1
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
2
|
Faheem S, Hameed H, Paiva-Santos AC, Khan MA, Ghumman SA, Hameed A. The role of chondroitin sulphate as a potential biomaterial for hepatic tissue regeneration: A comprehensive review. Int J Biol Macromol 2024; 280:136332. [PMID: 39482129 DOI: 10.1016/j.ijbiomac.2024.136332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/26/2024] [Accepted: 10/03/2024] [Indexed: 11/03/2024]
Abstract
Chondroitin sulphate is an anionic hetero-polysaccharide, having numerous structural affinities for building the bio-active components. In addition to biodegradable/biocompatible activities, chondroitin sulphate also possesses anti-coagulant/anti-thrombogenic, anti-inflammatory, anti-oxidant as well as anti-tumor activities. Chondroitin sulphate has an inherited affinity for glycosylation enzymes and receptors, which are overexpressed over degenerated cells and organelles. Because of this affinity, chondroitin sulphate is nominated as an active cellular/subcellular targeted biological macromolecule to assist in site-specific delivery. Chondroitin sulphate is mainly considered a promising biomaterial for drug targeting and tissue engineering due to its specific physicochemical, mechanical, bio-degradation, and biological characteristics. In this review, the fundamental applications of chondroitin sulphate in hepatic tissue engineering are discussed. Chondroitin sulphate along with mesenchymal stem cells (MSCs) based scaffold and hydrogels for biopharmaceuticals' delivery in hepatic tissue engineering are critically discussed. In addition, the manuscript also describes leading features and markers involved in hepatic damage, and the potential role of chondroitin sulphate-based delivery systems in hepatic tissue engineering.
Collapse
Affiliation(s)
- Saleha Faheem
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore 54000, Pakistan.
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore 54000, Pakistan.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore 54000, Pakistan.
| | | | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore 54000, Pakistan.
| |
Collapse
|
3
|
Kasturi M, Mathur V, Gadre M, Srinivasan V, Vasanthan KS. Three Dimensional Bioprinting for Hepatic Tissue Engineering: From In Vitro Models to Clinical Applications. Tissue Eng Regen Med 2024; 21:21-52. [PMID: 37882981 PMCID: PMC10764711 DOI: 10.1007/s13770-023-00576-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 10/27/2023] Open
Abstract
Fabrication of functional organs is the holy grail of tissue engineering and the possibilities of repairing a partial or complete liver to treat chronic liver disorders are discussed in this review. Liver is the largest gland in the human body and plays a responsible role in majority of metabolic function and processes. Chronic liver disease is one of the leading causes of death globally and the current treatment strategy of organ transplantation holds its own demerits. Hence there is a need to develop an in vitro liver model that mimics the native microenvironment. The developed model should be a reliable to understand the pathogenesis, screen drugs and assist to repair and replace the damaged liver. The three-dimensional bioprinting is a promising technology that recreates in vivo alike in vitro model for transplantation, which is the goal of tissue engineers. The technology has great potential due to its precise control and its ability to homogeneously distribute cells on all layers in a complex structure. This review gives an overview of liver tissue engineering with a special focus on 3D bioprinting and bioinks for liver disease modelling and drug screening.
Collapse
Affiliation(s)
- Meghana Kasturi
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mrunmayi Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Varadharajan Srinivasan
- Department of Civil Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
4
|
de Hoyos-Vega JM, Hong HJ, Stybayeva G, Revzin A. Hepatocyte cultures: From collagen gel sandwiches to microfluidic devices with integrated biosensors. APL Bioeng 2021; 5:041504. [PMID: 34703968 PMCID: PMC8519630 DOI: 10.1063/5.0058798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocytes are parenchymal cells of the liver responsible for drug detoxification, urea and bile production, serum protein synthesis, and glucose homeostasis. Hepatocytes are widely used for drug toxicity studies in bioartificial liver devices and for cell-based liver therapies. Because hepatocytes are highly differentiated cells residing in a complex microenvironment in vivo, they tend to lose hepatic phenotype and function in vitro. This paper first reviews traditional culture approaches used to rescue hepatic function in vitro and then discusses the benefits of emerging microfluidic-based culture approaches. We conclude by reviewing integration of hepatocyte cultures with bioanalytical or sensing approaches.
