51
|
Clayton ZS, Hutton DA, Mahoney SA, Seals DR. Anthracycline chemotherapy-mediated vascular dysfunction as a model of accelerated vascular aging. ACTA ACUST UNITED AC 2021; 2:45-69. [PMID: 34212156 DOI: 10.1002/aac2.12033] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide, and age is by far the greatest risk factor for developing CVD. Vascular dysfunction, including endothelial dysfunction and arterial stiffening, is responsible for much of the increase in CVD risk with aging. A key mechanism involved in vascular dysfunction with aging is oxidative stress, which reduces the bioavailability of nitric oxide (NO) and induces adverse changes to the extracellular matrix of the arterial wall (e.g., elastin fragmentation/degradation, collagen deposition) and an increase in advanced glycation end products, which form crosslinks in arterial wall structural proteins. Although vascular dysfunction and CVD are most prevalent in older adults, several conditions can "accelerate" these events at any age. One such factor is chemotherapy with anthracyclines, such as doxorubicin (DOXO), to combat common forms of cancer. Children, adolescents and young adults treated with these chemotherapeutic agents demonstrate impaired vascular function and an increased risk of future CVD development compared with healthy age-matched controls. Anthracycline treatment also worsens vascular dysfunction in mid-life (50-64 years of age) and older (65 and older) adults such that endothelial dysfunction and arterial stiffness are greater compared to age-matched controls. Collectively, these observations indicate that use of anthracycline chemotherapeutic agents induce a vascular aging-like phenotype and that the latter contributes to premature CVD in cancer survivors exposed to these agents. Here, we review the existing literature supporting these ideas, discuss potential mechanisms as well as interventions that may protect arteries from these adverse effects, identify research gaps and make recommendations for future research.
Collapse
|
52
|
Saadat M, Mostafaei F, Mahdinloo S, Abdi M, Zahednezhad F, Zakeri-Milani P, Valizadeh H. Drug delivery of pH-Sensitive nanoparticles into the liver cancer cells. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102557] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
53
|
Ellias SD, Larson EL, Taner T, Nyberg SL. Cell-Mediated Therapies to Facilitate Operational Tolerance in Liver Transplantation. Int J Mol Sci 2021; 22:ijms22084016. [PMID: 33924646 PMCID: PMC8069094 DOI: 10.3390/ijms22084016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
Cell therapies using immune cells or non-parenchymal cells of the liver have emerged as potential treatments to facilitate immunosuppression withdrawal and to induce operational tolerance in liver transplant (LT) recipients. Recent pre-clinical and clinical trials of cellular therapies including regulatory T cells, regulatory dendritic cells, and mesenchymal cells have shown promising results. Here we briefly summarize current concepts of cellular therapy for induction of operational tolerance in LT recipients.
Collapse
Affiliation(s)
- Samia D. Ellias
- Division of Transplant Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA; (S.D.E.); (E.L.L.); (T.T.)
| | - Ellen L. Larson
- Division of Transplant Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA; (S.D.E.); (E.L.L.); (T.T.)
| | - Timucin Taner
- Division of Transplant Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA; (S.D.E.); (E.L.L.); (T.T.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Scott L. Nyberg
- Division of Transplant Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA; (S.D.E.); (E.L.L.); (T.T.)
- Correspondence: ; Tel.: +1-507-266-6772; Fax: +1-507-266-2810
| |
Collapse
|
54
|
Tsai MY, Yang WC, Lin CF, Wang CM, Liu HY, Lin CS, Lin JW, Lin WL, Lin TC, Fan PS, Hung KH, Lu YW, Chang GR. The Ameliorative Effects of Fucoidan in Thioacetaide-Induced Liver Injury in Mice. Molecules 2021; 26:molecules26071937. [PMID: 33808318 PMCID: PMC8036993 DOI: 10.3390/molecules26071937] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023] Open
Abstract
Liver disorders have been recognized as one major health concern. Fucoidan, a sulfated polysaccharide extracted from the brown seaweed Fucus serratus, has previously been reported as an anti-inflammatory and antioxidant. However, the discovery and validation of its hepatoprotective properties and elucidation of its mechanisms of action are still unknown. The objective of the current study was to investigate the effect and possible modes of action of a treatment of fucoidan against thioacetamide (TAA)-induced liver injury in male C57BL/6 mice by serum biochemical and histological analyses. The mouse model for liver damage was developed by the administration of TAA thrice a week for six weeks. The mice with TAA-induced liver injury were orally administered fucoidan once a day for 42 days. The treated mice showed significantly higher body weights; food intakes; hepatic antioxidative enzymes (catalase, glutathione peroxidase (GPx), and superoxide dismutase (SOD)); and a lower serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and C-reactive protein (CRP) levels. Additionally, a reduced hepatic IL-6 level and a decreased expression of inflammatory-related genes, such as cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) mRNA was observed. These results demonstrated that fucoidan had a hepatoprotective effect on liver injury through the suppression of the inflammatory responses and acting as an antioxidant. In addition, here, we validated the use of fucoidan against liver disorders with supporting molecular data.
Collapse
Affiliation(s)
- Ming-Yang Tsai
- Animal Industry Division, Livestock Research Institute, Council of Agriculture, Executive Yuan, 112 Muchang, Xinhua Dist, Tainan 71246, Taiwan;
- Graduate Institute of Bioresources, National Pingtung University of Science and Technology, 1 Shuefu Road, Neipu, Pingtung 91201, Taiwan
| | - Wei-Cheng Yang
- School of Veterinary Medicine, National Taiwan University, 4 Section, 1 Roosevelt Road, Taipei 10617, Taiwan; (W.-C.Y.); (C.-S.L.)
| | - Chuen-Fu Lin
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Shuefu Road, Neipu, Pingtung 912301, Taiwan;
| | - Chao-Min Wang
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.); (P.-S.F.)
| | - Hsien-Yueh Liu
- Bachelor Degree Program in Animal Healthcare, Hungkuang University, 6 Section, 1018 Taiwan Boulevard, Shalu District, Taichung 433304, Taiwan; (H.-Y.L.); (J.-W.L.); (W.-L.L.)
| | - Chen-Si Lin
- School of Veterinary Medicine, National Taiwan University, 4 Section, 1 Roosevelt Road, Taipei 10617, Taiwan; (W.-C.Y.); (C.-S.L.)
| | - Jen-Wei Lin
- Bachelor Degree Program in Animal Healthcare, Hungkuang University, 6 Section, 1018 Taiwan Boulevard, Shalu District, Taichung 433304, Taiwan; (H.-Y.L.); (J.-W.L.); (W.-L.L.)
| | - Wei-Li Lin
- Bachelor Degree Program in Animal Healthcare, Hungkuang University, 6 Section, 1018 Taiwan Boulevard, Shalu District, Taichung 433304, Taiwan; (H.-Y.L.); (J.-W.L.); (W.-L.L.)
- General Education Center, Chaoyang University of Technology, 168 Jifeng Eastern Road, Taichung 413310, Taiwan
| | - Tzu-Chun Lin
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.); (P.-S.F.)
| | - Pei-Shan Fan
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.); (P.-S.F.)
| | - Kuo-Hsiang Hung
- Graduate Institute of Bioresources, National Pingtung University of Science and Technology, 1 Shuefu Road, Neipu, Pingtung 91201, Taiwan
- Correspondence: (K.-H.H.); (Y.-W.L.); (G.-R.C.)
| | - Yu-Wen Lu
- Department of Chinese Medicine, Show Chwan Memorial Hospital, 1 Section, 542 Chung-Shan Road, Changhua 50008, Taiwan
- Department of Chinese Medicine, Chang Bing Show Chwan Memorial Hospital, 6 Lugong Road, Changhua 50544, Taiwan
- Correspondence: (K.-H.H.); (Y.-W.L.); (G.-R.C.)
| | - Geng-Ruei Chang
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.); (P.-S.F.)
- Correspondence: (K.-H.H.); (Y.-W.L.); (G.-R.C.)
| |
Collapse
|
55
|
Freag MS, Namgung B, Reyna Fernandez ME, Gherardi E, Sengupta S, Jang HL. Human Nonalcoholic Steatohepatitis on a Chip. Hepatol Commun 2021; 5:217-233. [PMID: 33553970 PMCID: PMC7850303 DOI: 10.1002/hep4.1647] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/04/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH), an advanced stage of nonalcoholic fatty liver disease (NAFLD), is a rapidly growing and global health problem compounded by the current absence of specific treatments. A major limiting factor in the development of new NASH therapies is the absence of models that capture the unique cellular structure of the liver microenvironment and recapitulate the complexities of NAFLD progression to NASH. Organ-on-a-chip platforms have emerged as a powerful approach to dynamically model diseases and test drugs. Herein, we describe a NASH-on-a-chip platform. Four main types of human primary liver cells (hepatocytes [HCs], Kupffer cells, liver sinusoidal endothelial cells, and hepatic stellate cells [HSCs]) were cocultured under microfluidic dynamics. Our chip-based model successfully recapitulated a functional liver cellular microenvironment with stable albumin and urea secretion for at least 2 weeks. Exposing liver chips to a lipotoxic environment led to gradual development of NASH phenotypic characteristics, including intracellular lipid accumulation, hepatocellular ballooning, HSC activation, and elevation of inflammatory and profibrotic markers. Further, exposure of the chip to elafibranor, a drug under study for the therapy of NASH, inhibited the development of NASH-specific hallmarks, causing an ~8-fold decrease in intracellular lipids, a 3-fold reduction in number of ballooned HCs, a significant reduction in HSC activation, and a significant decrease in the levels of inflammatory and profibrotic markers compared with controls. Conclusion: We have successfully developed a microfluidic NASH-on-a-chip platform that recapitulates the main NASH histologic endpoints in a single chip and that can emerge as a powerful noninvasive, human-relevant, in vitro platform to study disease pathogenesis and develop novel anti-NASH drugs.
