy
| | - Luca Laghi
- Centre of Food-omics, Department of Agro-Food Science and Technology, University of Bologna, Cesena 47521, Italy
| | - Zhizhong Zhang
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda, Dujiangyan, Sichuan 611800, China
| | - Yongguo He
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda, Dujiangyan, Sichuan 611800, China
| | - Daifu Wu
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda, Dujiangyan, Sichuan 611800, China
| | - Hemin Zhang
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda, Dujiangyan, Sichuan 611800, China
| | - Yan Huang
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda, Dujiangyan, Sichuan 611800, China
| | - Caiwu Li
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda, Dujiangyan, Sichuan 611800, China
| | - Likou Zou
- College of Resources, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| |
Collapse
|
19
|
Ben Guebila M, Thiele I. Predicting gastrointestinal drug effects using contextualized metabolic models. PLoS Comput Biol 2019; 15:e1007100. [PMID: 31242176 PMCID: PMC6594586 DOI: 10.1371/journal.pcbi.1007100] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/13/2019] [Indexed: 12/28/2022] Open
Abstract
Gastrointestinal side effects are among the most common classes of adverse reactions associated with orally absorbed drugs. These effects decrease patient compliance with the treatment and induce undesirable physiological effects. The prediction of drug action on the gut wall based on in vitro data solely can improve the safety of marketed drugs and first-in-human trials of new chemical entities. We used publicly available data of drug-induced gene expression changes to build drug-specific small intestine epithelial cell metabolic models. The combination of measured in vitro gene expression and in silico predicted metabolic rates in the gut wall was used as features for a multilabel support vector machine to predict the occurrence of side effects. We showed that combining local gut wall-specific metabolism with gene expression performs better than gene expression alone, which indicates the role of small intestine metabolism in the development of adverse reactions. Furthermore, we reclassified FDA-labeled drugs with respect to their genetic and metabolic profiles to show hidden similarities between seemingly different drugs. The linkage of xenobiotics to their transcriptomic and metabolic profiles could take pharmacology far beyond the usual indication-based classifications.
Collapse
Affiliation(s)
- Marouen Ben Guebila
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ines Thiele
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- School of Medicine, National University of Ireland, Galway, University Road, Galway, Ireland
- Discipline of Microbiology, School of Natural Sciences, National University of Ireland, Galway, University Road, Galway, Ireland
- * E-mail:
| |
Collapse
|
20
|
Sen P, Orešič M. Metabolic Modeling of Human Gut Microbiota on a Genome Scale: An Overview. Metabolites 2019; 9:E22. [PMID: 30695998 PMCID: PMC6410263 DOI: 10.3390/metabo9020022] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
There is growing interest in the metabolic interplay between the gut microbiome and host metabolism. Taxonomic and functional profiling of the gut microbiome by next-generation sequencing (NGS) has unveiled substantial richness and diversity. However, the mechanisms underlying interactions between diet, gut microbiome and host metabolism are still poorly understood. Genome-scale metabolic modeling (GSMM) is an emerging approach that has been increasingly applied to infer diet⁻microbiome, microbe⁻microbe and host⁻microbe interactions under physiological conditions. GSMM can, for example, be applied to estimate the metabolic capabilities of microbes in the gut. Here, we discuss how meta-omics datasets such as shotgun metagenomics, can be processed and integrated to develop large-scale, condition-specific, personalized microbiota models in healthy and disease states. Furthermore, we summarize various tools and resources available for metagenomic data processing and GSMM, highlighting the experimental approaches needed to validate the model predictions.
Collapse
Affiliation(s)
- Partho Sen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland.
- School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden.
| | - Matej Orešič
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland.
- School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden.
| |
Collapse
|
21
|
Sahoo S, Ravi Kumar RK, Nicolay B, Mohite O, Sivaraman K, Khetan V, Rishi P, Ganesan S, Subramanyan K, Raman K, Miles W, Elchuri SV. Metabolite systems profiling identifies exploitable weaknesses in retinoblastoma. FEBS Lett 2018; 593:23-41. [PMID: 30417337 DOI: 10.1002/1873-3468.13294] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/25/2018] [Accepted: 11/06/2018] [Indexed: 11/06/2022]
Abstract
Retinoblastoma (RB) is a childhood eye cancer. Currently, chemotherapy, local therapy, and enucleation are the main ways in which these tumors are managed. The present work is the first study that uses constraint-based reconstruction and analysis approaches to identify and explain RB-specific survival strategies, which are RB tumor specific. Importantly, our model-specific secretion profile is also found in RB1-depleted human retinal cells in vitro and suggests that novel biomarkers involved in lipid metabolism may be important. Finally, RB-specific synthetic lethals have been predicted as lipid and nucleoside transport proteins that can aid in novel drug target development.
