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
|
Martini T, Gobet C, Salati A, Blanc J, Mookhoek A, Reinehr M, Knott G, Sordet-Dessimoz J, Naef F. A sexually dimorphic hepatic cycle of periportal VLDL generation and subsequent pericentral VLDLR-mediated re-uptake. Nat Commun 2024; 15:8422. [PMID: 39341814 PMCID: PMC11438914 DOI: 10.1038/s41467-024-52751-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Recent single-cell transcriptomes revealed spatiotemporal programmes of liver function on the sublobular scale. However, how sexual dimorphism affected this space-time logic remained poorly understood. We addressed this by performing scRNA-seq in the mouse liver, which revealed that sex, space and time together markedly influence xenobiotic detoxification and lipoprotein metabolism. The very low density lipoprotein receptor (VLDLR) exhibits a pericentral expression pattern, with significantly higher mRNA and protein levels in female mice. Conversely, VLDL assembly is periportally biased, suggesting a sexually dimorphic hepatic cycle of periportal formation and pericentral uptake of VLDL. In humans, VLDLR expression is also pericentral, with higher mRNA and protein levels in premenopausal women compared to similarly aged men. Individuals with low hepatic VLDLR expression show a high prevalence of atherosis in the coronary artery already at an early age and an increased incidence of heart attack.
Collapse
Affiliation(s)
- Tomaz Martini
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cédric Gobet
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Andrea Salati
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jérôme Blanc
- Bioelectron Microscopy Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Aart Mookhoek
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Michael Reinehr
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Graham Knott
- Bioelectron Microscopy Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jessica Sordet-Dessimoz
- Histology Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
3
|
Seubnooch P, Montani M, Dufour JF, Masoodi M. Spatial lipidomics reveals zone-specific hepatic lipid alteration and remodeling in metabolic dysfunction-associated steatohepatitis. J Lipid Res 2024; 65:100599. [PMID: 39032559 PMCID: PMC11388789 DOI: 10.1016/j.jlr.2024.100599] [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: 03/06/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
Alteration in lipid metabolism plays a pivotal role in developing metabolic dysfunction-associated steatohepatitis (MASH). However, our understanding of alteration in lipid metabolism across liver zonation in MASH remains limited. Within this study, we investigated MASH-associated zone-specific lipid metabolism in a diet and chemical-induced MASH mouse model. Spatial lipidomics using mass spectrometry imaging in a MASH mouse model revealed 130 lipids from various classes altered across liver zonation and exhibited zone-specific lipid signatures in MASH. Triacylglycerols, diacylglycerols, sphingolipids and ceramides showed distinct zone-specific changes and re-distribution from pericentral to periportal localization in MASH. Saturated and monounsaturated fatty acids (FA) were the primary FA composition of increased lipids in MASH, while polyunsaturated FAs were the major FA composition of decreased lipids. We observed elevated fibrosis in the periportal region, which could be the result of observed metabolic alteration across zonation. Our study provides valuable insights into zone-specific hepatic lipid metabolism and demonstrates the significance of spatial lipidomics in understanding liver lipid metabolism. Identifying unique lipid distribution patterns may offer valuable insights into the pathophysiology of MASH and facilitate the discovery of diagnostic markers associated with liver zonation.
Collapse
Affiliation(s)
- Patcharamon Seubnooch
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Matteo Montani
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jean-Francois Dufour
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - Mojgan Masoodi
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland.
| |
Collapse
|
4
|
Santos AA, Delgado TC, Marques V, Ramirez-Moncayo C, Alonso C, Vidal-Puig A, Hall Z, Martínez-Chantar ML, Rodrigues CM. Spatial metabolomics and its application in the liver. Hepatology 2024; 79:1158-1179. [PMID: 36811413 PMCID: PMC11020039 DOI: 10.1097/hep.0000000000000341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/05/2023] [Indexed: 02/24/2023]
Abstract
Hepatocytes work in highly structured, repetitive hepatic lobules. Blood flow across the radial axis of the lobule generates oxygen, nutrient, and hormone gradients, which result in zoned spatial variability and functional diversity. This large heterogeneity suggests that hepatocytes in different lobule zones may have distinct gene expression profiles, metabolic features, regenerative capacity, and susceptibility to damage. Here, we describe the principles of liver zonation, introduce metabolomic approaches to study the spatial heterogeneity of the liver, and highlight the possibility of exploring the spatial metabolic profile, leading to a deeper understanding of the tissue metabolic organization. Spatial metabolomics can also reveal intercellular heterogeneity and its contribution to liver disease. These approaches facilitate the global characterization of liver metabolic function with high spatial resolution along physiological and pathological time scales. This review summarizes the state of the art for spatially resolved metabolomic analysis and the challenges that hinder the achievement of metabolome coverage at the single-cell level. We also discuss several major contributions to the understanding of liver spatial metabolism and conclude with our opinion on the future developments and applications of these exciting new technologies.
Collapse
Affiliation(s)
- André A. Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Teresa C. Delgado
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Bizkaia, Spain
- Congenital Metabolic Disorders, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Vanda Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Carmen Ramirez-Moncayo
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | | | - Antonio Vidal-Puig
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Centro Investigation Principe Felipe, Valencia, Spain
| | - Zoe Hall
- Division of Systems Medicine, Imperial College London, London, UK
| | - María Luz Martínez-Chantar
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Cecilia M.P. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
5
|
Okada J, Landgraf A, Xiaoli AM, Liu L, Horton M, Schuster VL, Yang F, Sidoli S, Qiu Y, Kurland IJ, Eliscovich C, Shinoda K, Pessin JE. Spatial hepatocyte plasticity of gluconeogenesis during the metabolic transitions between fed, fasted and starvation states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591168. [PMID: 38746329 PMCID: PMC11092462 DOI: 10.1101/2024.04.29.591168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The liver acts as a master regulator of metabolic homeostasis in part by performing gluconeogenesis. This process is dysregulated in type 2 diabetes, leading to elevated hepatic glucose output. The parenchymal cells of the liver (hepatocytes) are heterogeneous, existing on an axis between the portal triad and the central vein, and perform distinct functions depending on location in the lobule. Here, using single cell analysis of hepatocytes across the liver lobule, we demonstrate that gluconeogenic gene expression ( Pck1 and G6pc ) is relatively low in the fed state and gradually increases first in the periportal hepatocytes during the initial fasting period. As the time of fasting progresses, pericentral hepatocyte gluconeogenic gene expression increases, and following entry into the starvation state, the pericentral hepatocytes show similar gluconeogenic gene expression to the periportal hepatocytes. Similarly, pyruvate-dependent gluconeogenic activity is approximately 10-fold higher in the periportal hepatocytes during the initial fasting state but only 1.5-fold higher in the starvation state. In parallel, starvation suppresses canonical beta-catenin signaling and modulates expression of pericentral and periportal glutamine synthetase and glutaminase, resulting in an enhanced pericentral glutamine-dependent gluconeogenesis. These findings demonstrate that hepatocyte gluconeogenic gene expression and gluconeogenic activity are highly spatially and temporally plastic across the liver lobule, underscoring the critical importance of using well-defined feeding and fasting conditions to define the basis of hepatic insulin resistance and glucose production.
Collapse
|
6
|
Tian H, Rajbhandari P, Tarolli J, Decker AM, Neelakantan TV, Angerer T, Zandkarimi F, Remotti H, Frache G, Winograd N, Stockwell BR. Multimodal mass spectrometry imaging identifies cell-type-specific metabolic and lipidomic variation in the mammalian liver. Dev Cell 2024; 59:869-881.e6. [PMID: 38359832 DOI: 10.1016/j.devcel.2024.01.025] [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: 09/16/2022] [Revised: 05/11/2023] [Accepted: 01/26/2024] [Indexed: 02/17/2024]
Abstract
Spatial single-cell omics provides a readout of biochemical processes. It is challenging to capture the transient lipidome/metabolome from cells in a native tissue environment. We employed water gas cluster ion beam secondary ion mass spectrometry imaging ([H2O]n>28K-GCIB-SIMS) at ≤3 μm resolution using a cryogenic imaging workflow. This allowed multiple biomolecular imaging modes on the near-native-state liver at single-cell resolution. Our workflow utilizes desorption electrospray ionization (DESI) to build a reference map of metabolic heterogeneity and zonation across liver functional units at tissue level. Cryogenic dual-SIMS integrated metabolomics, lipidomics, and proteomics in the same liver lobules at single-cell level, characterizing the cellular landscape and metabolic states in different cell types. Lipids and metabolites classified liver metabolic zones, cell types and subtypes, highlighting the power of spatial multi-omics at high spatial resolution for understanding celluar and biomolecular organizations in the mammalian liver.
Collapse
Affiliation(s)
- Hua Tian
- Environmental and Occupational Health, Pitt Public Health, Pittsburgh, PA 15261, USA; Children's Neuroscience Institute, School of Medicine, Pittsburgh, PA 15224, USA.
| | - Presha Rajbhandari
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | - Aubrianna M Decker
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | - Tina Angerer
- The Luxembourg Institute of Science and Technology, 4362 Esch-sur-Alzette, Luxembourg; Department of Pharmaceutical Biosciences, Uppsala University, 751 05 Uppsala, Sweden
| | | | - Helen Remotti
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Gilles Frache
- The Luxembourg Institute of Science and Technology, 4362 Esch-sur-Alzette, Luxembourg
| | - Nicholas Winograd
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Chemistry, Columbia University, New York, NY 10027, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
7
|
Kumar BS. Desorption electrospray ionization mass spectrometry imaging (DESI-MSI) in disease diagnosis: an overview. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:3768-3784. [PMID: 37503728 DOI: 10.1039/d3ay00867c] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Tissue analysis, which is essential to histology and is considered the benchmark for the diagnosis and prognosis of many illnesses, including cancer, is significant. During surgery, the surgical margin of the tumor is assessed using the labor-intensive, challenging, and commonly subjective technique known as frozen section histopathology. In the biopsy section, large numbers of molecules can now be visualized at once (ion images) following recent developments in [MSI] mass spectrometry imaging under atmospheric conditions. This is vastly superior to and different from the single optical tissue image processing used in traditional histopathology. This review article will focus on the advancement of desorption electrospray ionization mass spectrometry imaging [DESI-MSI] technique, which is label-free and requires little to no sample preparation. Since the proportion of molecular species in normal and abnormal tissues is different, DESI-MSI can capture ion images of the distributions of lipids and metabolites on biopsy sections, which can provide rich diagnostic information. This is not a systematic review but a summary of well-known, cutting-edge and recent DESI-MSI applications in cancer research between 2018 and 2023.