Collapse
Affiliation(s)
- May S Freag
- Center for Engineered TherapeuticsDivision of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA.,Division of Health Sciences and TechnologyHarvard-Massachusetts Institute of TechnologyMassachusetts Institute of TechnologyBostonMAUSA
| | - Bumseok Namgung
- Center for Engineered TherapeuticsDivision of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA.,Division of Health Sciences and TechnologyHarvard-Massachusetts Institute of TechnologyMassachusetts Institute of TechnologyBostonMAUSA
| | - Maria E Reyna Fernandez
- Center for Engineered TherapeuticsDivision of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA.,Division of Health Sciences and TechnologyHarvard-Massachusetts Institute of TechnologyMassachusetts Institute of TechnologyBostonMAUSA
| | - Ermanno Gherardi
- Unit of Immunology and General PathologyDepartment of Molecular MedicineUniversity of PaviaPaviaItaly
| | - Shiladitya Sengupta
- Center for Engineered TherapeuticsDivision of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA.,Division of Health Sciences and TechnologyHarvard-Massachusetts Institute of TechnologyMassachusetts Institute of TechnologyBostonMAUSA.,Dana Farber Cancer InstituteBostonMAUSA
| | - Hae Lin Jang
- Center for Engineered TherapeuticsDivision of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
56
|
|
57
|
Aravalli RN. Generating liver using blastocyst complementation: Opportunities and challenges. Xenotransplantation 2020; 28:e12668. [PMID: 33372360 DOI: 10.1111/xen.12668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/05/2020] [Accepted: 11/26/2020] [Indexed: 12/28/2022]
Abstract
Orthotopic liver transplantation (OLT) is the only definitive treatment option for many patients with end-stage liver disease. Current supply of donor livers for OLT is not keeping up with the growing demand. To overcome this problem, a number of experimental strategies have been developed either to provide a bridge to transplant for patients on the waiting list or to bioengineer whole livers for OLT by replenishing them with fresh supplies of hepatic cells. In recent years, blastocyst complementation has emerged as the most promising approach for generating whole organs and, in combination with gene editing technology, it has revolutionized regenerative medicine. This methodology was successful in producing xenogeneic organs in animal hosts. Blastocyst complementation has the potential to produce whole livers in large animals that could be xenotransplanted in humans, thereby reducing the shortage of livers for OLT. However, significant experimental and ethical barriers remain for the production of human livers in domestic animals, such as the pig. This review summarizes the current knowledge and provides future perspectives for liver xenotransplantation in humans.
Collapse
Affiliation(s)
- Rajagopal N Aravalli
- Department of Electrical and Computer Engineering, College of Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
58
|
Schmitt MV, Reichel A, Liu X, Fricker G, Lienau P. Extension of the Mechanistic Tissue Distribution Model of Rodgers and Rowland by Systematic Incorporation of Lysosomal Trapping: Impact on Unbound Partition Coefficient and Volume of Distribution Predictions in the Rat. Drug Metab Dispos 2020; 49:53-61. [PMID: 33148688 DOI: 10.1124/dmd.120.000161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/28/2020] [Indexed: 11/22/2022] Open
Abstract
Physiologically based pharmacokinetic modeling has become a standard tool to predict drug distribution in early stages of drug discovery; however, this does not currently encompass lysosomal trapping. For basic lipophilic compounds, lysosomal sequestration is known to potentially influence intracellular as well as tissue distribution. The aim of our research was to reliably predict the lysosomal drug content and ultimately integrate this mechanism into pharmacokinetic prediction models. First, we further validated our previously presented method to predict the lysosomal drug content (Schmitt et al., 2019) for a larger set of compounds (n = 41) showing a very good predictivity. Using the lysosomal marker lipid bis(monoacylglycero)phosphate, we estimated the lysosomal volume fraction for all major tissues in the rat, ranging from 0.03% for adipose up to 5.3% for spleen. The pH-driven lysosomal trapping was then estimated and fully integrated into the mechanistic distribution model published by Rodgers et al. (2005) Predictions of Kpu improved for all lysosome-rich tissues. For instance, Kpu increased for nicotine 4-fold (spleen) and 2-fold (lung and kidney) and for quinidine 1.8-fold (brain), although for most other drugs the effects were much less (≤7%). Overall, the effect was strongest for basic compounds with a lower lipophilicity, such as nicotine, for which the unbound volume of distribution at steady-state prediction changed from 1.34 to 1.58 l/kg. For more lipophilic (basic) compounds or those that already show strong interactions with acidic phospholipids, the additional contribution of lysosomal trapping was less pronounced. Nevertheless, lysosomal trapping will also affect intracellular distribution of such compounds. SIGNIFICANCE STATEMENT: The estimation of the lysosomal content in all body tissues facilitated the incorporation of lysosomal sequestration into a general physiologically based pharmacokinetic model, leading to improved predictions as well as elucidating its influence on tissue and subcellular distribution in the rat.
Collapse
Affiliation(s)
- Maximilian V Schmitt
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Andreas Reichel
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Xiaohui Liu
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Gert Fricker
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Philip Lienau
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., A.R., P.L.); School of Life Sciences, Tsinghua University, Beijing, China (X.L.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| |
Collapse
|
59
|
Mak KM, Shin DW. Hepatic sinusoids versus central veins: Structures, markers, angiocrines, and roles in liver regeneration and homeostasis. Anat Rec (Hoboken) 2020; 304:1661-1691. [PMID: 33135318 DOI: 10.1002/ar.24560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/14/2020] [Accepted: 10/22/2020] [Indexed: 01/20/2023]
Abstract
The blood circulates through the hepatic sinusoids delivering nutrients and oxygen to the liver parenchyma and drains into the hepatic central vein, yet the structures and phenotypes of these vessels are distinctively different. Sinusoidal endothelial cells are uniquely fenestrated, lack basal lamina and possess organelles involved in endocytosis, pinocytosis, degradation, synthesis and secretion. Hepatic central veins are nonfenestrated but are also active in synthesis and secretion. Endothelial cells of sinusoids and central veins secrete angiocrines that play respective roles in hepatic regeneration and metabolic homeostasis. The list of markers for identifying sinusoidal endothelial cells is long and their terminologies are complex. Further, their uses vary in different investigations and, in some instances, could be confusing. Central vein markers are fewer but more distinctive. Here we analyze and categorize the molecular pathways/modules associated with the sinusoid-mediated liver regeneration in response to partial hepatectomy and chemical-induced acute or chronic injury. Similarly, we highlight the findings that central vein-derived angiocrines interact with Wnt/β-catenin in perivenous hepatocytes to direct gene expression and maintain pericentral metabolic zonation. The proposal that perivenous hepatocytes behave as stem/progenitor cells to provoke hepatic homeostatic cell renewal is reevaluated and newer concepts of broad zonal distribution of hepatocyte proliferation in liver homeostasis and regeneration are updated. Thus, this review integrates the structures, biology and physiology of liver sinusoids and central veins in mediating hepatic regeneration and metabolic homeostasis.
Collapse
Affiliation(s)
- Ki M Mak
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Da Wi Shin
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
60
|
Playing Jekyll and Hyde-The Dual Role of Lipids in Fatty Liver Disease. Cells 2020; 9:cells9102244. [PMID: 33036257 PMCID: PMC7601321 DOI: 10.3390/cells9102244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/27/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Lipids play Jekyll and Hyde in the liver. On the one hand, the lipid-laden status of hepatic stellate cells is a hallmark of healthy liver. On the other hand, the opposite is true for lipid-laden hepatocytes—they obstruct liver function. Neglected lipid accumulation in hepatocytes can progress into hepatic fibrosis, a condition induced by the activation of stellate cells. In their resting state, these cells store substantial quantities of fat-soluble vitamin A (retinyl esters) in large lipid droplets. During activation, these lipid organelles are gradually degraded. Hence, treatment of fatty liver disease is treading a tightrope—unsophisticated targeting of hepatic lipid accumulation might trigger problematic side effects on stellate cells. Therefore, it is of great importance to gain more insight into the highly dynamic lipid metabolism of hepatocytes and stellate cells in both quiescent and activated states. In this review, part of the special issue entitled “Cellular and Molecular Mechanisms underlying the Pathogenesis of Hepatic Fibrosis 2020”, we discuss current and highly versatile aspects of neutral lipid metabolism in the pathogenesis of non-alcoholic fatty liver disease (NAFLD).
Collapse
|
61
|
Gu S, Yan M, Wang C, Meng X, Xiang Z, Qiu Y, Han X. Microcystin-leucine-arginine induces liver fibrosis by activating the Hedgehog pathway in hepatic stellate cells. Biochem Biophys Res Commun 2020; 533:770-778. [PMID: 32988585 DOI: 10.1016/j.bbrc.2020.09.075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/20/2020] [Indexed: 12/14/2022]
Abstract
Microcystin-leucine-arginine (MC-LR), produced by cyanobacteria, accumulates in the liver through blood circulation. We investigated the impact of MC-LR on liver fibrosis. Mice received a daily injection of MC-LR at various concentrations for 14 consecutive days aa and then mouse liver was obtained for histopathological and immunoblot analysis. Next, a human hepatic stellate cell line (LX-2) was treated with MC-LR at various concentrations followed by measurement of cell viability, cell cycle and relevant protein expression levels. Our data confirmed the induction of mouse liver fibrosis after exposure to MC-LR at 15 μg/kg and 30 μg/kg. Furthermore, we demonstrated that LX-2 cells could uptake MC-LR, resulting in cell proliferation and differentiation through impacting the Hedgehog signaling after the treatment of MC-LR at 50 nM. Our data supported that MC-LR could induce liver fibrosis by modulating the expression of the transcription factor Gli2 in the Hedgehog signaling in hepatic stellate cells.
Collapse
Affiliation(s)
- Shen Gu
- Department of Hepatopancreatobiliary Surgery, Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, 210008, Jiangsu Province, China; Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China.
| | - Minghao Yan
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China.
| | - Cong Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| | - Xiannan Meng
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China.
| | - Zou Xiang
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Research Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.
| | - Yudong Qiu
- Department of Hepatopancreatobiliary Surgery, Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, 210008, Jiangsu Province, China.
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China.
| |
Collapse
|
62
|
Azparren-Angulo M, Royo F, Gonzalez E, Liebana M, Brotons B, Berganza J, Goñi-de-Cerio F, Manicardi N, Abad-Jordà L, Gracia-Sancho J, Falcon-Perez JM. Extracellular vesicles in hepatology: Physiological role, involvement in pathogenesis, and therapeutic opportunities. Pharmacol Ther 2020; 218:107683. [PMID: 32961265 DOI: 10.1016/j.pharmthera.2020.107683] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Since the first descriptions of hepatocyte-released exosome-like vesicles in 2008, the number of publications describing Extracellular Vesicles (EVs) released by liver cells in the context of hepatic physiology and pathology has grown exponentially. This growing interest highlights both the importance that cell-to-cell communication has in the organization of multicellular organisms from a physiological point of view, as well as the opportunity that these circulating organelles offer in diagnostics and therapeutics. In the present review, we summarize systematically and comprehensively the myriad of works that appeared in the last decade and lighted the discussion about the best opportunities for using EVs in liver disease therapeutics.
