1
|
Xiao G, Huang X, Huang T, Chen Z, Huang Y, Huang R, Wang X. Hepatitis B virus X protein differentially regulates the angiogenesis of Hepatocellular Carcinoma through p53-VEGF axis according to glucose levels. Ann Hepatol 2024; 29:101543. [PMID: 39216627 DOI: 10.1016/j.aohep.2024.101543] [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: 10/17/2023] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION AND OBJECTIVES Blood glucose fluctuates severely in the diabetes (DM) and tumor microenvironment. Our previous works have found Hepatitis B virus X protein (HBx) differentially regulated metastasis and apoptosis of hepatoma cells depending on glucose concentration. We here aimed to explore whether HBx played dual roles in the angiogenesis of hepatocellular carcinoma varying on different glucose levels. MATERIALS AND METHODS We collected conditioned medium from HBx-overexpressing cells cultured with two solubilities of glucose, and then applied to EA.hy926 cells. Alternatively, a co-culture cell system was established with hepatoma cells and EA.hy926 cells. We analyzed the angiogenesis of EA.hy926 cells with CCK8, wound-healing, transwell-migartion and tube formation experiment. ELISA was conducted to detect the secretion levels of angiogenesis-related factors. siRNAs were used to detect the P53-VEGF axis. RESULTS HBx expressed in hepatoma cells suppressed VEGF secretion, and subsequently inhibited the proliferation, migration and tube formation of EA.hy926 cells in a high glucose condition, while attenuating these in the lower glucose condition. Furthermore, the p53-VEGF axis was required for the dual role of HBx in angiogenesis. Additionally, HBx mainly regulated the nuclear p53. CONCLUSIONS These data suggest that the dual roles of HBx confer hepatoma cells to remain in a glucose-rich environment and escape from the glucose-low milieu through tumor vessels, promoting liver tumor progression overall. We exclusively revealed the dual role of HBx on the angiogenesis of liver tumors, which may shed new light on the mechanism and management strategy of HBV- and DM-related hepatocellular carcinoma.
Collapse
Affiliation(s)
- Guitao Xiao
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China; Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, 105, Jiuyibei Road, Xin Luo, Longyan, Fujian 364000, PR China
| | - Xiaoyun Huang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China; Fujian Medical University Cancer Center, Fujian Medical University, 1, Xuefubei Road, Minhou, Fuzhou, Fujian 350001, PR China
| | - Tingxuan Huang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China
| | - Zhixin Chen
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China
| | - Yuehong Huang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China
| | - Rongfeng Huang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China; Fujian Medical University Cancer Center, Fujian Medical University, 1, Xuefubei Road, Minhou, Fuzhou, Fujian 350001, PR China.
| | - Xiaozhong Wang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China; Fujian Medical University Cancer Center, Fujian Medical University, 1, Xuefubei Road, Minhou, Fuzhou, Fujian 350001, PR China.
| |
Collapse
|
2
|
Guo J, Krehl K, Safraou Y, Wallach I, Braun J, Meierhofer D, Sack I, Berndt N. Pregnancy alters fatty acid metabolism, glucose regulation, and detoxification of the liver in synchrony with biomechanical property changes. Heliyon 2024; 10:e39674. [PMID: 39506943 PMCID: PMC11538949 DOI: 10.1016/j.heliyon.2024.e39674] [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: 03/27/2024] [Revised: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Pregnancy places a metabolic burden on the body including the liver, which is responsible for ensuring adequate nutrition for the maternal and fetal systems. To gain a better understanding of liver adaptation, this study investigates metabolic shifts occurring in livers of pregnant rats. Metabolic capacities of the livers of pregnant and non-pregnant female Wistar rats were assessed using comprehensive metabolic models. Kinetic metabolic models were generated for each animal based on protein abundance data from proteomics analysis allowing for a subject-specific assessment of hepatic metabolic functions. Data are available via ProteomeXchange with identifier PXD050758. Additionally, tissue stiffness, viscosity, and water diffusion obtained from magnetic resonance imaging and elastography were correlated with metabolic capabilities to study the relationship between metabolic function and biophysical properties. Proteome profiling revealed differences in protein expression in the livers of pregnant and non-pregnant animals. Functional analysis showed significant variations in metabolic capacities. Livers of pregnant rats had reduced capacities in carbohydrate and fatty acid metabolism, along with altered urea synthesis. Additionally, there were associations between metabolic functions and biophysical properties highlighting potential links between changes in liver structure and metabolic capacities during pregnancy. In summary, our work reveals extensive hepatic metabolic changes in pregnant rats. The liver adapts its metabolic capacities to ensure whole-body metabolic homeostasis but may struggle to counteract nutritional challenges, such as hypoglycemia. The study, employing a personalized approach combining proteomics, kinetic modeling, and advanced imaging, sheds light on the intricate interplay between hepatic adaptations and medical imaging markers, providing a foundation for further investigations into the implications for maternal and fetal health.
