1
|
van de Graaf SFJ, Paulusma CC, In Het Panhuis W. Getting in the zone: Metabolite transport across liver zones. Acta Physiol (Oxf) 2024; 240:e14239. [PMID: 39364668 DOI: 10.1111/apha.14239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
The liver has many functions including the regulation of nutrient and metabolite levels in the systemic circulation through efficient transport into and out of hepatocytes. To sustain these functions, hepatocytes display large functional heterogeneity. This heterogeneity is reflected by zonation of metabolic processes that take place in different zones of the liver lobule, where nutrient-rich blood enters the liver in the periportal zone and flows through the mid-zone prior to drainage by a central vein in the pericentral zone. Metabolite transport plays a pivotal role in the division of labor across liver zones, being either transport into the hepatocyte or transport between hepatocytes through the blood. Signaling pathways that regulate zonation, such as Wnt/β-catenin, have been shown to play a causal role in the development of metabolic dysfunction-associated steatohepatitis (MASH) progression, but the (patho)physiological regulation of metabolite transport remains enigmatic. Despite the practical challenges to separately study individual liver zones, technological advancements in the recent years have greatly improved insight in spatially divided metabolite transport. This review summarizes the theories behind the regulation of zonation, diurnal rhythms and their effect on metabolic zonation, contemporary techniques used to study zonation and current technological challenges, and discusses the current view on spatial and temporal metabolite transport.
Collapse
Affiliation(s)
- Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Coen C Paulusma
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Wietse In Het Panhuis
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Abo SMC, Layton AT. Modeling sex-specific whole-body metabolic responses to feeding and fasting. Comput Biol Med 2024; 181:109024. [PMID: 39178806 DOI: 10.1016/j.compbiomed.2024.109024] [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: 05/21/2024] [Revised: 07/27/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024]
Abstract
Men generally favor carbohydrate metabolism, while women lean towards lipid metabolism, resulting in significant sex-based differences in energy oxidation across various metabolic states such as fasting and feeding. These differences are influenced by body composition and inherent metabolic fluxes, including increased lipolysis rates in women. However, understanding how sex influences organ-specific metabolism and systemic manifestations remains incomplete. To address these gaps, we developed a sex-specific, whole-body metabolic model for feeding and fasting scenarios in healthy young adults. Our model integrates organ metabolism with whole-body responses to mixed meals, particularly high-carbohydrate and high-fat meals. Our predictions suggest that differences in liver and adipose tissue nutrient storage and oxidation patterns drive systemic metabolic disparities. We propose that sex differences in fasting hepatic glucose output may result from the different handling of free fatty acids, glycerol, and glycogen. We identified a metabolic pathway, possibly more prevalent in female livers, redirecting lipids towards carbohydrate metabolism to support hepatic glucose production. This mechanism is facilitated by the TG-FFA cycle between adipose tissue and the liver. Incorporating sex-specific data into multi-scale frameworks offers insights into how sex modulates human metabolism.
Collapse
Affiliation(s)
- Stéphanie M C Abo
- Department of Applied Mathematics, University of Waterloo, 200 University Ave W, Waterloo, N2L 3G1, Ontario, Canada.
| | - Anita T Layton
- Department of Applied Mathematics, University of Waterloo, 200 University Ave W, Waterloo, N2L 3G1, Ontario, Canada; Cheriton School of Computer Science, Department of Biology, and School of Pharmacy, 200 University Ave W, Waterloo, N2L 3G1, Ontario, Canada.
| |
Collapse
|
3
|
Simonsson C, Nyman E, Gennemark P, Gustafsson P, Hotz I, Ekstedt M, Lundberg P, Cedersund G. A unified framework for prediction of liver steatosis dynamics in response to different diet and drug interventions. Clin Nutr 2024; 43:1532-1543. [PMID: 38754305 DOI: 10.1016/j.clnu.2024.05.017] [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: 11/03/2023] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) is a common metabolic disorder, characterized by the accumulation of excess fat in the liver, and is a driving factor for various severe liver diseases. These multi-factorial and multi-timescale changes are observed in different clinical studies, but these studies have not been integrated into a unified framework. In this study, we aim to present such a unified framework in the form of a dynamic mathematical model. METHODS For model training and validation, we collected data for dietary or drug-induced interventions aimed at reducing or increasing liver fat. The model was formulated using ordinary differential equations (ODEs) and the mathematical analysis, model simulation, model formulation and the model parameter estimation were all performed in MATLAB. RESULTS Our mathematical model describes accumulation of fat in the liver and predicts changes in lipid fluxes induced by both dietary and drug interventions. The model is validated using data from a wide range of drug and dietary intervention studies and can predict both short-term (days) and long-term (weeks) changes in liver fat. Importantly, the model computes the contribution of each individual lipid flux to the total liver fat dynamics. Furthermore, the model can be combined with an established bodyweight model, to simulate even longer scenarios (years), also including the effects of insulin resistance and body weight. To help prepare for corresponding eHealth applications, we also present a way to visualize the simulated changes, using dynamically changing lipid droplets, seen in images of liver biopsies. CONCLUSION In conclusion, we believe that the minimal model presented herein might be a useful tool for future applications, and to further integrate and understand data regarding changes in dietary and drug induced changes in ectopic TAG in the liver. With further development and validation, the minimal model could be used as a disease progression model for steatosis.
Collapse
Affiliation(s)
- Christian Simonsson
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden; Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden; Department of Radiation Physics, Radiology, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Elin Nyman
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Peter Gennemark
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden; Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Peter Gustafsson
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden; Department of Media and Information Technology, Linköping University, Norrköping, Sweden
| | - Ingrid Hotz
- Department of Media and Information Technology, Linköping University, Norrköping, Sweden
| | - Mattias Ekstedt
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden; Department of Gastroenterology and Hepatology, Department of Health, Medicine and Caring Sciences, Linköping University, Sweden
| | - Peter Lundberg
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden; Department of Radiation Physics, Radiology, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Gunnar Cedersund
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden; Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.
| |
Collapse
|
4
|
Lambers L, Waschinsky N, Schleicher J, König M, Tautenhahn HM, Albadry M, Dahmen U, Ricken T. Quantifying fat zonation in liver lobules: an integrated multiscale in silico model combining disturbed microperfusion and fat metabolism via a continuum biomechanical bi-scale, tri-phasic approach. Biomech Model Mechanobiol 2024; 23:631-653. [PMID: 38402347 DOI: 10.1007/s10237-023-01797-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/22/2023] [Indexed: 02/26/2024]
Abstract
Metabolic zonation refers to the spatial separation of metabolic functions along the sinusoidal axes of the liver. This phenomenon forms the foundation for adjusting hepatic metabolism to physiological requirements in health and disease (e.g., metabolic dysfunction-associated steatotic liver disease/MASLD). Zonated metabolic functions are influenced by zonal morphological abnormalities in the liver, such as periportal fibrosis and pericentral steatosis. We aim to analyze the interplay between microperfusion, oxygen gradient, fat metabolism and resulting zonated fat accumulation in a liver lobule. Therefore we developed a continuum biomechanical, tri-phasic, bi-scale, and multicomponent in silico model, which allows to numerically simulate coupled perfusion-function-growth interactions two-dimensionally in liver lobules. The developed homogenized model has the following specifications: (i) thermodynamically consistent, (ii) tri-phase model (tissue, fat, blood), (iii) penta-substances (glycogen, glucose, lactate, FFA, and oxygen), and (iv) bi-scale approach (lobule, cell). Our presented in silico model accounts for the mutual coupling between spatial and time-dependent liver perfusion, metabolic pathways and fat accumulation. The model thus allows the prediction of fat development in the liver lobule, depending on perfusion, oxygen and plasma concentration of free fatty acids (FFA), oxidative processes, the synthesis and the secretion of triglycerides (TGs). The use of a bi-scale approach allows in addition to focus on scale bridging processes. Thus, we will investigate how changes at the cellular scale affect perfusion at the lobular scale and vice versa. This allows to predict the zonation of fat distribution (periportal or pericentral) depending on initial conditions, as well as external and internal boundary value conditions.