Collapse
Affiliation(s)
- Maria Azparren-Angulo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Felix Royo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain; Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Esperanza Gonzalez
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Marc Liebana
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Bruno Brotons
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Jesús Berganza
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Parque Tecnológico, Edificio 202, 48170 Zamudio, Bizkaia, Spain
| | - Felipe Goñi-de-Cerio
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Parque Tecnológico, Edificio 202, 48170 Zamudio, Bizkaia, Spain
| | - Nicoló Manicardi
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Laia Abad-Jordà
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain; Hepatology, Department of Biomedical Research, Inselspital & University of Bern, Switzerland
| | - Juan M Falcon-Perez
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain; Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid 28029, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia 48015, Spain.
| |
Collapse
|
63
|
Wilkinson AL, Qurashi M, Shetty S. The Role of Sinusoidal Endothelial Cells in the Axis of Inflammation and Cancer Within the Liver. Front Physiol 2020; 11:990. [PMID: 32982772 PMCID: PMC7485256 DOI: 10.3389/fphys.2020.00990] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSEC) form a unique barrier between the liver sinusoids and the underlying parenchyma, and thus play a crucial role in maintaining metabolic and immune homeostasis, as well as actively contributing to disease pathophysiology. Whilst their endocytic and scavenging function is integral for nutrient exchange and clearance of waste products, their capillarisation and dysfunction precedes fibrogenesis. Furthermore, their ability to promote immune tolerance and recruit distinct immunosuppressive leukocyte subsets can allow persistence of chronic viral infections and facilitate tumour development. In this review, we present the immunological and barrier functions of LSEC, along with their role in orchestrating fibrotic processes which precede tumourigenesis. We also summarise the role of LSEC in modulating the tumour microenvironment, and promoting development of a pre-metastatic niche, which can drive formation of secondary liver tumours. Finally, we summarise closely inter-linked disease pathways which collectively perpetuate pathogenesis, highlighting LSEC as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Shishir Shetty
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
64
|
Pandey E, Nour AS, Harris EN. Prominent Receptors of Liver Sinusoidal Endothelial Cells in Liver Homeostasis and Disease. Front Physiol 2020; 11:873. [PMID: 32848838 PMCID: PMC7396565 DOI: 10.3389/fphys.2020.00873] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) are the most abundant non-parenchymal cells lining the sinusoidal capillaries of the hepatic system. LSECs are characterized with numerous fenestrae and lack basement membrane as well as a diaphragm. These unique morphological characteristics of LSECs makes them the most permeable endothelial cells of the mammalian vasculature and aid in regulating flow of macromolecules and small lipid-based structures between sinusoidal blood and parenchymal cells. LSECs have a very high endocytic capacity aided by scavenger receptors (SR), such as SR-A, SR-B (SR-B1 and CD-36), SR-E (Lox-1 and mannose receptors), and SR-H (Stabilins). Other high-affinity receptors for mediating endocytosis include the FcγRIIb, which assist in the antibody-mediated removal of immune complexes. Complemented with intense lysosomal activity, LSECs play a vital role in the uptake and degradation of many blood borne waste macromolecules and small (<280 nm) colloids. Currently, seven Toll-like receptors have been investigated in LSECs, which are involved in the recognition and clearance of pathogen-associated molecular pattern (PAMPs) as well as damage associated molecular pattern (DAMP). Along with other SRs, LSECs play an essential role in maintaining lipid homeostasis with the low-density lipoprotein receptor-related protein-1 (LRP-1), in juxtaposition with hepatocytes. LSECs co-express two surface lectins called L-Specific Intercellular adhesion molecule-3 Grabbing Non-integrin Receptor (L-SIGN) and liver sinusoidal endothelial cell lectin (LSECtin). LSECs also express several adhesion molecules which are involved in the recruitment of leukocytes at the site of inflammation. Here, we review these cell surface receptors as well as other components expressed by LSECs and their functions in the maintenance of liver homeostasis. We further discuss receptor expression and activity and dysregulation associated with the initiation and progression of many liver diseases, such as hepatocellular carcinoma, liver fibrosis, and cirrhosis, alcoholic and non-alcoholic fatty liver diseases and pseudocapillarization with aging.
Collapse
Affiliation(s)
- Ekta Pandey
- Department of Biochemistry, Universityof Nebraska, Lincoln, NE, United States
| | - Aiah S Nour
- Department of Biochemistry, Universityof Nebraska, Lincoln, NE, United States
| | - Edward N Harris
- Department of Biochemistry, Universityof Nebraska, Lincoln, NE, United States
| |
Collapse
|
65
|
Targeting mitochondrial fitness as a strategy for healthy vascular aging. Clin Sci (Lond) 2020; 134:1491-1519. [PMID: 32584404 DOI: 10.1042/cs20190559] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide and aging is the primary risk factor for CVD. The development of vascular dysfunction, including endothelial dysfunction and stiffening of the large elastic arteries (i.e., the aorta and carotid arteries), contribute importantly to the age-related increase in CVD risk. Vascular aging is driven in large part by oxidative stress, which reduces bioavailability of nitric oxide and promotes alterations in the extracellular matrix. A key upstream driver of vascular oxidative stress is age-associated mitochondrial dysfunction. This review will focus on vascular mitochondria, mitochondrial dysregulation and mitochondrial reactive oxygen species (ROS) production and discuss current evidence for prevention and treatment of vascular aging via lifestyle and pharmacological strategies that improve mitochondrial health. We will also identify promising areas and important considerations ('research gaps') for future investigation.
Collapse
|
66
|
Sun X, Harris EN. New aspects of hepatic endothelial cells in physiology and nonalcoholic fatty liver disease. Am J Physiol Cell Physiol 2020; 318:C1200-C1213. [PMID: 32374676 DOI: 10.1152/ajpcell.00062.2020] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The liver is the central metabolic hub for carbohydrate, lipid, and protein metabolism. It is composed of four major types of cells, including hepatocytes, endothelial cells (ECs), Kupffer cells, and stellate cells. Hepatic ECs are highly heterogeneous in both mice and humans, representing the second largest population of cells in liver. The majority of them line hepatic sinusoids known as liver sinusoidal ECs (LSECs). The structure and biology of LSECs and their roles in physiology and liver disease were reviewed recently. Here, we do not give a comprehensive review of LSEC structure, function, or pathophysiology. Instead, we focus on the recent progress in LSEC research and other hepatic ECs in physiology and nonalcoholic fatty liver disease and other hepatic fibrosis-related conditions. We discuss several current areas of interest, including capillarization, scavenger function, autophagy, cellular senescence, paracrine effects, and mechanotransduction. In addition, we summarize the strengths and weaknesses of evidence for the potential role of endothelial-to-mesenchymal transition in liver fibrosis.
Collapse
Affiliation(s)
- Xinghui Sun
- Department of Biochemistry, University of Nebraska-Lincoln, Beadle Center, Lincoln, Nebraska.,Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska.,Nebraska Center for the Prevention of Obesity Diseases through Dietary Molecules, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Edward N Harris
- Department of Biochemistry, University of Nebraska-Lincoln, Beadle Center, Lincoln, Nebraska.,Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska.,Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
67
|
Sugahara G, Ishida Y, Sun J, Tateno C, Saito T. Art of Making Artificial Liver: Depicting Human Liver Biology and Diseases in Mice. Semin Liver Dis 2020; 40:189-212. [PMID: 32074631 PMCID: PMC8629128 DOI: 10.1055/s-0040-1701444] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Advancement in both bioengineering and cell biology of the liver led to the establishment of the first-generation humanized liver chimeric mouse (HLCM) model in 2001. The HLCM system was initially developed to satisfy the necessity for a convenient and physiologically representative small animal model for studies of hepatitis B virus and hepatitis C virus infection. Over the last two decades, the HLCM system has substantially evolved in quality, production capacity, and utility, thereby growing its versatility beyond the study of viral hepatitis. Hence, it has been increasingly employed for a variety of applications including, but not limited to, the investigation of drug metabolism and pharmacokinetics and stem cell biology. To date, more than a dozen distinctive HLCM systems have been established, and each model system has similarities as well as unique characteristics, which are often perplexing for end-users. Thus, this review aims to summarize the history, evolution, advantages, and pitfalls of each model system with the goal of providing comprehensive information that is necessary for researchers to implement the ideal HLCM system for their purposes. Furthermore, this review article summarizes the contribution of HLCM and its derivatives to our mechanistic understanding of various human liver diseases, its potential for novel applications, and its current limitations.
Collapse
Affiliation(s)
- Go Sugahara
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California,Research & Development Department, PhoenixBio, Co., Ltd, Higashi-Hiroshima, Hiroshima, Japan
| | - Yuji Ishida
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California,Research & Development Department, PhoenixBio, Co., Ltd, Higashi-Hiroshima, Hiroshima, Japan
| | - Jeffrey Sun
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Chise Tateno
- Research & Development Department, PhoenixBio, Co., Ltd, Higashi-Hiroshima, Hiroshima, Japan
| | - Takeshi Saito
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California,USC Research Center for Liver Diseases, Los Angeles, California
| |
Collapse
|
68
|
Effect of Open-Ended Coaxial Probe-to-Tissue Contact Pressure on Dielectric Measurements. SENSORS 2020; 20:s20072060. [PMID: 32268598 PMCID: PMC7181017 DOI: 10.3390/s20072060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/21/2022]
Abstract
Open-ended coaxial probes are widely used to gather dielectric properties of biological tissues. Due to the lack of an agreed data acquisition protocol, several environmental conditions can cause inaccuracies when comparing dielectric data. In this work, the effect of a different measurement probe-to-tissue contact pressure was monitored in the frequency range from 0.5 to 20 GHz. Therefore, we constructed a controlled lifting platform with an integrated pressure sensor to exert a constant pressure on the tissue sample during the dielectric measurement. In the pressure range from 7.74 kPa to 77.4 kPa, we observed a linear correlation of - 0 . 31 ± 0 . 09 % and - 0 . 32 ± 0 . 14 % per kPa for, respectively, the relative real and imaginary complex permittivity. These values are statistically significant compared with the reported measurement uncertainty. Following the literature in different biology-related disciplines regarding pressure-induced variability in measurements, we hypothesize that these changes originate from squeezing out the interstitial and extracellular fluid. This process locally increases the concentration of membranes, cellular organelles, and proteins in the sensed volume. Finally, we suggest moving towards a standardized probe-to-tissue contact pressure, since the literature has already demonstrated that reprobing at the same pressure can produce repeatable data within a 1% uncertainty interval.