Collapse
Affiliation(s)
- Swagatika Sahoo
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, India.,Initiative for Biological Systems Engineering, Indian Institute of Technology Madras, Chennai, India
| | | | - Brandon Nicolay
- Department of Molecular Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, USA.,Agios Pharmaceutical, 88 Sidney Street, Cambridge, MA, USA
| | - Omkar Mohite
- Initiative for Biological Systems Engineering, Indian Institute of Technology Madras, Chennai, India.,Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India.,The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Vikas Khetan
- Shri Bhagwan Mahavir Vitreoretinal Services and Ocular Oncology Services, Sankara Nethralaya, Chennai, India
| | - Pukhraj Rishi
- Shri Bhagwan Mahavir Vitreoretinal Services and Ocular Oncology Services, Sankara Nethralaya, Chennai, India
| | - Suganeswari Ganesan
- Department of Histopathology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | | | - Karthik Raman
- Initiative for Biological Systems Engineering, Indian Institute of Technology Madras, Chennai, India.,Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India.,Robert Bosch Centre for Data Science and Artificial Intelligence (RBC-DSAI), Indian Institute of Technology Madras, Chennai, India
| | - Wayne Miles
- Department of Molecular Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, USA.,Department of Molecular Genetics, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Sailaja V Elchuri
- Department of Nanotechnology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| |
Collapse
|
22
|
Wegrzyn AB, Stolle S, Rienksma RA, Martins Dos Santos VAP, Bakker BM, Suarez-Diez M. Cofactors revisited - Predicting the impact of flavoprotein-related diseases on a genome scale. Biochim Biophys Acta Mol Basis Dis 2018; 1865:360-370. [PMID: 30385409 DOI: 10.1016/j.bbadis.2018.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/10/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022]
Abstract
Flavin adenine dinucleotide (FAD) and its precursor flavin mononucleotide (FMN) are redox cofactors that are required for the activity of more than hundred human enzymes. Mutations in the genes encoding these proteins cause severe phenotypes, including a lack of energy supply and accumulation of toxic intermediates. Ideally, patients should be diagnosed before they show symptoms so that treatment and/or preventive care can start immediately. This can be achieved by standardized newborn screening tests. However, many of the flavin-related diseases lack appropriate biomarker profiles. Genome-scale metabolic models can aid in biomarker research by predicting altered profiles of potential biomarkers. Unfortunately, current models, including the most recent human metabolic reconstructions Recon and HMR, typically treat enzyme-bound flavins incorrectly as free metabolites. This in turn leads to artificial degrees of freedom in pathways that are strictly coupled. Here, we present a reconstruction of human metabolism with a curated and extended flavoproteome. To illustrate the functional consequences, we show that simulations with the curated model - unlike simulations with earlier Recon versions - correctly predict the metabolic impact of multiple-acyl-CoA-dehydrogenase deficiency as well as of systemic flavin-depletion. Moreover, simulations with the new model allowed us to identify a larger number of biomarkers in flavoproteome-related diseases, without loss of accuracy. We conclude that adequate inclusion of cofactors in constraint-based modelling contributes to higher precision in computational predictions.
Collapse
Affiliation(s)
- Agnieszka B Wegrzyn
- Systems Medicine of Metabolism and Signaling, Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, 9713, AV, Groningen, the Netherlands; Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, 9713, AV, Groningen, the Netherlands
| | - Sarah Stolle
- Systems Medicine of Metabolism and Signaling, Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, 9713, AV, Groningen, the Netherlands; Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, 9713, AV, Groningen, the Netherlands
| | - Rienk A Rienksma
- Systems and Synthetic Biology, Wageningen University & Research, 6708, WE, Wageningen, the Netherlands
| | - Vítor A P Martins Dos Santos
- Systems and Synthetic Biology, Wageningen University & Research, 6708, WE, Wageningen, the Netherlands; Lifeglimmer GmbH., 12163 Berlin, Germany
| | - Barbara M Bakker
- Systems Medicine of Metabolism and Signaling, Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, 9713, AV, Groningen, the Netherlands; Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, 9713, AV, Groningen, the Netherlands.
| | - Maria Suarez-Diez
- Systems and Synthetic Biology, Wageningen University & Research, 6708, WE, Wageningen, the Netherlands.
| |
Collapse
|
23
|
Piñero J, Furlong LI, Sanz F. In silico models in drug development: where we are. Curr Opin Pharmacol 2018; 42:111-121. [PMID: 30205360 DOI: 10.1016/j.coph.2018.08.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/30/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
Abstract
The use and utility of computational models in drug development has significantly grown in the last decades, fostered by the availability of high throughput datasets and new data analysis strategies. These in silico approaches are demonstrating their ability to generate reliable predictions as well as new knowledge on the mode of action of drugs and the mechanisms underlying their side effects, altogether helping to reduce the costs of drug development. The aim of this review is to provide a panorama of developments in the field in the last two years.
Collapse
Affiliation(s)
- Janet Piñero
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Laura I Furlong
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Ferran Sanz
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain.
| |
Collapse
|
24
|
Sertbas M, Ulgen KO. Unlocking Human Brain Metabolism by Genome-Scale and Multiomics Metabolic Models: Relevance for Neurology Research, Health, and Disease. ACTA ACUST UNITED AC 2018; 22:455-467. [DOI: 10.1089/omi.2018.0088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mustafa Sertbas
- Department of Chemical Engineering, Bogazici University, Istanbul, Turkey
| | - Kutlu O. Ulgen
- Department of Chemical Engineering, Bogazici University, Istanbul, Turkey
| |
Collapse
|
25
|
Constraint-based modeling in microbial food biotechnology. Biochem Soc Trans 2018; 46:249-260. [PMID: 29588387 PMCID: PMC5906707 DOI: 10.1042/bst20170268] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 12/19/2022]
Abstract
Genome-scale metabolic network reconstruction offers a means to leverage the value of the exponentially growing genomics data and integrate it with other biological knowledge in a structured format. Constraint-based modeling (CBM) enables both the qualitative and quantitative analyses of the reconstructed networks. The rapid advancements in these areas can benefit both the industrial production of microbial food cultures and their application in food processing. CBM provides several avenues for improving our mechanistic understanding of physiology and genotype–phenotype relationships. This is essential for the rational improvement of industrial strains, which can further be facilitated through various model-guided strain design approaches. CBM of microbial communities offers a valuable tool for the rational design of defined food cultures, where it can catalyze hypothesis generation and provide unintuitive rationales for the development of enhanced community phenotypes and, consequently, novel or improved food products. In the industrial-scale production of microorganisms for food cultures, CBM may enable a knowledge-driven bioprocess optimization by rationally identifying strategies for growth and stability improvement. Through these applications, we believe that CBM can become a powerful tool for guiding the areas of strain development, culture development and process optimization in the production of food cultures. Nevertheless, in order to make the correct choice of the modeling framework for a particular application and to interpret model predictions in a biologically meaningful manner, one should be aware of the current limitations of CBM.