Collapse
Affiliation(s)
- Bharath Sampath Kumar
- Independent Researcher, 21, B2, 27th Street, Nanganallur, Chennai 61, TamilNadu, India.
| |
Collapse
|
8
|
Seubnooch P, Montani M, Tsouka S, Claude E, Rafiqi U, Perren A, Dufour JF, Masoodi M. Characterisation of hepatic lipid signature distributed across the liver zonation using mass spectrometry imaging. JHEP Rep 2023; 5:100725. [PMID: 37284141 PMCID: PMC10240278 DOI: 10.1016/j.jhepr.2023.100725] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/03/2023] [Accepted: 02/27/2023] [Indexed: 06/08/2023] Open
Abstract
Background & Aims Lipid metabolism plays an important role in liver pathophysiology. The liver lobule asymmetrically distributes oxygen and nutrition, resulting in heterogeneous metabolic functions. Periportal and pericentral hepatocytes have different metabolic functions, which lead to generating liver zonation. We developed spatial metabolic imaging using desorption electrospray ionisation mass spectrometry to investigate lipid distribution across liver zonation with high reproducibility and accuracy. Methods Fresh frozen livers from healthy mice with control diet were analysed using desorption electrospray ionisation mass spectrometry imaging. Imaging was performed at 50 μm × 50 μm pixel size. Regions of interest (ROIs) were manually created by co-registering with histological data to determine the spatial hepatic lipids across liver zonation. The ROIs were confirmed by double immunofluorescence. The mass list of specific ROIs was automatically created, and univariate and multivariate statistical analysis were performed to identify statistically significant lipids across liver zonation. Results A wide range of lipid species was identified, including fatty acids, phospholipids, triacylglycerols, diacylglycerols, ceramides, and sphingolipids. We characterised hepatic lipid signatures in three different liver zones (periportal zone, midzone, and pericentral zone) and validated the reproducibility of our method for measuring a wide range of lipids. Fatty acids were predominantly detected in the periportal region, whereas phospholipids were distributed in both the periportal and pericentral zones. Interestingly, phosphatidylinositols, PI(36:2), PI(36:3), PI(36:4), PI(38:5), and PI(40:6) were located predominantly in the midzone (zone 2). Triacylglycerols and diacylglycerols were detected mainly in the pericentral region. De novo triacylglycerol biosynthesis appeared to be the most influenced pathway across the three zones. Conclusions The ability to accurately assess zone-specific hepatic lipid distribution in the liver could lead to a better understanding of lipid metabolism during the progression of liver disease. Impact and Implications Zone-specific hepatic lipid metabolism could play an important role in lipid homoeostasis during disease progression. Herein, we defined the zone-specific references of hepatic lipid species in the three liver zones using molecular imaging. The de novo triacylglycerol biosynthesis was highlighted as the most influenced pathway across the three zones.
Collapse
Affiliation(s)
- Patcharamon Seubnooch
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - Matteo Montani
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Sofia Tsouka
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
| | | | - Umara Rafiqi
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jean-Francois Dufour
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - Mojgan Masoodi
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
9
|
Jeon YG, Kim YY, Lee G, Kim JB. Physiological and pathological roles of lipogenesis. Nat Metab 2023; 5:735-759. [PMID: 37142787 DOI: 10.1038/s42255-023-00786-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/15/2023] [Indexed: 05/06/2023]
Abstract
Lipids are essential metabolites, which function as energy sources, structural components and signalling mediators. Most cells are able to convert carbohydrates into fatty acids, which are often converted into neutral lipids for storage in the form of lipid droplets. Accumulating evidence suggests that lipogenesis plays a crucial role not only in metabolic tissues for systemic energy homoeostasis but also in immune and nervous systems for their proliferation, differentiation and even pathophysiological roles. Thus, excessive or insufficient lipogenesis is closely associated with aberrations in lipid homoeostasis, potentially leading to pathological consequences, such as dyslipidaemia, diabetes, fatty liver, autoimmune diseases, neurodegenerative diseases and cancers. For systemic energy homoeostasis, multiple enzymes involved in lipogenesis are tightly controlled by transcriptional and post-translational modifications. In this Review, we discuss recent findings regarding the regulatory mechanisms, physiological roles and pathological importance of lipogenesis in multiple tissues such as adipose tissue and the liver, as well as the immune and nervous systems. Furthermore, we briefly introduce the therapeutic implications of lipogenesis modulation.
Collapse
Affiliation(s)
- Yong Geun Jeon
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Ye Young Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Gung Lee
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jae Bum Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
10
|
Cunningham RP, Porat-Shliom N. Liver Zonation - Revisiting Old Questions With New Technologies. Front Physiol 2021; 12:732929. [PMID: 34566696 PMCID: PMC8458816 DOI: 10.3389/fphys.2021.732929] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the ever-increasing prevalence of non-alcoholic fatty liver disease (NAFLD), the etiology and pathogenesis remain poorly understood. This is due, in part, to the liver's complex physiology and architecture. The liver maintains glucose and lipid homeostasis by coordinating numerous metabolic processes with great efficiency. This is made possible by the spatial compartmentalization of metabolic pathways a phenomenon known as liver zonation. Despite the importance of zonation to normal liver function, it is unresolved if and how perturbations to liver zonation can drive hepatic pathophysiology and NAFLD development. While hepatocyte heterogeneity has been identified over a century ago, its examination had been severely hindered due to technological limitations. Recent advances in single cell analysis and imaging technologies now permit further characterization of cells across the liver lobule. This review summarizes the advances in examining liver zonation and elucidating its regulatory role in liver physiology and pathology. Understanding the spatial organization of metabolism is vital to further our knowledge of liver disease and to provide targeted therapeutic avenues.
Collapse
Affiliation(s)
- Rory P Cunningham
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Natalie Porat-Shliom
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
11
|
Rodimova SA, Kuznetsova DS, Bobrov NV, Gulin AA, Vasin AA, Gubina MV, Scheslavsky VI, Elagin VV, Karabut MM, Zagainov VE, Zagaynova EV. Multiphoton Microscopy and Mass Spectrometry for Revealing Metabolic Heterogeneity of Hepatocytes in vivo. Sovrem Tekhnologii Med 2021; 13:18-29. [PMID: 34513073 PMCID: PMC8353720 DOI: 10.17691/stm2021.13.2.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of the investigation was to study the possibility of revealing the heterogeneity of normal liver hepatocytes in terms of metabolic status using the modern methods of multiphoton microscopy and mass spectrometry.
Collapse
Affiliation(s)
- S A Rodimova
- Junior Researcher, Laboratory of Regenerative Medicine, Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; PhD Student, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603950, Russia
| | - D S Kuznetsova
- Researcher, Laboratory of Regenerative Medicine, Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - N V Bobrov
- Assistant, Department of Theoretical Surgery and Transplantology, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Surgeon, Oncology Department, Volga District Medical Centre of Federal Medical Biological Agency of Russia, 14 Ilyinskaya St., Nizhny Novgorod, 603109, Russia
| | - A A Gulin
- Senior Researcher, Acting Head of the Laboratory of Biophotonics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 4 Kosygina St., Moscow, 119991, Russia; Researcher, Faculty of Chemistry, Lomonosov Moscow State University, 1 Leninskiye Gory, Moscow, 119991, Russia
| | - A A Vasin
- Research Engineer, Laboratory of Nanophotonics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 4 Kosygina St., Moscow, 119991, Russia; Student, Faculty of Chemistry, Lomonosov Moscow State University, 1 Leninskiye Gory, Moscow, 119991, Russia
| | - M V Gubina
- Research Engineer, Laboratory of Nanophotonics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 4 Kosygina St., Moscow, 119991, Russia; Student, Phystech School of Electronics, Photonics and Molecular Physics, Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141701, Russia
| | - V I Scheslavsky
- Senior Researcher, Becker & Hickl, GmbH, Nunsdorfer Ring 7-9, Berlin, 12277, Germany; Head of the Laboratory of High-Resolution Microscopy, Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - V V Elagin
- Researcher, Laboratory of High-Resolution Microscopy, Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - M M Karabut
- Researcher, Laboratory of Genomics and Adaptive Antitumor Immunity, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - V E Zagainov
- Head of the Department of Theoretical Surgery and Transplantology, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Chief Specialist in Surgery, Volga District Medical Centre of Federal Medical Biological Agency of Russia, 14 Ilyinskaya St., Nizhny Novgorod, 603109, Russia
| | - E V Zagaynova
- Rector, National Research Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603950, Russia; Senior Researcher, Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| |
Collapse
|
12
|
Li T, Yang H, Li X, Hou Y, Zhao Y, Wu W, Zhao L, Wang F, Zhao Z. Open-flow microperfusion combined with mass spectrometry for in vivo liver lipidomic analysis. Analyst 2021; 146:1915-1923. [PMID: 33481970 DOI: 10.1039/d0an02189j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
At present, conventional microdialysis (MD) techniques cannot efficiently sample lipids in vivo, possibly due to the high mass transfer resistance and/or the serious adsorption of lipids onto the semi-permeable membrane of a MD probe. The in vivo monitoring of lipids could be of great significance for the study of disease development and mechanisms. In this work, an open-flow microperfusion (OFM) probe was fabricated, and the conditions for sampling lipids via OFM were optimized. Using OFM, the recovery of lipid standards was improved to more than 34.7%. OFM is used for the in vivo sampling of lipids in mouse liver tissue with fibrosis, and it is then combined with mass spectrometry (MS) to perform lipidomic analysis. 156 kinds of lipids were identified in the dialysate collected via OFM, and it was found that the phospholipid levels, including PC, PE, and SM, were significantly higher in a liver suffering from fibrosis. For the first time, OFM combined with MS to sample and analyze lipids has provided a promising platform for in vivo lipidomic studies.
Collapse
Affiliation(s)
- Tuo Li
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing 100190, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Hui Yang
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Xing Li
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing 100190, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yinzhu Hou
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing 100190, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yao Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing 100190, China.
| | - Wenjing Wu
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing 100190, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Lingyu Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing 100190, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Fuyi Wang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing 100190, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Zhenwen Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing 100190, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
13
|
Tatsushima K, Hasuzawa N, Wang L, Hiasa M, Sakamoto S, Ashida K, Sudo N, Moriyama Y, Nomura M. Vesicular ATP release from hepatocytes plays a role in the progression of nonalcoholic steatohepatitis. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166013. [PMID: 33212187 DOI: 10.1016/j.bbadis.2020.166013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 12/18/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is becoming a growing public health problem along with the increase of metabolic syndrome worldwide. Extracellular nucleotides are known to serve as a danger signal by initiating purinergic signaling in many inflammatory disorders, although the role of purinergic signaling in the progression of NASH remains to be clarified. Vesicular nucleotide transporter (VNUT) is a key molecule responsible for vesicular ATP release to initiate purinergic signaling. Here, we studied the role of VNUT in the progression of nonalcoholic steatohepatitis. VNUT was expressed in mouse hepatocytes and associated, at least in part, with apolipoprotein B (apoB)-containing vesicles. High glucose stimulation evoked release of appreciable amount of ATP from hepatocytes, which disappeared in hepatocytes of Vnut knockout (Vnut-/-) mice. Glucose treatment also stimulated triglyceride secretion from hepatocytes, which was inhibited by PPADS and MRS211, antagonists of P2Y receptors, and clodronate, a VNUT inhibitor, and was significantly reduced in Vnut-/- mice. In vivo, postprandial secretion of triglyceride from hepatocytes was observed, while the serum triglyceride level was significantly reduced in Vnut-/- mice. On a high-fat diet, the liver of wild type mice exhibited severe inflammation, fibrosis, and macrophage infiltration, which is similar to NASH in humans, while this NASH pathology was not observed in Vnut-/- mice. These results suggest that VNUT-mediated vesicular ATP release regulates triglyceride secretion and involves in chronic inflammation in hepatocytes. Since blockade of vesicular ATP release protects against progression of steatohepatitis, VNUT may be a pharmacological target for NASH.