Collapse
Affiliation(s)
- Jing Guo
- Department of Radiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Karolina Krehl
- Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Berlin, Germany
| | - Yasmine Safraou
- Department of Radiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Iwona Wallach
- Institute of Computer-assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jürgen Braun
- Institute of Medical Informatics, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Meierhofer
- Mass Spectrometry Facility, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Ingolf Sack
- Department of Radiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nikolaus Berndt
- Institute of Computer-assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| |
Collapse
|
3
|
Dey S, Bhat A, Janani G, Shandilya V, Gupta R, Mandal BB. Microfluidic human physiomimetic liver model as a screening platform for drug induced liver injury. Biomaterials 2024; 310:122627. [PMID: 38823194 DOI: 10.1016/j.biomaterials.2024.122627] [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: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
The pre-clinical animal models often fail to predict intrinsic and idiosyncratic drug induced liver injury (DILI), thus contributing to drug failures in clinical trials, black box warnings and withdrawal of marketed drugs. This suggests a critical need for human-relevant in vitro models to predict diverse DILI phenotypes. In this study, a porcine liver extracellular matrix (ECM) based biomaterial ink with high printing fidelity, biocompatibility and tunable rheological and mechanical properties is formulated for supporting both parenchymal and non-parenchymal cells. Further, we applied 3D printing and microfluidic technology to bioengineer a human physiomimetic liver acinus model (HPLAM), recapitulating the radial hepatic cord-like structure with functional sinusoidal microvasculature network, biochemical and biophysical properties of native liver acinus. Intriguingly, the human derived hepatic cells incorporated HPLAM cultured under physiologically relevant microenvironment, acts as metabolic biofactories manifesting enhanced hepatic functionality, secretome levels and biomarkers expression over several weeks. We also report that the matured HPLAM reproduces dose- and time-dependent hepatotoxic response of human clinical relevance to drugs typically recognized for inducing diverse DILI phenotypes as compared to conventional static culture. Overall, the developed HPLAM emulates in vivo like functions and may provide a useful platform for DILI risk assessment to better determine safety and human risk.
Collapse
Affiliation(s)
- Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Amritha Bhat
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - G Janani
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Vartik Shandilya
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Raghvendra Gupta
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Biman B Mandal
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
4
|
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
|
5
|
Liotta A, Loroch S, Wallach I, Klewe K, Marcus K, Berndt N. Metabolic Adaptation in Epilepsy: From Acute Response to Chronic Impairment. Int J Mol Sci 2024; 25:9640. [PMID: 39273587 PMCID: PMC11395010 DOI: 10.3390/ijms25179640] [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: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Epilepsy is characterized by hypersynchronous neuronal discharges, which are associated with an increased cerebral metabolic rate of oxygen and ATP demand. Uncontrolled seizure activity (status epilepticus) results in mitochondrial exhaustion and ATP depletion, which potentially generate energy mismatch and neuronal loss. Many cells can adapt to increased energy demand by increasing metabolic capacities. However, acute metabolic adaptation during epileptic activity and its relationship to chronic epilepsy remains poorly understood. We elicited seizure-like events (SLEs) in an in vitro model of status epilepticus for eight hours. Electrophysiological recording and tissue oxygen partial pressure recordings were performed. After eight hours of ongoing SLEs, we used proteomics-based kinetic modeling to evaluate changes in metabolic capacities. We compared our findings regarding acute metabolic adaptation to published proteomic and transcriptomic data from chronic epilepsy patients. Epileptic tissue acutely responded to uninterrupted SLEs by upregulating ATP production capacity. This was achieved by a coordinated increase in the abundance of proteins from the respiratory chain and oxidative phosphorylation system. In contrast, chronic epileptic tissue shows a 25-40% decrease in ATP production capacity. In summary, our study reveals that epilepsy leads to dynamic metabolic changes. Acute epileptic activity boosts ATP production, while chronic epilepsy reduces it significantly.