Collapse
|
26
|
Magnúsdóttir S, Thiele I. Modeling metabolism of the human gut microbiome. Curr Opin Biotechnol 2017; 51:90-96. [PMID: 29258014 DOI: 10.1016/j.copbio.2017.12.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/15/2022]
Abstract
The human gut microbiome plays an important part in human health. The complexity of the microbiome makes it difficult to determine the detailed metabolic functions and cross-talk occurs between the individual species. In silico systems biology studies of the microbiome can help to identify metabolite exchanges among gut microbes. Constraint-based reconstruction and analysis methods use biochemically accurate genome-scale metabolic networks of microorganisms to simulate metabolism between species in a given microbiome and help generate novel hypotheses on microbial interactions. Here, we review metabolic modeling studies that have investigated metabolic functions of the gut microbiome.
Collapse
Affiliation(s)
- Stefanía Magnúsdóttir
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ines Thiele
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
27
|
Predicting the murine enterocyte metabolic response to diets that differ in lipid and carbohydrate composition. Sci Rep 2017; 7:8784. [PMID: 28821741 PMCID: PMC5562867 DOI: 10.1038/s41598-017-07350-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/27/2017] [Indexed: 11/09/2022] Open
Abstract
The small intestine serves as gatekeeper at the interface between body and diet and is thought to play an important role in the etiology of obesity and associated metabolic disorders. A computational modelling approach was used to improve our understanding of the metabolic responses of epithelial cells to different diets. A constraint based, mouse-specific enterocyte metabolic model (named mmu_ENT717) was constructed to describe the impact of four fully characterized semi-purified diets, that differed in lipid and carbohydrate composition, on uptake, metabolism, as well as secretion of carbohydrates and lipids. Our simulation results predicted luminal sodium as a limiting factor for active glucose absorption; necessity of apical localization of glucose transporter GLUT2 for absorption of all glucose in the postprandial state; potential for gluconeogenesis in enterocytes; and the requirement of oxygen for the formation of endogenous cholesterol needed for chylomicron formation under luminal cholesterol-free conditions. In addition, for a number of enzymopathies related to intestinal carbohydrate and lipid metabolism it was found that their effects might be ameliorated through dietary interventions. In conclusion, our improved enterocyte-specific model was shown to be a suitable platform to study effects of dietary interventions on enterocyte metabolism, and provided novel and deeper insights into enterocyte metabolism.
Collapse
|
28
|
Thiele I, Clancy CM, Heinken A, Fleming RM. Quantitative systems pharmacology and the personalized drug-microbiota-diet axis. CURRENT OPINION IN SYSTEMS BIOLOGY 2017; 4:43-52. [PMID: 32984662 PMCID: PMC7493425 DOI: 10.1016/j.coisb.2017.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Precision medicine is an emerging paradigm that aims at maximizing the benefits and minimizing the adverse effects of drugs. Realistic mechanistic models are needed to understand and limit heterogeneity in drug responses. While pharmacokinetic models describe in detail a drug's absorption and metabolism, they generally do not account for individual variations in response to environmental influences, in addition to genetic variation. For instance, the human gut microbiota metabolizes drugs and is modulated by diet, and it exhibits significant variation among individuals. However, the influence of the gut microbiota on drug failure or drug side effects is under-researched. Here, we review recent advances in computational modeling approaches that could contribute to a better, mechanism-based understanding of drug-microbiota-diet interactions and their contribution to individual drug responses. By integrating systems biology and quantitative systems pharmacology with microbiology and nutrition, the conceptually and technologically demand for novel approaches could be met to enable the study of individual variability, thereby providing breakthrough support for progress in precision medicine.
Collapse
Affiliation(s)
- Ines Thiele
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - Catherine M. Clancy
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - Almut Heinken
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - Ronan M.T. Fleming
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
29
|
Venkatasubramanian PB, Toydemir G, de Wit N, Saccenti E, Martins Dos Santos VAP, van Baarlen P, Wells JM, Suarez-Diez M, Mes JJ. Use of Microarray Datasets to generate Caco-2-dedicated Networks and to identify Reporter Genes of Specific Pathway Activity. Sci Rep 2017; 7:6778. [PMID: 28755007 PMCID: PMC5533711 DOI: 10.1038/s41598-017-06355-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/09/2017] [Indexed: 12/30/2022] Open
Abstract
Intestinal epithelial cells, like Caco-2, are commonly used to study the interaction between food, other luminal factors and the host, often supported by microarray analysis to study the changes in gene expression as a result of the exposure. However, no compiled dataset for Caco-2 has ever been initiated and Caco-2-dedicated gene expression networks are barely available. Here, 341 Caco-2-specific microarray samples were collected from public databases and from in-house experiments pertaining to Caco-2 cells exposed to pathogens, probiotics and several food compounds. Using these datasets, a gene functional association network specific for Caco-2 was generated containing 8937 nodes 129711 edges. Two in silico methods, a modified version of biclustering and the new Differential Expression Correlation Analysis, were developed to identify Caco-2-specific gene targets within a pathway of interest. These methods were subsequently applied to the AhR and Nrf2 signalling pathways and altered expression of the predicted target genes was validated by qPCR in Caco-2 cells exposed to coffee extracts, known to activate both AhR and Nrf2 pathways. The datasets and in silico method(s) to identify and predict responsive target genes can be used to more efficiently design experiments to study Caco-2/intestinal epithelial-relevant biological processes.