Collapse
Affiliation(s)
- Lena Lambers
- Institute of Structural Mechanics and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Pfaffenwaldring 27, Stuttgart, 70191, Germany
| | - Navina Waschinsky
- Institute of Structural Mechanics and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Pfaffenwaldring 27, Stuttgart, 70191, Germany
| | - Jana Schleicher
- Friedrich-Schiller-Universität Jena, Fürstengraben 27, Jena, 07743, Germany
| | - Matthias König
- Systems Medicine of Liver, Institute for Theoretical Biology, Institute for Biology, Humboldt-University Berlin, Philippstraße 13, 10115 Berlin, Germany
| | - Hans-Michael Tautenhahn
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Liebigstraße 20, Leipzig, 04103, Germany
| | - Mohamed Albadry
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, Drackendorfer Straße 1, Jena, 07747, Germany
- Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkom, Menoufia, Egypt
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, Drackendorfer Straße 1, Jena, 07747, Germany
| | - Tim Ricken
- Institute of Structural Mechanics and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Pfaffenwaldring 27, Stuttgart, 70191, Germany.
| |
Collapse
|
5
|
Liao Y, Davies NA, Bogle IDL. A process systems Engineering approach to analysis of fructose consumption in the liver system and consequences for Non-Alcoholic fatty liver disease. Chem Eng Sci 2022. [DOI: 10.1016/j.ces.2022.118131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
6
|
Sové RJ, Verma BK, Wang H, Ho WJ, Yarchoan M, Popel AS. Virtual clinical trials of anti-PD-1 and anti-CTLA-4 immunotherapy in advanced hepatocellular carcinoma using a quantitative systems pharmacology model. J Immunother Cancer 2022; 10:e005414. [PMID: 36323435 PMCID: PMC9639136 DOI: 10.1136/jitc-2022-005414] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer and is the third-leading cause of cancer-related death worldwide. Most patients with HCC are diagnosed at an advanced stage, and the median survival for patients with advanced HCC treated with modern systemic therapy is less than 2 years. This leaves the advanced stage patients with limited treatment options. Immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1) or its ligand, are widely used in the treatment of HCC and are associated with durable responses in a subset of patients. ICIs targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) also have clinical activity in HCC. Combination therapy of nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4) is the first treatment option for HCC to be approved by Food and Drug Administration that targets more than one immune checkpoints. METHODS In this study, we used the framework of quantitative systems pharmacology (QSP) to perform a virtual clinical trial for nivolumab and ipilimumab in HCC patients. Our model incorporates detailed biological mechanisms of interactions of immune cells and cancer cells leading to antitumor response. To conduct virtual clinical trial, we generate virtual patient from a cohort of 5,000 proposed patients by extending recent algorithms from literature. The model was calibrated using the data of the clinical trial CheckMate 040 (ClinicalTrials.gov number, NCT01658878). RESULTS Retrospective analyses were performed for different immune checkpoint therapies as performed in CheckMate 040. Using machine learning approach, we predict the importance of potential biomarkers for immune blockade therapies. CONCLUSIONS This is the first QSP model for HCC with ICIs and the predictions are consistent with clinically observed outcomes. This study demonstrates that using a mechanistic understanding of the underlying pathophysiology, QSP models can facilitate patient selection and design clinical trials with improved success.
Collapse
Affiliation(s)
- Richard J Sové
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Babita K Verma
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Won Jin Ho
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark Yarchoan
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Casas B, Vilén L, Bauer S, Kanebratt KP, Wennberg Huldt C, Magnusson L, Marx U, Andersson TB, Gennemark P, Cedersund G. Integrated experimental-computational analysis of a HepaRG liver-islet microphysiological system for human-centric diabetes research. PLoS Comput Biol 2022; 18:e1010587. [PMID: 36260620 PMCID: PMC9621595 DOI: 10.1371/journal.pcbi.1010587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 10/31/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
Microphysiological systems (MPS) are powerful tools for emulating human physiology and replicating disease progression in vitro. MPS could be better predictors of human outcome than current animal models, but mechanistic interpretation and in vivo extrapolation of the experimental results remain significant challenges. Here, we address these challenges using an integrated experimental-computational approach. This approach allows for in silico representation and predictions of glucose metabolism in a previously reported MPS with two organ compartments (liver and pancreas) connected in a closed loop with circulating medium. We developed a computational model describing glucose metabolism over 15 days of culture in the MPS. The model was calibrated on an experiment-specific basis using data from seven experiments, where HepaRG single-liver or liver-islet cultures were exposed to both normal and hyperglycemic conditions resembling high blood glucose levels in diabetes. The calibrated models reproduced the fast (i.e. hourly) variations in glucose and insulin observed in the MPS experiments, as well as the long-term (i.e. over weeks) decline in both glucose tolerance and insulin secretion. We also investigated the behaviour of the system under hypoglycemia by simulating this condition in silico, and the model could correctly predict the glucose and insulin responses measured in new MPS experiments. Last, we used the computational model to translate the experimental results to humans, showing good agreement with published data of the glucose response to a meal in healthy subjects. The integrated experimental-computational framework opens new avenues for future investigations toward disease mechanisms and the development of new therapies for metabolic disorders. Microphysiological systems (MPS) are powerful tools to unravel biological knowledge underlying disease. MPS provide a physiologically relevant, human-based in vitro setting, which can potentially yield better translatability to humans than current animal models and traditional cell cultures. However, mechanistic interpretation and extrapolation of the experimental results to human outcome remain significant challenges. In this study, we confront these challenges using an integrated experimental-computational approach. We present a computational model describing glucose metabolism in a previously reported MPS integrating liver and pancreas. This MPS supports a homeostatic feedback loop between HepaRG/HHSteC spheroids and pancreatic islets, and allows for detailed investigations of mechanisms underlying type 2 diabetes in humans. We show that the computational model captures the complex dynamics of glucose-insulin regulation observed in the system, and can provide mechanistic insight into disease progression features, such as insulin resistance and β-cell dynamics. Furthermore, the computational model can explain key differences in temporal dynamics between MPS and human responses, and thus provides a tool for translating experimental insights into human outcome. The integrated experimental-computational framework opens new avenues for future investigations toward disease mechanisms and the development of new therapies for metabolic disorders.