Collapse
Affiliation(s)
- Jose M. de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| |
Collapse
|
5
|
Roy HS, Singh R, Ghosh D. SARS-CoV-2 and tissue damage: current insights and biomaterial-based therapeutic strategies. Biomater Sci 2021; 9:2804-2824. [PMID: 33666206 DOI: 10.1039/d0bm02077j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The effect of SARS-CoV-2 infection on humanity has gained worldwide attention and importance due to the rapid transmission, lack of treatment options and high mortality rate of the virus. While scientists across the world are searching for vaccines/drugs that can control the spread of the virus and/or reduce the risks associated with infection, patients infected with SARS-CoV-2 have been reported to have tissue/organ damage. With most tissues/organs having limited regenerative potential, interventions that prevent further damage or facilitate healing would be helpful. In the past few decades, biomaterials have gained prominence in the field of tissue engineering, in view of their major role in the regenerative process. Here we describe the effect of SARS-CoV-2 on multiple tissues/organs, and provide evidence for the positive role of biomaterials in aiding tissue repair. These findings are further extrapolated to explore their prospects as a therapeutic platform to address the tissue/organ damage that is frequently observed during this viral outbreak. This study suggests that the biomaterial-based approach could be an effective strategy for regenerating tissues/organs damaged by SARS-CoV-2.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Rupali Singh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Deepa Ghosh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| |
Collapse
|
6
|
Labour MN, Le Guilcher C, Aid-Launais R, El Samad N, Lanouar S, Simon-Yarza T, Letourneur D. Development of 3D Hepatic Constructs Within Polysaccharide-Based Scaffolds with Tunable Properties. Int J Mol Sci 2020; 21:ijms21103644. [PMID: 32455711 PMCID: PMC7279349 DOI: 10.3390/ijms21103644] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Organoids production is a key tool for in vitro studies of physiopathological conditions, drug-induced toxicity assays, and for a potential use in regenerative medicine. Hence, it prompted studies on hepatic organoids and liver regeneration. Numerous attempts to produce hepatic constructs had often limited success due to a lack of viability or functionality. Moreover, most products could not be translated for clinical studies. The aim of this study was to develop functional and viable hepatic constructs using a 3D porous scaffold with an adjustable structure, devoid of any animal component, that could also be used as an in vivo implantable system. We used a combination of pharmaceutical grade pullulan and dextran with different porogen formulations to form crosslinked scaffolds with macroporosity ranging from 30 µm to several hundreds of microns. Polysaccharide scaffolds were easy to prepare and to handle, and allowed confocal observations thanks to their transparency. A simple seeding method allowed a rapid impregnation of the scaffolds with HepG2 cells and a homogeneous cell distribution within the scaffolds. Cells were viable over seven days and form spheroids of various geometries and sizes. Cells in 3D express hepatic markers albumin, HNF4α and CYP3A4, start to polarize and were sensitive to acetaminophen in a concentration-dependant manner. Therefore, this study depicts a proof of concept for organoid production in 3D scaffolds that could be prepared under GMP conditions for reliable drug-induced toxicity studies and for liver tissue engineering.
Collapse
Affiliation(s)
- Marie-Noëlle Labour
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
- École Pratique des Hautes Études, Paris Sciences et Lettres (PSL) Research University, 4-14 rue Ferrus, 75014 Paris, France
| | - Camile Le Guilcher
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
| | - Rachida Aid-Launais
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM UMS-34, FRIM Université de Paris, X Bichat School of Medicine, F-75018 Paris, France
| | - Nour El Samad
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
| | - Soraya Lanouar
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
| | - Teresa Simon-Yarza
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
| | - Didier Letourneur
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
- Correspondence:
| |
Collapse
|
7
|
Krüger M, Oosterhoff LA, van Wolferen ME, Schiele SA, Walther A, Geijsen N, De Laporte L, van der Laan LJW, Kock LM, Spee B. Cellulose Nanofibril Hydrogel Promotes Hepatic Differentiation of Human Liver Organoids. Adv Healthc Mater 2020; 9:e1901658. [PMID: 32090504 DOI: 10.1002/adhm.201901658] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/01/2020] [Indexed: 12/16/2022]
Abstract
To replicate functional liver tissue in vitro for drug testing or transplantation, 3D tissue engineering requires representative cell models as well as scaffolds that not only promote tissue production but also are applicable in a clinical setting. Recently, adult liver-derived liver organoids are found to be of much interest due to their genetic stability, expansion potential, and ability to differentiate toward a hepatocyte-like fate. The current standard for culturing these organoids is a basement membrane hydrogel like Matrigel (MG), which is derived from murine tumor material and apart from its variability and high costs, possesses an undefined composition and is therefore not clinically applicable. Here, a cellulose nanofibril (CNF) hydrogel is investigated with regard to its potential to serve as an alternative clinical grade scaffold to differentiate liver organoids. The results show that its mechanical properties are suitable for differentiation with overall, either equal or improved, functionality of the hepatocyte-like cells compared to MG. Therefore, and because of its defined and tunable chemical definition, the CNF hydrogel presents a viable alternative to MG for liver tissue engineering with the option for clinical use.
Collapse
Affiliation(s)
- Melanie Krüger
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht University Uppsalalaan 8 3584 CT Utrecht The Netherlands
- LifeTec Group BV Kennedyplein 10‐11 5611 ZS Eindhoven The Netherlands
| | - Loes A. Oosterhoff
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht University Uppsalalaan 8 3584 CT Utrecht The Netherlands
| | - Monique E. van Wolferen
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht University Uppsalalaan 8 3584 CT Utrecht The Netherlands
| | - Simon A. Schiele
- DWI – Leibniz‐Institut für Interaktive Materialien e.V.Advanced Materials for BiomedicineITMC – Institute of Technical and Macromolecular ChemistryRWTH University Aachen Forckenbeckstr. 50 52056 Aachen Germany
| | - Andreas Walther
- Albert‐Ludwigs‐Universität FreiburgInstitute for Macromolecular Chemistry Stefan‐Meier‐Strasse 3, Hermann Staudinger Building 79104 Freiburg Germany
| | - Niels Geijsen
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht University Uppsalalaan 8 3584 CT Utrecht The Netherlands
- Hubrecht Institute for Developmental Biology and Stem Cell ResearchUniversity Medical Center Utrecht Uppsalalaan 8 3584 CT Utrecht The Netherlands
| | - Laura De Laporte
- DWI – Leibniz‐Institut für Interaktive Materialien e.V.Advanced Materials for BiomedicineITMC – Institute of Technical and Macromolecular ChemistryRWTH University Aachen Forckenbeckstr. 50 52056 Aachen Germany
| | | | - Linda M. Kock