Collapse
Affiliation(s)
| | - Gamze Toydemir
- Alanya Alaaddin Keykubat University, Faculty of Engineering, Food Engineering Department, Kestel-Alanya, 07450, Antalya, Turkey
| | - Nicole de Wit
- Wageningen University & Research, Food & Biobased Research, Bornse Weilanden 9, 6708 WG, Wageningen, The Netherlands
| | - Edoardo Saccenti
- Wageningen University & Research, Systems and Synthetic Biology, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Vitor A P Martins Dos Santos
- Wageningen University & Research, Systems and Synthetic Biology, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- LifeGlimmerGmbH, Markelstrasse 38, 12163, Berlin, Germany
| | - Peter van Baarlen
- Wageningen University & Research, Host-Microbe Interactomics, De Elst 1, 6708 WD, Wageningen, The Netherlands
| | - Jerry M Wells
- Wageningen University & Research, Host-Microbe Interactomics, De Elst 1, 6708 WD, Wageningen, The Netherlands
| | - Maria Suarez-Diez
- Wageningen University & Research, Systems and Synthetic Biology, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Jurriaan J Mes
- Wageningen University & Research, Food & Biobased Research, Bornse Weilanden 9, 6708 WG, Wageningen, The Netherlands.
| |
Collapse
|
30
|
Martin FP, Su MM, Xie GX, Guiraud SP, Kussmann M, Godin JP, Jia W, Nydegger A. Urinary metabolic insights into host-gut microbial interactions in healthy and IBD children. World J Gastroenterol 2017; 23:3643-3654. [PMID: 28611517 PMCID: PMC5449421 DOI: 10.3748/wjg.v23.i20.3643] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 03/29/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To identify metabolic signatures in urine samples from healthy and inflammatory bowel disease (IBD) children.
METHODS We applied liquid chromatography and gas chromatography coupled to targeted mass spectrometry (MS)-based metabolite profiling to identify and quantify bile acids and host-gut microbial metabolites in urine samples collected from 21 pediatric IBD patients monitored three times over one year (baseline, 6 and 12 mo), and 27 age- and gender-matched healthy children.
RESULTS urinary metabolic profiles of IBD children differ significantly from healthy controls. Such metabolic differences encompass central energy metabolism, amino acids, bile acids and gut microbial metabolites. In particular, levels of pyroglutamic acid, glutamic acid, glycine and cysteine, were significantly higher in IBD children in the course of the study. This suggests that glutathione cannot be optimally synthesized and replenished. Whilst alterations of the enterohepatic circulation of bile acids in pediatric IBD patients is known, we show here that non-invasive urinary bile acid profiling can assess those altered hepatic and intestinal barrier dysfunctions.
CONCLUSION The present study shows how non-invasive sampling of urine followed by targeted MS-based metabonomic analysis can elucidate and monitor the metabolic status of children with different GI health/disease status.
Collapse
|
31
|
Biomedical applications of cell- and tissue-specific metabolic network models. J Biomed Inform 2017; 68:35-49. [DOI: 10.1016/j.jbi.2017.02.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 12/17/2022]
|
32
|
In silico analysis of human metabolism: Reconstruction, contextualization and application of genome-scale models. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.coisb.2017.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
33
|
Analysing Algorithms and Data Sources for the Tissue-Specific Reconstruction of Liver Healthy and Cancer Cells. Interdiscip Sci 2017; 9:36-45. [PMID: 28255832 DOI: 10.1007/s12539-017-0214-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/12/2016] [Accepted: 01/02/2017] [Indexed: 01/27/2023]
Abstract
Genome-Scale Metabolic Models (GSMMs), mathematical representations of the cell metabolism in different organisms including humans, are resourceful tools to simulate metabolic phenotypes and understand associated diseases, such as obesity, diabetes and cancer. In the last years, different algorithms have been developed to generate tissue-specific metabolic models that simulate different phenotypes for distinct cell types. Hepatocyte cells are one of the main sites of metabolic conversions, mainly due to their diverse physiological functions. Most of the liver's tissue is formed by hepatocytes, being one of the largest and most important organs regarding its biological functions. Hepatocellular carcinoma is, also, one of the most important human cancers with high mortality rates. In this study, we will analyze four different algorithms (MBA, mCADRE, tINIT and FASTCORE) for tissue-specific model reconstruction, based on a template model and two types of data sources: transcriptomics and proteomics. These methods will be applied to the reconstruction of metabolic models for hepatocyte cells and HepG2 cancer cell line. The models will be analyzed and compared under different perspectives, emphasizing their functional analysis considering a set of metabolic liver tasks. The results show that there is no "ideal" algorithm. However, with the current analysis, we were able to retrieve knowledge about the metabolism of the liver.
Collapse
|
34
|
Robinson JL, Nielsen J. Integrative analysis of human omics data using biomolecular networks. MOLECULAR BIOSYSTEMS 2016; 12:2953-64. [PMID: 27510223 DOI: 10.1039/c6mb00476h] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
High-throughput '-omics' technologies have given rise to an increasing abundance of genome-scale data detailing human biology at the molecular level. Although these datasets have already made substantial contributions to a more comprehensive understanding of human physiology and diseases, their interpretation becomes increasingly cryptic and nontrivial as they continue to expand in size and complexity. Systems biology networks offer a scaffold upon which omics data can be integrated, facilitating the extraction of new and physiologically relevant information from the data. Two of the most prevalent networks that have been used for such integrative analyses of omics data are genome-scale metabolic models (GEMs) and protein-protein interaction (PPI) networks, both of which have demonstrated success among many different omics and sample types. This integrative approach seeks to unite 'top-down' omics data with 'bottom-up' biological networks in a synergistic fashion that draws on the strengths of both strategies. As the volume and resolution of high-throughput omics data continue to grow, integrative network-based analyses are expected to play an increasingly important role in their interpretation.