Collapse
Affiliation(s)
- Agustin Liotta
- Department of Anesthesiology and Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Institute of Computer-Assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), 13353 Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Stefan Loroch
- Medizinisches Proteom-Center, Center for Protein Diagnostics (PRODI), Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany
- QC-MS/Fa. Dr. Loroch, BioMedizinZentrum, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Iwona Wallach
- Institute of Computer-Assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), 13353 Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Kristoffer Klewe
- QC-MS/Fa. Dr. Loroch, BioMedizinZentrum, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Center for Protein Diagnostics (PRODI), Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Nikolaus Berndt
- Institute of Computer-Assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), 13353 Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Department of Molecular Toxicology, 14558 Nuthetal, Germany
| |
Collapse
|
6
|
Scheidecker B, Poulain S, Sugimoto M, Arakawa H, Kim SH, Kawanishi T, Kato Y, Danoy M, Nishikawa M, Sakai Y. Mechanobiological stimulation in organ-on-a-chip systems reduces hepatic drug metabolic capacity in favor of regenerative specialization. Biotechnol Bioeng 2024; 121:1435-1452. [PMID: 38184801 DOI: 10.1002/bit.28653] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/08/2024]
Abstract
Hepatic physiology depends on the liver's complex structural composition which among others, provides high oxygen supply rates, locally differential oxygen tension, endothelial paracrine signaling, as well as residual hemodynamic shear stress to resident hepatocytes. While functional improvements were shown by implementing these factors into hepatic culture systems, direct cause-effect relationships are often not well characterized-obfuscating their individual contribution in more complex microphysiological systems. By comparing increasingly complex hepatic in vitro culture systems that gradually implement these parameters, we investigate the influence of the cellular microenvironment to overall hepatic functionality in pharmacological applications. Here, hepatocytes were modulated in terms of oxygen tension and supplementation, endothelial coculture, and exposure to fluid shear stress delineated from oxygen influx. Results from transcriptomic and metabolomic evaluation indicate that particularly oxygen supply rates are critical to enhance cellular functionality-with cellular drug metabolism remaining comparable to physiological conditions after prolonged static culture. Endothelial signaling was found to be a major contributor to differential phenotype formation known as metabolic zonation, indicated by WNT pathway activity. Lastly, oxygen-delineated shear stress was identified to direct cellular fate towards increased hepatic plasticity and regenerative phenotypes at the cost of drug metabolic functionality - in line with regenerative effects observed in vivo. With these results, we provide a systematic evaluation of critical parameters and their impact in hepatic systems. Given their adherence to physiological effects in vivo, this highlights the importance of their implementation in biomimetic devices, such as organ-on-a-chip systems. Considering recent advances in basic liver biology, direct translation of physiological structures into in vitro models is a promising strategy to expand the capabilities of pharmacological models.
Collapse
Affiliation(s)
| | - Stéphane Poulain
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Arakawa
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Soo H Kim
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Takumi Kawanishi
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yukio Kato
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Mathieu Danoy
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Masaki Nishikawa
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:1-95. [DOI: 10.1016/b978-0-7020-8228-3.00001-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
8
|
Li S, Li C, Khan MI, Liu J, Shi Z, Gao D, Qiu B, Ding W. Microneedle array facilitates hepatic sinusoid construction in a large-scale liver-acinus-chip microsystem. MICROSYSTEMS & NANOENGINEERING 2023; 9:75. [PMID: 37303831 PMCID: PMC10247758 DOI: 10.1038/s41378-023-00544-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/03/2023] [Accepted: 02/24/2023] [Indexed: 06/13/2023]
Abstract
Hepatic sinusoids play a key role in maintaining high activities of liver cells in the hepatic acinus. However, the construction of hepatic sinusoids has always been a challenge for liver chips, especially for large-scale liver microsystems. Herein, we report an approach for the construction of hepatic sinusoids. In this approach, hepatic sinusoids are formed by demolding a self-developed microneedle array from a photocurable cell-loaded matrix in a large-scale liver-acinus-chip microsystem with a designed dual blood supply. Primary sinusoids formed by demolded microneedles and spontaneously self-organized secondary sinusoids can be clearly observed. Benefiting from significantly enhanced interstitial flows by formed hepatic sinusoids, cell viability is witnessed to be considerably high, liver microstructure formation occurs, and hepatocyte metabolism is enhanced. In addition, this study preliminarily demonstrates the effects of the resulting oxygen and glucose gradients on hepatocyte functions and the application of the chip in drug testing. This work paves the way for the biofabrication of fully functionalized large-scale liver bioreactors.