Collapse
Affiliation(s)
- Belén Casas
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Liisa Vilén
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Kajsa P. Kanebratt
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Charlotte Wennberg Huldt
- Bioscience, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lisa Magnusson
- Bioscience, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Tommy B. Andersson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Peter Gennemark
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Gunnar Cedersund
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
8
|
Silfvergren O, Simonsson C, Ekstedt M, Lundberg P, Gennemark P, Cedersund G. Digital twin predicting diet response before and after long-term fasting. PLoS Comput Biol 2022; 18:e1010469. [PMID: 36094958 PMCID: PMC9499255 DOI: 10.1371/journal.pcbi.1010469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 09/22/2022] [Accepted: 08/04/2022] [Indexed: 11/30/2022] Open
Abstract
Today, there is great interest in diets proposing new combinations of macronutrient compositions and fasting schedules. Unfortunately, there is little consensus regarding the impact of these different diets, since available studies measure different sets of variables in different populations, thus only providing partial, non-connected insights. We lack an approach for integrating all such partial insights into a useful and interconnected big picture. Herein, we present such an integrating tool. The tool uses a novel mathematical model that describes mechanisms regulating diet response and fasting metabolic fluxes, both for organ-organ crosstalk, and inside the liver. The tool can mechanistically explain and integrate data from several clinical studies, and correctly predict new independent data, including data from a new study. Using this model, we can predict non-measured variables, e.g. hepatic glycogen and gluconeogenesis, in response to fasting and different diets. Furthermore, we exemplify how such metabolic responses can be successfully adapted to a specific individual’s sex, weight, height, as well as to the individual’s historical data on metabolite dynamics. This tool enables an offline digital twin technology. Fasting and diet are central components of prevention against cardiovascular disease. Unfortunately, there is little consensus regarding which diet schemes are optimal. This is partially because different clinical studies contribute with different non-connected pieces of knowledge, which have not been fully integrated into a useful and interconnected big picture. In principle, mathematical models describing meal responses could be used for such an integration. However, today’s models still lack critical mechanisms, such as protein metabolism and a dynamic glycogen regulation. Herein, we present a) a new expanded model structure including these mechanisms; b) a set of parameters which can simultaneously describe a wide array of complementary estimation data, in both healthy and diabetic populations; c) a personalisation-script, which allows these generic parameters to be tuned to an individual/sub-population, using demographics (age, weight, height, diabetes status) and historic metabolic data. We exemplify how this personalisation can be used to predict new independent data, including a new clinical study, where a qualitatively new prediction is validated: that an oral protein tolerance test gives a clear response in plasma glucose, after, but not before, a 48h fasting period. Our combined model, parameters, and fitting script lay the foundation for an offline digital twin.
Collapse
Affiliation(s)
- Oscar Silfvergren
- Department of Biomedical Engineering, IMT, Linköping University, Linköping, Sweden
| | - Christian Simonsson
- Department of Biomedical Engineering, IMT, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualisation, Linköping University, Linköping, Sweden
| | - Mattias Ekstedt
- Center for Medical Image Science and Visualisation, Linköping University, Linköping, Sweden
- Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Peter Lundberg
- Center for Medical Image Science and Visualisation, Linköping University, Linköping, Sweden
- Department of Medical Radiation Physics, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Peter Gennemark
- Department of Biomedical Engineering, IMT, Linköping University, Linköping, Sweden
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Gunnar Cedersund
- Department of Biomedical Engineering, IMT, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualisation, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
9
|
Siler SQ. Applications of Quantitative Systems Pharmacology (QSP) in Drug Development for NAFLD and NASH and Its Regulatory Application. Pharm Res 2022; 39:1789-1802. [PMID: 35610402 PMCID: PMC9314276 DOI: 10.1007/s11095-022-03295-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/17/2022] [Indexed: 02/08/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a widely prevalent disease, but approved pharmaceutical treatments are not available. As such, there is great activity within the pharmaceutical industry to accelerate drug development in this area and improve the quality of life and reduce mortality for NASH patients. The use of quantitative systems pharmacology (QSP) can help make this overall process more efficient. This mechanism-based mathematical modeling approach describes both the pathophysiology of a disease and how pharmacological interventions can modify pathophysiologic mechanisms. Multiple capabilities are provided by QSP modeling, including the use of model predictions to optimize clinical studies. The use of this approach has grown over the last 20 years, motivating discussions between modelers and regulators to agree upon methodologic standards. These include model transparency, documentation, and inclusion of clinical pharmacodynamic biomarkers. Several QSP models have been developed that describe NASH pathophysiology to varying extents. One specific application of NAFLDsym, a QSP model of NASH, is described in this manuscript. Simulations were performed to help understand if patient behaviors could help explain the relatively high rate of fibrosis stage reductions in placebo cohorts. Simulated food intake and body weight fluctuated periodically over time. The relatively slow turnover of liver collagen allowed persistent reductions in predicted fibrosis stage despite return to baseline for liver fat, plasma ALT, and the NAFLD activity score. Mechanistic insights such as this that have been derived from QSP models can help expedite the development of safe and effective treatments for NASH patients.
Collapse
Affiliation(s)
- Scott Q Siler
- DILIsym Services, a Division of Simulations Plus, 510-862-6027, 6 Davis Drive, PO Box 12317, Research Triangle Park, North Carolina, 27709, USA.
| |
Collapse
|
10
|
Christ B, Dahmen U, Radde N, Ricken T. Editorial: Computational Modeling for Liver Surgery and Interventions. Front Physiol 2022; 13:859522. [PMID: 35242057 PMCID: PMC8886156 DOI: 10.3389/fphys.2022.859522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Bruno Christ
- Cell Transplantation/Molecular Hepatology Lab, Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig Medical Center, Leipzig, Germany
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | - Nicole Radde
- Institute for Systems Theory and Automatic Control, Faculty of Engineering Design, Production Engineering and Automotive Engineering, University of Stuttgart, Stuttgart, Germany
| | - Tim Ricken
- Institute of Mechanics, Structural Analysis and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
11
|
A biologically based model to quantitatively assess the role of the nuclear receptors liver X (LXR), and pregnane X (PXR) on chemically induced hepatic steatosis. Toxicol Lett 2022; 359:46-54. [PMID: 35143881 PMCID: PMC9644840 DOI: 10.1016/j.toxlet.2022.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/24/2022] [Accepted: 02/03/2022] [Indexed: 11/21/2022]
Abstract
Hepatic steatosis is characterized by the intracellular increase of free fatty acids (FFAs) in the form of triglycerides in hepatocytes. This hepatic adverse outcome can be caused by many factors, including exposure to drugs or environmental toxicants. Mechanistically, accumulation of lipids in the liver can take place via several mechanisms such as de novo synthesis and/or uptake of FFAs from serum via high fat content diets. De novo synthesis of FFAs within the liver is mediated by the liver X receptor (LXR), and their uptake into the liver is mediated through the pregnane X receptor (PXR). We investigated the impact of chemical exposure on FFAs hepatic content via activation of LXR and PXR by integrating chemical-specific physiologically based pharmacokinetic (PBPK) models with a quantitative toxicology systems (QTS) model of hepatic lipid homeostasis. Three known agonists of LXR and/or PXR were modeled: T0901317 (antagonist for both receptors), GW3965 (LXR only), and Rifampicin (PXR only). Model predictions showed that T0901317 caused the most FFAs build-up in the liver, followed by Rifampicin and then GW3965. These modeling results highlight the importance of PXR activation for serum FFAs uptake into the liver while suggesting that increased hepatic FAAs de novo synthesis alone may not be enough to cause appreciable accumulation of lipids in the liver under normal environmental exposure levels. Moreover, the overall PBPK-hepatic lipids quantitative model can be used to screen chemicals for their potential to cause in vivo hepatic lipid content buildup in view of their in vitro potential to activate the nuclear receptors and their exposure levels.