Collapse
Affiliation(s)
- Jonathan L Robinson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE412 96 Gothenburg, Sweden.
| | | |
Collapse
|
35
|
Model-based dietary optimization for late-stage, levodopa-treated, Parkinson's disease patients. NPJ Syst Biol Appl 2016; 2:16013. [PMID: 28725472 PMCID: PMC5516849 DOI: 10.1038/npjsba.2016.13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 03/10/2016] [Accepted: 03/25/2016] [Indexed: 01/14/2023] Open
Abstract
Levodopa has been the gold standard for Parkinson’s disease treatment for more than 40 years. Its bioavailability is hindered by dietary amino acids, leading to fluctuations in the motor response particularly in late-stage (stage 3 and 4 on Hoehn and Yahr scale) patients. The routine dietary intervention consists of low-protein (<0.8 g/kg) diets or the redistribution of daily protein allowance to the last meal. Computational modeling was used to examine the fluctuation of gastrointestinal levodopa absorption under consideration of the diet by (i) identifying the group of patients that could benefit from dietary interventions, (ii) comparing existing diet recommendations for their impact on levodopa bioavailability, and (iii) suggesting a mechanism-based dietary intervention. We developed a multiscale computational model consisting of an ordinary differential equations-based advanced compartmentalized absorption and transit (ACAT) gut model and metabolic genome-scale small intestine epithelial cell model. We used this model to investigate complex spatiotemporal relationship between dietary amino acids and levodopa absorption. Our model predicted an improvement in bioavailability, as reflected by blood concentrations of levodopa with protein redistribution diet by 34% compared with a low-protein diet and by 11% compared with the ante cibum (a.c.) administration. These results are consistent with the reported better outcome in late-stage patients. A systematic analysis of the effect of different amino acids in the diet suggested that a serine-rich diet could improve the bioavailability by 22% compared with the a.c. administration. In addition, the slower gastric emptying rate in PD patients exacerbates the loss of levodopa due to competition. Optimizing dietary recommendations in quantity, composition, and intake time holds the promise to improve levodopa efficiency and patient’s quality of life based on highly detailed, mechanistic models of gut physiology endowed with improved extrapolative properties, thus paving the way for precision medical treatment.
Collapse
|
36
|
Ryu JY, Kim HU, Lee SY. Reconstruction of genome-scale human metabolic models using omics data. Integr Biol (Camb) 2016; 7:859-68. [PMID: 25730289 DOI: 10.1039/c5ib00002e] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The impact of genome-scale human metabolic models on human systems biology and medical sciences is becoming greater, thanks to increasing volumes of model building platforms and publicly available omics data. The genome-scale human metabolic models started with Recon 1 in 2007, and have since been used to describe metabolic phenotypes of healthy and diseased human tissues and cells, and to predict therapeutic targets. Here we review recent trends in genome-scale human metabolic modeling, including various generic and tissue/cell type-specific human metabolic models developed to date, and methods, databases and platforms used to construct them. For generic human metabolic models, we pay attention to Recon 2 and HMR 2.0 with emphasis on data sources used to construct them. Draft and high-quality tissue/cell type-specific human metabolic models have been generated using these generic human metabolic models. Integration of tissue/cell type-specific omics data with the generic human metabolic models is the key step, and we discuss omics data and their integration methods to achieve this task. The initial version of the tissue/cell type-specific human metabolic models can further be computationally refined through gap filling, reaction directionality assignment and the subcellular localization of metabolic reactions. We review relevant tools for this model refinement procedure as well. Finally, we suggest the direction of further studies on reconstructing an improved human metabolic model.
Collapse
Affiliation(s)
- Jae Yong Ryu
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Plus Program), Center for Systems and Synthetic Biotechnology, Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea.
| | | | | |
Collapse
|
37
|
van Beek JHGM, Kirkwood TBL, Bassingthwaighte JB. Understanding the physiology of the ageing individual: computational modelling of changes in metabolism and endurance. Interface Focus 2016; 6:20150079. [PMID: 27051508 DOI: 10.1098/rsfs.2015.0079] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Ageing and lifespan are strongly affected by metabolism. The maximal possible uptake of oxygen is not only a good predictor of performance in endurance sports, but also of life expectancy. Figuratively speaking, healthy ageing is a competitive sport. Although the root cause of ageing is damage to macromolecules, it is the balance with repair processes that is decisive. Reduced or intermittent nutrition, hormones and intracellular signalling pathways that regulate metabolism have strong effects on ageing. Homeostatic regulatory processes tend to keep the environment of the cells within relatively narrow bounds. On the other hand, the body is constantly adapting to physical activity and food consumption. Spontaneous fluctuations in heart rate and other processes indicate youth and health. A (homeo)dynamic aspect of homeostasis deteriorates with age. We are now in a position to develop computational models of human metabolism and the dynamics of heart rhythm and oxygen transport that will advance our understanding of ageing. Computational modelling of the connections between dietary restriction, metabolism and protein turnover may increase insight into homeostasis of the proteins in our body. In this way, the computational reconstruction of human physiological processes, the Physiome, can help prevent frailty and age-related disease.
Collapse
Affiliation(s)
- Johannes H G M van Beek
- Section Functional Genomics, Department of Clinical Genetics , VU University medical centre , Amsterdam , The Netherlands
| | - Thomas B L Kirkwood
- Newcastle University Institute for Ageing , Newcastle upon Tyne NE4 5PL , UK
| | | |
Collapse
|
38
|
Schultz A, Qutub AA. Reconstruction of Tissue-Specific Metabolic Networks Using CORDA. PLoS Comput Biol 2016; 12:e1004808. [PMID: 26942765 PMCID: PMC4778931 DOI: 10.1371/journal.pcbi.1004808] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 02/13/2016] [Indexed: 01/07/2023] Open
Abstract
Human metabolism involves thousands of reactions and metabolites. To interpret this complexity, computational modeling becomes an essential experimental tool. One of the most popular techniques to study human metabolism as a whole is genome scale modeling. A key challenge to applying genome scale modeling is identifying critical metabolic reactions across diverse human tissues. Here we introduce a novel algorithm called Cost Optimization Reaction Dependency Assessment (CORDA) to build genome scale models in a tissue-specific manner. CORDA performs more efficiently computationally, shows better agreement to experimental data, and displays better model functionality and capacity when compared to previous algorithms. CORDA also returns reaction associations that can greatly assist in any manual curation to be performed following the automated reconstruction process. Using CORDA, we developed a library of 76 healthy and 20 cancer tissue-specific reconstructions. These reconstructions identified which metabolic pathways are shared across diverse human tissues. Moreover, we identified changes in reactions and pathways that are differentially included and present different capacity profiles in cancer compared to healthy tissues, including up-regulation of folate metabolism, the down-regulation of thiamine metabolism, and tight regulation of oxidative phosphorylation.