Collapse
|
69
|
Recent Advances in Practical Methods for Liver Cell Biology: A Short Overview. Int J Mol Sci 2020; 21:ijms21062027. [PMID: 32188134 PMCID: PMC7139397 DOI: 10.3390/ijms21062027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022] Open
Abstract
Molecular and cellular research modalities for the study of liver pathologies have been tremendously improved over the recent decades. Advanced technologies offer novel opportunities to establish cell isolation techniques with excellent purity, paving the path for 2D and 3D microscopy and high-throughput assays (e.g., bulk or single-cell RNA sequencing). The use of stem cell and organoid research will help to decipher the pathophysiology of liver diseases and the interaction between various parenchymal and non-parenchymal liver cells. Furthermore, sophisticated animal models of liver disease allow for the in vivo assessment of fibrogenesis, portal hypertension and hepatocellular carcinoma (HCC) and for the preclinical testing of therapeutic strategies. The purpose of this review is to portray in detail novel in vitro and in vivo methods for the study of liver cell biology that had been presented at the workshop of the 8th meeting of the European Club for Liver Cell Biology (ECLCB-8) in October of 2018 in Bonn, Germany.
Collapse
|
70
|
Ahn SY, Maeng YS, Kim YR, Choe YH, Hwang HS, Hyun YM. In vivo monitoring of dynamic interaction between neutrophil and human umbilical cord blood-derived mesenchymal stem cell in mouse liver during sepsis. Stem Cell Res Ther 2020; 11:44. [PMID: 32014040 PMCID: PMC6998265 DOI: 10.1186/s13287-020-1559-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/30/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Background Sepsis is a global inflammatory disease that causes death. It has been reported that mesenchymal stem cell (MSC) treatment can attenuate inflammatory and septic symptoms. In this study, we investigated how interactions between neutrophils and human umbilical cord blood (hUCB)-MSCs in the liver of septic mice are involved in mitigating sepsis that is mediated by MSCs. Accordingly, we aimed to determine whether hUCB-MSC application could be an appropriate treatment for sepsis. Methods To induce septic condition, lipopolysaccharide (LPS) was intraperitoneally (i.p.) injected into mice 24 h after the intravenous (i.v.) injection of saline or hUCB-MSCs. To determine the effect of hUCB-MSCs on the immune response during sepsis, histologic analysis, immunoassays, and two-photon intravital imaging were performed 6 h post-LPS injection. For the survival study, mice were monitored for 6 days after LPS injection. Results The injection (i.v.) of hUCB-MSCs alleviated the severity of LPS-induced sepsis by increasing IL-10 levels (p < 0.001) and decreasing mortality (p < 0.05) in septic mice. In addition, this significantly reduced the recruitment of neutrophils (p < 0.001) to the liver. In hUCB-MSC-treated condition, we also observed several distinct patterns of dynamic interactions between neutrophils and hUCB-MSCs in the inflamed mouse liver, as well as vigorous interactions between hepatic stellate cells (HSCs or ito cells) and hUCB-MSCs. Interestingly, hUCB-MSCs that originated from humans were not recognized as foreign in the mouse body and consequently did not cause graft rejection. Conclusions These distinct interaction patterns between innate immune cells and hUCB-MSCs demonstrated that hUCB-MSCs have beneficial effects against LPS-induced sepsis through associations with neutrophils. In addition, the immunomodulatory properties of hUCB-MSCs might enable immune evasion in the host. Taken together, our results suggest the prospects of hUCB-MSCs as a therapeutic tool to inhibit inflammation and alleviate pathological immune responses such as sepsis.
Collapse
Affiliation(s)
- Sung Yong Ahn
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Sun Maeng
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Rim Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Ho Choe
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Han Sung Hwang
- Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea.
| | - Young-Min Hyun
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea. .,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
71
|
Török G, Erdei Z, Lilienberg J, Apáti Á, Homolya L. The importance of transporters and cell polarization for the evaluation of human stem cell-derived hepatic cells. PLoS One 2020; 15:e0227751. [PMID: 31971960 PMCID: PMC6977753 DOI: 10.1371/journal.pone.0227751] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 12/27/2019] [Indexed: 12/20/2022] Open
Abstract
One of the most promising applications of human pluripotent stem cells is their utilization for human-based pharmacological models. Despite the fact that membrane transporters expressed in the liver play pivotal role in various hepatic functions, thus far only little attention was devoted to the membrane transporter composition of the stem cell-derived liver models. In the present work, we have differentiated HUES9, a human embryonic stem cell line, toward the hepatic lineage, and monitored the expression levels of numerous differentiation marker and liver transporter genes with special focus on ABC transporters. In addition, the effect of bile acid treatment and polarizing culturing conditions on hepatic maturation has been assessed. We found that most transporter genes crucial for hepatic functions are markedly induced during hepatic differentiation; however, as regards the transporter composition the end-stage cells still exhibited dual, hepatocyte and cholangiocyte character. Although the bile acid treatment and sandwich culturing only slightly influenced the gene expressions, the stimulated cell polarization resulted in formation of bile canaliculi and proper localization of transporters. Our results point to the importance of membrane transporters in human stem cell-derived hepatic models and demonstrate the relevance of cell polarization in generation of applicable cellular models with correctly localized transporters. On the basis of our observations we suggest that conventional criteria for the evaluation of the quality of stem cell-derived hepatocyte-like cells ought to be augmented with additional elements, such as polarized and functional expression of hepatic transporters.
Collapse
Affiliation(s)
- György Török
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
| | - Zsuzsa Erdei
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
| | - Julianna Lilienberg
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
| | - Ágota Apáti
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
| | - László Homolya
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
- * E-mail:
| |
Collapse
|
72
|
Böttger R, Pauli G, Chao PH, AL Fayez N, Hohenwarter L, Li SD. Lipid-based nanoparticle technologies for liver targeting. Adv Drug Deliv Rev 2020; 154-155:79-101. [PMID: 32574575 DOI: 10.1016/j.addr.2020.06.017] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022]
Abstract
Liver diseases such as hepatitis, cirrhosis, and hepatocellular carcinoma are global health problems accounting for approximately 800 million cases and over 2 million deaths per year worldwide. Major drawbacks of standard pharmacological therapies are the inability to deliver a sufficient concentration of a therapeutic agent to the diseased liver, and nonspecific drug delivery leading to undesirable systemic side effects. Additionally, depending on the specific liver disease, drug delivery to a subset of liver cells is required. In recent years, lipid nanoparticles have been developed to passively and actively target drugs to the liver. The success of this approach has been highlighted by the FDA-approval of the first liver-targeting lipid nanoparticle, ONPATTRO, in 2018 and many other promising candidate technologies are expected to follow. This review summarizes recent developments of various lipid-based liver-targeting technologies, namely solid-lipid nanoparticles, liposomes, niosomes and micelles, and discusses the challenges and future perspectives in this field.
Collapse
|
73
|
Flow-Based Three-Dimensional Co-Culture Model for Long-Term Hepatotoxicity Prediction. MICROMACHINES 2019; 11:mi11010036. [PMID: 31892214 PMCID: PMC7019533 DOI: 10.3390/mi11010036] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/13/2019] [Accepted: 12/25/2019] [Indexed: 12/30/2022]
Abstract
We developed concave microwell arrays to establish a size-controllable 3-D co-culture liver model for in vitro drug toxicity testing, to predict hepatotoxicity. The interaction of hepatocytes with hepatic stellate cells (HSCs) was investigated by co-culturing primary 3-D hepatocyte spheroids and HSCs (heterosphere), using 3-D liver-on-a-chip. The effect of HSCs was investigated during spheroid formation; they were involved in controlling the organization of spheroidal aggregates and the formation of tight cell–cell contacts. Scanning electron microscopy (SEM) images showed that co-cultured spheroids with smoother surfaces in the flow chip aggregated more tightly and rapidly, compared to mono-cultured spheroids, until 13 days. Metabolic function analysis revealed that heterospheres secreted 40% more albumin and urea than hepatospheres on day 13. Additionally, an acetaminophen (AAP) and isoniazid (INH) concentration-dependent increase in CYP3A4 expression was detected in the 3-D cultures, and an increase in Lactate dehydrogenase (LDH) release after AAP and INH treatment was observed. CYP1A2, Mrp1 and UGT1A5 mRNA expression levels in the heterospheres and hepatospheres were evaluated from days 3 to 13. To examine the potential for toxicity testing in the flow-conditioned culture of the heterospheres, we evaluated cytotoxicity using the endpoint LDH release in the heterospheres and hepatospheres. IC50 values for AAP and INH after 24 h of exposure were calculated from the dose–response curves of the compounds. Flow-conditioned heterosphere culture results suggest that it may be suitable for long-term culture and cytotoxicity testing. Thus, our co-culture system closely resembles the in vivo environment and allows long-term in vitro hepatotoxicity prediction.
Collapse
|
74
|
Reiterer M, Branco CM. Endothelial cells and organ function: applications and implications of understanding unique and reciprocal remodelling. FEBS J 2019; 287:1088-1100. [PMID: 31736207 PMCID: PMC7155104 DOI: 10.1111/febs.15143] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/21/2019] [Accepted: 11/15/2019] [Indexed: 12/16/2022]
Abstract
The microvasculature is a heterogeneous, dynamic and versatile component of the systemic circulation, with a unique ability to locally self-regulate and to respond to organ demand and environmental stimuli. Endothelial cells from different organs display considerable variation, but it is currently unclear to what extent functional properties of organ-specific endothelial cells are intrinsic, acquired and/or reprogrammable. Vascular function is a fundamental pillar of homeostasis, and dysfunction results in systemic consequences for the organism. Additionally, vascular failure can occur downstream of organ disease or environmental stress, often driving an exacerbation of symptoms and pathologies originally independent of the local circulation. The understanding of the molecular mechanisms underlying endothelial physiology and metabolism holds the promise to inform and improve diagnosis, prognosis and treatment options for a myriad of conditions as unrelated as cancer, neurodegeneration or pulmonary hypertension, and likely everything in between, if we consider that also treatments for such conditions are primarily distributed via the bloodstream. However, studying endothelial function has its challenges: the origin, isolation, culture conditions and preconditioning stimuli make this an extremely variable cell type to study and difficult to source. Animal models exist but are neither trivial to generate, nor necessarily adequately translatable to human disease. In this article, we aim to illustrate the breadth of microvascular functions in different environments, highlighting current and pioneering studies that have advanced our insight into the importance of the integrity of this tissue, as well as the limitations posed by its heterogeneity and plasticity.