Collapse
Affiliation(s)
- Shibo Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027 China
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Chengpan Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Muhammad Imran Khan
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Jing Liu
- School of Biology, Food and Environment, Hefei University, Hefei, Anhui 230601 China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195 USA
| | - Bensheng Qiu
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Weiping Ding
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| |
Collapse
|
9
|
Shahryari M, Keller S, Meierhofer D, Wallach I, Safraou Y, Guo J, Marticorena Garcia SR, Braun J, Makowski MR, Sack I, Berndt N. On the relationship between metabolic capacities and in vivo viscoelastic properties of the liver. Front Bioeng Biotechnol 2023; 10:1042711. [PMID: 36698634 PMCID: PMC9868178 DOI: 10.3389/fbioe.2022.1042711] [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: 09/12/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
The liver is the central metabolic organ. It constantly adapts its metabolic capacity to current physiological requirements. However, the relationship between tissue structure and hepatic function is incompletely understood; this results in a lack of diagnostic markers in medical imaging that can provide information about the liver's metabolic capacity. Therefore, using normal rabbit livers, we combined magnetic resonance elastography (MRE) with proteomics-based kinetic modeling of central liver metabolism to investigate the potential role of MRE for predicting the liver's metabolic function in vivo. Nineteen New Zealand white rabbits were investigated by multifrequency MRE and positron emission tomography (PET). This yielded maps of shear wave speed (SWS), penetration rate (PR) and standardized uptake value (SUV). Proteomic analysis was performed after the scans. Hepatic metabolic functions were assessed on the basis of the HEPATOKIN1 model in combination with a model of hepatic lipid-droplet metabolism using liquid chromatography-mass spectrometry. Our results showed marked differences between individual livers in both metabolic functions and stiffness properties, though not in SUV. When livers were divided into 'stiff' and 'soft' subgroups (cutoff SWS = 1.6 m/s), stiff livers showed a lower capacity for triacylglycerol storage, while at the same time showing an increased capacity for gluconeogenesis and cholesterol synthesis. Furthermore, SWS was correlated with gluconeogenesis and PR with urea production and glutamine exchange. In conclusion, our study indicates a close relationship between the viscoelastic properties of the liver and metabolic function. This could be used in future studies to predict non-invasively the functional reserve capacity of the liver in patients.
Collapse
Affiliation(s)
- Mehrgan Shahryari
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sarah Keller
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Meierhofer
- Mass Spectrometry Facility, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Iwona Wallach
- Institute of Computer-Assisted Cardiovascular Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yasmine Safraou
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jing Guo
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stephan R. Marticorena Garcia
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jürgen Braun
- Institute of Medical Informatics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcus R. Makowski
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Faculty of Medicine, Munich, Germany
| | - Ingolf Sack
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nikolaus Berndt
- Institute of Computer-Assisted Cardiovascular Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,*Correspondence: Nikolaus Berndt,
| |
Collapse
|
10
|
Maeda K, Hagimori S, Sugimoto M, Sakai Y, Nishikawa M. Simulation of the crosstalk between glucose and acetaminophen metabolism in a liver zonation model. Front Pharmacol 2022; 13:995597. [PMID: 36210818 PMCID: PMC9537759 DOI: 10.3389/fphar.2022.995597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The liver metabolizes a variety of substances that sometimes interact and regulate each other. The modeling of a single cell or a single metabolic pathway does not represent the complexity of the organ, including metabolic zonation (heterogeneity of functions) along with liver sinusoids. Here, we integrated multiple metabolic pathways into a single numerical liver zonation model, including drug and glucose metabolism. The model simulated the time-course of metabolite concentrations by the combination of dynamic simulation and metabolic flux analysis and successfully reproduced metabolic zonation and localized hepatotoxicity induced by acetaminophen (APAP). Drug metabolism was affected by nutritional status as the glucuronidation reaction rate changed. Moreover, sensitivity analysis suggested that the reported metabolic characteristics of obese adults and healthy infants in glucose metabolism could be associated with the metabolic features of those in drug metabolism. High activities of phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphate phosphatase in obese adults led to increased APAP oxidation by cytochrome P450 2E1. In contrast, the high activity of glycogen synthase and low activities of PEPCK and glycogen phosphorylase in healthy infants led to low glucuronidation and high sulfation rates of APAP. In summary, this model showed the effects of glucose metabolism on drug metabolism by integrating multiple pathways into a single liver metabolic zonation model.
Collapse
Affiliation(s)
- Kazuhiro Maeda
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, Japan
| | - Shuta Hagimori
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Masahiro Sugimoto
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
- *Correspondence: Masahiro Sugimoto,
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Masaki Nishikawa
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Alterations of Central Liver Metabolism of Pediatric Patients with Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:ijms231911072. [PMID: 36232372 PMCID: PMC9570193 DOI: 10.3390/ijms231911072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 12/02/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in children and is associated with overweight and insulin resistance (IR). Almost nothing is known about in vivo alterations of liver metabolism in NAFLD, especially in the early stages of non-alcoholic steatohepatitis (NASH). Here, we used a complex mathematical model of liver metabolism to quantify the central hepatic metabolic functions of 71 children with biopsy-proven NAFLD. For each patient, a personalized model variant was generated based on enzyme abundances determined by mass spectroscopy. Our analysis revealed statistically significant alterations in the hepatic carbohydrate, lipid, and ammonia metabolism, which increased with the degree of obesity and severity of NAFLD. Histologic features of NASH and IR displayed opposing associations with changes in carbohydrate and lipid metabolism but synergistically decreased urea synthesis in favor of the increased release of glutamine, a driver of liver fibrosis. Taken together, our study reveals already significant alterations in the NASH liver of pediatric patients, which, however, are differently modulated by the simultaneous presence of IR.