- LifeTec Group BV Kennedyplein 10‐11 5611 ZS Eindhoven The Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht University Uppsalalaan 8 3584 CT Utrecht The Netherlands
| |
Collapse
|
8
|
da Silva Morais A, Oliveira JM, Reis RL. Biomaterials and Microfluidics for Liver Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1230:65-86. [DOI: 10.1007/978-3-030-36588-2_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
9
|
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: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
10
|
Ban Q, Chen W, Du S, Wang H, Li J, You R, Jin Y, Xue Y, Guan YQ. The preparation of the ordered pores colloidal crystal scaffold and its role in promoting growth of lung cells. Colloids Surf B Biointerfaces 2019; 173:907-917. [PMID: 30551309 DOI: 10.1016/j.colsurfb.2018.10.055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/04/2018] [Accepted: 10/21/2018] [Indexed: 01/27/2023]
Affiliation(s)
- Qing Ban
- School of Life Science, South China Normal University, Guangzhou 510631, China; Institute for Advanced Materials and Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Wuya Chen
- School of Life Science, South China Normal University, Guangzhou 510631, China; Institute for Advanced Materials and Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Shiwei Du
- School of Life Science, South China Normal University, Guangzhou 510631, China; Institute for Advanced Materials and Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Huimin Wang
- School of Life Science, South China Normal University, Guangzhou 510631, China; Institute for Advanced Materials and Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Jian Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Rong You
- School of Life Science, South China Normal University, Guangzhou 510631, China; Institute for Advanced Materials and Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Ying Jin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yongyong Xue
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University, Guangzhou 510631, China; Institute for Advanced Materials and Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
11
|
Luo X, Gupta K, Ananthanarayanan A, Wang Z, Xia L, Li A, Sakban RB, Liu S, Yu H. Directed Differentiation of Adult Liver Derived Mesenchymal Like Stem Cells into Functional Hepatocytes. Sci Rep 2018; 8:2818. [PMID: 29434311 PMCID: PMC5809507 DOI: 10.1038/s41598-018-20304-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 01/09/2018] [Indexed: 01/09/2023] Open
Abstract
Shortage of functional hepatocytes hampers drug safety testing and therapeutic applications because mature hepatocytes cannot be expanded and maintain functions in vitro. Recent studies have reported that liver progenitor cells can originate from mature hepatocytes in vivo. Derivation of proliferating progenitor cells from mature hepatocytes, and re-differentiation into functional hepatocytes in vitro has not been successful. Here we report the derivation of novel mesenchymal-like stem cells (arHMSCs) from adult rat hepatocytes. Immunofluorescence and flow cytometry characterization of arHMSCs found expression of mesenchymal markers CD29, CD44, CD90, vimentin and alpha smooth muscle actin. These arHMSCs proliferated in vitro for 4 passages yielding 104 fold increase in cell number in 28 days, and differentiated into hepatocyte-like cells (arHMSC-H). The arHMSC-H expressed significantly higher level of hepatocyte-specific markers (200 fold for albumin and 6 fold for Cyp450 enzymes) than arHMSCs. The arHMSC-H also demonstrated dose response curves similar to primary hepatocytes for 3 of the 6 paradigm hepatotoxicants tested, demonstrating utility in drug safety testing applications.
Collapse
Affiliation(s)
- Xiaobei Luo
- Department of Gastroenterology, Nanfang hospital, Southern Medical University, Guangzhou, China
| | - Kapish Gupta
- Mechanobiology Institute, National University of, Singapore, Singapore
| | - Abhishek Ananthanarayanan
- Invitrocue Pte Ltd, Singapore, Singapore.,Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences (CeLS), Singapore, Singapore
| | - Zenan Wang
- Department of Gastroenterology, Nanfang hospital, Southern Medical University, Guangzhou, China.,Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lei Xia
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Aimin Li
- Department of Gastroenterology, Nanfang hospital, Southern Medical University, Guangzhou, China
| | - Rashidah Binte Sakban
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Side Liu
- Department of Gastroenterology, Nanfang hospital, Southern Medical University, Guangzhou, China.
| | - Hanry Yu
- Department of Gastroenterology, Nanfang hospital, Southern Medical University, Guangzhou, China. .,Mechanobiology Institute, National University of, Singapore, Singapore. .,Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore. .,NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences (CeLS), Singapore, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore.
| |
Collapse
|
12
|
Calejo MT, Ilmarinen T, Skottman H, Kellomäki M. Breath figures in tissue engineering and drug delivery: State-of-the-art and future perspectives. Acta Biomater 2018; 66:44-66. [PMID: 29183847 DOI: 10.1016/j.actbio.2017.11.043] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/09/2017] [Accepted: 11/22/2017] [Indexed: 12/11/2022]
Abstract
The breath figure (BF) method is an easy, low-cost method to prepare films with a highly organized honeycomb-like porous surface. The particular surface topography and porous nature of these materials makes them valuable substrates for studying the complex effects of topography on cell fate, and to produce biomimetic materials with high performance in tissue engineering. Numerous researchers over the last two decades have studied the effects of the honeycomb topography on a variety of primary and immortalized cell lines, and drew important conclusions that can be translated to the construction of optimal biomaterials for cell culture. The literature also encouragingly shows the potential of honeycomb films to induce differentiation of stem cells down a specific lineage without the need for biochemical stimuli. Here, we review the main studies where BF honeycomb films are used as substrates for tissue engineering applications. Furthermore, we highlight the numerous advantages of the porous nature of the films, such as the enhanced, spatially controlled adsorption of proteins, the topographical cues influencing cellular behavior, and the enhanced permeability which is essential both in vitro and in vivo. Finally, this review highlights the elegant use of honeycomb films as drug-eluting biomaterials or as reservoirs for distinct drug delivery systems. STATEMENT OF SIGNIFICANCE Combining biocompatible surfaces and 3D nano/microscale topographies, such as pores or grooves, is an effective strategy for manufacturing tissue engineering scaffolds. The breath figure (BF) method is an easy technique to prepare cell culture substrates with an organized, honeycomb-like porous surface. These surface features make these scaffolds valuable for studying how the cells interact with the biomaterials. Their unique surface topography can also resemble the natural environment of the tissues in the human body. For that reason, numerous studies, using different cell types, have shown that honeycomb films can constitute high performance substrates for cell culture. Here, we review those studies, we highlight the advantages of honeycomb films in tissue engineering and we discuss their potential as unique drug-eluting systems.