Collapse
Affiliation(s)
- Moritz Reiterer
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, UK.,Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Cristina M Branco
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, UK
| |
Collapse
|
75
|
Boyman L, Karbowski M, Lederer WJ. Regulation of Mitochondrial ATP Production: Ca 2+ Signaling and Quality Control. Trends Mol Med 2019; 26:21-39. [PMID: 31767352 DOI: 10.1016/j.molmed.2019.10.007] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023]
Abstract
Cardiac ATP production primarily depends on oxidative phosphorylation in mitochondria and is dynamically regulated by Ca2+ levels in the mitochondrial matrix as well as by cytosolic ADP. We discuss mitochondrial Ca2+ signaling and its dysfunction which has recently been linked to cardiac pathologies including arrhythmia and heart failure. Similar dysfunction in other excitable and long-lived cells including neurons is associated with neurodegenerative diseases such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD). Central to this new understanding is crucial Ca2+ regulation of both mitochondrial quality control and ATP production. Mitochondria-associated membrane (MAM) signaling from the sarcoplasmic reticulum (SR) and the endoplasmic reticulum (ER) to mitochondria is discussed. We propose future research directions that emphasize a need to define quantitatively the physiological roles of MAMs, as well as mitochondrial quality control and ATP production.
Collapse
Affiliation(s)
- Liron Boyman
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mariusz Karbowski
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - W Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
76
|
Fernando DH, Forbes JM, Angus PW, Herath CB. Development and Progression of Non-Alcoholic Fatty Liver Disease: The Role of Advanced Glycation End Products. Int J Mol Sci 2019; 20:E5037. [PMID: 31614491 PMCID: PMC6834322 DOI: 10.3390/ijms20205037] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/19/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects up to 30% of the adult population and is now a major cause of liver disease-related premature illness and deaths in the world. Treatment is largely based on lifestyle modification, which is difficult to achieve in most patients. Progression of simple fatty liver or steatosis to its severe form non-alcoholic steatohepatitis (NASH) and liver fibrosis has been explained by a 'two-hit hypothesis'. Whilst simple steatosis is considered the first hit, its transformation to NASH may be driven by a second hit. Of several factors that constitute the second hit, advanced glycation end products (AGEs), which are formed when reducing-sugars react with proteins or lipids, have been implicated as major candidates that drive steatosis to NASH via the receptor for AGEs (RAGE). Both endogenous and processed food-derived (exogenous) AGEs can activate RAGE, mainly present on Kupffer cells and hepatic stellate cells, thus propagating NAFLD progression. This review focuses on the pathophysiology of NAFLD with special emphasis on the role of food-derived AGEs in NAFLD progression to NASH and liver fibrosis. Moreover, the effect of dietary manipulation to reduce AGE content in food or the therapies targeting AGE/RAGE pathway on disease progression is also discussed.
Collapse
Affiliation(s)
- Dinali H Fernando
- Department of Medicine, The University of Melbourne, Melbourne 3084, Australia.
| | | | - Peter W Angus
- Liver transplant unit, Austin Health, Heidelberg 3084, Australia.
| | - Chandana B Herath
- Department of Medicine, The University of Melbourne, Melbourne 3084, Australia.
| |
Collapse
|
77
|
Falkenberg KD, Rohlenova K, Luo Y, Carmeliet P. The metabolic engine of endothelial cells. Nat Metab 2019; 1:937-946. [PMID: 32694836 DOI: 10.1038/s42255-019-0117-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023]
Abstract
Endothelial cells (ECs) line the quiescent vasculature but can form new blood vessels (a process termed angiogenesis) in disease. Strategies targeting angiogenic growth factors have been clinically developed for the treatment of malignant and ocular diseases. Studies over the past decade have documented that several pathways of central carbon metabolism are necessary for EC homeostasis and growth, and that strategies that stimulate or block EC metabolism can be used to promote or inhibit vessel growth, respectively. In this Review, we provide an updated overview of the growing understanding of central carbon metabolic pathways in ECs and the therapeutic opportunities for targeting EC metabolism.
Collapse
Affiliation(s)
- Kim D Falkenberg
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Katerina Rohlenova
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Yonglun Luo
- Lars Bolund Institute of Regenerative Medicine, BGI-Qindao, Qindao, China
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium.
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium.
| |
Collapse
|
78
|
Zhai X, Wang W, Dou D, Ma Y, Gang D, Jiang Z, Shi B, Jin B. A novel technique to prepare a single cell suspension of isolated quiescent human hepatic stellate cells. Sci Rep 2019; 9:12757. [PMID: 31485000 PMCID: PMC6726602 DOI: 10.1038/s41598-019-49287-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 08/20/2019] [Indexed: 01/17/2023] Open
Abstract
To explore a simple and easy-to-learn procedure for the isolation of human quiescent hepatic stellate cells (HSCs) that requires no advanced training. Thus reducing costs and increasing efficiency. This protocol will provide sufficient primary cells with minimal contaminants for future basic research on diseases associated with human HSCs. Normal liver tissues were isolated from patients undergoing hepatic hemangioma resection, and a single cell suspension of these tissues was prepared using the Gentle MACS tissue processor. By using this method, the difficulty of the procedure was reduced, fewer cells were lost during the preparation treatments, and the maximal activity of single cells was maintained. Following preparation of the cell suspension, the HSCs were further isolated using a Nycodenz density gradient. Cell viability was examined by trypan blue staining, and the purity of the quiescent human HSCs was determined by autofluorescence and oil red O staining. Activated and quiescent human HSCs were identified using immunofluorescence and Western blotting. The cell cycle distribution in activated and quiescent human HSCs was analyzed by flow cytometry.The recovery rate of the HSCs was approximately (2.1 ± 0.23) × 106 of tissue, with 94.43 ± 1.89% cell viability and 93.8 ± 1.52% purity. The technique used in this study is a simple, high-yield, and repeatable method for HSC isolation that is worthy of recommendation.
Collapse
Affiliation(s)
- Xiangyu Zhai
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Wei Wang
- School of medicine, Shandong University, Jinan, China
| | - Dandan Dou
- School of basic medical sciences, Shandong University, Jinan, China
| | - Yunlong Ma
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Du Gang
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Zhengchen Jiang
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Binyao Shi
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Bin Jin
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China.
| |
Collapse
|
79
|
Li X, Sun X, Carmeliet P. Hallmarks of Endothelial Cell Metabolism in Health and Disease. Cell Metab 2019; 30:414-433. [PMID: 31484054 DOI: 10.1016/j.cmet.2019.08.011] [Citation(s) in RCA: 270] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 01/13/2023]
Abstract
In 2009, it was postulated that endothelial cells (ECs) would only be able to execute the orders of growth factors if these cells would accordingly adapt their metabolism. Ten years later, it has become clear that ECs, often differently from other cell types, rely on distinct metabolic pathways to survive and form new blood vessels; that manipulation of EC metabolic pathways alone (even without changing angiogenic signaling) suffices to alter vessel sprouting; and that perturbations of these metabolic pathways can underlie excess formation of new blood vessels (angiogenesis) in cancer and ocular diseases. Initial proof of evidence has been provided that targeting (normalizing) these metabolic perturbations in diseased ECs and delivery of metabolites deserve increasing attention as novel therapeutic approaches for inhibiting or stimulating vessel growth in multiple disorders.
Collapse
Affiliation(s)
- Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China.
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Peter Carmeliet
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven B-3000, Belgium.
| |
Collapse
|
80
|
Ye S, Boeter JWB, Penning LC, Spee B, Schneeberger K. Hydrogels for Liver Tissue Engineering. Bioengineering (Basel) 2019; 6:E59. [PMID: 31284412 PMCID: PMC6784004 DOI: 10.3390/bioengineering6030059] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
Bioengineered livers are promising in vitro models for drug testing, toxicological studies, and as disease models, and might in the future be an alternative for donor organs to treat end-stage liver diseases. Liver tissue engineering (LTE) aims to construct liver models that are physiologically relevant. To make bioengineered livers, the two most important ingredients are hepatic cells and supportive materials such as hydrogels. In the past decades, dozens of hydrogels have been developed to act as supportive materials, and some have been used for in vitro models and formed functional liver constructs. However, currently none of the used hydrogels are suitable for in vivo transplantation. Here, the histology of the human liver and its relationship with LTE is introduced. After that, significant characteristics of hydrogels are described focusing on LTE. Then, both natural and synthetic materials utilized in hydrogels for LTE are reviewed individually. Finally, a conclusion is drawn on a comparison of the different hydrogels and their characteristics and ideal hydrogels are proposed to promote LTE.
Collapse
Affiliation(s)
- Shicheng Ye
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Jochem W B Boeter
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Louis C Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Kerstin Schneeberger
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
81
|
Schulze RJ, Schott MB, Casey CA, Tuma PL, McNiven MA. The cell biology of the hepatocyte: A membrane trafficking machine. J Cell Biol 2019; 218:2096-2112. [PMID: 31201265 PMCID: PMC6605791 DOI: 10.1083/jcb.201903090] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/24/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022] Open
Abstract
The liver performs numerous vital functions, including the detoxification of blood before access to the brain while simultaneously secreting and internalizing scores of proteins and lipids to maintain appropriate blood chemistry. Furthermore, the liver also synthesizes and secretes bile to enable the digestion of food. These diverse attributes are all performed by hepatocytes, the parenchymal cells of the liver. As predicted, these cells possess a remarkably well-developed and complex membrane trafficking machinery that is dedicated to moving specific cargos to their correct cellular locations. Importantly, while most epithelial cells secrete nascent proteins directionally toward a single lumen, the hepatocyte secretes both proteins and bile concomitantly at its basolateral and apical domains, respectively. In this Beyond the Cell review, we will detail these central features of the hepatocyte and highlight how membrane transport processes play a key role in healthy liver function and how they are affected by disease.