Collapse
|
12
|
Verma A, Manchel A, Narayanan R, Hoek JB, Ogunnaike BA, Vadigepalli R. A Spatial Model of Hepatic Calcium Signaling and Glucose Metabolism Under Autonomic Control Reveals Functional Consequences of Varying Liver Innervation Patterns Across Species. Front Physiol 2021; 12:748962. [PMID: 34899380 PMCID: PMC8662697 DOI: 10.3389/fphys.2021.748962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/11/2021] [Indexed: 11/25/2022] Open
Abstract
Rapid breakdown of hepatic glycogen stores into glucose plays an important role during intense physical exercise to maintain systemic euglycemia. Hepatic glycogenolysis is governed by several different liver-intrinsic and systemic factors such as hepatic zonation, circulating catecholamines, hepatocellular calcium signaling, hepatic neuroanatomy, and the central nervous system (CNS). Of the factors regulating hepatic glycogenolysis, the extent of lobular innervation varies significantly between humans and rodents. While rodents display very few autonomic nerve terminals in the liver, nearly every hepatic layer in the human liver receives neural input. In the present study, we developed a multi-scale, multi-organ model of hepatic metabolism incorporating liver zonation, lobular scale calcium signaling, hepatic innervation, and direct and peripheral organ-mediated communication between the liver and the CNS. We evaluated the effect of each of these governing factors on the total hepatic glucose output and zonal glycogenolytic patterns within liver lobules during simulated physical exercise. Our simulations revealed that direct neuronal stimulation of the liver and an increase in circulating catecholamines increases hepatic glucose output mediated by mobilization of intracellular calcium stores and lobular scale calcium waves. Comparing simulated glycogenolysis between human-like and rodent-like hepatic innervation patterns (extensive vs. minimal) suggested that propagation of calcium transients across liver lobules acts as a compensatory mechanism to improve hepatic glucose output in sparsely innervated livers. Interestingly, our simulations suggested that catecholamine-driven glycogenolysis is reduced under portal hypertension. However, increased innervation coupled with strong intercellular communication can improve the total hepatic glucose output under portal hypertension. In summary, our modeling and simulation study reveals a complex interplay of intercellular and multi-organ interactions that can lead to differing calcium dynamics and spatial distributions of glycogenolysis at the lobular scale in the liver.
Collapse
Affiliation(s)
- Aalap Verma
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States.,Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexandra Manchel
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Rahul Narayanan
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jan B Hoek
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Babatunde A Ogunnaike
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
13
|
Citrus Flavanone Narirutin, In Vitro and In Silico Mechanistic Antidiabetic Potential. Pharmaceutics 2021; 13:pharmaceutics13111818. [PMID: 34834233 PMCID: PMC8619962 DOI: 10.3390/pharmaceutics13111818] [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: 09/09/2021] [Revised: 10/16/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
Citrus fruits and juices have been studied extensively for their potential involvement in the prevention of various diseases. Flavanones, the characteristic polyphenols of citrus species, are the primarily compounds responsible for these studied health benefits. Using in silico and in vitro methods, we are exploring the possible antidiabetic action of narirutin, a flavanone family member. The goal of the in silico research was to anticipate how narirutin would interact with eight distinct receptors implicated in diabetes control and complications, namely, dipeptidyl-peptidase 4 (DPP4), protein tyrosine phosphatase 1B (PTP1B), free fatty acid receptor 1 (FFAR1), aldose reductase (AldR), glycogen phosphorylase (GP), alpha-amylase (AAM), peroxisome proliferator-activated receptor gamma (PPAR-γ), alpha-glucosidase (AGL), while the in vitro study looked into narirutin’s possible inhibitory impact on alpha-amylase and alpha-glucosidase. The results indicate that the studied citrus flavanone interacted remarkably with most of the receptors and had an excellent inhibitory activity during the in vitro tests suggesting its potent role among the different constituent of the citrus compounds in the management of diabetes and also its complications.
Collapse
|
14
|
Seyedpour SM, Nabati M, Lambers L, Nafisi S, Tautenhahn HM, Sack I, Reichenbach JR, Ricken T. Application of Magnetic Resonance Imaging in Liver Biomechanics: A Systematic Review. Front Physiol 2021; 12:733393. [PMID: 34630152 PMCID: PMC8493836 DOI: 10.3389/fphys.2021.733393] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022] Open
Abstract
MRI-based biomechanical studies can provide a deep understanding of the mechanisms governing liver function, its mechanical performance but also liver diseases. In addition, comprehensive modeling of the liver can help improve liver disease treatment. Furthermore, such studies demonstrate the beginning of an engineering-level approach to how the liver disease affects material properties and liver function. Aimed at researchers in the field of MRI-based liver simulation, research articles pertinent to MRI-based liver modeling were identified, reviewed, and summarized systematically. Various MRI applications for liver biomechanics are highlighted, and the limitations of different viscoelastic models used in magnetic resonance elastography are addressed. The clinical application of the simulations and the diseases studied are also discussed. Based on the developed questionnaire, the papers' quality was assessed, and of the 46 reviewed papers, 32 papers were determined to be of high-quality. Due to the lack of the suitable material models for different liver diseases studied by magnetic resonance elastography, researchers may consider the effect of liver diseases on constitutive models. In the future, research groups may incorporate various aspects of machine learning (ML) into constitutive models and MRI data extraction to further refine the study methodology. Moreover, researchers should strive for further reproducibility and rigorous model validation and verification.