Collapse
Affiliation(s)
- Maria Teresa Calejo
- BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland.
| | - Tanja Ilmarinen
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Heli Skottman
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Minna Kellomäki
- BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland; BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| |
Collapse
|
13
|
Zhang J, Zhao X, Liang L, Li J, Demirci U, Wang S. A decade of progress in liver regenerative medicine. Biomaterials 2017; 157:161-176. [PMID: 29274550 DOI: 10.1016/j.biomaterials.2017.11.027] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 11/05/2017] [Accepted: 11/21/2017] [Indexed: 12/15/2022]
Abstract
Liver diseases can be caused by viral infection, metabolic disorder, alcohol consumption, carcinoma or injury, chronically progressing to end-stage liver disease or rapidly resulting in acute liver failure. In either situation, liver transplantation is most often sought for life saving, which is, however, significantly limited by severe shortage of organ donors. Until now, tremendous multi-disciplinary efforts have been dedicated to liver regenerative medicine, aiming at providing transplantable cells, microtissues, or bioengineered whole liver via tissue engineering, or maintaining partial liver functions via extracorporeal support. In both directions, new compatible biomaterials, stem cell sources, and bioengineering approaches have fast-forwarded liver regenerative medicine towards potential clinical applications. Another important progress in this field is the development of liver-on-a-chip technologies, which enable tissue engineering, disease modeling, and drug testing under biomimetic extracellular conditions. In this review, we aim to highlight the last decade's progress in liver regenerative medicine from liver tissue engineering, bioartificial liver devices (BAL), to liver-on-a-chip platforms, and then to present challenges ahead for further advancement.
Collapse
Affiliation(s)
- Jingwei Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China; Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Liguo Liang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China; Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Jun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China.
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University, School of Medicine, Palo Alto, CA 94304, USA; Department of Electrical Engineering (By courtesy), Stanford University, Stanford, CA 94305, USA.
| | - ShuQi Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China; Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China; Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University, School of Medicine, Palo Alto, CA 94304, USA.
| |
Collapse
|
14
|
Affiliation(s)
- Milica Radisic
- University of Toronto and Toronto General Research Institute
| | | |
Collapse
|
15
|
刘 煜, 黄 沙, 姚 斌, 李 曌, 李 想, 付 小, 吴 旭. [Three-dimensional bioprinted microstructure promotes proliferation and viability of murine epithelial stem cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:761-766. [PMID: 28669949 PMCID: PMC6744142 DOI: 10.3969/j.issn.1673-4254.2017.06.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To evaluate the effect of different microstructures prepared by three-dimensional (3D) bioprinting on proliferation and viability of the murine epithelial stem cells in vitro. METHODS 3D cell-laden microstructures were constructed using 3 different printing nozzles with diameters of 210, 340, and 420 µm. Fluorescence microscopy and the live/dead assay kit were used to observe the proliferation and viability of the murine epithelial stem cells in the microstructures. RESULTS All the 3D cell-laden micro-structures were capable of promoting the proliferation of murine epithelial stem cells. In the 3 groups of micro-structures, the cell viability decreased significantly with time until 7 days after printing (P<0.01), but at 14 days after the printing, the cell viability increased significantly as compared with that at 7 days (P<0.01). The viability of the cells was significantly higher in the microstructure printed using a 420 µm nozzle than in the microstructures printed with 210 µm and 340 µm nozzles (P<0.01). CONCLUSION The microstructure printed with a 420 µm nozzle can stably promote the proliferation of murine epithelial stem cells and maintain a high level of cell viability, suggesting the feasibility of constructing tissue-engineered epidermis and full-thickness skin graft using 3D bioprinting technique.