Collapse
Affiliation(s)
- Keita Tatsushima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Endocrine Center, Toranomon Hospital, Tokyo 105-8470, Japan
| | - Nao Hasuzawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Lixiang Wang
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Miki Hiasa
- Department of Membrane Biochemistry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Shohei Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kenji Ashida
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Nobuyuki Sudo
- Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshinori Moriyama
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan; Department of Membrane Biochemistry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan.
| | - Masatoshi Nomura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan.
| |
Collapse
|
14
|
Pek NMQ, Liu KJ, Nichane M, Ang LT. Controversies Surrounding the Origin of Hepatocytes in Adult Livers and the in Vitro Generation or Propagation of Hepatocytes. Cell Mol Gastroenterol Hepatol 2020; 11:273-290. [PMID: 32992051 PMCID: PMC7695885 DOI: 10.1016/j.jcmgh.2020.09.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/21/2022]
Abstract
Epithelial cells in the liver (known as hepatocytes) are high-performance engines of myriad metabolic functions and versatile responders to liver injury. As hepatocytes metabolize amino acids, alcohol, drugs, and other substrates, they produce and are exposed to a milieu of toxins and harmful byproducts that can damage themselves. In the healthy liver, hepatocytes generally divide slowly. However, after liver injury, hepatocytes can ramp up proliferation to regenerate the liver. Yet, on extensive injury, regeneration falters, and liver failure ensues. It is therefore critical to understand the mechanisms underlying liver regeneration and, in particular, which liver cells are mobilized during liver maintenance and repair. Controversies continue to surround the very existence of hepatic stem cells and, if they exist, their spatial location, multipotency, degree of contribution to regeneration, ploidy, and susceptibility to tumorigenesis. This review discusses these controversies. Finally, we highlight how insights into hepatocyte regeneration and biology in vivo can inform in vitro studies to propagate primary hepatocytes with liver regeneration-associated signals and to generate hepatocytes de novo from pluripotent stem cells.
Collapse
Affiliation(s)
| | | | | | - Lay Teng Ang
- Correspondence Address correspondence to: Lay Teng Ang, PhD, Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford-UC Berkeley Siebel Stem Cell Institute, Stanford University School of Medicine, Stanford, California 94305.
| |
Collapse
|
15
|
Reproducibility across single-cell RNA-seq protocols for spatial ordering analysis. PLoS One 2020; 15:e0239711. [PMID: 32986734 PMCID: PMC7521718 DOI: 10.1371/journal.pone.0239711] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/12/2020] [Indexed: 01/12/2023] Open
Abstract
As newer single-cell protocols generate increasingly more cells at reduced sequencing depths, the value of a higher read depth may be overlooked. Using data from three different single-cell RNA-seq protocols that lend themselves to having either higher read depth (Smart-seq) or many cells (MARS-seq and 10X), we evaluate their ability to recapitulate biological signals in the context of spatial reconstruction. Overall, we find gene expression profiles after spatial reconstruction analysis are highly reproducible between datasets despite being generated by different protocols and using different computational algorithms. While UMI-based protocols such as 10X and MARS-seq allow for capturing more cells, Smart-seq's higher sensitivity and read-depth allow for analysis of lower expressed genes and isoforms. Additionally, we evaluate trade-offs for each protocol by performing subsampling analyses and find that optimizing the balance between sequencing depth and number of cells within a protocol is necessary for efficient use of resources. Our analysis emphasizes the importance of selecting a protocol based on the biological questions and features of interest.
Collapse
|
16
|
Immune-Deficient Pfp/Rag2 -/- Mice Featured Higher Adipose Tissue Mass and Liver Lipid Accumulation with Growing Age than Wildtype C57BL/6N Mice. Cells 2019; 8:cells8080775. [PMID: 31349725 PMCID: PMC6721582 DOI: 10.3390/cells8080775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/13/2019] [Accepted: 07/22/2019] [Indexed: 02/08/2023] Open
Abstract
Aging is a risk factor for adipose tissue dysfunction, which is associated with inflammatory innate immune mechanisms. Since the adipose tissue/liver axis contributes to hepatosteatosis, we sought to determine age-related adipose tissue dysfunction in the context of the activation of the innate immune system fostering fatty liver phenotypes. Using wildtype and immune-deficient mice, we compared visceral adipose tissue and liver mass as well as hepatic lipid storage in young (ca. 14 weeks) and adult (ca. 30 weeks) mice. Adipocyte size was determined as an indicator of adipocyte function and liver steatosis was quantified by hepatic lipid content. Further, lipid storage was investigated under normal and steatosis-inducing culture conditions in isolated hepatocytes. The physiological age-related increase in body weight was associated with a disproportionate increase in adipose tissue mass in immune-deficient mice, which coincided with higher triglyceride storage in the liver. Lipid storage was similar in isolated hepatocytes from wildtype and immune-deficient mice under normal culture conditions but was significantly higher in immune-deficient than in wildtype hepatocytes under steatosis-inducing culture conditions. Immune-deficient mice also displayed increased inflammatory, adipogenic, and lipogenic markers in serum and adipose tissue. Thus, the age-related increase in body weight coincided with an increase in adipose tissue mass and hepatic steatosis. In association with a (pro-)inflammatory milieu, aging thus promotes hepatosteatosis, especially in immune-deficient mice.
Collapse
|
17
|
Galsgaard KD, Pedersen J, Knop FK, Holst JJ, Wewer Albrechtsen NJ. Glucagon Receptor Signaling and Lipid Metabolism. Front Physiol 2019; 10:413. [PMID: 31068828 PMCID: PMC6491692 DOI: 10.3389/fphys.2019.00413] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 03/26/2019] [Indexed: 01/04/2023] Open
Abstract
Glucagon is secreted from the pancreatic alpha cells upon hypoglycemia and stimulates hepatic glucose production. Type 2 diabetes is associated with dysregulated glucagon secretion, and increased glucagon concentrations contribute to the diabetic hyperglycemia. Antagonists of the glucagon receptor have been considered as glucose-lowering therapy in type 2 diabetes patients, but their clinical applicability has been questioned because of reports of therapy-induced increments in liver fat content and increased plasma concentrations of low-density lipoprotein. Conversely, in animal models, increased glucagon receptor signaling has been linked to improved lipid metabolism. Glucagon acts primarily on the liver and by regulating hepatic lipid metabolism glucagon may reduce hepatic lipid accumulation and decrease hepatic lipid secretion. Regarding whole-body lipid metabolism, it is controversial to what extent glucagon influences lipolysis in adipose tissue, particularly in humans. Glucagon receptor agonists combined with glucagon-like peptide 1 receptor agonists (dual agonists) improve dyslipidemia and reduce hepatic steatosis. Collectively, emerging data support an essential role of glucagon for lipid metabolism.
Collapse
Affiliation(s)
- Katrine D Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Cardiology, Nephrology and Endocrinology, Nordsjællands Hospital Hillerød, University of Copenhagen, Hillerød, Denmark
| | - Filip K Knop
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
Nair J, Velpandian T, Das US, Sharma P, Nag T, Mathur SR, Mathur R. Molecular and Metabolic Markers of Fructose Induced Hepatic Insulin Resistance in Developing and Adult Rats are Distinct and Aegle marmelos is an Effective Modulator. Sci Rep 2018; 8:15950. [PMID: 30374065 PMCID: PMC6206063 DOI: 10.1038/s41598-018-33503-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 09/26/2018] [Indexed: 12/21/2022] Open
Abstract
The time course of pathogenesis of fructose mediated hepatic insulin resistance (HepIR) is not well-delineated and we chronicle it here from post-weaning to adulthood stages. Weaned rats were provided for either 4 or 8 weeks, i.e., upto adolescence or adulthood, chow + drinking water, chow + fructose, 15% or chow + fructose, 15% + hydroalcoholic extract of leaves of Aegle marmelos (AM-HM, 500 mg/kg/d, po) and assessed for feed intake, fructose intake, body weight, fasting blood sugar, oral glucose tolerance test, HOMA-IR, insulin tolerance test and lipid profile. Activities of enzymes (glucose-6-phosphatase, hexokinase, phosphofructokinase, aldehyde dehydrogenase), hormones (leptin, ghrelin, insulin), insulin signaling molecules (Akt-PI3k, AMPK, JNK) hallmarks of inflammation (TNF-α), angiogenesis (VEGF), hypoxia (HIF-1), lipogenesis (mTOR) and regulatory nuclear transcription factors of de novo lipogenesis and hepatic insulin resistance gene (SREBP-1, FoxO1) that together govern the hepatic fructose metabolism, were also studied. The effect of fructose-rich environment on metabolic milieu of hepatocytes was confirmed using (human hepatocellular carcinoma) HepG2 cells. Using in vitro model, fructose uptake and glucose output from isolated murine hepatocytes were measured to establish the HepIR under fructose environment and delineate the effect of AM-HM. The leaves from the plant Aegle marmelos (L) Correa were extracted, fractionated and validated for rutin content using LC-MS/MS. The rutin content of extract was quantified and correlated with oral pharmacokinetic parameters in rat. The outcomes of the study suggest that the molecular and metabolic markers of fructose induced HepIR in developing and adult rats are distinct. Further, AM-HM exerts a multi-pronged attack by raising insulin secretion, augmenting insulin action, improving downstream signaling of insulin, reducing overall requirement of insulin and modulating hepatic expression of glucose transporter (Glut2). The butanol fraction of AM-HM holds promise for future development.
Collapse
Affiliation(s)
- Jayachandran Nair
- Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences and Research, Pushp Vihar, Sec III, MB Road, New Delhi, 110017, India
| | - Thirumurthy Velpandian
- Department of Ocular Pharmacology, Dr. R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar East, Aurobindo Marg, New Delhi, 110029, India
| | - Ujjalkumar Subhash Das
- Department of Ocular Pharmacology, Dr. R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar East, Aurobindo Marg, New Delhi, 110029, India
| | - Prateek Sharma
- Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences and Research, Pushp Vihar, Sec III, MB Road, New Delhi, 110017, India
| | - Tapas Nag
- Department of Anatomy, All India Institute of Medical Sciences, Ansari Nagar East, Aurobindo Marg, New Delhi, 110029, India
| | - Sandeep R Mathur
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar East, Aurobindo Marg, New Delhi, 110029, India
| | - Rajani Mathur
- Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences and Research, Pushp Vihar, Sec III, MB Road, New Delhi, 110017, India.
| |
Collapse
|
19
|
Schaarschmidt B, Vlaic S, Medyukhina A, Neugebauer S, Nietzsche S, Gonnert FA, Rödel J, Singer M, Kiehntopf M, Figge MT, Jacobsen ID, Bauer M, Press AT. Molecular signatures of liver dysfunction are distinct in fungal and bacterial infections in mice. Theranostics 2018; 8:3766-3780. [PMID: 30083258 PMCID: PMC6071540 DOI: 10.7150/thno.24333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 04/16/2018] [Indexed: 12/29/2022] Open
Abstract
Rationale: The liver is a central organ not only for metabolism but also immune function. Life-threatening infections of both bacterial and fungal origin can affect liver function but it is yet unknown whether molecular changes differ depending on the pathogen. We aimed to determine whether the hepatic host response to bacterial and fungal infections differs in terms of hepatic metabolism and liver function. Methods: We compared murine models of infection, including bacterial peritoneal contamination and infection (PCI), intraperitoneal and systemic C. albicans infection, at 6 and 24 h post-infection, to sham controls. The molecular hepatic host response was investigated by the detection of regulatory modules based on large-scale protein-protein interaction networks and expression data. Topological analysis of these regulatory modules was used to reveal infection-specific biological processes and molecular mechanisms. Intravital microscopy and immunofluorescence microscopy were used to further analyze specific aspects of pathophysiology such as cholestasis. Results: Down-regulation of lipid catabolism and bile acid synthesis was observed after 6 h in all infection groups. Alterations in lipid catabolism were characterized by accumulation of long chain acylcarnitines and defective beta-oxidation, which affected metabolism by 6 h. While PCI led to an accumulation of unconjugated bile acids (BA), C. albicans infection caused accumulation of conjugated BA independent of the route of infection. Hepatic dye clearance and transporter expression revealed reduced hepatic uptake in fungal infections vs. defects in secretion following polybacterial infection. Conclusion: Molecular phenotypes of lipid accumulation and cholestasis allow differentiation between pathogens as well as routes of infection at early stages in mice. Targeted metabolomics could be a useful tool for the profiling of infected/septic patients and the type of pathogen, with subsequent customization and targeting of therapy.