Collapse
Affiliation(s)
- Seyed M. Seyedpour
- Institute of Mechanics, Structural Analysis and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Stuttgart, Germany
- Biomechanics Lab, Institute of Mechanics, Structural Analysis and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Stuttgart, Germany
| | - Mehdi Nabati
- Department of Mechanical Engineering, Faculty of Engineering, Boğaziçi University, Istanbul, Turkey
| | - Lena Lambers
- Institute of Mechanics, Structural Analysis and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Stuttgart, Germany
- Biomechanics Lab, Institute of Mechanics, Structural Analysis and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Stuttgart, Germany
| | - Sara Nafisi
- Faculty of Pharmacy, Istinye University, Istanbul, Turkey
| | - Hans-Michael Tautenhahn
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | - Ingolf Sack
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Charité Mitte, Berlin, Germany
| | - Jürgen R. Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
- Center of Medical Optics and Photonics, Friedrich Schiller University, Jena, Germany
- Michael Stifel Center for Data-driven and Simulation Science Jena, Friedrich Schiller University, Jena, Germany
| | - Tim Ricken
- Institute of Mechanics, Structural Analysis and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Stuttgart, Germany
- Biomechanics Lab, Institute of Mechanics, Structural Analysis and Dynamics, Faculty of Aerospace Engineering and Geodesy, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
15
|
Zuo C, Zhang H, Liang S, Teng W, Bao C, Li D, Hu Y, Wang Q, Li Z, Li Y. The alleviation of lipid deposition in steatosis hepatocytes by capsaicin-loaded α-lactalbumin nanomicelles via promoted endocytosis and synergetic multiple signaling pathways. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
16
|
Holzhütter HG, Berndt N. Computational Hypothesis: How Intra-Hepatic Functional Heterogeneity May Influence the Cascading Progression of Free Fatty Acid-Induced Non-Alcoholic Fatty Liver Disease (NAFLD). Cells 2021; 10:cells10030578. [PMID: 33808045 PMCID: PMC7999144 DOI: 10.3390/cells10030578] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is the most common type of chronic liver disease in developed nations, affecting around 25% of the population. Elucidating the factors causing NAFLD in individual patients to progress in different rates and to different degrees of severity, is a matter of active medical research. Here, we aim to provide evidence that the intra-hepatic heterogeneity of rheological, metabolic and tissue-regenerating capacities plays a central role in disease progression. We developed a generic mathematical model that constitutes the liver as ensemble of small liver units differing in their capacities to metabolize potentially cytotoxic free fatty acids (FFAs) and to repair FFA-induced cell damage. Transition from simple steatosis to more severe forms of NAFLD is described as self-amplifying process of cascading liver failure, which, to stop, depends essentially on the distribution of functional capacities across the liver. Model simulations provided the following insights: (1) A persistently high plasma level of FFAs is sufficient to drive the liver through different stages of NAFLD; (2) Presence of NAFLD amplifies the deleterious impact of additional tissue-damaging hits; and (3) Coexistence of non-steatotic and highly steatotic regions is indicative for the later occurrence of severe NAFLD stages.
Collapse
Affiliation(s)
- Hermann-Georg Holzhütter
- Institute of Biochemistry, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Correspondence:
| | - Nikolaus Berndt
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| |
Collapse
|
17
|
Virtual metabolic human dynamic model for pathological analysis and therapy design for diabetes. iScience 2021; 24:102101. [PMID: 33615200 PMCID: PMC7878987 DOI: 10.1016/j.isci.2021.102101] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/21/2020] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
A virtual metabolic human model is a valuable complement to experimental biology and clinical studies, because in vivo research involves serious ethical and technical problems. I have proposed a multi-organ and multi-scale kinetic model that formulates the reactions of enzymes and transporters with the regulation of hormonal actions at postprandial and postabsorptive states. The computational model consists of 202 ordinary differential equations for metabolites with 217 reaction rates and 1,140 kinetic parameter constants. It is the most comprehensive, largest, and highly predictive model of the whole-body metabolism. Use of the model revealed the mechanisms by which individual disorders, such as steatosis, β cell dysfunction, and insulin resistance, were combined to cause diabetes. The model predicted a glycerol kinase inhibitor to be an effective medicine for type 2 diabetes, which not only decreased hepatic triglyceride but also reduced plasma glucose. The model also enabled us to rationally design combination therapy. A standard of virtual metabolic human dynamic models is proposed It integrates the three scales of molecules, organs, and whole body It gets insight into pathological mechanisms of type 1 and type 2 diabetes It enables the computer-aided design of medication treatment for diabetes
Collapse
|
18
|
Dong K, Zhao Q, Xue Y, Wei Y, Zhang Y, Yang Y. TCTP participates in hepatic metabolism by regulating gene expression involved in insulin resistance. Gene 2020; 768:145263. [PMID: 33122078 DOI: 10.1016/j.gene.2020.145263] [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: 02/05/2020] [Revised: 09/04/2020] [Accepted: 10/20/2020] [Indexed: 10/23/2022]
Abstract
Translationally controlled tumor protein (TCTP) has various cellular functions and molecular interactions, many related to its growth-promoting and antiapoptotic properties. Recently, TCTP expression was reported to increases in insulin-resistant mice fed with high-fat diet. TCTP is a multifunctional protein, but its role in liver metabolism is unclear. Here, we investigated the function and mechanism of TCTP in HepG2 cells. Knock-down of TCTP led to 287 differentially expressed genes (DEGs) that were highly associated with cellular apoptosis and signal response, TNF and NF-κB signaling pathways, glycolysis/gluconeogenesis, insulin resistance, FoxO and insulin signaling pathways, adipocytokine and AMPK signaling pathways. shTCTP downregulated the expression of the key gluconeogenesis enzyme phosphoenolpyruvate carboxykinase (PCK1). Furthermore, TCTP regulated the alternative splicing of genes enriched in the phospholipid biosynthetic process and glycerophospholipid metabolism. We further showed that shTCTP down-regulated the intracellular levels of triglyceride and total cholesterol. Our results showed that TCTP regulates the liver cell transcriptome at both the transcriptional and alternative splicing levels. The TCTP regulatory network predicts the biological functions of TCTP in glucose and lipid metabolism, and also insulin resistance, which may be associated with liver metabolism and diseases such as nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Kun Dong
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jie Fang Avenue, Hankou, Wuhan, Hubei 430030, China.
| | - Qiuchen Zhao
- College of Life Sciences, Wuhan University, NO.299 Ba Yi Avenue, Wuchang, Wuhan 430072, China.
| | - Yaqiang Xue
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, Building 18-2, East Lake High-Tech Development Zone, 388 Gaoxin 2nd Road, Wuhan, Hubei 430075, China; Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, Building 18-1, East Lake High-Tech Development Zone, 388 Gaoxin 2nd Road, Wuhan, Hubei 430075, China.
| | - Yaxun Wei
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, Building 18-1, East Lake High-Tech Development Zone, 388 Gaoxin 2nd Road, Wuhan, Hubei 430075, China.
| | - Yi Zhang
- ABLife BioBigData Institute, Optics Valley International Biomedical Park, Building 18-1, East Lake High-Tech Development Zone, 388 Gaoxin 2nd Road, Wuhan, Hubei 430075, China.
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jie Fang Avenue, Hankou, Wuhan, Hubei 430030, China.
| |
Collapse
|
19
|
Liao Y, Davies NA, Bogle IDL. Computational Modeling of Fructose Metabolism and Development in NAFLD. Front Bioeng Biotechnol 2020; 8:762. [PMID: 32775322 PMCID: PMC7388684 DOI: 10.3389/fbioe.2020.00762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcohol fatty liver disease (NAFLD) is a common disorder that has increased in prevalence 20-fold over the last three decades. It covers a spectrum of conditions resulting from excess lipid accumulation in the liver without alcohol abuse. Among all the risk factors, over-consumption of fructose has been repeatedly reported in both clinical and experimental studies to be highly associated with the development of NAFLD. However, studying in vivo systems is complicated, time consuming and expensive. A detailed kinetic model of fructose metabolism was constructed to investigate the metabolic mechanisms whereby fructose consumption can induce dyslipidaemia associated with NAFLD and to explore whether the pathological conditions can be reversed during the early stages of disease. The model contains biochemical components and reactions identified from the literature, including ~120 parameters, 25 variables, and 25 first order differential equations. Three scenarios were presented to demonstrate the behavior of the model. Scenario one predicts the acute effects of a change in carbohydrate input in lipid profiles. The results present progressive triglyceride accumulations in the liver and plasma for three diets. The rate of accumulation was greater in the fructose diet than that of the mixed or glucose only models. Scenario two explores the variability of metabolic reaction rate within the general population. Sensitivity analysis reveals that hepatic triglyceride concentration is most sensitive to the rate constant of pyruvate kinase and fructokinase. Scenario three tests the effect of one specific inhibitor that might be potentially administered. The simulations of fructokinase suppression provide a good model for potentially reversing simple steatosis induced by high fructose consumption, which can be corroborated by experimental studies. The predictions in these three scenarios suggest that the model is robust and it has sufficient detail to present the kinetic relationship between fructose and lipid in the liver.