Collapse
Affiliation(s)
- 煜凡 刘
- 南方医科大学,广东 广州 510515Southern Medical University, Guangzhou 510515, China
- 南方医科大学南方医院胸外科,广东 广州 510515Southern Medical University, Guangzhou 510515, China
| | - 沙 黄
- 解放军总医院基础医学研究所,北京 100853Institute of Basic Medical Sciences, General Hospital of PLA, Beijing 100853, China
| | - 斌 姚
- 解放军总医院第一附属医院全军创伤修复与组织再生重点实验室暨皮肤损伤修复与组织再生北京市重点实验室,北京 100048Key Laboratory of Tissue Repair and Regeneration of PLA, and Beijing Key Laboratory of Skin Injury, Repair and Regeneration, First Hospital Affiliated to General Hospital of PLA, Beijing 100048, China
- 南开大学医学院,天津 300071School of Medicine, Nankai University, Tianjin 300071, China
| | - 曌 李
- 解放军总医院第一附属医院全军创伤修复与组织再生重点实验室暨皮肤损伤修复与组织再生北京市重点实验室,北京 100048Key Laboratory of Tissue Repair and Regeneration of PLA, and Beijing Key Laboratory of Skin Injury, Repair and Regeneration, First Hospital Affiliated to General Hospital of PLA, Beijing 100048, China
| | - 想 李
- 南方医科大学,广东 广州 510515Southern Medical University, Guangzhou 510515, China
- 南方医科大学南方医院胸外科,广东 广州 510515Southern Medical University, Guangzhou 510515, China
| | - 小兵 付
- 解放军总医院第一附属医院全军创伤修复与组织再生重点实验室暨皮肤损伤修复与组织再生北京市重点实验室,北京 100048Key Laboratory of Tissue Repair and Regeneration of PLA, and Beijing Key Laboratory of Skin Injury, Repair and Regeneration, First Hospital Affiliated to General Hospital of PLA, Beijing 100048, China
| | - 旭 吴
- 南方医科大学,广东 广州 510515Southern Medical University, Guangzhou 510515, China
- 南方医科大学南方医院胸外科,广东 广州 510515Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
16
|
Mattei G, Magliaro C, Pirone A, Ahluwalia A. Decellularized Human Liver Is Too Heterogeneous for Designing a Generic Extracellular Matrix Mimic Hepatic Scaffold. Artif Organs 2017; 41:E347-E355. [PMID: 28543403 DOI: 10.1111/aor.12925] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/24/2016] [Accepted: 01/04/2017] [Indexed: 12/12/2022]
Abstract
Decellularized human livers are considered the perfect extracellular matrix (ECM) surrogate because both three-dimensional architecture and biological features of the hepatic microenvironment are thought to be preserved. However, donor human livers are in chronically short supply, both for transplantation or as decellularized scaffolds, and will become even scarcer as life expectancy increases. It is hence of interest to determine the structural and biochemical properties of human hepatic ECM to derive design criteria for engineering biomimetic scaffolds. The intention of this work was to obtain quantitative design specifications for fabricating scaffolds for hepatic tissue engineering using human livers as a template. To this end, hepatic samples from five patients scheduled for hepatic resection were decellularized using a protocol shown to reproducibly conserve matrix composition and microstructure in porcine livers. The decellularization outcome was evaluated through histological and quantitative image analyses to evaluate cell removal, protein, and glycosaminoglycan content per unit area. Applying the same decellularization protocol to human liver samples obtained from five different patients yielded five different outcomes. Only one liver out of five was completely decellularized, while the other four showed different levels of remaining cells and matrix. Moreover, protein and glycosaminoglycan content per unit area after decellularization were also found to be patient- (or donor-) dependent. This donor-to-donor variability of human livers thus precludes their use as templates for engineering a generic "one-size fits all" ECM-mimic hepatic scaffold.
Collapse
Affiliation(s)
| | | | - Andrea Pirone
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | | |
Collapse
|
17
|
Li B, Ren K, Zhang H, Jia F, Wang J, Chang H, Wang Y, Ji J. Nanostructured Multilayer Films Assembled from Poly(dopamine)‐Coated Carbon Nanotubes for Controlling Cell Behavior. CHEMNANOMAT 2017; 3:319-327. [DOI: 10.1002/cnma.201700024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
AbstractNano‐topographic surfaces have been used as an effective tool to control cell behavior such as adhesion and proliferation. In this study, multilayer films with nano‐topographic features were fabricated by alternatively assembling poly(l‐lysine) (PLL) and poly(dopamine)‐coated carbon nanotubes (CNTs@PDA) layers. The growth of PLL/CNTs@PDA film presented a perfect linear relationship with the number of bilayers. A nanostructured morphology with interpenetrating CNT networks was observed by scanning electron microscopy (SEM). Adhesion and proliferation of endothelial cells (ECs) and smooth muscle cells (SMCs) on the PLL/CNTs@PDA multilayer films have been evaluated. The films support initial adhesion of both ECs and SMCs. Interestingly, the PLL/CNTs@PDA multilayer films were found to promote proliferation of SMCs and inhibited proliferation of ECs. Further, pheochromocytoma (PC12) cells were employed to evaluate the influence of PLL/CNTs@PDA multilayer films on the outgrowth of synapses. We found that the nanostructured surface significantly promoted the synapses of PC12 cell growth and formation. Our findings suggest that cytophilic surfaces with the nanostructured morphology have diverse effects on different cells, which sheds light on new design of biomaterial surfaces in cell‐based applications.
Collapse
Affiliation(s)
- Bo‐chao Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 P.R. China
| | - Ke‐feng Ren
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 P.R. China
| | - He Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 P.R. China
| | - Fan Jia
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 P.R. China
| | - Jin‐lei Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 P.R. China
| | - Hao Chang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 P.R. China
| | - Yun‐bing Wang
- National Engineering Research Center for Biomaterials Sichuan University Chengdu 610064 P.R. China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 P.R. China
| |
Collapse
|
18
|
Narmada BC, Goh YT, Li H, Sinha S, Yu H, Cheung C. Human Stem Cell-Derived Endothelial-Hepatic Platform for Efficacy Testing of Vascular-Protective Metabolites from Nutraceuticals. Stem Cells Transl Med 2017; 6:851-863. [PMID: 28297582 PMCID: PMC5442778 DOI: 10.5966/sctm.2016-0129] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/09/2016] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis underlies many cardiovascular and cerebrovascular diseases. Nutraceuticals are emerging as a therapeutic moiety for restoring vascular health. Unlike small-molecule drugs, the complexity of ingredients in nutraceuticals often confounds evaluation of their efficacy in preclinical evaluation. It is recognized that the liver is a vital organ in processing complex compounds into bioactive metabolites. In this work, we developed a coculture system of human pluripotent stem cell-derived endothelial cells (hPSC-ECs) and human pluripotent stem cell-derived hepatocytes (hPSC-HEPs) for predicting vascular-protective effects of nutraceuticals. To validate our model, two compounds (quercetin and genistein), known to have anti-inflammatory effects on vasculatures, were selected. We found that both quercetin and genistein were ineffective at suppressing inflammatory activation by interleukin-1β owing to limited metabolic activity of hPSC-ECs. Conversely, hPSC-HEPs demonstrated metabolic capacity to break down both nutraceuticals into primary and secondary metabolites. When hPSC-HEPs were cocultured with hPSC-ECs to permit paracrine interactions, the continuous turnover of metabolites mitigated interleukin-1β stimulation on hPSC-ECs. We observed significant reductions in inflammatory gene expressions, nuclear translocation of nuclear factor κB, and interleukin-8 production. Thus, integration of hPSC-HEPs could accurately reproduce systemic effects involved in drug metabolism in vivo to unravel beneficial constituents in nutraceuticals. This physiologically relevant endothelial-hepatic platform would be a great resource in predicting the efficacy of complex nutraceuticals and mechanistic interrogation of vascular-targeting candidate compounds. Stem Cells Translational Medicine 2017;6:851-863.