Collapse
Affiliation(s)
- Ryan J Schulze
- Division of Gastroenterology and Hepatology, Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Micah B Schott
- Division of Gastroenterology and Hepatology, Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Carol A Casey
- Research Service, Department of Veterans Affairs, Nebraska-Western Iowa Health Care System, Omaha, NE
- Departments of Internal Medicine and Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE
| | | | - Mark A McNiven
- Division of Gastroenterology and Hepatology, Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| |
Collapse
|
82
|
Prior N, Hindley CJ, Rost F, Meléndez E, Lau WWY, Göttgens B, Rulands S, Simons BD, Huch M. Lgr5 + stem and progenitor cells reside at the apex of a heterogeneous embryonic hepatoblast pool. Development 2019; 146:dev.174557. [PMID: 31142540 PMCID: PMC6602348 DOI: 10.1242/dev.174557] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/15/2019] [Indexed: 12/12/2022]
Abstract
During mouse embryogenesis, progenitors within the liver known as hepatoblasts give rise to adult hepatocytes and cholangiocytes. Hepatoblasts, which are specified at E8.5-E9.0, have been regarded as a homogeneous progenitor population that initiate differentiation from E13.5. Recently, scRNA-seq analysis has identified sub-populations of transcriptionally distinct hepatoblasts at E11.5. Here, we show that hepatoblasts are not only transcriptionally but also functionally heterogeneous, and that a subpopulation of E9.5-E10.0 hepatoblasts exhibit a previously unidentified early commitment to cholangiocyte fate. Importantly, we also identify a subpopulation constituting 2% of E9.5-E10.0 hepatoblasts that express the adult stem cell marker Lgr5, and generate both hepatocyte and cholangiocyte progeny that persist for the lifespan of the mouse. Combining lineage tracing and scRNA-seq, we show that Lgr5 marks E9.5-E10.0 bipotent liver progenitors residing at the apex of a hepatoblast hierarchy. Furthermore, isolated Lgr5+ hepatoblasts can be clonally expanded in vitro into embryonic liver organoids, which can commit to either hepatocyte or cholangiocyte fates. Our study demonstrates functional heterogeneity within E9.5 hepatoblasts and identifies Lgr5 as a marker for a subpopulation of bipotent liver progenitors.
Collapse
Affiliation(s)
- Nicole Prior
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
| | - Christopher J Hindley
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK.,The Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thompson Avenue, Cambridge, CB3 0HE, UK
| | - Fabian Rost
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187 Dresden, Germany
| | - Elena Meléndez
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
| | - Winnie W Y Lau
- Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Berthold Göttgens
- Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Steffen Rulands
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK.,The Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thompson Avenue, Cambridge, CB3 0HE, UK.,Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187 Dresden, Germany.,Center for Systems Biology Dresden, Pfotenhauer Strasse 108, 01307 Dresden, Germany
| | - Benjamin D Simons
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK.,The Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thompson Avenue, Cambridge, CB3 0HE, UK.,Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QR, UK
| | - Meritxell Huch
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK .,Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QR, UK.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
| |
Collapse
|
83
|
Haemmerle G, Lass A. Genetically modified mouse models to study hepatic neutral lipid mobilization. Biochim Biophys Acta Mol Basis Dis 2019; 1865:879-894. [PMID: 29883718 PMCID: PMC6887554 DOI: 10.1016/j.bbadis.2018.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/25/2018] [Accepted: 06/01/2018] [Indexed: 02/07/2023]
Abstract
Excessive accumulation of triacylglycerol is the common denominator of a wide range of clinical pathologies of liver diseases, termed non-alcoholic fatty liver disease. Such excessive triacylglycerol deposition in the liver is also referred to as hepatic steatosis. Although liver steatosis often resolves over time, it eventually progresses to steatohepatitis, liver fibrosis and cirrhosis, with associated complications, including liver failure, hepatocellular carcinoma and ultimately death of affected individuals. From the disease etiology it is obvious that a tight regulation between lipid uptake, triacylglycerol synthesis, hydrolysis, secretion and fatty acid oxidation is required to prevent triacylglycerol deposition in the liver. In addition to triacylglycerol, also a tight control of other neutral lipid ester classes, i.e. cholesteryl esters and retinyl esters, is crucial for the maintenance of a healthy liver. Excessive cholesteryl ester accumulation is a hallmark of cholesteryl ester storage disease or Wolman disease, which is associated with premature death. The loss of hepatic vitamin A stores (retinyl ester stores of hepatic stellate cells) is incidental to the onset of liver fibrosis. Importantly, this more advanced stage of liver disease usually does not resolve but progresses to life threatening stages, i.e. liver cirrhosis and cancer. Therefore, understanding the enzymes and pathways that mobilize hepatic neutral lipid esters is crucial for the development of strategies and therapies to ameliorate pathophysiological conditions associated with derangements of hepatic neutral lipid ester stores, including liver steatosis, steatohepatitis, and fibrosis. This review highlights the physiological roles of enzymes governing the mobilization of neutral lipid esters at different sites in liver cells, including cytosolic lipid droplets, endoplasmic reticulum, and lysosomes. This article is part of a Special Issue entitled Molecular Basis of Disease: Animal models in liver disease.
Collapse
Affiliation(s)
- Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31/II, 8010 Graz, Austria.
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31/II, 8010 Graz, Austria; BioTechMed-Graz, Austria.
| |
Collapse
|
84
|
Sensitivity of multifrequency magnetic resonance elastography and diffusion-weighted imaging to cellular and stromal integrity of liver tissue. J Biomech 2019; 88:201-208. [DOI: 10.1016/j.jbiomech.2019.03.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/27/2019] [Accepted: 03/27/2019] [Indexed: 12/16/2022]
|
85
|
Szafraniec E, Kus E, Wislocka A, Kukla B, Sierka E, Untereiner V, Sockalingum GD, Chlopicki S, Baranska M. Raman spectroscopy-based insight into lipid droplets presence and contents in liver sinusoidal endothelial cells and hepatocytes. JOURNAL OF BIOPHOTONICS 2019; 12:e201800290. [PMID: 30578586 DOI: 10.1002/jbio.201800290] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 06/09/2023]
Abstract
Liver sinusoidal endothelial cells (LSECs), a type of endothelial cells with unique morphology and function, play an important role in the liver hemostasis, and LSECs dysfunction is involved in the development of nonalcoholic fatty liver disease (NAFLD). Here, we employed Raman imaging and chemometric data analysis in order to characterize the presence of lipid droplets (LDs) and their lipid content in primary murine LSECs, in comparison with hepatocytes, isolated from mice on high-fat diet. On NAFLD development, LDs content in LSECs changed toward more unsaturated lipids, and this response was associated with an increased expression of stearylo-CoA desaturase-1. To the best of our knowledge, this is a first report characterizing LDs in LSECs, where their chemical composition is analyzed along the progression of NAFLD at the level of single LD using Raman imaging.
Collapse
Affiliation(s)
- Ewelina Szafraniec
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Edyta Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Adrianna Wislocka
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Bozena Kukla
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Ewa Sierka
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Valérie Untereiner
- Plateforme d'Imagerie Cellulaire et Tissulaire (PICT), Université de Reims Champagne-Ardenne, Reims, France
| | - Ganesh D Sockalingum
- BioSpecT-BioSpectroscopie Translationnelle, Université de Reims Champagne-Ardenne, Reims, France
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Malgorzata Baranska
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| |
Collapse
|
86
|
Sakai K, Katsumi H, Kamano K, Yamauchi K, Hajima A, Morishita M, Sakane T, Yamamoto A. Hepatic and Intrahepatic Targeting of Hydrogen Sulfide Prodrug by Bioconjugation. Biol Pharm Bull 2019; 42:273-279. [PMID: 30713258 DOI: 10.1248/bpb.b18-00773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hydrogen sulfide (H2S) is an endogenous gaseous transmitter known to play an important role in biological functions. For the hepatic and intrahepatic targeting of H2S prodrug at the cellular level, we developed two types of sulfo-albumins, in which five sulfide groups (source of H2S) were covalently bound to succinylated (Suc) or galactosylated (Gal) bovine serum albumin (BSA). Sulfo-BSA-Suc and polyethylene glycol (PEG)-Sulfo-BSA-Gal, both released H2S in the 5 mM glutathione solution, but not in the plasma. Sulfo-BSA-Suc and PEG-Sulfo-BSA-Gal were taken up by RAW264.7 cells (mouse macrophage-like cells) and Hep G2 cells (human hepatocellular carcinoma cells), respectively, and H2S was released. These results indicate that Sulfo-BSA-Suc and PEG -Sulfo-BSA-Gal selectively released H2S intracellularly. In a biodistribution study, up to 80% of 111In-labeled Sulfo-BSA-Suc and PEG-Sulfo-BSA-Gal rapidly accumulated in the liver, 30 min after intravenous injection in mice. Furthermore, 111In-labeled Sulfo-BSA-Suc and PEG-Sulfo-BSA-Gal predominantly accumulated in liver nonparenchymal (endothelial cells and Kupffer cells) and parenchymal cells (hepatocytes), respectively. These findings suggest that targeted delivery of H2S prodrug to a specific type of liver cells was successfully achieved by bioconjugation.
Collapse
Affiliation(s)
- Kosuke Sakai
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | | | - Kentaro Kamano
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | - Kiyo Yamauchi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | - Ayuko Hajima
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | | | - Toshiyasu Sakane
- Department of Biopharmaceutics, Kyoto Pharmaceutical University.,Department of Pharmaceutical Technology, Kobe Pharmaceutical University
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| |
Collapse
|
87
|
Immune modulation of liver sinusoidal endothelial cells by melittin nanoparticles suppresses liver metastasis. Nat Commun 2019; 10:574. [PMID: 30718511 PMCID: PMC6361944 DOI: 10.1038/s41467-019-08538-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/30/2018] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) are responsible for the immunologic tolerance of liver which is a common site for visceral metastases, suggesting its potential role as an target for cancer immunotherapy. However, targeted modulation of LSECs is still not achieved thus far. Here, we report LSECs are specifically targeted and modulated by melittin nanoparticles (α-melittin-NPs). Intravital imaging shows that LSECs fluoresce within 20 s after intravenous injection of α-melittin-NPs. α-melittin-NPs trigger the activation of LSECs and lead to dramatic changes of cytokine/chemokine milieu in the liver, which switches the hepatic immunologic environment to the activated state. As a result, α-melittin-NPs resist the formation of metastatic lesions with high efficiency. More strikingly, the survival rate reaches 80% in the spontaneous liver metastatic tumor model. Our research provides support for the use of α-melittin-NPs to break LSEC-mediated immunologic tolerance, which opens an avenue to control liver metastasis through the immunomodulation of LSECs. Liver sinusoidal endothelial cells are known to promote immune tolerance in liver. Here, the authors target these cells using melittin nanoparticles and show alterations in the liver immune environment and suppression of liver metastases.