Collapse
Affiliation(s)
- Yunjie Liao
- Department of Chemical Engineering, Center for Process Systems Engineering, University College London, London, United Kingdom.,Division of Medicine, Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Nathan A Davies
- Division of Medicine, Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - I David L Bogle
- Department of Chemical Engineering, Center for Process Systems Engineering, University College London, London, United Kingdom
| |
Collapse
|
20
|
Nikmaneshi MR, Firoozabadi B, Munn LL. A mechanobiological mathematical model of liver metabolism. Biotechnol Bioeng 2020; 117:2861-2874. [PMID: 32501531 DOI: 10.1002/bit.27451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/30/2020] [Accepted: 06/04/2020] [Indexed: 02/01/2023]
Abstract
The liver plays a complex role in metabolism and detoxification, and better tools are needed to understand its function and to develop liver-targeted therapies. In this study, we establish a mechanobiological model of liver transport and hepatocyte biology to elucidate the metabolism of urea and albumin, the production/detoxification of ammonia, and consumption of oxygen and nutrients. Since hepatocellular shear stress (SS) can influence the enzymatic activities of liver, the effect of SS on the urea and albumin synthesis are empirically modeled through the mechanotransduction mechanisms. The results demonstrate that the rheology and dynamics of the sinusoid flow can significantly affect liver metabolism. We show that perfusate rheology and blood hematocrit can affect urea and albumin production by changing hepatocyte mechanosensitive metabolism. The model can also simulate enzymatic diseases of the liver such as hyperammonemia I, hyperammonemia II, hyperarginemia, citrollinemia, and argininosuccinicaciduria, which disrupt the urea metabolism and ammonia detoxification. The model is also able to predict how aggregate cultures of hepatocytes differ from single cell cultures. We conclude that in vitro perfusable devices for the study of liver metabolism or personalized medicine should be designed with similar morphology and fluid dynamics as patient liver tissue. This robust model can be adapted to any type of hepatocyte culture to determine how hepatocyte viability, functionality, and metabolism are influenced by liver pathologies and environmental conditions.
Collapse
Affiliation(s)
- Mohammad R Nikmaneshi
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran.,Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bahar Firoozabadi
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Lance L Munn
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
21
|
Sahoo S, Singh D, Chakraborty P, Jolly MK. Emergent Properties of the HNF4α-PPARγ Network May Drive Consequent Phenotypic Plasticity in NAFLD. J Clin Med 2020; 9:E870. [PMID: 32235813 PMCID: PMC7141525 DOI: 10.3390/jcm9030870] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/15/2020] [Accepted: 03/18/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease in adults and children. It is characterized by excessive accumulation of lipids in the hepatocytes of patients without any excess alcohol intake. With a global presence of 24% and limited therapeutic options, the disease burden of NAFLD is increasing. Thus, it becomes imperative to attempt to understand the dynamics of disease progression at a systems-level. Here, we decoded the emergent dynamics of underlying gene regulatory networks that were identified to drive the initiation and the progression of NAFLD. We developed a mathematical model to elucidate the dynamics of the HNF4α-PPARγ gene regulatory network. Our simulations reveal that this network can enable multiple co-existing phenotypes under certain biological conditions: an adipocyte, a hepatocyte, and a "hybrid" adipocyte-like state of the hepatocyte. These phenotypes may also switch among each other, thus enabling phenotypic plasticity and consequently leading to simultaneous deregulation of the levels of molecules that maintain a hepatic identity and/or facilitate a partial or complete acquisition of adipocytic traits. These predicted trends are supported by the analysis of clinical data, further substantiating the putative role of phenotypic plasticity in driving NAFLD. Our results unravel how the emergent dynamics of underlying regulatory networks can promote phenotypic plasticity, thereby propelling the clinically observed changes in gene expression often associated with NAFLD.
Collapse
Affiliation(s)
- Sarthak Sahoo
- Undergraduate Programme, Indian Institute of Science, Bangalore 560012, India
| | - Divyoj Singh
- Undergraduate Programme, Indian Institute of Science, Bangalore 560012, India
| | - Priyanka Chakraborty
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
22
|
Liver Bioreactor Design Issues of Fluid Flow and Zonation, Fibrosis, and Mechanics: A Computational Perspective. J Funct Biomater 2020; 11:jfb11010013. [PMID: 32121053 PMCID: PMC7151609 DOI: 10.3390/jfb11010013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/27/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering, with the goal of repairing or replacing damaged tissue and organs, has continued to make dramatic science-based advances since its origins in the late 1980’s and early 1990’s. Such advances are always multi-disciplinary in nature, from basic biology and chemistry through physics and mathematics to various engineering and computer fields. This review will focus its attention on two topics critical for tissue engineering liver development: (a) fluid flow, zonation, and drug screening, and (b) biomechanics, tissue stiffness, and fibrosis, all within the context of 3D structures. First, a general overview of various bioreactor designs developed to investigate fluid transport and tissue biomechanics is given. This includes a mention of computational fluid dynamic methods used to optimize and validate these designs. Thereafter, the perspective provided by computer simulations of flow, reactive transport, and biomechanics responses at the scale of the liver lobule and liver tissue is outlined, in addition to how bioreactor-measured properties can be utilized in these models. Here, the fundamental issues of tortuosity and upscaling are highlighted, as well as the role of disease and fibrosis in these issues. Some idealized simulations of the effects of fibrosis on lobule drug transport and mechanics responses are provided to further illustrate these concepts. This review concludes with an outline of some practical applications of tissue engineering advances and how efficient computational upscaling techniques, such as dual continuum modeling, might be used to quantify the transition of bioreactor results to the full liver scale.
Collapse
|
23
|
Li, J, Wang, T, Xia J, Yao W, Huang F. Enzymatic and nonenzymatic protein acetylations control glycolysis process in liver diseases. FASEB J 2019; 33:11640-11654. [PMID: 31370704 PMCID: PMC6902721 DOI: 10.1096/fj.201901175r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022]
Abstract
Impaired glycolysis has pathologic effects on the occurrence and progression of liver diseases, and it appears that glycolysis is increased to different degrees in different liver diseases. As an important post-translational modification, reversible lysine acetylation regulates almost all cellular processes, including glycolysis. Lysine acetylation can occur enzymatically with acetyltransferases or nonenzymatically with acetyl-coenzyme A. Accompanied by the progression of liver diseases, there seems to be a temporal and spatial variation between enzymatic and nonenzymatic acetylations in the regulation of glycolysis. Here, we summarize the most recent findings on the functions and targets of acetylation in controlling glycolysis in the different stages of liver diseases. In addition, we discuss the differences and causes between enzymatic and nonenzymatic acetylations in regulating glycolysis throughout the progression of liver diseases. Then, we review these new discoveries to provide the potential implications of these findings for therapeutic interventions in liver diseases.-Li, J., Wang, T., Xia, J., Yao, W., Huang, F. Enzymatic and nonenzymatic protein acetylations control glycolysis process in liver diseases.