Collapse
Affiliation(s)
- André Schultz
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Amina A. Qutub
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
39
|
Aurich MK, Thiele I. Computational Modeling of Human Metabolism and Its Application to Systems Biomedicine. Methods Mol Biol 2016; 1386:253-81. [PMID: 26677187 DOI: 10.1007/978-1-4939-3283-2_12] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Modern high-throughput techniques offer immense opportunities to investigate whole-systems behavior, such as those underlying human diseases. However, the complexity of the data presents challenges in interpretation, and new avenues are needed to address the complexity of both diseases and data. Constraint-based modeling is one formalism applied in systems biology. It relies on a genome-scale reconstruction that captures extensive biochemical knowledge regarding an organism. The human genome-scale metabolic reconstruction is increasingly used to understand normal cellular and disease states because metabolism is an important factor in many human diseases. The application of human genome-scale reconstruction ranges from mere querying of the model as a knowledge base to studies that take advantage of the model's topology and, most notably, to functional predictions based on cell- and condition-specific metabolic models built based on omics data.An increasing number and diversity of biomedical questions are being addressed using constraint-based modeling and metabolic models. One of the most successful biomedical applications to date is cancer metabolism, but constraint-based modeling also holds great potential for inborn errors of metabolism or obesity. In addition, it offers great prospects for individualized approaches to diagnostics and the design of disease prevention and intervention strategies. Metabolic models support this endeavor by providing easy access to complex high-throughput datasets. Personalized metabolic models have been introduced. Finally, constraint-based modeling can be used to model whole-body metabolism, which will enable the elucidation of metabolic interactions between organs and disturbances of these interactions as either causes or consequence of metabolic diseases. This chapter introduces constraint-based modeling and describes some of its contributions to systems biomedicine.
Collapse
Affiliation(s)
- Maike K Aurich
- Luxembourg Center for Systems Biomedicine, University of Luxembourg, Campus Belval, 7, Avenue des Hauts-Fourneaux, Esch-sur-alzette, L-4362, Luxembourg
| | - Ines Thiele
- Luxembourg Center for Systems Biomedicine, University of Luxembourg, Campus Belval, 7, Avenue des Hauts-Fourneaux, Esch-sur-alzette, L-4362, Luxembourg.
| |
Collapse
|
40
|
Sengupta A, Grover M, Chakraborty A, Saxena S. HEPNet: A Knowledge Base Model of Human Energy Pool Network for Predicting the Energy Availability Status of an Individual. PLoS One 2015; 10:e0127918. [PMID: 26053019 PMCID: PMC4460090 DOI: 10.1371/journal.pone.0127918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/20/2015] [Indexed: 11/18/2022] Open
Abstract
HEPNet is an electronic representation of metabolic reactions occurring within human cellular organization focusing on inflow and outflow of the energy currency ATP, GTP and other energy associated moieties. The backbone of HEPNet consists of primary bio-molecules such as carbohydrates, proteins and fats which ultimately constitute the chief source for the synthesis and obliteration of energy currencies in a cell. A series of biochemical pathways and reactions constituting the catabolism and anabolism of various metabolites are portrayed through cellular compartmentalization. The depicted pathways function synchronously toward an overarching goal of producing ATP and other energy associated moieties to bring into play a variety of cellular functions. HEPNet is manually curated with raw data from experiments and is also connected to KEGG and Reactome databases. This model has been validated by simulating it with physiological states like fasting, starvation, exercise and disease conditions like glycaemia, uremia and dihydrolipoamide dehydrogenase deficiency (DLDD). The results clearly indicate that ATP is the master regulator under different metabolic conditions and physiological states. The results also highlight that energy currencies play a minor role. However, the moiety creatine phosphate has a unique character, since it is a ready-made source of phosphoryl groups for the rapid synthesis of ATP from ADP. HEPNet provides a framework for further expanding the network diverse age groups of both the sexes, followed by the understanding of energetics in more complex metabolic pathways that are related to human disorders.
Collapse
Affiliation(s)
- Abhishek Sengupta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, U.P., India
| | - Monendra Grover
- Centre for Agricultural Bioinformatics (CABin), Indian Agricultural Statistics Research Institute (IASRI), ICAR, New Delhi, India
| | - Amlan Chakraborty
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, U.P., India
| | - Sarika Saxena
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, U.P., India
| |
Collapse
|
41
|
Anoxic Conditions Promote Species-Specific Mutualism between Gut Microbes In Silico. Appl Environ Microbiol 2015; 81:4049-61. [PMID: 25841013 DOI: 10.1128/aem.00101-15] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/31/2015] [Indexed: 12/31/2022] Open
Abstract
The human gut is inhabited by thousands of microbial species, most of which are still uncharacterized. Gut microbes have adapted to each other's presence as well as to the host and engage in complex cross feeding. Constraint-based modeling has been successfully applied to predicting microbe-microbe interactions, such as commensalism, mutualism, and competition. Here, we apply a constraint-based approach to model pairwise interactions between 11 representative gut microbes. Microbe-microbe interactions were computationally modeled in conjunction with human small intestinal enterocytes, and the microbe pairs were subjected to three diets with various levels of carbohydrate, fat, and protein in normoxic or anoxic environments. Each microbe engaged in species-specific commensal, parasitic, mutualistic, or competitive interactions. For instance, Streptococcus thermophilus efficiently outcompeted microbes with which it was paired, in agreement with the domination of streptococci in the small intestinal microbiota. Under anoxic conditions, the probiotic organism Lactobacillus plantarum displayed mutualistic behavior toward six other species, which, surprisingly, were almost entirely abolished under normoxic conditions. This finding suggests that the anoxic conditions in the large intestine drive mutualistic cross feeding, leading to the evolvement of an ecosystem more complex than that of the small intestinal microbiota. Moreover, we predict that the presence of the small intestinal enterocyte induces competition over host-derived nutrients. The presented framework can readily be expanded to a larger gut microbial community. This modeling approach will be of great value for subsequent studies aiming to predict conditions favoring desirable microbes or suppressing pathogens.