Collapse
|
88
|
Baselet B, Sonveaux P, Baatout S, Aerts A. Pathological effects of ionizing radiation: endothelial activation and dysfunction. Cell Mol Life Sci 2019; 76:699-728. [PMID: 30377700 PMCID: PMC6514067 DOI: 10.1007/s00018-018-2956-z] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/19/2018] [Accepted: 10/23/2018] [Indexed: 01/13/2023]
Abstract
The endothelium, a tissue that forms a single layer of cells lining various organs and cavities of the body, especially the heart and blood as well as lymphatic vessels, plays a complex role in vascular biology. It contributes to key aspects of vascular homeostasis and is also involved in pathophysiological processes, such as thrombosis, inflammation, and hypertension. Epidemiological data show that high doses of ionizing radiation lead to cardiovascular disease over time. The aim of this review is to summarize the current knowledge on endothelial cell activation and dysfunction after ionizing radiation exposure as a central feature preceding the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Bjorn Baselet
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK•CEN), Mol, Belgium
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics, Université catholique de Louvain (UCL), Brussels, Belgium
| | - Pierre Sonveaux
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics, Université catholique de Louvain (UCL), Brussels, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK•CEN), Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - An Aerts
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK•CEN), Mol, Belgium.
| |
Collapse
|
89
|
Choi WM, Eun HS, Lee YS, Kim SJ, Kim MH, Lee JH, Shim YR, Kim HH, Kim YE, Yi HS, Jeong WI. Experimental Applications of in situ Liver Perfusion Machinery for the Study of Liver Disease. Mol Cells 2019; 42:45-55. [PMID: 30665288 PMCID: PMC6354060 DOI: 10.14348/molcells.2018.0330] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/29/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
The liver is involved in a wide range of activities in vertebrates and some other animals, including metabolism, protein synthesis, detoxification, and the immune system. Until now, various methods have been devised to study liver diseases; however, each method has its own limitations. In situ liver perfusion machinery, originally developed in rats, has been successfully adapted to mice, enabling the study of liver diseases. Here we describe the protocol, which is a simple but widely applicable method for investigating the liver diseases. The liver is perfused in situ by cannulation of the portal vein and suprahepatic inferior vena cava (IVC), with antegrade closed circuit circulation completed by clamping the infrahepatic IVC. In situ liver perfusion can be utilized to evaluate immune cell migration and function, hemodynamics and related cellular reactions in each type of hepatic cells, and the metabolism of toxic or other compounds by changing the composition of the circulating media. In situ liver perfusion method maintains liver function and cell viability for up to 2 h. This study also describes an optional protocol using density-gradient centrifugation for the separation of different types of hepatic cells, allowing the determination of changes in each cell type. In summary, this method of in situ liver perfusion will be useful for studying liver diseases as a complement to other established methods.
Collapse
Affiliation(s)
- Won-Mook Choi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon,
Korea
| | - Hyuk Soo Eun
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon,
Korea
| | - Young-Sun Lee
- Department of Internal Medicine, Korea University College of Medicine,
Korea
| | - Sun Jun Kim
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon,
Korea
| | - Myung-Ho Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon,
Korea
| | - Jun-Hee Lee
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon,
Korea
| | - Young-Ri Shim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon,
Korea
| | - Hee-Hoon Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon,
Korea
| | - Ye Eun Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon,
Korea
| | - Hyon-Seung Yi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon,
Korea
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon,
Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon,
Korea
| |
Collapse
|
90
|
Bonnaventure P, Cusin F, Pastor CM. Hepatocyte Concentrations of Imaging Compounds Associated with Transporter Inhibition: Evidence in Perfused Rat Livers. Drug Metab Dispos 2019; 47:412-418. [PMID: 30674615 DOI: 10.1124/dmd.118.084624] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/18/2019] [Indexed: 12/15/2022] Open
Abstract
In the liver, several approaches are used to investigate and predict the complex issue of drug-induced transporter inhibition. These approaches include in vitro assays and pharmacokinetic models that predict how inhibitors modify the systemic and liver concentrations of the victim drugs. Imaging is another approach that shows how inhibitors might alter liver concentrations stronger than systemic concentrations. In perfused rat livers associated with a gamma counter that measures liver concentrations continuously, we previously showed how fluxes across transporters generate the hepatocyte concentrations of two clinical imaging compounds, one with a low extraction ratio [gadobenate dimeglumine (BOPTA)] and one with a high extraction ratio [mebrofenin (MEB)]. BOPTA and MEB are transported by rat organic anion transporting polypeptide and multiple resistance-associated protein 2, which are both inhibited by rifampicin. The aim of the study is to measure how rifampicin modifies the hepatocyte concentrations and membrane clearances of BOPTA and MEB and to determine whether these compounds might be used to investigate transporter-mediated drug-drug interactions in clinical studies. We show that rifampicin coperfusion greatly decreases BOPTA hepatocyte concentrations, but increases those of MEB. Rifampicin strongly decreases BOPTA hepatic clearance. In contrast, rifampicin decreases moderately MEB hepatic clearance and blocks the biliary intrinsic clearance, increasing MEB hepatocyte concentrations. In conclusion, low concentrations prevent the quantification of BOPTA biliary intrinsic clearance, while MEB is a promising imaging probe substrate to evidence transporter-mediated drug-drug interactions when inhibitors act on influx and efflux transporters.
Collapse
Affiliation(s)
- Pierre Bonnaventure
- Department of Radiology, Hôpitaux Universitaires de Genève, Geneva, Switzerland (P.B., F.C., C.M.P.); and Laboratory of Imaging Biomarkers, Centre of Research on Inflammation, Unité Mixte de Recherche 1149, Institut National de la santé et de la Recherche Médicale and University Paris Diderot, Paris, France (C.M.P.)
| | - Fabien Cusin
- Department of Radiology, Hôpitaux Universitaires de Genève, Geneva, Switzerland (P.B., F.C., C.M.P.); and Laboratory of Imaging Biomarkers, Centre of Research on Inflammation, Unité Mixte de Recherche 1149, Institut National de la santé et de la Recherche Médicale and University Paris Diderot, Paris, France (C.M.P.)
| | - Catherine M Pastor
- Department of Radiology, Hôpitaux Universitaires de Genève, Geneva, Switzerland (P.B., F.C., C.M.P.); and Laboratory of Imaging Biomarkers, Centre of Research on Inflammation, Unité Mixte de Recherche 1149, Institut National de la santé et de la Recherche Médicale and University Paris Diderot, Paris, France (C.M.P.)
| |
Collapse
|
91
|
Schmitt MV, Lienau P, Fricker G, Reichel A. Quantitation of Lysosomal Trapping of Basic Lipophilic Compounds Using In Vitro Assays and In Silico Predictions Based on the Determination of the Full pH Profile of the Endo-/Lysosomal System in Rat Hepatocytes. Drug Metab Dispos 2018; 47:49-57. [PMID: 30409837 DOI: 10.1124/dmd.118.084541] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/22/2018] [Indexed: 12/28/2022] Open
Abstract
Lysosomal sequestration may affect the pharmacokinetics, efficacy, and safety of new basic lipophilic drug candidates potentially impacting their intracellular concentrations and tissue distribution. It may also be involved in drug-drug interactions, drug resistance, and phospholipidosis. However, currently there are no assays to evaluate the lysosomotropic behavior of compounds in a setting fully meeting the needs of drug discovery. We have, therefore, integrated a set of methods to reliably rank order, quantify, and calculate the extent of lysosomal sequestration in rat hepatocytes. An indirect fluorescence-based assay monitors the displacement of the fluorescence probe LysoTracker Red by test compounds. Using a lysosomal-specific evaluation algorithm allows one to generate IC50 values at lower than previously reported concentrations. The concentration range directly agrees with the concentration dependency of the lysosomal drug content itself directly quantified by liquid chromatography-tandem mass spectrometry and thus permits a quantitative link between the indirect and the direct trapping assay. Furthermore, we have determined the full pH profile and corresponding volume fractions of the endo-/lysosomal system in plated rat hepatocytes, enabling a more accurate in silico prediction of the extent of lysosomal trapping based only on pK a values as input, allowing early predictions even prior to chemical synthesis. The concentration dependency-i.e., the saturability of the trapping-can then be determined by the IC50 values generated in vitro. Thereby, a more quantitative assessment of the susceptibility of basic lipophilic compounds for lysosomal trapping is possible.
Collapse
Affiliation(s)
- Maximilian V Schmitt
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., P.L., A.R.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Philip Lienau
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., P.L., A.R.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Gert Fricker
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., P.L., A.R.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| | - Andreas Reichel
- Bayer AG, Pharmaceuticals R&D, Translational Sciences, Research Pharmacokinetics, Berlin, Germany (M.V.S., P.L., A.R.); and Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany (M.V.S., G.F.)
| |
Collapse
|
92
|
Bousseau S, Vergori L, Soleti R, Lenaers G, Martinez MC, Andriantsitohaina R. Glycosylation as new pharmacological strategies for diseases associated with excessive angiogenesis. Pharmacol Ther 2018; 191:92-122. [DOI: 10.1016/j.pharmthera.2018.06.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/01/2018] [Indexed: 02/07/2023]
|
93
|
Yurt KK, Kivrak EG, Altun G, Mohamed H, Ali F, Gasmalla HE, Kaplan S. A brief update on physical and optical disector applications and sectioning-staining methods in neuroscience. J Chem Neuroanat 2018; 93:16-29. [DOI: 10.1016/j.jchemneu.2018.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/25/2018] [Accepted: 02/25/2018] [Indexed: 02/06/2023]
|
94
|
Fu X, Sluka JP, Clendenon SG, Dunn KW, Wang Z, Klaunig JE, Glazier JA. Modeling of xenobiotic transport and metabolism in virtual hepatic lobule models. PLoS One 2018; 13:e0198060. [PMID: 30212461 PMCID: PMC6136710 DOI: 10.1371/journal.pone.0198060] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/23/2018] [Indexed: 12/29/2022] Open
Abstract
Computational models of normal liver function and xenobiotic induced liver damage are increasingly being used to interpret in vitro and in vivo data and as an approach to the de novo prediction of the liver’s response to xenobiotics. The microdosimetry (dose at the level of individual cells) of xenobiotics vary spatially within the liver because of both compound-independent and compound-dependent factors. In this paper, we build model liver lobules to investigate the interplay between vascular structure, blood flow and cellular transport that lead to regional variations in microdosimetry. We then compared simulation results obtained using this complex spatial model with a simpler linear pipe model of a sinusoid and a very simple single box model. We found that variations in diffusive transport, transporter-mediated transport and metabolism, coupled with complex liver sinusoid architecture and blood flow distribution, led to three essential patterns of xenobiotic exposure within the virtual liver lobule: (1) lobular-wise uniform, (2) radially varying and (3) both radially and azimuthally varying. We propose to use these essential patterns of exposure as a reference for selection of model representations when a computational study involves modeling detailed hepatic responses to xenobiotics.