Collapse
Affiliation(s)
| | - Yeek Teck Goh
- Institute of Molecular and Cell Biology, Proteos, Singapore
| | - Huan Li
- Institute of Bioengineering and Nanotechnology, Nanos, Singapore
| | - Sanjay Sinha
- The Anne McLaren Laboratory of Regenerative Medicine, Wellcome Trust‐Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Division of Cardiovascular Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, Nanos, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Mechanobiology Institute, Singapore
- Singapore‐MIT Alliance for Research and Technology, BioSyM, Singapore
| | - Christine Cheung
- Institute of Molecular and Cell Biology, Proteos, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
19
|
Liu Y, Wang S, Wang Y. Patterned Fibers Embedded Microfluidic Chips Based on PLA and PDMS for Ag Nanoparticle Safety Testing. Polymers (Basel) 2016; 8:E402. [PMID: 30974676 PMCID: PMC6431932 DOI: 10.3390/polym8110402] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 01/26/2023] Open
Abstract
A new method to integrate poly-dl-lactide (PLA) patterned electrospun fibers with a polydimethylsiloxane (PDMS) microfluidic chip was successfully developed via lithography. Hepatocyte behavior under static and dynamic conditions was investigated. Immunohistochemical analyses indicated good hepatocyte survival under the dynamic culture system with effective hepatocyte spheroid formation in the patterned microfluidic chip vs. static culture conditions and tissue culture plate (TCP). In particular, hepatocytes seeded in this microfluidic chip under a flow rate of 10 μL/min could re-establish hepatocyte polarity to support biliary excretion and were able to maintain high levels of albumin and urea secretion over 15 days. Furthermore, the optimized system could produce sensitive and consistent responses to nano-Ag-induced hepatotoxicity during culture. Thus, this microfluidic chip device provides a new means of fabricating complex liver tissue-engineered scaffolds, and may be of considerable utility in the toxicity screening of nanoparticles.
Collapse
Affiliation(s)
- Yaowen Liu
- College of Food Science, Sichuan Agricultural University, Yaan 625014, China.
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Shuyao Wang
- College of Food Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Yihao Wang
- College of Food Science, Sichuan Agricultural University, Yaan 625014, China.
| |
Collapse
|
20
|
Yang R, Wu L, Chen J, Chen W, Zhang L, Zhang L, You R, Yin L, Li CH, Guan YQ. Effects of Differentiation and Antisenescence from BMSCs to Hepatocy-Like Cells of the PAAm-IGF-1/TNF-α Biomaterial. ACS APPLIED MATERIALS & INTERFACES 2016; 8:26638-26647. [PMID: 27668443 DOI: 10.1021/acsami.6b10377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Aiming at the cells' differentiation phenomenon and senescence problem in liver tissue engineering, this work is designed to synthesize three different chargeable polymers (polypropylene acid (PAAc), polyethylene glycol (PEG), and polypropylene amine (PAAm)) coimmobilized by the insulin-like growth factor 1 (IGF-1) and tumor necrosis factor-α (TNF-α). We explore the hepatocyte differentiation effect and the antisenecence effect of PSt-PAAm-IGF-1/TNF-α biomaterial which was selected from the three different chargeable polymers in bone marrow mesenchymal stem cells (BMSCs). Our work will establish a model for studying the biochemical molecular regulation mechanism and signal transduction pathway of cell senescence in liver tissue engineering, which provide a molecular basis for developing biomaterials for liver tissue engineering.