Collapse
|
42
|
Resendis-Antonio O, González-Torres C, Jaime-Muñoz G, Hernandez-Patiño CE, Salgado-Muñoz CF. Modeling metabolism: A window toward a comprehensive interpretation of networks in cancer. Semin Cancer Biol 2015; 30:79-87. [DOI: 10.1016/j.semcancer.2014.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 04/01/2014] [Accepted: 04/04/2014] [Indexed: 12/01/2022]
|
43
|
Oberhardt MA, Gianchandani EP. Genome-scale modeling and human disease: an overview. Front Physiol 2015; 5:527. [PMID: 25667572 PMCID: PMC4304257 DOI: 10.3389/fphys.2014.00527] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 12/23/2014] [Indexed: 12/11/2022] Open
Affiliation(s)
- Matthew A Oberhardt
- Department of Molecular Microbiology and Biotechnology, Faculty of Life Sciences, School of Computer Sciences, and Sackler School of Medicine, Tel Aviv University Tel Aviv, Israel
| | - Erwin P Gianchandani
- Division of Computer and Network Systems, United States National Science Foundation Arlington, VA, USA
| |
Collapse
|
44
|
Aurich MK, Paglia G, Rolfsson Ó, Hrafnsdóttir S, Magnúsdóttir M, Stefaniak MM, Palsson BØ, Fleming RMT, Thiele I. Prediction of intracellular metabolic states from extracellular metabolomic data. Metabolomics 2015; 11:603-619. [PMID: 25972769 PMCID: PMC4419158 DOI: 10.1007/s11306-014-0721-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/31/2014] [Indexed: 11/03/2022]
Abstract
Metabolic models can provide a mechanistic framework to analyze information-rich omics data sets, and are increasingly being used to investigate metabolic alternations in human diseases. An expression of the altered metabolic pathway utilization is the selection of metabolites consumed and released by cells. However, methods for the inference of intracellular metabolic states from extracellular measurements in the context of metabolic models remain underdeveloped compared to methods for other omics data. Herein, we describe a workflow for such an integrative analysis emphasizing on extracellular metabolomics data. We demonstrate, using the lymphoblastic leukemia cell lines Molt-4 and CCRF-CEM, how our methods can reveal differences in cell metabolism. Our models explain metabolite uptake and secretion by predicting a more glycolytic phenotype for the CCRF-CEM model and a more oxidative phenotype for the Molt-4 model, which was supported by our experimental data. Gene expression analysis revealed altered expression of gene products at key regulatory steps in those central metabolic pathways, and literature query emphasized the role of these genes in cancer metabolism. Moreover, in silico gene knock-outs identified unique control points for each cell line model, e.g., phosphoglycerate dehydrogenase for the Molt-4 model. Thus, our workflow is well-suited to the characterization of cellular metabolic traits based on extracellular metabolomic data, and it allows the integration of multiple omics data sets into a cohesive picture based on a defined model context.
Collapse
Affiliation(s)
- Maike K. Aurich
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, Esch-Sur-Alzette, Luxembourg
| | - Giuseppe Paglia
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
| | - Óttar Rolfsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
| | | | | | - Magdalena M. Stefaniak
- School of Health Science, Faculty of Food Science and Nutrition, University of Iceland, Reykjavik, Iceland
| | - Bernhard Ø. Palsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
- Department of Bioengineering, University of California San Diego, La Jolla, CA USA
| | - Ronan M. T. Fleming
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, Esch-Sur-Alzette, Luxembourg
| | - Ines Thiele
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, Esch-Sur-Alzette, Luxembourg
| |
Collapse
|
45
|
Correia S, Rocha M. A Critical Evaluation of Methods for the Reconstruction of Tissue-Specific Models. PROGRESS IN ARTIFICIAL INTELLIGENCE 2015. [DOI: 10.1007/978-3-319-23485-4_35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
46
|
Sahoo S, Haraldsdóttir HS, Fleming RMT, Thiele I. Modeling the effects of commonly used drugs on human metabolism. FEBS J 2014; 282:297-317. [PMID: 25345908 DOI: 10.1111/febs.13128] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 09/21/2014] [Accepted: 10/13/2014] [Indexed: 02/02/2023]
Abstract
Metabolism contributes significantly to the pharmacokinetics and pharmacodynamics of a drug. In addition, diet and genetics have a profound effect on cellular metabolism with respect to both health and disease. In the present study, we assembled a comprehensive, literature-based drug metabolic reconstruction of the 18 most highly prescribed drug groups, including statins, anti-hypertensives, immunosuppressants and analgesics. This reconstruction captures in detail our current understanding of their absorption, intracellular distribution, metabolism and elimination. We combined this drug module with the most comprehensive reconstruction of human metabolism, Recon 2, yielding Recon2_DM1796, which accounts for 2803 metabolites and 8161 reactions. By defining 50 specific drug objectives that captured the overall drug metabolism of these compounds, we investigated the effects of dietary composition and inherited metabolic disorders on drug metabolism and drug-drug interactions. Our main findings include: (a) a shift in dietary patterns significantly affects statins and acetaminophen metabolism; (b) disturbed statin metabolism contributes to the clinical phenotype of mitochondrial energy disorders; and (c) the interaction between statins and cyclosporine can be explained by several common metabolic and transport pathways other than the previously established CYP3A4 connection. This work holds the potential for studying adverse drug reactions and designing patient-specific therapies.