Collapse
Affiliation(s)
- Barbara Schaarschmidt
- Department for Anesthesiology and Intensive Care Medicine, AG Nanophysiology, Jena University Hospital, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Sebastian Vlaic
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, Jena University Hospital, Jena, Germany
- Research Group Systems Biology/Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany
- Department of Bioinformatics, Friedrich-Schiller-University, Jena, Germany
| | - Anna Medyukhina
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany
| | - Sophie Neugebauer
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| | - Sandor Nietzsche
- Electron Microscopy Center, Jena University Hospital, Jena, Germany
| | - Falk A. Gonnert
- Department for Anesthesiology and Intensive Care Medicine, AG Nanophysiology, Jena University Hospital, Jena, Germany
| | - Jürgen Rödel
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, UK
| | - Michael Kiehntopf
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| | - Marc Thilo Figge
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany
- Friedrich-Schiller-University, Jena, Germany
| | - Ilse D. Jacobsen
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany
- Friedrich-Schiller-University, Jena, Germany
| | - Michael Bauer
- Department for Anesthesiology and Intensive Care Medicine, AG Nanophysiology, Jena University Hospital, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Adrian T. Press
- Department for Anesthesiology and Intensive Care Medicine, AG Nanophysiology, Jena University Hospital, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| |
Collapse
|
20
|
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
|
21
|
|
22
|
Africa JA, Behling CA, Brunt EM, Zhang N, Luo Y, Wells A, Hou J, Belt PH, Kohil R, Lavine JE, Molleston JP, Newton KP, Whitington PF, Schwimmer JB. In Children With Nonalcoholic Fatty Liver Disease, Zone 1 Steatosis Is Associated With Advanced Fibrosis. Clin Gastroenterol Hepatol 2018; 16:438-446.e1. [PMID: 28286193 PMCID: PMC5589478 DOI: 10.1016/j.cgh.2017.02.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 01/26/2017] [Accepted: 02/14/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Focal zone 1 steatosis, although rare in adults with nonalcoholic fatty liver disease (NAFLD), does occur in children with NAFLD. We investigated whether focal zone 1 steatosis and focal zone 3 steatosis are distinct subphenotypes of pediatric NAFLD. We aimed to determine associations between the zonality of steatosis and demographic, clinical, and histologic features in children with NAFLD. METHODS We performed a cross-sectional study of baseline data from 813 children (age <18 years; mean age, 12.8 ± 2.7 years). The subjects had biopsy-proven NAFLD and were enrolled in the Nonalcoholic Steatohepatitis Clinical Research Network. Liver histology was reviewed using the Nonalcoholic Steatohepatitis Clinical Research Network scoring system. RESULTS Zone 1 steatosis was present in 18% of children with NAFLD (n = 146) and zone 3 steatosis was present in 32% (n = 244). Children with zone 1 steatosis were significantly younger (10 vs 14 years; P < .001) and a significantly higher proportion had any fibrosis (81% vs 51%; P < .001) or advanced fibrosis (13% vs 5%; P < .001) compared with children with zone 3 steatosis. In contrast, children with zone 3 steatosis were significantly more likely to have steatohepatitis (30% vs 6% in children with zone 1 steatosis; P < .001). CONCLUSIONS Children with zone 1 or zone 3 distribution of steatosis have an important subphenotype of pediatric NAFLD. Children with zone 1 steatosis are more likely to have advanced fibrosis and children with zone 3 steatosis are more likely to have steatohepatitis. To achieve a comprehensive understanding of pediatric NAFLD, studies of pathophysiology, natural history, and response to treatment should account for the zonality of steatosis.
Collapse
Affiliation(s)
- Jonathan A. Africa
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla, California,Department of Gastroenterology, Rady Children’s Hospital San Diego, San Diego, California
| | - Cynthia A. Behling
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla, California,Department of Pathology, Sharp Memorial Hospital, San Diego, CA
| | | | - Nan Zhang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla, California
| | - Yunjun Luo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla, California
| | - Alan Wells
- Clinical and Translational Research Institute, University of California San Diego School of Medicine, La Jolla, California
| | - Jiayi Hou
- Clinical and Translational Research Institute, University of California San Diego School of Medicine, La Jolla, California
| | | | - Rohit Kohil
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Joel E. Lavine
- Department of Pediatrics, Columbia University, New York, NY
| | - Jean P. Molleston
- Department of Pediatrics, Indiana University, Indianapolis, IN,James Whitcomb Riley Hospital, Indianapolis, IN
| | - Kimberly P. Newton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla, California,Department of Gastroenterology, Rady Children’s Hospital San Diego, San Diego, California
| | | | - Jeffrey B. Schwimmer
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla, California,Department of Gastroenterology, Rady Children’s Hospital San Diego, San Diego, California
| | | |
Collapse
|
23
|
Lemberger UJ, Fuchs CD, Schöfer C, Bileck A, Gerner C, Stojakovic T, Taketo MM, Trauner M, Egger G, Österreicher CH. Hepatocyte specific expression of an oncogenic variant of β-catenin results in lethal metabolic dysfunction in mice. Oncotarget 2018. [PMID: 29541410 PMCID: PMC5834276 DOI: 10.18632/oncotarget.24346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Wnt/β-catenin signaling plays a crucial role in embryogenesis, tissue homeostasis, metabolism and malignant transformation of different organs including the liver. Continuous β-catenin signaling due to somatic mutations in exon 3 of the Ctnnb1 gene is associated with different liver diseases including cancer and cholestasis. Results Expression of a degradation resistant form of β-catenin in hepatocytes resulted in 100% mortality within 31 days after birth. Ctnnb1CAhep mice were characterized by reduced body weight, significantly enlarged livers with hepatocellular fat accumulation around central veins and increased hepatic triglyceride content. Proteomics analysis using whole liver tissue revealed significant deregulation of proteins involved in fat, glucose and mitochondrial energy metabolism, which was also reflected in morphological anomalies of hepatocellular mitochondria. Key enzymes involved in transport and synthesis of fatty acids and cholesterol were significantly deregulated in livers of Ctnnb1CAhep mice. Furthermore, carbohydrate metabolism was substantially disturbed in mutant mice. Conclusion Continuous β-catenin signaling in hepatocytes results in premature death due to severe disturbances of liver associated metabolic pathways and mitochondrial dysfunction. Methods To investigate the influence of permanent β-catenin signaling on liver biology we analyzed mice with hepatocyte specific expression of a dominant stable form of β-catenin (Ctnnb1CAhep) and their WT littermates by serum biochemistry, histology, electron microscopy, mRNA profiling and proteomic analysis of the liver.
Collapse
Affiliation(s)
- Ursula J Lemberger
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.,Hans Popper Laboratory for Molecular Hepatology, Department of Internal Medicine, Medical University of Vienna, Vienna, Austria
| | - Claudia D Fuchs
- Hans Popper Laboratory for Molecular Hepatology, Department of Internal Medicine, Medical University of Vienna, Vienna, Austria
| | - Christian Schöfer
- Department of Cell and Developmental Biology, Medical University of Vienna, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Makoto M Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michael Trauner
- Hans Popper Laboratory for Molecular Hepatology, Department of Internal Medicine, Medical University of Vienna, Vienna, Austria
| | - Gerda Egger
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | | |
Collapse
|
24
|
Abstract
Endocrine is an important and tightly regulated system for maintaining body homeostasis. Endocrine glands produce hormones, which are released into blood stream to guide the target cells responding to all sorts of stimulations. For maintaining body homeostasis, the secretion and activity of a particular hormone needs to be adjusted in responding to environmental challenges such as changes in nutritional status or chronic stress. Hypoxia, a status caused by reduced oxygen availability or imbalance of oxygen consumption/supply in an organ or within a cell, is a stress that affects many physiological and pathological processes. Hypoxic stress in endocrine organs is especially critical because endocrine glands control body homeostasis. Local hypoxia affects not only the particular gland but also the downstream cells/organs regulated by hormones secreted from this gland. Hypoxia-inducible factors (HIFs) are transcription factors that function as master regulators of oxygen homeostasis. Recent studies report that aberrant expression of HIFs in endocrine organs may result in the development and/or progression of diseases including diabetes, endometriosis, infertility and cancers. In this article, we will review recent findings in HIF-mediated endocrine organ dysfunction and the systemic syndromes caused by these disorders.
Collapse
Affiliation(s)
- Hsiu-Chi Lee
- Institute of Basic Medical SciencesCollege of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shaw-Jenq Tsai
- Institute of Basic Medical SciencesCollege of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of PhysiologyCollege of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
25
|
Kubota N, Kubota T, Kajiwara E, Iwamura T, Kumagai H, Watanabe T, Inoue M, Takamoto I, Sasako T, Kumagai K, Kohjima M, Nakamuta M, Moroi M, Sugi K, Noda T, Terauchi Y, Ueki K, Kadowaki T. Differential hepatic distribution of insulin receptor substrates causes selective insulin resistance in diabetes and obesity. Nat Commun 2016; 7:12977. [PMID: 27708333 PMCID: PMC5059684 DOI: 10.1038/ncomms12977] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/23/2016] [Indexed: 12/20/2022] Open
Abstract
Hepatic insulin signalling involves insulin receptor substrates (Irs) 1/2, and is normally associated with the inhibition of gluconeogenesis and activation of lipogenesis. In diabetes and obesity, insulin no longer suppresses hepatic gluconeogenesis, while continuing to activate lipogenesis, a state referred to as 'selective insulin resistance'. Here, we show that 'selective insulin resistance' is caused by the differential expression of Irs1 and Irs2 in different zones of the liver. We demonstrate that hepatic Irs2-knockout mice develop 'selective insulin resistance', whereas mice lacking in Irs1, or both Irs1 and Irs2, develop 'total insulin resistance'. In obese diabetic mice, Irs1/2-mediated insulin signalling is impaired in the periportal zone, which is the primary site of gluconeogenesis, but enhanced in the perivenous zone, which is the primary site of lipogenesis. While hyperinsulinaemia reduces Irs2 expression in both the periportal and perivenous zones, Irs1 expression, which is predominantly in the perivenous zone, remains mostly unaffected. These data suggest that 'selective insulin resistance' is induced by the differential distribution, and alterations of hepatic Irs1 and Irs2 expression.