Collapse
Affiliation(s)
- Runcai Yang
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Lifang Wu
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Jiehong Chen
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Wuya Chen
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Lin Zhang
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Li Zhang
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Rong You
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Liang Yin
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Chu-Hua Li
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University , Guangzhou 510631, China
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University , Guangzhou 510631, China
| |
Collapse
|
21
|
Kanninen LK, Harjumäki R, Peltoniemi P, Bogacheva MS, Salmi T, Porola P, Niklander J, Smutný T, Urtti A, Yliperttula ML, Lou YR. Laminin-511 and laminin-521-based matrices for efficient hepatic specification of human pluripotent stem cells. Biomaterials 2016; 103:86-100. [DOI: 10.1016/j.biomaterials.2016.06.054] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 06/21/2016] [Indexed: 12/11/2022]
|
22
|
Integrated Circuit-Based Biofabrication with Common Biomaterials for Probing Cellular Biomechanics. Trends Biotechnol 2016; 34:171-186. [DOI: 10.1016/j.tibtech.2015.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/03/2015] [Accepted: 11/18/2015] [Indexed: 01/10/2023]
|
23
|
Ma C, Zhao L, Zhou EM, Xu J, Shen S, Wang J. On-Chip Construction of Liver Lobule-like Microtissue and Its Application for Adverse Drug Reaction Assay. Anal Chem 2016; 88:1719-27. [DOI: 10.1021/acs.analchem.5b03869] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Chao Ma
- Colleges of Veterinary Medicine and Science, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Lei Zhao
- Colleges of Veterinary Medicine and Science, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - En-Min Zhou
- Colleges of Veterinary Medicine and Science, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Juan Xu
- Colleges of Veterinary Medicine and Science, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Shaofei Shen
- Colleges of Veterinary Medicine and Science, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Jinyi Wang
- Colleges of Veterinary Medicine and Science, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| |
Collapse
|
24
|
3D bioprinting of tissues and organs. Nat Biotechnol 2015; 32:773-85. [PMID: 25093879 DOI: 10.1038/nbt.2958] [Citation(s) in RCA: 3677] [Impact Index Per Article: 367.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 06/12/2014] [Indexed: 02/07/2023]
Abstract
Additive manufacturing, otherwise known as three-dimensional (3D) printing, is driving major innovations in many areas, such as engineering, manufacturing, art, education and medicine. Recent advances have enabled 3D printing of biocompatible materials, cells and supporting components into complex 3D functional living tissues. 3D bioprinting is being applied to regenerative medicine to address the need for tissues and organs suitable for transplantation. Compared with non-biological printing, 3D bioprinting involves additional complexities, such as the choice of materials, cell types, growth and differentiation factors, and technical challenges related to the sensitivities of living cells and the construction of tissues. Addressing these complexities requires the integration of technologies from the fields of engineering, biomaterials science, cell biology, physics and medicine. 3D bioprinting has already been used for the generation and transplantation of several tissues, including multilayered skin, bone, vascular grafts, tracheal splints, heart tissue and cartilaginous structures. Other applications include developing high-throughput 3D-bioprinted tissue models for research, drug discovery and toxicology.
Collapse
|
25
|
Neiman JAS, Raman R, Chan V, Rhoads MG, Raredon MSB, Velazquez JJ, Dyer RL, Bashir R, Hammond PT, Griffith LG. Photopatterning of hydrogel scaffolds coupled to filter materials using stereolithography for perfused 3D culture of hepatocytes. Biotechnol Bioeng 2015; 112:777-87. [PMID: 25384798 DOI: 10.1002/bit.25494] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 11/05/2014] [Indexed: 12/17/2022]
Abstract
In vitro models that recapitulate the liver's structural and functional complexity could prolong hepatocellular viability and function to improve platforms for drug toxicity studies and understanding liver pathophysiology. Here, stereolithography (SLA) was employed to fabricate hydrogel scaffolds with open channels designed for post-seeding and perfused culture of primary hepatocytes that form 3D structures in a bioreactor. Photopolymerizable polyethylene glycol-based hydrogels were fabricated coupled to chemically activated, commercially available filters (polycarbonate and polyvinylidene fluoride) using a chemistry that permitted cell viability, and was robust enough to withstand perfused culture of up to 1 µL/s for at least 7 days. SLA energy dose, photoinitiator concentrations, and pretreatment conditions were screened to determine conditions that maximized cell viability and hydrogel bonding to the filter. Multiple open channel geometries were readily achieved, and included ellipses and rectangles. Rectangular open channels employed for subsequent studies had final dimensions on the order of 350 µm by 850 µm. Cell seeding densities and flow rates that promoted cell viability were determined. Perfused culture of primary hepatocytes in hydrogel scaffolds in the presence of soluble epidermal growth factor (EGF) prolonged the maintenance of albumin production throughout the 7-day culture relative to 2D controls. This technique of bonding hydrogel scaffolds can be employed to fabricate soft scaffolds for a number of bioreactor configurations and applications.
Collapse
Affiliation(s)
- Jaclyn A Shepard Neiman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wang X, Jiao Q, Zhang S, Ye Z, Zhou Y, Tan WS. Perfusion culture-induced template-assisted assembling of cell-laden microcarriers is a promising route for fabricating macrotissues. Biotechnol J 2014; 9:1425-34. [DOI: 10.1002/biot.201400238] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/23/2014] [Accepted: 09/05/2014] [Indexed: 11/06/2022]
|
27
|
Abstract
Interest in "engineering liver" arises from multiple communities: therapeutic replacement; mechanistic models of human processes; and drug safety and efficacy studies. An explosion of micro- and nanofabrication, biomaterials, microfluidic, and other technologies potentially affords unprecedented opportunity to create microphysiological models of the human liver, but engineering design principles for how to deploy these tools effectively toward specific applications, including how to define the essential constraints of any given application (available sources of cells, acceptable cost, and user-friendliness), are still emerging. Arguably less appreciated is the parallel growth in computational systems biology approaches toward these same problems-particularly in parsing complex disease processes from clinical material, building models of response networks, and in how to interpret the growing compendium of data on drug efficacy and toxicology in patient populations. Here, we provide insight into how the complementary paths of engineering liver-experimental and computational-are beginning to interplay toward greater illumination of human disease states and technologies for drug development.