Collapse
Affiliation(s)
- Swagatika Sahoo
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | | | | | | |
Collapse
|
47
|
Abstract
Motivation: Genome-scale metabolic reconstructions summarize current knowledge about a target organism in a structured manner and as such highlight missing information. Such gaps can be filled algorithmically. Scalability limitations of available algorithms for gap filling hinder their application to compartmentalized reconstructions. Results: We present fastGapFill, a computationally efficient tractable extension to the COBRA toolbox that permits the identification of candidate missing knowledge from a universal biochemical reaction database (e.g. Kyoto Encyclopedia of Genes and Genomes) for a given (compartmentalized) metabolic reconstruction. The stoichiometric consistency of the universal reaction database and of the metabolic reconstruction can be tested for permitting the computation of biologically more relevant solutions. We demonstrate the efficiency and scalability of fastGapFill on a range of metabolic reconstructions. Availability and implementation: fastGapFill is freely available from http://thielelab.eu. Contact:ines.thiele@uni.lu Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ines Thiele
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, L-4362
| | - Nikos Vlassis
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, L-4362
| | - Ronan M T Fleming
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, L-4362
| |
Collapse
|
48
|
Shoaie S, Nielsen J. Elucidating the interactions between the human gut microbiota and its host through metabolic modeling. Front Genet 2014; 5:86. [PMID: 24795748 PMCID: PMC4000997 DOI: 10.3389/fgene.2014.00086] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 03/31/2014] [Indexed: 01/03/2023] Open
Abstract
Increased understanding of the interactions between the gut microbiota, diet and environmental effects may allow us to design efficient treatment strategies for addressing global health problems. Existence of symbiotic microorganisms in the human gut provides different functions for the host such as conversion of nutrients, training of the immune system, and resistance to pathogens. The gut microbiome also plays an influential role in maintaining human health, and it is a potential target for prevention and treatment of common disorders including obesity, type 2 diabetes, and atherosclerosis. Due to the extreme complexity of such disorders, it is necessary to develop mathematical models for deciphering the role of its individual elements as well as the entire system and such models may assist in better understanding of the interactions between the bacteria in the human gut and the host by use of genome-scale metabolic models (GEMs). Recently, GEMs have been employed to explore the interactions between predominant bacteria in the gut ecosystems. Additionally, these models enabled analysis of the contribution of each species to the overall metabolism of the microbiota through the integration of omics data. The outcome of these studies can be used for proposing optimal conditions for desired microbiome phenotypes. Here, we review the recent progress and challenges for elucidating the interactions between the human gut microbiota and host through metabolic modeling. We discuss how these models may provide scaffolds for analyzing high-throughput data, developing probiotics and prebiotics, evaluating the effects of probiotics and prebiotics and eventually designing clinical interventions.
Collapse
Affiliation(s)
- Saeed Shoaie
- Department of Chemical and Biological Engineering, Chalmers University of Technology Gothenburg, Sweden
| | - Jens Nielsen
- Department of Chemical and Biological Engineering, Chalmers University of Technology Gothenburg, Sweden
| |
Collapse
|
49
|
Sahoo S, Aurich MK, Jonsson JJ, Thiele I. Membrane transporters in a human genome-scale metabolic knowledgebase and their implications for disease. Front Physiol 2014; 5:91. [PMID: 24653705 PMCID: PMC3949408 DOI: 10.3389/fphys.2014.00091] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 02/17/2014] [Indexed: 01/18/2023] Open
Abstract
Membrane transporters enable efficient cellular metabolism, aid in nutrient sensing, and have been associated with various diseases, such as obesity and cancer. Genome-scale metabolic network reconstructions capture genomic, physiological, and biochemical knowledge of a target organism, along with a detailed representation of the cellular metabolite transport mechanisms. Since the first reconstruction of human metabolism, Recon 1, published in 2007, progress has been made in the field of metabolite transport. Recently, we published an updated reconstruction, Recon 2, which significantly improved the metabolic coverage and functionality. Human metabolic reconstructions have been used to investigate the role of metabolism in disease and to predict biomarkers and drug targets. Given the importance of cellular transport systems in understanding human metabolism in health and disease, we analyzed the coverage of transport systems for various metabolite classes in Recon 2. We will review the current knowledge on transporters (i.e., their preferred substrates, transport mechanisms, metabolic relevance, and disease association for each metabolite class). We will assess missing coverage and propose modifications and additions through a transport module that is functional when combined with Recon 2. This information will be valuable for further refinements. These data will also provide starting points for further experiments by highlighting areas of incomplete knowledge. This review represents the first comprehensive overview of the transporters involved in central metabolism and their transport mechanisms, thus serving as a compendium of metabolite transporters specific for human metabolic reconstructions.
Collapse
Affiliation(s)
- Swagatika Sahoo
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| | - Maike K Aurich
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| | - Jon J Jonsson
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Iceland Reykjavik, Iceland ; Department of Genetics and Molecular Medicine, Landspitali, National University Hospital of Iceland Reykjavik, Iceland
| | - Ines Thiele
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| |
Collapse
|
50
|
Oberhardt MA, Yizhak K, Ruppin E. Metabolically re-modeling the drug pipeline. Curr Opin Pharmacol 2013; 13:778-85. [DOI: 10.1016/j.coph.2013.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 05/04/2013] [Accepted: 05/06/2013] [Indexed: 02/07/2023]
|