Collapse
Affiliation(s)
- Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Department of Clinical Nutrition Therapy, The University of Tokyo, Tokyo 113-8655, Japan.,Clinical Nutrition Program, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 162-8636, Japan
| | - Tetsuya Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Clinical Nutrition Program, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 162-8636, Japan.,Division of Cardiovascular Medicine, Toho University, Ohashi Hospital, Tokyo 153-8515, Japan
| | - Eiji Kajiwara
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tomokatsu Iwamura
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroki Kumagai
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Taku Watanabe
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Hokkaido 060-8648, Japan
| | - Mariko Inoue
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Clinical Nutrition Program, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 162-8636, Japan
| | - Iseki Takamoto
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Clinical Nutrition Program, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 162-8636, Japan
| | - Takayoshi Sasako
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | | | - Motoyuki Kohjima
- Department of Gastroenterology, Clinical Research Center, National Hospital Organization Kyushu Medical Center, Fukuoka 810-8563, Japan
| | - Makoto Nakamuta
- Department of Gastroenterology, Clinical Research Center, National Hospital Organization Kyushu Medical Center, Fukuoka 810-8563, Japan
| | - Masao Moroi
- Division of Cardiovascular Medicine, Toho University, Ohashi Hospital, Tokyo 153-8515, Japan
| | - Kaoru Sugi
- Division of Cardiovascular Medicine, Toho University, Ohashi Hospital, Tokyo 153-8515, Japan
| | - Tetsuo Noda
- Department of Cell Biology, Japanese Foundation for Cancer Research-Cancer Institute, Tokyo 135-8550, Japan
| | - Yasuo Terauchi
- Department of Diabetes and Endocrinology, Yokohama City University, School of Medicine, Kanagawa 236-0004, Japan
| | - Kohjiro Ueki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
26
|
Ashworth WB, Davies NA, Bogle IDL. A Computational Model of Hepatic Energy Metabolism: Understanding Zonated Damage and Steatosis in NAFLD. PLoS Comput Biol 2016; 12:e1005105. [PMID: 27632189 PMCID: PMC5025084 DOI: 10.1371/journal.pcbi.1005105] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 08/12/2016] [Indexed: 12/20/2022] Open
Abstract
In non-alcoholic fatty liver disease (NAFLD), lipid build-up and the resulting damage is known to occur more severely in pericentral cells. Due to the complexity of studying individual regions of the sinusoid, the causes of this zone specificity and its implications on treatment are largely ignored. In this study, a computational model of liver glucose and lipid metabolism is presented which treats the sinusoid as the repeating unit of the liver rather than the single hepatocyte. This allows for inclusion of zonated enzyme expression by splitting the sinusoid into periportal to pericentral compartments. By simulating insulin resistance (IR) and high intake diets leading to the development of steatosis in the model, we identify key differences between periportal and pericentral cells accounting for higher susceptibility to pericentral steatosis. Secondly, variation between individuals is seen in both susceptibility to steatosis and in its development across the sinusoid. Around 25% of obese individuals do not show excess liver fat, whilst 16% of lean individuals develop NAFLD. Furthermore, whilst pericentral cells tend to show higher lipid levels, variation is seen in the predominant location of steatosis from pericentral to pan-sinusoidal or azonal. Sensitivity analysis was used to identify the processes which have the largest effect on both total hepatic triglyceride levels and on the sinusoidal location of steatosis. As is seen in vivo, steatosis occurs when simulating IR in the model, predominantly due to increased uptake, along with an increase in de novo lipogenesis. Additionally, concentrations of glucose intermediates including glycerol-3-phosphate increased when simulating IR due to inhibited glycogen synthesis. Several differences between zones contributed to a higher susceptibility to steatosis in pericentral cells in the model simulations. Firstly, the periportal zonation of both glycogen synthase and the oxidative phosphorylation enzymes meant that the build-up of glucose intermediates was less severe in the periportal hepatocyte compartments. Secondly, the periportal zonation of the enzymes mediating β-oxidation and oxidative phosphorylation resulted in excess fats being metabolised more rapidly in the periportal hepatocyte compartments. Finally, the pericentral expression of de novo lipogenesis contributed to pericentral steatosis when additionally simulating the increase in sterol-regulatory element binding protein 1c (SREBP-1c) seen in NAFLD patients in vivo. The hepatic triglyceride concentration was predicted to be most sensitive to inter-individual variation in the activity of enzymes which, either directly or indirectly, determine the rate of free fatty acid (FFA) oxidation. The concentration was most strongly dependent on the rate constants for β-oxidation and oxidative phosphorylation. It also showed moderate sensitivity to the rate constants for processes which alter the allosteric inhibition of β-oxidation by acetyl-CoA. The predominant sinusoidal location of steatosis meanwhile was most sensitive variations in the zonation of proteins mediating FFA uptake or triglyceride release as very low density lipoproteins (VLDL). Neither the total hepatic concentration nor the location of steatosis showed strong sensitivity to variations in the lipogenic rate constants.
Collapse
Affiliation(s)
- William B. Ashworth
- Institute of Liver and Digestive Health, University College London, London, United Kingdom
- Department of Chemical Engineering, University College London, London, United Kingdom
- CoMPLEX, University College London, London, United Kingdom
| | - Nathan A. Davies
- Institute of Liver and Digestive Health, University College London, London, United Kingdom
| | - I. David L. Bogle
- Department of Chemical Engineering, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
27
|
Selective Insulin Resistance in the Kidney. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5825170. [PMID: 27247938 PMCID: PMC4876201 DOI: 10.1155/2016/5825170] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/04/2016] [Accepted: 04/07/2016] [Indexed: 12/12/2022]
Abstract
Insulin resistance has been characterized as attenuation of insulin sensitivity at target organs and tissues, such as muscle and fat tissues and the liver. The insulin signaling cascade is divided into major pathways such as the PI3K/Akt pathway and the MAPK/MEK pathway. In insulin resistance, however, these pathways are not equally impaired. For example, in the liver, inhibition of gluconeogenesis by the insulin receptor substrate (IRS) 2 pathway is impaired, while lipogenesis by the IRS1 pathway is preserved, thus causing hyperglycemia and hyperlipidemia. It has been recently suggested that selective impairment of insulin signaling cascades in insulin resistance also occurs in the kidney. In the renal proximal tubule, insulin signaling via IRS1 is inhibited, while insulin signaling via IRS2 is preserved. Insulin signaling via IRS2 continues to stimulate sodium reabsorption in the proximal tubule and causes sodium retention, edema, and hypertension. IRS1 signaling deficiency in the proximal tubule may impair IRS1-mediated inhibition of gluconeogenesis, which could induce hyperglycemia by preserving glucose production. In the glomerulus, the impairment of IRS1 signaling deteriorates the structure and function of podocyte and endothelial cells, possibly causing diabetic nephropathy. This paper mainly describes selective insulin resistance in the kidney, focusing on the proximal tubule.
Collapse
|
28
|
Zhang P, Li L, Bao Z, Huang F. Role of BAF60a/BAF60c in chromatin remodeling and hepatic lipid metabolism. Nutr Metab (Lond) 2016; 13:30. [PMID: 27127533 PMCID: PMC4848843 DOI: 10.1186/s12986-016-0090-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/19/2016] [Indexed: 02/07/2023] Open
Abstract
The switching defective/sucrose non-fermenting (SWI/SNF) complexes play an important role in hepatic lipid metabolism regulating both transcriptional activation and repression. BAF60a is a core subunit of the SWI/SNF chromatin-remodeling complexes that activates the transcription of fatty acid oxidation genes during fasting/glucagon. BAF60c, another subunit of SWI/SNF complexes, is recruited to form the lipoBAF complex that activates lipogenic genes, promoting lipogenesis and increasing the triglyceride level in response to feeding/insulin. Interestingly, hepatocytes located in the periportal and perivenous zones of the liver display a remarkable heterogeneity in the activity of various enzymes, metabolic functions and gene expression. Especially, fatty-acid oxidation was shown to be mostly periportal, whereas lipogenesis was mostly perivenous. Therefore, the present review highlights the role of of SWI/SNF regulating lipid metabolism under nutritional and hormonal control, which may be associated with hepatocyte heterogeneity.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Lulu Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Zhengxi Bao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Feiruo Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| |
Collapse
|
29
|
Ishikawa M, Saito K, Yamada H, Nakatsu N, Maekawa K, Saito Y. Plasma lipid profiling of different types of hepatic fibrosis induced by carbon tetrachloride and lomustine in rats. Lipids Health Dis 2016; 15:74. [PMID: 27068811 PMCID: PMC4828842 DOI: 10.1186/s12944-016-0244-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/06/2016] [Indexed: 12/15/2022] Open
Abstract
Background Plasma lipid profiling has emerged as a useful tool for understanding the pathophysiology of hepatic injury and disease. Hepatic fibrosis results from chronic, progressive damage to the liver and can lead, in turn, to more serious conditions such as hepatic cirrhosis and hepatocellular carcinoma. Thus, the present study aimed to investigate the plasma lipid profiles of two types of hepatic fibrosis in order to aid the understanding of the pathophysiology of hepatic fibrosis. Methods A liquid chromatography and mass spectrometry platform was used to reveal and compare the plasma lipid profiles of two types of chemical-induced hepatic fibrosis. Rat models of centrilobular fibrosis and bile duct fibrosis were established via chronic exposure to the known fibrogenic hepatotoxins, carbon tetrachloride (CCl4) or lomustine (LS), respectively, over a 28-day period. To delineate the specific alterations in the lipid profiles as a result of the hepatic fibrosis, we also employed non-fibrogenic hepatotoxicants (2-acetamidofluorene, N-nitrosodiethylamine, and ethambutol) as well as 3-day treatment of CCl4 and LS, which did not induce fibrosis. Results Our assay platform identified 228 lipids in the rat plasma, and the global lipid profile clearly distinguished these models from the control via principal component analysis. In addition, the alteration of the plasma lipid profile caused by CCl4 and LS were clearly different. Furthermore, a number of lipids were identified as specific alterations caused by fibrosis induced only by CCl4 and LS, respectively. Three lysophosphatidylcholines (LPC[18:3], LPC[20:4], and LPC[22:6]), and three phosphatidylcholines (PC[18:2/20:4], PC[40:8], and PC[20:4/22:6]) are specific circulating lipids, the levels of which were altered by both CCl4 and LS treatment; however, their levels were decreased by chronic exposure to CCl4 and increased by chronic exposure to LS. Conclusions These results suggest that different types of chemical-induced hepatic fibrosis demonstrate clear differences in their plasma lipid profiles. Our study provides insights into the alteration of plasma lipidomic profiles as a result of the fibrosis of different parts of the hepatic lobule, and may help to understand the pathophysiology of different types of hepatic fibrosis. Electronic supplementary material The online version of this article (doi:10.1186/s12944-016-0244-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masaki Ishikawa
- Division of Medical Safety Science, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya, Tokyo, 158-8501, Japan
| | - Kosuke Saito
- Division of Medical Safety Science, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya, Tokyo, 158-8501, Japan.
| | - Hiroshi Yamada
- Toxicogenomics Informatics Project, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saitoazagi, Ibaraki, Osaka, 567-0085, Japan
| | - Noriyuki Nakatsu
- Toxicogenomics Informatics Project, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saitoazagi, Ibaraki, Osaka, 567-0085, Japan
| | - Keiko Maekawa
- Division of Medical Safety Science, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya, Tokyo, 158-8501, Japan
| | - Yoshiro Saito
- Division of Medical Safety Science, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya, Tokyo, 158-8501, Japan
| |
Collapse
|
30
|
van den Beukel JC, Boon MR, Steenbergen J, Rensen PCN, Meijer OC, Themmen APN, Grefhorst A. Cold Exposure Partially Corrects Disturbances in Lipid Metabolism in a Male Mouse Model of Glucocorticoid Excess. Endocrinology 2015; 156:4115-28. [PMID: 26372178 DOI: 10.1210/en.2015-1092] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
High glucocorticoid concentrations are accompanied by metabolic side effects such as high plasma triglyceride (TG) concentrations. Liver, brown adipose tissue (BAT) and white adipose tissue are important regulators of plasma TG. Exposure to 4°C reduces plasma TG concentrations, and we therefore aimed to study the interaction between glucocorticoid excess and 24 hours of exposure to 4°C on lipid metabolism. For this, mice were implanted with 50-mg corticosterone or control pellets and housed for 24 hours at 23°C or 4°C 1 week later, after which various aspects of TG metabolism in liver, BAT, and white adipose tissue were studied. Corticosterone treatment resulted in a 3.8-fold increase of plasma TG concentrations. Increased TG was normalized by cold exposure, an effect still present 24 hours after cold exposure. Corticosterone treatment increased hepatic TG content by 3.5-fold and provoked secretion of large, TG-rich very low density lipoprotein particles. Cold exposure reduced very low density lipoprotein-TG secretion by approximately 50%. Corticosterone strongly decreased BAT activity: BAT weight increased by 3.5-fold, whereas uncoupling protein 1 (Ucp1) mRNA expression and Ucp1 protein content of BAT were reduced by 75% and 60%, respectively. Cold exposure partially normalized these parameters of BAT activity. The uptake of TG by BAT was not affected by corticosterone treatment but was increased 4.5-fold upon cold exposure. In conclusion, cold exposure normalizes corticosterone-induced hypertriglyceridemia, at least partly via activating BAT.