Collapse
Affiliation(s)
- Juan Li,
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Tongxin Wang,
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jun Xia
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Weilei Yao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Feiruo Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
24
|
Taylor DL, Gough A, Schurdak ME, Vernetti L, Chennubhotla CS, Lefever D, Pei F, Faeder JR, Lezon TR, Stern AM, Bahar I. Harnessing Human Microphysiology Systems as Key Experimental Models for Quantitative Systems Pharmacology. Handb Exp Pharmacol 2019; 260:327-367. [PMID: 31201557 PMCID: PMC6911651 DOI: 10.1007/164_2019_239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Two technologies that have emerged in the last decade offer a new paradigm for modern pharmacology, as well as drug discovery and development. Quantitative systems pharmacology (QSP) is a complementary approach to traditional, target-centric pharmacology and drug discovery and is based on an iterative application of computational and systems biology methods with multiscale experimental methods, both of which include models of ADME-Tox and disease. QSP has emerged as a new approach due to the low efficiency of success in developing therapeutics based on the existing target-centric paradigm. Likewise, human microphysiology systems (MPS) are experimental models complementary to existing animal models and are based on the use of human primary cells, adult stem cells, and/or induced pluripotent stem cells (iPSCs) to mimic human tissues and organ functions/structures involved in disease and ADME-Tox. Human MPS experimental models have been developed to address the relatively low concordance of human disease and ADME-Tox with engineered, experimental animal models of disease. The integration of the QSP paradigm with the use of human MPS has the potential to enhance the process of drug discovery and development.
Collapse
Affiliation(s)
- D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark E Schurdak
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence Vernetti
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chakra S Chennubhotla
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Lefever
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
| | - Fen Pei
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - James R Faeder
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy R Lezon
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew M Stern
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivet Bahar
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Misener R, Allenby MC, Fuentes-Garí M, Gupta K, Wiggins T, Panoskaltsis N, Pistikopoulos EN, Mantalaris A. Stem cell biomanufacturing under uncertainty: A case study in optimizing red blood cell production. AIChE J 2018; 64:3011-3022. [PMID: 30166646 PMCID: PMC6108044 DOI: 10.1002/aic.16042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/08/2017] [Indexed: 12/12/2022]
Abstract
As breakthrough cellular therapy discoveries are translated into reliable, commercializable applications, effective stem cell biomanufacturing requires systematically developing and optimizing bioprocess design and operation. This article proposes a rigorous computational framework for stem cell biomanufacturing under uncertainty. Our mathematical tool kit incorporates: high‐fidelity modeling, single variate and multivariate sensitivity analysis, global topological superstructure optimization, and robust optimization. The advantages of the proposed bioprocess optimization framework using, as a case study, a dual hollow fiber bioreactor producing red blood cells from progenitor cells were quantitatively demonstrated. The optimization phase reduces the cost by a factor of 4, and the price of insuring process performance against uncertainty is approximately 15% over the nominal optimal solution. Mathematical modeling and optimization can guide decision making; the possible commercial impact of this cellular therapy using the disruptive technology paradigm was quantitatively evaluated. © 2017 American Institute of Chemical Engineers AIChE J, 64: 3011–3022, 2018
Collapse
Affiliation(s)
- Ruth Misener
- Dept. of Computing; Imperial College London; South Kensington London SW7 2AZ U.K
| | - Mark C. Allenby
- Dept. of Haematology; Imperial College London; Harrow London HA1 3UJ U. K
| | - María Fuentes-Garí
- Dept. of Haematology; Imperial College London; Harrow London HA1 3UJ U. K
| | - Karan Gupta
- Dept. of Haematology; Imperial College London; Harrow London HA1 3UJ U. K
| | - Thomas Wiggins
- Dept. of Haematology; Imperial College London; Harrow London HA1 3UJ U. K
| | - Nicki Panoskaltsis
- Artie McFerrin Dept. of Chemical Engineering; Texas A&M University; College Station TX 77843
| | | | | |
Collapse
|
26
|
Schleicher J, Dahmen U, Guthke R, Schuster S. Zonation of hepatic fat accumulation: insights from mathematical modelling of nutrient gradients and fatty acid uptake. J R Soc Interface 2018; 14:rsif.2017.0443. [PMID: 28835543 DOI: 10.1098/rsif.2017.0443] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/28/2017] [Indexed: 02/07/2023] Open
Abstract
Intrinsic of non-alcoholic fatty liver diseases is an aberrant accumulation of triglycerides (steatosis), which occurs inhomogeneously within lobules. To improve our understanding of the mechanisms involved in this zonation patterning, we developed a mathematical multicompartment model of hepatic fatty acid metabolism accompanied by blood flow simulations. A model analysis determines the influence of the uptake process of fatty acids, the porto-central gradient of plasma fatty acid concentration, and the oxygen supply via blood on the zonation of triglyceride accumulation. From this theoretical perspective, the plasma oxygen gradient, but not the fatty acid gradient, leads the way to a zonated triglyceride accumulation by its decisive role in oxidative processes. In addition, the uptake mechanism of fatty acids seems to be fundamental for a pericentral dominance of steatosis. However, the mechanism of cellular fatty acid uptake from the blood is still under debate. Our theoretical approach supports the transporter-mediated uptake mechanism and reveals that the maximal velocity of fatty acid uptake affects the switching between a periportal and a pericentral triglyceride accumulation. Further research on hepatic fatty acid uptake is needed to push forward our understanding of aberrant triglyceride accumulation in diet-induced steatosis.
Collapse
Affiliation(s)
- Jana Schleicher
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany .,Department of Bioinformatics, Friedrich-Schiller-University Jena, Jena, Germany
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Reinhard Guthke
- Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| | - Stefan Schuster
- Department of Bioinformatics, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
27
|
Berndt N, Horger MS, Bulik S, Holzhütter HG. A multiscale modelling approach to assess the impact of metabolic zonation and microperfusion on the hepatic carbohydrate metabolism. PLoS Comput Biol 2018; 14:e1006005. [PMID: 29447152 PMCID: PMC5841820 DOI: 10.1371/journal.pcbi.1006005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/07/2018] [Accepted: 01/26/2018] [Indexed: 12/11/2022] Open
Abstract
The capacity of the liver to convert the metabolic input received from the incoming portal and arterial blood into the metabolic output of the outgoing venous blood has three major determinants: The intra-hepatic blood flow, the transport of metabolites between blood vessels (sinusoids) and hepatocytes and the metabolic capacity of hepatocytes. These determinants are not constant across the organ: Even in the normal organ, but much more pronounced in the fibrotic and cirrhotic liver, regional variability of the capillary blood pressure, tissue architecture and the expression level of metabolic enzymes (zonation) have been reported. Understanding how this variability may affect the regional metabolic capacity of the liver is important for the interpretation of functional liver tests and planning of pharmacological and surgical interventions. Here we present a mathematical model of the sinusoidal tissue unit (STU) that is composed of a single sinusoid surrounded by the space of Disse and a monolayer of hepatocytes. The total metabolic output of the liver (arterio-venous glucose difference) is obtained by integration across the metabolic output of a representative number of STUs. Application of the model to the hepatic glucose metabolism provided the following insights: (i) At portal glucose concentrations between 6–8 mM, an intra-sinusoidal glucose cycle may occur which is constituted by glucose producing periportal hepatocytes and glucose consuming pericentral hepatocytes, (ii) Regional variability of hepatic blood flow is higher than the corresponding regional variability of the metabolic output, (iii) a spatially resolved metabolic functiogram of the liver is constructed. Variations of tissue parameters are equally important as variations of enzyme activities for the control of the arterio-venous glucose difference. Glucose homeostasis is one of the central liver functions. The liver extracts glucose from the blood when plasma glucose levels are high and produces glucose when plasma glucose levels are low. To fulfill this function the liver is organized in smallest functional units, the sinusoidal tissue units (STUs). These STUs consist of a single sinusoid surrounded by linear arranged hepatocytes. Liver zonation describes the spatial separation of metabolic pathways along the STUs. As blood flows through the sinusoid the plasma nutrient and hormone composition changes and in conjunction with the heterogeneous endowment of metabolic enzymes this leads to big differences in the metabolic performance of hepatocytes depending on their position within the sinusoid. This makes liver zonation and blood flow two central determinants for the functional output of the liver. In this work we present a tissue model of hepatic carbohydrate metabolism that combines liver zonation and microperfusion within the STU. We show that structural properties, enzymatic properties and regional bloodflow are equally important for the understanding of liver functionality. With our work we provide a true multi-scale model bridging the scale from the cellular to the tissue level.