Collapse
|
12
|
Builes-Montaño CE, Lema-Perez L, Garcia-Tirado J, Alvarez H. Main glucose hepatic fluxes in healthy subjects predicted from a phenomenological-based model. Comput Biol Med 2022; 142:105232. [DOI: 10.1016/j.compbiomed.2022.105232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/08/2022] [Accepted: 01/09/2022] [Indexed: 11/28/2022]
|
13
|
Chalhoub ER, Belovich JM. Quantitative analysis of the interaction of ethanol metabolism with gluconeogenesis and fatty acid oxidation in the perfused liver of fasted rats. Arch Biochem Biophys 2022; 718:109148. [PMID: 35143783 DOI: 10.1016/j.abb.2022.109148] [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: 10/27/2021] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 12/12/2022]
Abstract
Ethanol is known to significantly affect gluconeogenesis and lipid metabolism in the liver, primarily by altering the redox ratio in both cytosol and mitochondria. The effect of ethanol was analyzed using a comprehensive, dynamic model of liver metabolism that takes into account sub-cellular compartmentation, detailed kinetics for the citric acid cycle, ethanol and acetaldehyde oxidation, and gluconeogenesis, and inter-compartmental transport of metabolites, including the malate-aspartate shuttle. The kinetic expression for alcohol dehydrogenase takes into account inhibition by ethanol and NADH. Simulations of perfusions of the rat liver were performed with various combinations of substrates (lactate, pyruvate, and fatty acids), with subsequent addition of ethanol to the perfusate. The model successfully predicts NADH/NAD+, in both cytosol and mitochondria, the expected directional flux of reducing equivalents between the two compartments during perfusion with different gluconeogenic precursors, and the effect of ethanol on glucose and ketone body production. This model can serve as a platform for in silico experiments investigating the effects of ethanol on the many dehydrogenases, and thus the major carbohydrate and lipid metabolic pathways in the liver, as well as potential effects of various drugs that may interact with ethanol.
Collapse
Affiliation(s)
- Elie R Chalhoub
- Department of Chemical Engineering, University of Balamand, Faculty of Engineering, P.O.Box 100, Tripoli, Lebanon.
| | - Joanne M Belovich
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44115, USA
| |
Collapse
|
14
|
Berndt N, Eckstein J, Wallach I, Nordmeyer S, Kelm M, Kirchner M, Goubergrits L, Schafstedde M, Hennemuth A, Kraus M, Grune T, Mertins P, Kuehne T, Holzhütter HG. CARDIOKIN1: Computational Assessment of Myocardial Metabolic Capability in Healthy Controls and Patients With Valve Diseases. Circulation 2021; 144:1926-1939. [PMID: 34762513 PMCID: PMC8663543 DOI: 10.1161/circulationaha.121.055646] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Many heart diseases can result in reduced pumping capacity of the heart muscle. A mismatch between ATP demand and ATP production of cardiomyocytes is one of the possible causes. Assessment of the relation between myocardial ATP production (MVATP) and cardiac workload is important for better understanding disease development and choice of nutritional or pharmacologic treatment strategies. Because there is no method for measuring MVATP in vivo, the use of physiology-based metabolic models in conjunction with protein abundance data is an attractive approach. METHOD: We developed a comprehensive kinetic model of cardiac energy metabolism (CARDIOKIN1) that recapitulates numerous experimental findings on cardiac metabolism obtained with isolated cardiomyocytes, perfused animal hearts, and in vivo studies with humans. We used the model to assess the energy status of the left ventricle of healthy participants and patients with aortic stenosis and mitral valve insufficiency. Maximal enzyme activities were individually scaled by means of protein abundances in left ventricle tissue samples. The energy status of the left ventricle was quantified by the ATP consumption at rest (MVATP[rest]), at maximal workload (MVATP[max]), and by the myocardial ATP production reserve, representing the span between MVATP(rest) and MVATP(max). Results: Compared with controls, in both groups of patients, MVATP(rest) was increased and MVATP(max) was decreased, resulting in a decreased myocardial ATP production reserve, although all patients had preserved ejection fraction. The variance of the energetic status was high, ranging from decreased to normal values. In both patient groups, the energetic status was tightly associated with mechanic energy demand. A decrease of MVATP(max) was associated with a decrease of the cardiac output, indicating that cardiac functionality and energetic performance of the ventricle are closely coupled. Conclusions: Our analysis suggests that the ATP-producing capacity of the left ventricle of patients with valvular dysfunction is generally diminished and correlates positively with mechanical energy demand and cardiac output. However, large differences exist in the energetic state of the myocardium even in patients with similar clinical or image-based markers of hypertrophy and pump function. Registration: URL: https://www.clinicaltrials.gov; Unique identifiers: NCT03172338 and NCT04068740.