Collapse
Affiliation(s)
- Johanna C van den Beukel
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Mariëtte R Boon
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jacobie Steenbergen
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Patrick C N Rensen
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Onno C Meijer
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Axel P N Themmen
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Aldo Grefhorst
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
31
|
Abstract
In insulin-resistant states (obesity, pre-diabetes, and type 2 diabetes), hepatic production of glucose and lipid synthesis are heightened in concert, implying that insulin deficiency and insulin excess coexists in this setting. The fact that insulin may be inadequate or excessive at any one point in differing organs and tissues has many biologic ramifications. In this context the concept of metabolic compartmentalization in the liver is offered herein as one perspective of this paradox. In particular, we focus on the hypothesis that insulin resistance accentuates differences in periportal and perivenous hepatocytes, namely periportal glucose production and perivenous lipid synthesis. Subsequently, excessive production of glucose and accumulation of lipids could be expected in the livers of patients with obesity and insulin resistance. Overall, in this review, we provide our integrative perspective regarding how excessive production of glucose in periportal hepatocytes and accumulation of lipids in perivenous hepatocytes interact in insulin resistant states.
Collapse
Affiliation(s)
- Roberto B Bazotte
- a Department of Pharmacology and Therapeutics ; State University of Maringá ; Maringá , Paraná , PR Brazil
| | | | | |
Collapse
|
32
|
Carotti S, Vespasiani-Gentilucci U, Perrone G, Picardi A, Morini S. Portal inflammation during NAFLD is frequent and associated with the early phases of putative hepatic progenitor cell activation. J Clin Pathol 2015; 68:883-90. [PMID: 26124313 DOI: 10.1136/jclinpath-2014-202717] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 06/11/2015] [Indexed: 12/19/2022]
Abstract
AIMS We investigated whether portal tract inflammation observed in non-alcoholic fatty liver disease (NAFLD) is associated with hepatic progenitor cell compartment activation, as thoroughly evaluated with different markers of the staminal lineage. METHODS Fifty-two patients with NAFLD were studied. NAFLD activity score, fibrosis and portal inflammation were histologically evaluated. Putative hepatic progenitor cells, intermediate hepatobiliary cells and bile ductules/interlobular bile ducts were evaluated by immunohistochemistry for cytokeratin (CK)-7, CK-19 and epithelial cell adhesion molecule (EpCAM), and a hepatic progenitor cell compartment score was derived. Hepatic stellate cell and myofibroblast activity was determined by immunohistochemistry for α-smooth muscle actin. RESULTS Portal inflammation was absent in a minority of patients, mild in 40% of cases and more than mild in about half of patients, showing a strong correlation with fibrosis (r=0.76, p<0.001). Portal inflammation correlated with CK-7-counted putative hepatic progenitor cells (r=0.48, p<0.001), intermediate hepatobiliary cells (r=0.6, p<0.001) and bile ductules/interlobular bile ducts (r=0.6, p<0.001), and with the activity of myofibroblasts (r=0.5, p<0.001). Correlations were confirmed when elements were counted by immunostaining for CK-19 and EpCAM. Lobular inflammation, ballooning, myofibroblast activity and hepatic progenitor cell compartment activation were associated with portal inflammation by univariate analysis. In the multivariate model, the only variable independently associated with portal inflammation was hepatic progenitor cell compartment activation (OR 3.7, 95% CI 1.1 to 12.6). CONCLUSIONS Portal inflammation is frequent during NAFLD and strongly associated with activation of putative hepatic progenitor cells since the first steps of their differentiation, portal myofibroblast activity and fibrosis.
Collapse
Affiliation(s)
- Simone Carotti
- Laboratory of Microscopic and Ultrastructural Anatomy, CIR, University Campus Bio-Medico of Rome, Rome, Italy
| | | | - Giuseppe Perrone
- Department of Anatomical Pathology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Antonio Picardi
- Clinical Medicine and Hepatology Unit, University Campus Bio-Medico of Rome, Rome, Italy
| | - Sergio Morini
- Laboratory of Microscopic and Ultrastructural Anatomy, CIR, University Campus Bio-Medico of Rome, Rome, Italy
| |
Collapse
|
33
|
Schleicher J, Tokarski C, Marbach E, Matz-Soja M, Zellmer S, Gebhardt R, Schuster S. Zonation of hepatic fatty acid metabolism - The diversity of its regulation and the benefit of modeling. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:641-56. [PMID: 25677822 DOI: 10.1016/j.bbalip.2015.02.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/26/2015] [Accepted: 02/03/2015] [Indexed: 02/07/2023]
Abstract
A pronounced heterogeneity between hepatocytes in subcellular structure and enzyme activities was discovered more than 50years ago and initiated the idea of metabolic zonation. In the last decades zonation patterns of liver metabolism were extensively investigated for carbohydrate, nitrogen and lipid metabolism. The present review focuses on zonation patterns of the latter. We review recent findings regarding the zonation of fatty acid uptake and oxidation, ketogenesis, triglyceride synthesis and secretion, de novo lipogenesis, as well as bile acid and cholesterol metabolism. In doing so, we expose knowledge gaps and discuss contradictory experimental results, for example on the zonation pattern of fatty acid oxidation and de novo lipogenesis. Thus, possible rewarding directions of further research are identified. Furthermore, recent findings about the regulation of metabolic zonation are summarized, especially regarding the role of hormones, nerve innervation, morphogens, gender differences and the influence of the circadian clock. In the last part of the review, a short collection of models considering hepatic lipid metabolism is provided. We conclude that modeling, despite its proven benefit for understanding of hepatic carbohydrate and ammonia metabolisms, has so far been largely disregarded in the study of lipid metabolism; therefore some possible fields of modeling interest are presented.
Collapse
Affiliation(s)
- J Schleicher
- Department of Bioinformatics, University of Jena, Jena, Germany.
| | - C Tokarski
- Department of Bioinformatics, University of Jena, Jena, Germany
| | - E Marbach
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - M Matz-Soja
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - S Zellmer
- Department of Chemicals and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - R Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - S Schuster
- Department of Bioinformatics, University of Jena, Jena, Germany
| |
Collapse
|
34
|
Vespasiani-Gentilucci U, Carotti S, Perrone G, Mazzarelli C, Galati G, Onetti-Muda A, Picardi A, Morini S. Hepatic toll-like receptor 4 expression is associated with portal inflammation and fibrosis in patients with NAFLD. Liver Int 2015; 35:569-81. [PMID: 24649857 DOI: 10.1111/liv.12531] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 03/09/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Notwithstanding evidences implicating the lipopolysaccharides (LPS)/toll-like receptor-4 (TLR4) axis in the pathogenesis of NAFLD, there are no studies aimed to characterize hepatic TLR4 expression in NAFLD patients. We aimed to analyse hepatic TLR4 expression and to verify its relationship with disease activity/evolution in NAFLD patients. METHODS Liver tissue from 74 patients with NAFLD and 12 controls was analysed by immunohistochemistry (IHC) for TLR4, α-smooth muscle actin (α-SMA) and cytokeratin-7. IHC for α-SMA was used to evaluate activation of fibrogenic cells (hepatic stellate cells and portal/septal myofibroblasts), that for cytokeratin-7 to count hepatic progenitor cells and bile ducts/ductules, and that for CD68, in a subgroup of 27 patients, for detecting macrophages. Serum LPS-binding protein (LBP), a sensitive marker of LPS activity, was determined in 36 patients and 32 controls. RESULTS As confirmed by double-labelling experiments, the highest level of TLR4 expression was observed in hepatic progenitor cells, biliary cells and portal/septal macrophages. TLR4-positive hepatic progenitor cells and bile ducts/ductules correlated with portal/interface inflammation, activity of fibrogenic cells and fibrosis (P < 0.001). Also the score of TLR4 positivity of porto-septal inflammatory infiltrate correlated with number of hepatic progenitor cells and bile ducts/ductules, activity of fibrogenic cells and fibrosis (P < 0.01). Serum LBP was increased in patients compared to controls (P < 0.001), and correlated with portal/interface inflammation, activity of portal/septal myofibroblasts and fibrosis (all P < 0.05). CONCLUSIONS TLR4 expression by regenerating and inflammatory cells at the porto-septal and interface level, favoured by increased LPS activity, is associated with activation of fibrogenic cells and the degree of fibrosis.
Collapse
|
35
|
Wilson GK, Tennant DA, McKeating JA. Hypoxia inducible factors in liver disease and hepatocellular carcinoma: current understanding and future directions. J Hepatol 2014; 61:1397-406. [PMID: 25157983 DOI: 10.1016/j.jhep.2014.08.025] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 08/07/2014] [Accepted: 08/17/2014] [Indexed: 02/07/2023]
Abstract
Hypoxia inducible transcription factors (HIFs) activate diverse pathways that regulate cellular metabolism, angiogenesis, proliferation, and migration, enabling a cell to respond to a low oxygen or hypoxic environment. HIFs are regulated by oxygen-dependent and independent signals including: mitochondrial dysfunction, reactive oxygen species, endoplasmic reticular stress, and viral infection. HIFs have been reported to play a role in the pathogenesis of liver disease of diverse aetiologies. This review explores the impact of HIFs on hepatocellular biology and inflammatory responses, highlighting the therapeutic potential of targeting HIFs for an array of liver pathologies.