Collapse
Affiliation(s)
- Nikolaus Berndt
- Computational Biochemistry Group, Institute of Biochemistry, Charite—University Medicine Berlin, Charitéplatz 1, Berlin
- * E-mail:
| | - Marius Stefan Horger
- Department of Diagnostic and Interventional Radiology, Eberhard-Karls-University Tubingen, Tuebingen, Germany
| | - Sascha Bulik
- Computational Biochemistry Group, Institute of Biochemistry, Charite—University Medicine Berlin, Charitéplatz 1, Berlin
- German Federal Institute for Risk Assessment, Junior Research Group Supply-Chain-Models, Max-Dohrn-Straße 8–10, Berlin, Germany
| | - Hermann-Georg Holzhütter
- Computational Biochemistry Group, Institute of Biochemistry, Charite—University Medicine Berlin, Charitéplatz 1, Berlin
| |
Collapse
|
28
|
Christ B, Dahmen U, Herrmann KH, König M, Reichenbach JR, Ricken T, Schleicher J, Ole Schwen L, Vlaic S, Waschinsky N. Computational Modeling in Liver Surgery. Front Physiol 2017; 8:906. [PMID: 29249974 PMCID: PMC5715340 DOI: 10.3389/fphys.2017.00906] [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: 08/31/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022] Open
Abstract
The need for extended liver resection is increasing due to the growing incidence of liver tumors in aging societies. Individualized surgical planning is the key for identifying the optimal resection strategy and to minimize the risk of postoperative liver failure and tumor recurrence. Current computational tools provide virtual planning of liver resection by taking into account the spatial relationship between the tumor and the hepatic vascular trees, as well as the size of the future liver remnant. However, size and function of the liver are not necessarily equivalent. Hence, determining the future liver volume might misestimate the future liver function, especially in cases of hepatic comorbidities such as hepatic steatosis. A systems medicine approach could be applied, including biological, medical, and surgical aspects, by integrating all available anatomical and functional information of the individual patient. Such an approach holds promise for better prediction of postoperative liver function and hence improved risk assessment. This review provides an overview of mathematical models related to the liver and its function and explores their potential relevance for computational liver surgery. We first summarize key facts of hepatic anatomy, physiology, and pathology relevant for hepatic surgery, followed by a description of the computational tools currently used in liver surgical planning. Then we present selected state-of-the-art computational liver models potentially useful to support liver surgery. Finally, we discuss the main challenges that will need to be addressed when developing advanced computational planning tools in the context of liver surgery.
Collapse
Affiliation(s)
- Bruno Christ
- Molecular Hepatology Lab, Clinics of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Matthias König
- Department of Biology, Institute for Theoretical Biology, Humboldt University of Berlin, Berlin, Germany
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Tim Ricken
- Mechanics, Structural Analysis, and Dynamics, TU Dortmund University, Dortmund, Germany
| | - Jana Schleicher
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany.,Department of Bioinformatics, Friedrich Schiller University Jena, Jena, Germany
| | | | - Sebastian Vlaic
- Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Navina Waschinsky
- Mechanics, Structural Analysis, and Dynamics, TU Dortmund University, Dortmund, Germany
| |
Collapse
|
29
|
Improving the economics of NASH/NAFLD treatment through the use of systems biology. Drug Discov Today 2017; 22:1532-1538. [DOI: 10.1016/j.drudis.2017.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/27/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022]
|
30
|
Qi S, Xu D, Li Q, Xie N, Xia J, Huo Q, Li P, Chen Q, Huang S. Metabonomics screening of serum identifies pyroglutamate as a diagnostic biomarker for nonalcoholic steatohepatitis. Clin Chim Acta 2017; 473:89-95. [PMID: 28842175 DOI: 10.1016/j.cca.2017.08.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 08/11/2017] [Accepted: 08/20/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE A key step in managing non-alcoholic fatty liver disease (NAFLD) is to differentiate nonalcoholic steatohepatitis (NASH) from simple steatosis (SS). METHOD Serum samples were collected from three groups: NASH patients (N=21), SS patients (N=38) and healthy controls (N=31). High performance liquid chromatography-mass spectrometry (HPLC-MS) was used to analyse the metabolic profile of the serum samples. The acquired data were processed by multivariate principal component analysis (PCA) and orthogonal partial least-squares-discriminant analysis (OPLS-DA) to identify novel metabolites. The potential biomarkers were quantitatively determined and their diagnostic power was further validated. RESULTS A total of 56 metabolites were capable of distinguishing NASH from SS samples based on the OPLS-DA model. Pyroglutamate was found to be the most promising factor in distinguishing the NASH from SS groups. With an optimal cut-off value of 4.82mmol/L, the sensitivity and specificity of the diagnosis of NASH were 72% and 85%, respectively. The area under the receiver operating characteristic (AUROC) of the pyroglutamate levels of NASH versus SS patients was more than those of tumor necrosis factor-α, adiponectin and interleukin-8. CONCLUSION These data suggest that pyroglutamate may be a new and useful biomarker for the diagnosis of NASH.
Collapse
Affiliation(s)
- Suwen Qi
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Depeng Xu
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Qiaoliang Li
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China.
| | - Ni Xie
- Shenzhen Second Hospital, The First Affiliated hospital of Shenzhen University, Shenzhen 518060, China
| | - Jun Xia
- Shenzhen Second Hospital, The First Affiliated hospital of Shenzhen University, Shenzhen 518060, China
| | - Qin Huo
- Shenzhen Second Hospital, The First Affiliated hospital of Shenzhen University, Shenzhen 518060, China
| | - Pu Li
- Department of Laboratory Medicine, The Second Hospital Affiliated to Chongqing Medical University, 410006 Chongqing, China
| | - Qiwen Chen
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Si Huang
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|