Collapse
Affiliation(s)
- Nikolaus Berndt
- Institute of Computer-assisted Cardiovascular Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Johannes Eckstein
- Institute of Computer-assisted Cardiovascular Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Institute of Biochemistry, Charitá - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Iwona Wallach
- Institute of Computer-assisted Cardiovascular Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Institute of Biochemistry, Charitá - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sarah Nordmeyer
- Institute of Computer-assisted Cardiovascular Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Congenital Heart Disease - Pediatric Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany
| | - Marcus Kelm
- Institute of Computer-assisted Cardiovascular Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Congenital Heart Disease - Pediatric Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung e. V. (DZHK), Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Marieluise Kirchner
- Berlin Institute of Health (BIH), Berlin, Germany; Proteomics Platform, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Leonid Goubergrits
- Institute of Computer-assisted Cardiovascular Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Einstein Center Digital Future, Berlin, Germany
| | - Marie Schafstedde
- Institute of Computer-assisted Cardiovascular Medicine, Charité; Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Congenital Heart Disease - Pediatric Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Anja Hennemuth
- Institute of Computer-assisted Cardiovascular Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Milena Kraus
- Digital Health Center, Hasso Plattner Institute, University of Potsdam, Germany
| | - Tilman Grune
- Deutsches Zentrum für Herz-Kreislauf-Forschung e. V. (DZHK), Berlin, Germany; Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Philipp Mertins
- Berlin Institute of Health (BIH), Berlin, Germany; Proteomics Platform, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Titus Kuehne
- Institute of Computer-assisted Cardiovascular Medicine, Charité; Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Congenital Heart Disease - Pediatric Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung e. V. (DZHK), Berlin, Germany
| | - Hermann-Georg Holzhütter
- Institute of Biochemistry, Charitá - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
15
|
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: 1.3] [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
|
16
|
Berndt N, Kolbe E, Gajowski R, Eckstein J, Ott F, Meierhofer D, Holzhütter HG, Matz-Soja M. Functional Consequences of Metabolic Zonation in Murine Livers: Insights for an Old Story. Hepatology 2021; 73:795-810. [PMID: 32286709 DOI: 10.1002/hep.31274] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 03/13/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Zone-dependent differences in expression of metabolic enzymes along the portocentral axis of the acinus are a long-known feature of liver metabolism. A prominent example is the preferential localization of the enzyme, glutamine synthetase, in pericentral hepatocytes, where it converts potentially toxic ammonia to the valuable amino acid, glutamine. However, with the exception of a few key regulatory enzymes, a comprehensive and quantitative assessment of zonal differences in the abundance of metabolic enzymes and, much more important, an estimation of the associated functional differences between portal and central hepatocytes is missing thus far. APPROACH AND RESULTS We addressed this problem by establishing a method for the separation of periportal and pericentral hepatocytes that yields sufficiently pure fractions of both cell populations. Quantitative shotgun proteomics identified hundreds of differentially expressed enzymes in the two cell populations. We used zone-specific proteomics data for scaling of the maximal activities to generate portal and central instantiations of a comprehensive kinetic model of central hepatic metabolism (Hepatokin1). CONCLUSIONS The model simulations revealed significant portal-to-central differences in almost all metabolic pathways involving carbohydrates, fatty acids, amino acids, and detoxification.
Collapse
Affiliation(s)
- Nikolaus Berndt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt-Universität zu Berlin, and Berlin Institute of HealthInstitute for Imaging Science and Computational Modelling in Cardiovascular MedicineBerlinGermany
| | - Erik Kolbe
- Rudolf-Schönheimer-Institute of BiochemistryFaculty of MedicineLeipzig UniversityLeipzigGermany
| | - Robert Gajowski
- Max Planck Institute for Molecular GeneticsBerlinGermany.,Department of Biology, Chemistry, PharmacyFreie UniversitätBerlinGermany
| | - Johannes Eckstein
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt-Universität zu Berlin, and Berlin Institute of HealthInstitute of BiochemistryBerlinGermany
| | - Fritzi Ott
- Rudolf-Schönheimer-Institute of BiochemistryFaculty of MedicineLeipzig UniversityLeipzigGermany
| | | | - Hermann-Georg Holzhütter
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt-Universität zu Berlin, and Berlin Institute of HealthInstitute of BiochemistryBerlinGermany
| | - Madlen Matz-Soja
- Rudolf-Schönheimer-Institute of BiochemistryFaculty of MedicineLeipzig UniversityLeipzigGermany.,Division of Hepatology, Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious DiseasesLeipzig University Medical CenterLeipzigGermany
| |
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: 2.3] [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
|
Chen T, Oh S, Gregory S, Shen X, Diehl AM. Single-cell omics analysis reveals functional diversification of hepatocytes during liver regeneration. JCI Insight 2020. [DOI: 10.1172/jci.insight.141024 33208554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
19
|
Chen T, Oh S, Gregory S, Shen X, Diehl AM. Single-cell omics analysis reveals functional diversification of hepatocytes during liver regeneration. JCI Insight 2020; 5:141024. [PMID: 33208554 PMCID: PMC7710279 DOI: 10.1172/jci.insight.141024] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/08/2020] [Indexed: 01/07/2023] Open
Abstract
Adult liver has enormous regenerative capacity; it can regenerate after losing two-thirds of its mass while sustaining essential metabolic functions. How the liver balances dual demands for increased proliferative activity with maintenance of organ function is unknown but essential to prevent liver failure. Using partial hepatectomy (PHx) in mice to model liver regeneration, we integrated single-cell RNA- and ATAC-Seq to map state transitions in approximately 13,000 hepatocytes at single-cell resolution as livers regenerated, and validated key findings with IHC, to uncover how the organ regenerates hepatocytes while simultaneously fulfilling its vital tissue-specific functions. After PHx, hepatocytes rapidly and transiently diversified into multiple distinct populations with distinct functional bifurcation: some retained the chromatin landscapes and transcriptomes of hepatocytes in undamaged adult livers, whereas others transitioned to acquire chromatin landscapes and transcriptomes of fetal hepatocytes. Injury-related signaling pathways known to be critical for regeneration were activated in transitioning hepatocytes, and the most fetal-like hepatocytes exhibited chromatin landscapes that were enriched with transcription factors regulated by those pathways.