Collapse
Affiliation(s)
- Garrick K Wilson
- Viral Hepatitis Research Group, Centre for Human Virology, University of Birmingham, Birmingham, UK
| | - Daniel A Tennant
- School of Cancer Sciences, University of Birmingham, Birmingham, UK
| | - Jane A McKeating
- Viral Hepatitis Research Group, Centre for Human Virology, University of Birmingham, Birmingham, UK; NIHR Liver Biomedical Research Unit, University of Birmingham, Birmingham, UK.
| |
Collapse
|
36
|
Hijmans BS, Grefhorst A, Oosterveer MH, Groen AK. Zonation of glucose and fatty acid metabolism in the liver: mechanism and metabolic consequences. Biochimie 2013; 96:121-9. [PMID: 23792151 DOI: 10.1016/j.biochi.2013.06.007] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 06/12/2013] [Indexed: 12/13/2022]
Abstract
The liver is generally considered as a relatively homogeneous organ containing four different cell types. It is however well-known that the liver is not homogeneous and consists of clearly demarcated metabolic zones. Hepatocytes from different zones show phenotypical heterogeneity in metabolic features, leading to zonation of metabolic processes across the liver acinus. Zonation of processes involved in glucose and fatty acid metabolism is rather flexible and therefore prone to change under (patho)physiological conditions. Hepatic zonation appears to play an important role in the segregation of the different metabolic pathways in the liver. As a consequence, perturbations in metabolic zonation may be a part of metabolic liver diseases. The metabolic syndrome is characterized by the inability of insulin to adequately suppress hepatic gluconeogenesis, leading to hyperglycemia, hyperinsulinemia and eventually to type II diabetes. As insulin promotes lipogenesis through the transcription factor sterol regulatory element binding protein (SREBP)-1c, one would expect that lipogenesis should also be impaired in insulin-resistant states. However, in the metabolic syndrome hepatic de novo lipogenesis is increased, leading to hyperlipidemia and hepatosteatosis, primarily in the pericentral zone. These observations suggest the co-existence of insulin resistant glucose metabolism and insulin sensitive lipid metabolism in the metabolic syndrome. Here we provide a theoretical framework to explain this so-called 'insulin signaling paradox' in the context of metabolic zonation of the liver.
Collapse
Affiliation(s)
- Brenda S Hijmans
- Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, The Netherlands.
| | | | | | | |
Collapse
|
37
|
Wattacheril J, Seeley EH, Angel P, Chen H, Bowen BP, Lanciault C, M.Caprioli R, Abumrad N, Flynn CR. Differential intrahepatic phospholipid zonation in simple steatosis and nonalcoholic steatohepatitis. PLoS One 2013; 8:e57165. [PMID: 23451176 PMCID: PMC3581520 DOI: 10.1371/journal.pone.0057165] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/18/2013] [Indexed: 12/24/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) occurs frequently in a setting of obesity, dyslipidemia and insulin resistance, but the etiology of the disease, particularly the events favoring progression to nonalcoholic steatohepatitis (NASH) as opposed to simple steatosis (SS), are not fully understood. Based on known zonation patterns in protein, glucose and lipid metabolism, coupled with evidence that phosphatidylcholine may play a role in NASH pathogenesis, we hypothesized that phospholipid zonation exists in liver and that specific phospholipid abundance and distribution may be associated with histologic disease. A survey of normal hepatic protein expression profiles in the Human Protein Atlas revealed pronounced zonation of enzymes involved in lipid utilization and storage, particularly those facilitating phosphatidylcholine (PC) metabolism. Immunohistochemistry of obese normal, SS and NASH liver specimens with anti-phosphatidylethanomine N-methyltransferase (PEMT) antibodies showed a progressive decrease in the zonal distribution of this PC biosynthetic enzyme. Phospholipid quantitation by liquid chromatography mass spectrometry (LC-MS) in hepatic extracts of Class III obese patients with increasing NAFLD severity revealed that most PC species with 32, 34 and 36 carbons as well as total PC abundance was decreased with SS and NASH. Matrix assisted laser desorption ionization - imaging mass spectrometry (MALDI-IMS) imaging revealed strong zonal distributions for 32, 34 and 36 carbon PCs in controls (minimal histologic findings) and SS that was lost in NASH specimens. Specific lipid species such as PC 34∶1 and PC 36∶2 best illustrated this phenomenon. These findings suggest that phospholipid zonation may be associated with the presence of an intrahepatic proinflammatory phenotype and thus have broad implications in the etiopathogenesis of NASH.
Collapse
Affiliation(s)
- Julia Wattacheril
- Columbia University, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, United States of America
| | - Erin H. Seeley
- Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Peggi Angel
- Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Benjamin P. Bowen
- GTL Bioenergy and Structural Biology, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Christian Lanciault
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Richard M.Caprioli
- Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Naji Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Charles Robb Flynn
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
38
|
Fukano Y, Tsuyama N, Mizuno H, Date S, Takano M, Masujima T. Drug metabolite heterogeneity in cultured single cells profiled by pico-trapping direct mass spectrometry. Nanomedicine (Lond) 2012; 7:1365-74. [DOI: 10.2217/nnm.12.34] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Aim: We investigated the heterogeneity of tafluprost metabolism in primary human hepatocytes at a single-cell level by live single-cell mass spectrometry (MS). Materials & methods: Picoliter volumes of cytoplasm were analyzed by nano-electrospray ionization MS in order to obtain single-cell metabolite profiles. The subcellular components of a single tafluprost-treated human hepatocyte were isolated and the single-cell metabolite profile was compared with those of traditional bulk hepatocyte analysis. Results: In the bulk hepatocyte analysis, liquid chromatography–MS showed the averaged metabolism of tafluprost to tafluprost acid (TA) and β-oxidized metabolites. However, live single-cell MS showed that tafluprost metabolism varied among individual cells. In addition, there was significant variation in the quantities of TA and a major metabolite, dinor-TA, among cells, whereas there was no significant variation in 7-ethoxycoumarin metabolism. Conclusion: Thus, live single-cell MS successfully detected the heterogeneity of drug metabolism in individual living hepatocytes. Original submitted 12 May 2011; Revised submitted 6 February 2012; Published online 14 May 2012
Collapse
Affiliation(s)
- Yasufumi Fukano
- Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
- Ophthalmic Research & Development Center, Santen Pharmaceutical Co., Ltd, 8916–16 Takayama-cho, Ikoma, Nara 630-0101, Japan
| | - Naohiro Tsuyama
- Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Hajime Mizuno
- Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Sachiko Date
- Quantitative Biology Center, RIKEN, 6-2-3 Furuedai, Suita, Osaka 565-0871, Japan
| | - Mikihisa Takano
- Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Tsutomu Masujima
- Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| |
Collapse
|
39
|
Hypoxia-inducible factors and their roles in energy metabolism. Int J Hematol 2012; 95:457-63. [PMID: 22535382 DOI: 10.1007/s12185-012-1069-y] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 03/29/2012] [Accepted: 03/29/2012] [Indexed: 01/03/2023]
Abstract
Over the course of evolution, aerobic organisms have developed sophisticated systems for responding to alterations in oxygen concentration, as oxygen acts as a final electron acceptor in oxidative phosphorylation for energy production. Hypoxia-inducible factor (HIF) plays a central role in the adaptive regulation of energy metabolism, by triggering a switch from mitochondrial oxidative phosphorylation to anaerobic glycolysis in hypoxic conditions. HIF also reduces oxygen consumption in mitochondria by inhibiting conversion of pyruvate to acetyl CoA, suppressing mitochondrial biogenesis and activating autophagy of mitochondria concomitantly with reduction in reactive oxygen species production. In addition, metabolic reprogramming in response to hypoxia through HIF activation is not limited to the regulation of carbohydrate metabolism; it occurs in lipid metabolism as well. Recent studies using in vivo gene-targeting technique have revealed unexpected, but novel functions of HIF in energy metabolism in a context- and cell type-specific manner, and shed light on the possibility of pharmaceutical targeting HIF as a new therapy against many diseases, including cancer, diabetes, and fatty liver.
Collapse
|
40
|
Orman MA, Mattick J, Androulakis IP, Berthiaume F, Ierapetritou MG. Stoichiometry based steady-state hepatic flux analysis: computational and experimental aspects. Metabolites 2012; 2:268-91. [PMID: 24957379 PMCID: PMC3901202 DOI: 10.3390/metabo2010268] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 03/05/2012] [Accepted: 03/06/2012] [Indexed: 11/16/2022] Open
Abstract
: The liver has many complex physiological functions, including lipid, protein and carbohydrate metabolism, as well as bile and urea production. It detoxifies toxic substances and medicinal products. It also plays a key role in the onset and maintenance of abnormal metabolic patterns associated with various disease states, such as burns, infections and major traumas. Liver cells have been commonly used in in vitro experiments to elucidate the toxic effects of drugs and metabolic changes caused by aberrant metabolic conditions, and to improve the functions of existing systems, such as bioartificial liver. More recently, isolated liver perfusion systems have been increasingly used to characterize intrinsic metabolic changes in the liver caused by various perturbations, including systemic injury, hepatotoxin exposure and warm ischemia. Metabolic engineering tools have been widely applied to these systems to identify metabolic flux distributions using metabolic flux analysis or flux balance analysis and to characterize the topology of the networks using metabolic pathway analysis. In this context, hepatic metabolic models, together with experimental methodologies where hepatocytes or perfused livers are mainly investigated, are described in detail in this review. The challenges and opportunities are also discussed extensively.
Collapse
Affiliation(s)
- Mehmet A Orman
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - John Mattick
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ioannis P Androulakis
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Marianthi G Ierapetritou
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
41
|
Nishiyama Y, Goda N, Kanai M, Niwa D, Osanai K, Yamamoto Y, Senoo-Matsuda N, Johnson RS, Miura S, Kabe Y, Suematsu M. HIF-1α induction suppresses excessive lipid accumulation in alcoholic fatty liver in mice. J Hepatol 2012; 56:441-7. [PMID: 21896344 DOI: 10.1016/j.jhep.2011.07.024] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 07/22/2011] [Accepted: 07/25/2011] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Chronic alcohol intake stimulates hepatic oxygen consumption and subsequently causes liver hypoxia, leading to activation of hypoxia inducible factor-1 (HIF-1). Although HIF-1 plays a crucial role in the metabolic switch from aerobic to anaerobic metabolism in response to hypoxia, its roles in the regulation of lipid metabolism in alcoholic fatty liver remain unknown. METHODS Wild-type and hepatocyte-specific HIF-1α-null mice were subjected to a 6% ethanol-containing liquid diet for 4 weeks, and functional effects of loss of the HIF-1α gene on lipid metabolism were examined in the liver. RESULTS Hepatocyte-specific HIF-1α-null mice developed severe hypertriglyceridemia with enhanced accumulation of lipids in the liver of mice exposed to a 6% ethanol-containing liquid diet for 4 weeks. Sterol regulatory element-binding protein 1c (SREBP-1c) and its downstream target acetyl-CoA carboxylase were greatly activated as the hepatic steatosis progressed, and these alterations were inversely correlated with the expression of the HIF-1-regulated gene DEC1. Overexpression of DEC1 in the mutant liver abrogated the detrimental effects of loss of HIF-1α gene on ethanol-induced fatty liver with reduced SREBP-1c expression. Conversely, co-administration of the HIF hydroxylase inhibitor dimethyloxalylglycine for the last 2 weeks improved markedly the ethanol-induced fatty liver in mice. CONCLUSIONS The current results provide direct evidence for protective roles of HIF-1 induction in the development of ethanol-induced fatty liver via activation of the HIF-1-regulated transcriptional repressor DEC1.
Collapse
Affiliation(s)
- Yasumasa Nishiyama
- Department of Biochemistry, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Amacher DE. The mechanistic basis for the induction of hepatic steatosis by xenobiotics. Expert Opin Drug Metab Toxicol 2011; 7:949-65. [PMID: 21510823 DOI: 10.1517/17425255.2011.577740] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Hepatic steatosis is the histological observation of numerous lipid inclusions due to an excess accumulation of triacylglycerols. They are a concern with new therapeutic candidates because they signify altered lipid metabolism that can progress to more serious liver toxicity. AREAS COVERED This article is based on an article search using the PubMed database from 1987 to 2011 and confirms associations for several previously marketed drugs with four basic hepatocellular mechanisms. The article also describes how these mechanisms are controlled by master regulators of lipid metabolism, which include gene transcription factors, nuclear receptors, hormonal signaling, energy sensing proteins, endoplasmic reticulum stress signaling and certain key metabolic intermediates. EXPERT OPINION Drug-induced hepatic steatosis is typically not detectable by conventional means other than invasive histological examinations. By understanding the basic mechanisms, key regulators and energy signaling systems of the liver, the investigator is better equipped to avoid xenobiotics with steatogenic potential in the drug discovery or early development process. There are now a number of methods for detecting this potential, specifically gene expression or metabolomic profiling and pathway analysis or mechanism-based in vitro systems.