Collapse
Affiliation(s)
- Xiao Fu
- Biocomplexity Institute, Indiana University, Bloomington, IN, United States of America
- Department of Physics, Indiana University, Bloomington, IN, United States of America
| | - James P. Sluka
- Biocomplexity Institute, Indiana University, Bloomington, IN, United States of America
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States of America
- * E-mail:
| | - Sherry G. Clendenon
- Biocomplexity Institute, Indiana University, Bloomington, IN, United States of America
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States of America
| | - Kenneth W. Dunn
- School of Medicine, Indiana University, Indianapolis, IN, United States of America
| | - Zemin Wang
- School of Public Health, Indiana University, Bloomington, IN, United States of America
| | - James E. Klaunig
- School of Public Health, Indiana University, Bloomington, IN, United States of America
| | - James A. Glazier
- Biocomplexity Institute, Indiana University, Bloomington, IN, United States of America
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States of America
| |
Collapse
|
95
|
Yang L, Li LC, Wang X, Wang WH, Wang YC, Xu CR. The contributions of mesoderm-derived cells in liver development. Semin Cell Dev Biol 2018; 92:63-76. [PMID: 30193996 DOI: 10.1016/j.semcdb.2018.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/31/2018] [Accepted: 09/02/2018] [Indexed: 02/07/2023]
Abstract
The liver is an indispensable organ for metabolism and drug detoxification. The liver consists of endoderm-derived hepatobiliary lineages and various mesoderm-derived cells, and interacts with the surrounding tissues and organs through the ventral mesentery. Liver development, from hepatic specification to liver maturation, requires close interactions with mesoderm-derived cells, such as mesothelial cells, hepatic stellate cells, mesenchymal cells, liver sinusoidal endothelial cells and hematopoietic cells. These cells affect liver development through precise signaling events and even direct physical contact. Through the use of new techniques, emerging studies have recently led to a deeper understanding of liver development and its related mechanisms, especially the roles of mesodermal cells in liver development. Based on these developments, the current protocols for in vitro hepatocyte-like cell induction and liver-like tissue construction have been optimized and are of great importance for the treatment of liver diseases. Here, we review the roles of mesoderm-derived cells in the processes of liver development, hepatocyte-like cell induction and liver-like tissue construction.
Collapse
Affiliation(s)
- Li Yang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Lin-Chen Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xin Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China
| | - Wei-Hua Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yan-Chun Wang
- Haidian Maternal & Child Health Hospital, Beijing, 100080, China
| | - Cheng-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China.
| |
Collapse
|
96
|
Kiamehr M, Alexanova A, Viiri LE, Heiskanen L, Vihervaara T, Kauhanen D, Ekroos K, Laaksonen R, Käkelä R, Aalto-Setälä K. hiPSC-derived hepatocytes closely mimic the lipid profile of primary hepatocytes: A future personalised cell model for studying the lipid metabolism of the liver. J Cell Physiol 2018; 234:3744-3761. [PMID: 30146765 DOI: 10.1002/jcp.27131] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022]
Abstract
Hepatocyte-like cells (HLCs) differentiated from human-induced pluripotent stem cells offer an alternative platform to primary human hepatocytes (PHHs) for studying the lipid metabolism of the liver. However, despite their great potential, the lipid profile of HLCs has not yet been characterized. Here, we comprehensively studied the lipid profile and fatty acid (FA) metabolism of HLCs and compared them with the current standard hepatocyte models: HepG2 cells and PHHs. We differentiated HLCs by five commonly used methods from three cell lines and thoroughly characterized them by gene and protein expression. HLCs generated by each method were assessed for their functionality and the ability to synthesize, elongate, and desaturate FAs. In addition, lipid and FA profiles of HLCs were investigated by both mass spectrometry and gas chromatography and then compared with the profiles of PHHs and HepG2 cells. HLCs resembled PHHs by expressing hepatic markers: secreting albumin, lipoprotein particles, and urea, and demonstrating similarities in their lipid and FA profile. Unlike HepG2 cells, HLCs contained low levels of lysophospholipids similar to the content of PHHs. Furthermore, HLCs were able to efficiently use the exogenous FAs available in their medium and simultaneously modify simple lipids into more complex ones to fulfill their needs. In addition, we propose that increasing the polyunsaturated FA supply of the culture medium may positively affect the lipid profile and functionality of HLCs. In conclusion, our data showed that HLCs provide a functional and relevant model to investigate human lipid homeostasis at both molecular and cellular levels.
Collapse
Affiliation(s)
- Mostafa Kiamehr
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Anna Alexanova
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Leena E Viiri
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | | | | | | | - Kim Ekroos
- Lipidomics Consulting Ltd, Espoo, Finland
| | - Reijo Laaksonen
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Zora Biosciences, Espoo, Finland
| | - Reijo Käkelä
- Faculty of Biology and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Katriina Aalto-Setälä
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Heart Hospital, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
97
|
Theodorou K, Boon RA. Endothelial Cell Metabolism in Atherosclerosis. Front Cell Dev Biol 2018; 6:82. [PMID: 30131957 PMCID: PMC6090045 DOI: 10.3389/fcell.2018.00082] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 07/13/2018] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis and its sequelae, such as myocardial infarction and stroke, are the leading cause of death worldwide. Vascular endothelial cells (EC) play a critical role in vascular homeostasis and disease. Atherosclerosis as well as its independent risk factors including diabetes, obesity, and aging, are hallmarked by endothelial activation and dysfunction. Metabolic pathways have emerged as key regulators of many EC functions, including angiogenesis, inflammation, and barrier function, processes which are deregulated during atherogenesis. In this review, we highlight the role of glucose, fatty acid, and amino acid metabolism in EC functions during physiological and pathological states, specifically atherosclerosis, diabetes, obesity and aging.
Collapse
Affiliation(s)
- Kosta Theodorou
- Centre of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe-University, Frankfurt am Main, Germany
| | - Reinier A Boon
- Centre of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe-University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research DZHK, Partner Site Rhine-Main, Berlin, Germany.,Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
98
|
Isolated Perfused Rat Livers to Quantify the Pharmacokinetics and Concentrations of Gd-BOPTA. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:3839108. [PMID: 30116162 PMCID: PMC6079620 DOI: 10.1155/2018/3839108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/17/2018] [Indexed: 12/14/2022]
Abstract
With recent advances in liver imaging, the estimation of liver concentrations is now possible following the injection of hepatobiliary contrast agents and radiotracers. However, how these images are generated remains partially unknown. Most experiments that would be helpful to increase this understanding cannot be performed in vivo. For these reasons, we investigated the liver distribution of the magnetic resonance (MR) contrast agent gadobenate dimeglumine (Gd-BOPTA, MultiHance®, Bracco Imaging) in isolated perfused rat livers (IPRLs). In IPRL, we developed a new set up that quantifies simultaneously the Gd-BOPTA compartment concentrations and the transfer rates between these compartments. Concentrations were measured either by MR signal intensity or by count rates when the contrast agent was labelled by [153Gd]. With this experimental model, we show how the Gd-BOPTA hepatocyte concentrations are modified by temperature and liver flow rates. We define new pharmacokinetic parameters to quantify the canalicular transport of Gd-BOPTA. Finally, we present how transfer rates generate Gd-BOPTA concentrations in rat liver compartments. These findings better explain how liver imaging with hepatobiliary radiotracers and contrast agents is generated and improve the image interpretation by clinicians.
Collapse
|
99
|
Quantification of hepatic perfusion and hepatocyte function with dynamic gadoxetic acid-enhanced MRI in patients with chronic liver disease. Clin Sci (Lond) 2018; 132:813-824. [PMID: 29440620 DOI: 10.1042/cs20171131] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 02/05/2018] [Accepted: 02/09/2018] [Indexed: 02/06/2023]
Abstract
The purpose of the present study was to develop and perform initial validation of dynamic MRI enhanced with gadoxetic acid as hepatobiliary contrast agent to quantify hepatic perfusion and hepatocyte function in patients with chronic liver disease. Free-breathing, dynamic gadoxetic acid-enhanced MRI was performed at 3.0 T using a 3D time-resolved angiography sequence with stochastic trajectories during 38 min. A dual-input three-compartment model was developed to derive hepatic perfusion and hepatocyte function parameters. Method feasibility was assessed in 23 patients with biopsy-proven chronic liver disease. Parameter analysis could be performed in 21 patients (91%). The hepatocyte function parameters were more discriminant than the perfusion parameters to differentiate between patients with minimal fibrosis (METAVIR F0-F1), intermediate fibrosis (F2-F3) and cirrhosis (F4). The areas under the receiver operating characteristic curves (ROCs) to diagnose significant fibrosis (METAVIR F ≥ 2) were: 0.95 (95% CI: 0.87-1; P<0.001) for biliary efflux, 0.88 (95% CI: 0.73-1; P<0.01) for sinusoidal backflux, 0.81 (95% CI: 0.61-1; P<0.05) for hepatocyte uptake fraction and 0.75 (95% CI: 0.54-1; P<0.05) for hepatic perfusion index (HPI), respectively. These initial results in patients with chronic liver diseases show that simultaneous quantification of hepatic perfusion and hepatocyte function is feasible with free breathing dynamic gadoxetic acid-enhanced MRI. Hepatocyte function parameters may be relevant to assess liver fibrosis severity.
Collapse
|
100
|
Gao C, Zhu Z, Gao Y, Lo LJ, Chen J, Luo L, Peng J. Hepatocytes in a normal adult liver are derived solely from the embryonic hepatocytes. J Genet Genomics 2018; 45:173-175. [PMID: 29574114 DOI: 10.1016/j.jgg.2017.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/14/2017] [Accepted: 12/28/2017] [Indexed: 01/13/2023]
Affiliation(s)
- Ce Gao
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhihui Zhu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuqi Gao
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Li Jan Lo
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jun Chen
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lingfei Luo
- College of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jinrong Peng
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|