Collapse
Affiliation(s)
- Tianyi Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Simon Gregory
- Department of Neurology, Duke University, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina, USA
| | - Anna Mae Diehl
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine and
| |
Collapse
|
20
|
Osei-Bordom D, Bozward AG, Oo YH. The hepatic microenvironment and regulatory T cells. Cell Immunol 2020; 357:104195. [PMID: 32861844 DOI: 10.1016/j.cellimm.2020.104195] [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: 05/29/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022]
Abstract
The human liver is regarded as a lymphoid organ that contributes to both local and systemic immune response. Intrahepatic immune cells including regulatory T cells (Tregs) reside in the hepatic microenvironment which is enriched with proinflammatory cytokines, chemokines and metabolites. In addition, the hepatic microenvironment has the unique ability to establish and maintain immune tolerance despite the continuous influx of the gut derived microbial products via the portal vein. Regulatory T cells play a crucial role in maintaining the hepatic tolerogenic state; however, the phenotypic stability, function and survival of Tregs in the inflamed liver microenvironment is still poorly understood. Despite this, Tregs immunotherapy remains as an appealing therapeutic option in autoimmune and immune mediated liver diseases. In order to advance cell therapy, it is important for us to further our understanding of the hepatic microenvironment, with the aim of developing ways to modify the hostile, inflamed environment to one which is more favourable. By doing so, T cell stability and function would be enhanced, resulting in improved clinical outcomes.
Collapse
Affiliation(s)
- Daniel Osei-Bordom
- Centre for Liver Research and NIHR BRC, Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom; European Reference Network Centre: Rare Liver, United Kingdom; Queen Elizabeth Hospital, University Hospital of Birmingham NHS Foundation Trust, United Kingdom
| | - Amber G Bozward
- Centre for Liver Research and NIHR BRC, Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom; European Reference Network Centre: Rare Liver, United Kingdom
| | - Ye Htun Oo
- Centre for Liver Research and NIHR BRC, Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom; European Reference Network Centre: Rare Liver, United Kingdom; Queen Elizabeth Hospital, University Hospital of Birmingham NHS Foundation Trust, United Kingdom.
| |
Collapse
|
21
|
Kang YB(A, Eo J, Bulutoglu B, Yarmush ML, Usta OB. Progressive hypoxia-on-a-chip: An in vitro oxygen gradient model for capturing the effects of hypoxia on primary hepatocytes in health and disease. Biotechnol Bioeng 2020; 117:763-775. [PMID: 31736056 PMCID: PMC7015781 DOI: 10.1002/bit.27225] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/07/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022]
Abstract
Oxygen is vital to the function of all tissues including the liver and lack of oxygen, that is, hypoxia can result in both acute and chronic injuries to the liver in vivo and ex vivo. Furthermore, a permanent oxygen gradient is naturally present along the liver sinusoid, which plays a role in the metabolic zonation and the pathophysiology of liver diseases. Accordingly, here, we introduce an in vitro microfluidic platform capable of actively creating a series of oxygen concentrations on a single continuous microtissue, ranging from normoxia to severe hypoxia. This range approximately captures both the physiologically relevant oxygen gradient generated from the portal vein to the central vein in the liver, and the severe hypoxia occurring in ischemia and liver diseases. Primary rat hepatocytes cultured in this microfluidic platform were exposed to an oxygen gradient of 0.3-6.9%. The establishment of an ascending hypoxia gradient in hepatocytes was confirmed in response to the decreasing oxygen supply. The hepatocyte viability in this platform decreased to approximately 80% along the hypoxia gradient. Simultaneously, a progressive increase in accumulation of reactive oxygen species and expression of hypoxia-inducible factor 1α was observed with increasing hypoxia. These results demonstrate the induction of distinct metabolic and genetic responses in hepatocytes upon exposure to an oxygen (/hypoxia) gradient. This progressive hypoxia-on-a-chip platform can be used to study the role of oxygen and hypoxia-associated molecules in modeling healthy and injured liver tissues. Its use can be further expanded to the study of other hypoxic tissues such as tumors as well as the investigation of drug toxicity and efficacy under oxygen-limited conditions.