Collapse
|
43
|
Lerapetritou MG, Georgopoulos PG, Roth CM, Androulakis LP. Tissue-level modeling of xenobiotic metabolism in liver: An emerging tool for enabling clinical translational research. Clin Transl Sci 2010; 2:228-37. [PMID: 20443896 DOI: 10.1111/j.1752-8062.2009.00092.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
This review summarizes some of the recent developments and identifies critical challenges associated with in vitro and in silico representations of the liver and assesses the translational potential of these models in the quest of rationalizing the process of evaluating drug efficacy and toxicity. It discusses a wide range of research efforts that have produced, during recent years, quantitative descriptions and conceptual as well as computational models of hepatic processes such as biotransport and biotransformation, intra- and intercellular signal transduction, detoxification, etc. The above mentioned research efforts cover multiple scales of biological organization, from molecule-molecule interactions to reaction network and cellular and histological dynamics, and have resulted in a rapidly evolving knowledge base for a "systems biology of the liver." Virtual organ/organism formulations represent integrative implementations of particular elements of this knowledge base, usually oriented toward the study of specific biological endpoints, and provide frameworks for translating the systems biology concepts into computational tools for quantitative prediction of responses to stressors and hypothesis generation for experimental design.
Collapse
Affiliation(s)
- Marianthi G Lerapetritou
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | | | | | | |
Collapse
|
44
|
Abstract
While virtually absent in our diet a few hundred years ago, fructose has now become a major constituent of our modern diet. Our main sources of fructose are sucrose from beet or cane, high fructose corn syrup, fruits, and honey. Fructose has the same chemical formula as glucose (C(6)H(12)O(6)), but its metabolism differs markedly from that of glucose due to its almost complete hepatic extraction and rapid hepatic conversion into glucose, glycogen, lactate, and fat. Fructose was initially thought to be advisable for patients with diabetes due to its low glycemic index. However, chronically high consumption of fructose in rodents leads to hepatic and extrahepatic insulin resistance, obesity, type 2 diabetes mellitus, and high blood pressure. The evidence is less compelling in humans, but high fructose intake has indeed been shown to cause dyslipidemia and to impair hepatic insulin sensitivity. Hepatic de novo lipogenesis and lipotoxicity, oxidative stress, and hyperuricemia have all been proposed as mechanisms responsible for these adverse metabolic effects of fructose. Although there is compelling evidence that very high fructose intake can have deleterious metabolic effects in humans as in rodents, the role of fructose in the development of the current epidemic of metabolic disorders remains controversial. Epidemiological studies show growing evidence that consumption of sweetened beverages (containing either sucrose or a mixture of glucose and fructose) is associated with a high energy intake, increased body weight, and the occurrence of metabolic and cardiovascular disorders. There is, however, no unequivocal evidence that fructose intake at moderate doses is directly related with adverse metabolic effects. There has also been much concern that consumption of free fructose, as provided in high fructose corn syrup, may cause more adverse effects than consumption of fructose consumed with sucrose. There is, however, no direct evidence for more serious metabolic consequences of high fructose corn syrup versus sucrose consumption.
Collapse
Affiliation(s)
- Luc Tappy
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, CH-1005 Lausanne, Switzerland.
| | | |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Intrahepatic fat content is increasingly being recognized as an integral part of metabolic dysfunction. This article reviews available methods for the assessment of hepatic steatosis. RECENT FINDINGS Apart from liver biopsy, there are several noninvasive radiologic modalities for evaluating nonalcoholic fatty liver disease. Ultrasonography, computed tomography, and traditional MRI remain largely qualitative methods for detecting mild to severe degrees of steatosis rather than quantitative methods for measuring liver fat content, even though novel attempts to collect objective quantitative information have recently been developed. Still, their sensitivity at mild degrees of steatosis is poor. Undoubtedly, most methodological advances have occurred in the field of MRI and magnetic resonance spectroscopy, which currently enable the accurate quantification of intrahepatic fat even at normal or near normal levels. Xenon computed tomography was also recently shown to offer another objective tool for the quantitative assessment of steatosis, although more validation studies are required. SUMMARY Several modalities can be used for measuring intrahepatic fat and assessing steatosis; the choice will ultimately depend on the intended use and available resources.
Collapse
Affiliation(s)
- Elisa Fabbrini
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | |
Collapse
|
46
|
Debois D, Bralet MP, Le Naour F, Brunelle A, Laprévote O. In situ lipidomic analysis of nonalcoholic fatty liver by cluster TOF-SIMS imaging. Anal Chem 2009; 81:2823-31. [PMID: 19296690 DOI: 10.1021/ac900045m] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mass spectrometry imaging has been used to map liver biopsies of several patients suffering from nonalcoholic fatty liver disease. This steatosis is characterized by an accumulation of triacylglycerols and diacylglycerols in the liver. Using time-of-flight-secondary ion mass spectrometry (TOF-SIMS) with a bismuth cluster ion source, it has been possible to map lipids in situ at the micrometer scale and to simultaneously characterize their molecular distribution on liver sections. Accumulation of triacylglycerols, diacylglycerols, monoacylglycerols, fatty acids, with the apparition of myristic acid, together with a dramatic depletion of vitamin E and a selective macrovacuolar localization of cholesterol are observed in steatosis areas of fatty livers compared to control livers. These ion species are concentrated in small vesicles having a size of a few micrometers. Moreover, very fine differences in lipid localizations, depending on alkyl acid chain lengths of diacylglycerols and fatty acids, have been found after careful scrutiny of the ion images. Finally, TOF-SIMS has revealed lipid zonation in the normal human liver and accumulation of very similar lipids to those detected in areas of the fatty livers, which are not characterized as steatotic ones by the histological control performed on serial tissue sections.
Collapse
Affiliation(s)
- Delphine Debois
- Institut de Chimie des Substances Naturelles, CNRS, UPR 2301, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | | | | | | | | |
Collapse
|
47
|
Han C, Wang J, Li L, Wang L, Zhang Z. The role of LXRα in goose primary hepatocyte lipogenesis. Mol Cell Biochem 2008; 322:37-42. [DOI: 10.1007/s11010-008-9937-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 10/13/2008] [Indexed: 11/25/2022]
|
48
|
Tavares FL, Seelaender MCL. Hepatic denervation impairs the assembly and secretion of VLDL-TAG. Cell Biochem Funct 2008; 26:557-65. [PMID: 18543355 DOI: 10.1002/cbf.1476] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
VLDL secretion is a regulated process that depends on the availability of lipids, apoB and MTP. Our aim was to investigate the effect of liver denervation upon the secretion of VLDL and the expression of proteins involved in this process. Denervation was achieved by applying a 85% phenol solution onto the portal tract, while control animals were treated with 9% NaCl. VLDL secretion was evaluated by the Tyloxapol method. The hepatic concentration of TAG and cholesterol, and the plasma concentration of TAG, cholesterol, VLDL-TAG, VLDL-cholesterol and HDL-cholesterol were measured, as well as mRNA expression of proteins involved in the process of VLDL assembly. Hepatic acinar distribution of MTP and apoB was evaluated by immunohistochemistry. Denervation increased plasma concentration of cholesterol (125.3 +/- 10.1 vs. 67.1 +/- 4.9 mg dL(-1)) and VLDL-cholesterol (61.6 +/- 5.6 vs. 29.4 +/- 3.3 mg dL(-1)), but HDL-cholesterol was unchanged (45.5 +/- 6.1 vs. 36.9 +/- 3.9 mg dL(-1)). Secretion of VLDL-TAG (47.5 +/- 23.8 vs. 148.5 +/- 27.4 mg dL h(-1)) and mRNA expression of CPT I and apoB were reduced (p < 0.01) in the denervated animals. MTP and apoB acinar distribution was not altered in the denervated animals, but the intensity of the reaction was reduced in relation to controls.
Collapse
Affiliation(s)
- Fábio Luís Tavares
- Molecular Biology of the Cell Group, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, Brazil
| | | |
Collapse
|
49
|
Postic C, Girard J. Contribution of de novo fatty acid synthesis to hepatic steatosis and insulin resistance: lessons from genetically engineered mice. J Clin Invest 2008; 118:829-38. [PMID: 18317565 DOI: 10.1172/jci34275] [Citation(s) in RCA: 904] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is associated with obesity, insulin resistance, and type 2 diabetes. NAFLD represents a large spectrum of diseases ranging from (i) fatty liver (hepatic steatosis); (ii) steatosis with inflammation and necrosis; and (iii) cirrhosis. Although the molecular mechanism leading to the development of hepatic steatosis in the pathogenesis of NAFLD is complex, recent animal models have shown that modulating important enzymes in fatty acid synthesis in liver may be key for the treatment of NAFLD. This review discusses recent advances in the field.
Collapse
Affiliation(s)
- Catherine Postic
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France.
| | | |
Collapse
|
50
|
García-Villafranca J, Guillén A, Castro J. Ethanol consumption impairs regulation of fatty acid metabolism by decreasing the activity of AMP-activated protein kinase in rat liver. Biochimie 2007; 90:460-6. [PMID: 17997005 DOI: 10.1016/j.biochi.2007.09.019] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Accepted: 09/28/2007] [Indexed: 02/06/2023]
Abstract
The mechanisms by which ethanol consumption causes accumulation of hepatic triacylglycerols are complex. AMP-activated protein kinase (AMPK) plays a central role in the regulation of lipid metabolism. Therefore, in the present study we investigated whether AMPK may have a role in the development of ethanol-induced fatty liver. Hepatocytes isolated from rats fed with an ethanol-containing liquid diet showed higher rates of fatty acid and triacylglycerol syntheses, but a decreased rate of fatty acid oxidation, concomitant to a lower activity of carnitine palmitoyltransferase I. Hepatocytes from both ethanol-fed and pair-fed control rats were incubated with 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), an AMPK activator in intact cells. In both hepatocyte preparations AICAR strongly inhibited the activity of acetyl-CoA carboxylase in parallel to fatty acid synthesis, but cells from ethanol-fed rats showed significantly lower sensitivity to inhibition by AICAR. Moreover, AICAR strongly decreased triacylglycerol synthesis and increased fatty acid oxidation in control hepatocytes, but these effects were markedly attenuated in hepatocytes from ethanol-fed rats. In parallel, AMPK in liver of ethanol-fed rats showed a decreased specific activity and a lower sensitivity to changes in the AMP/ATP ratio, compared to the enzyme of control rats. These effects are consistent with the impairment of AMPK-mediated regulation of fatty acid metabolism after ethanol consumption, that will facilitate triacylglycerol accumulation. Taken together, these findings suggest that a decreased AMPK activity may have an important role in the development of alcoholic fatty liver.
Collapse
Affiliation(s)
- Javier García-Villafranca
- Departamento de Bioquímica y Biología Molecular I, Facultad de Biología, Universidad Complutense, Ciudad Universitaria, s/n, Madrid, Spain
| | | | | |
Collapse
|