Collapse
Affiliation(s)
- Young Bok (Abraham) Kang
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children-Boston, Boston, MA, USA
- College of Engineering, George Fox University, Newberg, OR, USA
| | - Jinsu Eo
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children-Boston, Boston, MA, USA
| | - Beyza Bulutoglu
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children-Boston, Boston, MA, USA
| | - Martin L. Yarmush
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children-Boston, Boston, MA, USA
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - O. Berk Usta
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children-Boston, Boston, MA, USA
| |
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: 1.0] [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
|
Kinetic modelling of quantitative proteome data predicts metabolic reprogramming of liver cancer. Br J Cancer 2019; 122:233-244. [PMID: 31819186 PMCID: PMC7052204 DOI: 10.1038/s41416-019-0659-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/17/2022] Open
Abstract
Background Metabolic alterations can serve as targets for diagnosis and cancer therapy. Due to the highly complex regulation of cellular metabolism, definite identification of metabolic pathway alterations remains challenging and requires sophisticated experimentation. Methods We applied a comprehensive kinetic model of the central carbon metabolism (CCM) to characterise metabolic reprogramming in murine liver cancer. Results We show that relative differences of protein abundances of metabolic enzymes obtained by mass spectrometry can be used to assess their maximal velocity values. Model simulations predicted tumour-specific alterations of various components of the CCM, a selected number of which were subsequently verified by in vitro and in vivo experiments. Furthermore, we demonstrate the ability of the kinetic model to identify metabolic pathways whose inhibition results in selective tumour cell killing. Conclusions Our systems biology approach establishes that combining cellular experimentation with computer simulations of physiology-based metabolic models enables a comprehensive understanding of deregulated energetics in cancer. We propose that modelling proteomics data from human HCC with our approach will enable an individualised metabolic profiling of tumours and predictions of the efficacy of drug therapies targeting specific metabolic pathways.
Collapse
|
24
|
Elsayed HRH, El Nashar EM, Abd-Elmonem MM. Is the hepatocyte ultrastructural zonal heterogeneity changed by overnight (16 h) fasting? Morphometric study. Ultrastruct Pathol 2019; 43:290-300. [PMID: 31791174 DOI: 10.1080/01913123.2019.1696906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Background and objectives: Hepatocyte ultra-structure is influenced by feeding status, circadian rhythm, and zone location. The goal of the present study was to study the effect of overnight fasting on the hepatocyte ultrastructure and the zonal heterogeneity and to discuss the functional correlation.Methods: A total of 14 male albino rats were divided into two groups: negative control group fed ad libitum and overnight fasting rats for 16 hours. The different subcellular structures of both centrilobular and periportal hepatocytes in both control and fasted groups were compared by transmission electron microscopy. Morphometric analysis of the electron micrographs was also done using imageJ software.Results: The lysosomes surface density, mitochondrial volume and surface densities were significantly higher in periportal hepatocytes however surface density of smooth endoplasmic reticulum (SER) and peroxisomes were significantly higher in centrilobular hepatocytes of the control group. Fasting caused a significant decrease in the surface density of rough endoplasmic reticulum and glycogen volume density but with significant increase in SER surface density with more mitochondrial fusion and stronger mitochondrial ER contacts, isolation membranes, and autophagosomes. The zonal differences were maintained after fasting. The organelles appeared normal with no signs of degeneration.Conclusion: The organelles appeared normal with no signs of degeneration and the zonal differences were maintained after fasting. The change in hepatocyte ultrastructure after fasting may be related to autophagy.
Collapse
Affiliation(s)
| | - Eman Mohammad El Nashar
- College of Medicine, Anatomy, King Khalid University, Abha, Saudi Arabia.,Faculty of Medicine, Histology and cell biology, Benha University, Benha, Egypt
| | | |
Collapse
|
25
|
Berndt N, Patzak A, Holzhütter HG. Metabolic modelling of kidney diseases: Lessons learned from the liver. Acta Physiol (Oxf) 2019; 227:e13350. [PMID: 31348847 DOI: 10.1111/apha.13350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Nikolaus Berndt
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute for Computational and Imaging Science in Cardiovascular Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Patzak
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|