Collapse
Affiliation(s)
- Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | | | | |
Collapse
|
28
|
|
29
|
Ananthanarayanan A, Nugraha B, Triyatni M, Hart S, Sankuratri S, Yu H. Scalable spheroid model of human hepatocytes for hepatitis C infection and replication. Mol Pharm 2014; 11:2106-14. [PMID: 24761996 DOI: 10.1021/mp500063y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Developing effective new drugs against hepatitis C (HCV) virus has been challenging due to the lack of appropriate small animal and in vitro models recapitulating the entire life cycle of the virus. Current in vitro models fail to recapitulate the complexity of human liver physiology. Here we present a method to study HCV infection and replication on spheroid cultures of Huh 7.5 cells and primary human hepatocytes. Spheroid cultures are constructed using a galactosylated cellulosic sponge with homogeneous macroporosity, enabling the formation and maintenance of uniformly sized spheroids. This facilitates easy handling of the tissue-engineered constructs and overcomes limitations inherent of traditional spheroid cultures. Spheroids formed in the galactosylated cellulosic sponge show enhanced hepatic functions in Huh 7.5 cells and maintain liver-specific functions of primary human hepatocytes for 2 weeks in culture. Establishment of apical and basolateral polarity along with the expression and localization of all HCV specific entry proteins allow for a 9-fold increase in viral entry in spheroid cultures over conventional monolayer cultures. Huh 7.5 cells cultured in the galactosylated cellulosic sponge also support replication of the HCV clone, JFH (Japanese fulminant hepatitis)-1 at higher levels than in monolayer cultures. The advantages of our system in maintaining liver-specific functions and allowing HCV infection together with its ease of handling make it suitable for the study of HCV biology in basic research and pharmaceutical R&D.
Collapse
|
30
|
Ebrahimkhani MR, Neiman JAS, Raredon MSB, Hughes DJ, Griffith LG. Bioreactor technologies to support liver function in vitro. Adv Drug Deliv Rev 2014; 69-70:132-57. [PMID: 24607703 PMCID: PMC4144187 DOI: 10.1016/j.addr.2014.02.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/18/2014] [Accepted: 02/24/2014] [Indexed: 02/08/2023]
Abstract
Liver is a central nexus integrating metabolic and immunologic homeostasis in the human body, and the direct or indirect target of most molecular therapeutics. A wide spectrum of therapeutic and technological needs drives efforts to capture liver physiology and pathophysiology in vitro, ranging from prediction of metabolism and toxicity of small molecule drugs, to understanding off-target effects of proteins, nucleic acid therapies, and targeted therapeutics, to serving as disease models for drug development. Here we provide perspective on the evolving landscape of bioreactor-based models to meet old and new challenges in drug discovery and development, emphasizing design challenges in maintaining long-term liver-specific function and how emerging technologies in biomaterials and microdevices are providing new experimental models.
Collapse
Affiliation(s)
- Mohammad R Ebrahimkhani
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jaclyn A Shepard Neiman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Micha Sam B Raredon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
31
|
Schotanus BA, Penning LC, Spee B. Potential of regenerative medicine techniques in canine hepatology. Vet Q 2014; 33:207-16. [PMID: 24422896 DOI: 10.1080/01652176.2013.875240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Liver cell turnover is very slow, especially compared to intestines and stomach epithelium and hair cells. Since the liver is the main detoxifying organ in the body, it does not come as a surprise that the liver has an unmatched regenerative capacity. After 70% partial hepatectomy, the liver size returns to normal in about two weeks due to replication of differentiated hepatocytes and cholangiocytes. Despite this, liver diseases are regularly encountered in the veterinary clinic. Dogs primarily present with parenchymal pathologies such as hepatitis. The estimated frequency of canine hepatitis depends on the investigated population and accounts for 1%-2% of our university clinic referral population, and up to 12% in a general population. In chronic and severe acute liver disease, the regenerative and replicative capacity of the hepatocytes and/or cholangiocytes falls short and the liver is not restored. In this situation, proliferation of hepatic stem cells or hepatic progenitor cells (HPCs), on histology called the ductular reaction, comes into play to replace the damaged hepatocytes or cholangiocytes. For unknown reasons the ductular reaction is often too little and too late, or differentiation into fully differentiated hepatocytes or cholangiocytes is hampered. In this way, HPCs fail to fully regenerate the liver. The presence and potential of HPCs does, however, provide great prospectives for their use in regenerative strategies. This review highlights the regulation of, and the interaction between, HPCs and other liver cell types and discusses potential regenerative medicine-oriented strategies in canine hepatitis, making use of (liver) stem cells.
Collapse
Affiliation(s)
- Baukje A Schotanus
- a Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine , Utrecht University , Utrecht , The Netherlands
| | | | | |
Collapse
|
32
|
Abstract
Liver extracellular matrix (ECM) composition, topography and biomechanical properties influence cell-matrix interactions. The ECM presents guiding cues for hepatocyte phenotype maintenance, differentiation and proliferation both in vitro and in vivo. Current understanding of such cell-guiding cues along with advancement of techniques for scaffold fabrication has led to evolution of matrices for liver tissue culture from simple porous scaffolds to more complex 3D matrices with microarchitecture similar to in vivo. Natural and synthetic polymeric biomaterials fabricated in different topographies and porous matrices have been used for hepatocyte culture. Heterotypic and homotypic cell interactions are necessary for developing an adult liver as well as an artificial liver. A high oxygen demand of hepatocytes as well as graded oxygen distribution in liver is another challenging attribute of the normal liver architecture that further adds to the complexity of engineered substrate design. A balanced interplay of cell-matrix interactions along with cell-cell interactions and adequate supply of oxygen and nutrient determines the success of an engineered substrate for liver cells. Techniques devised to incorporate these features of hepatic function and mimic liver architecture range from maintaining liver cells in mm-sized tailor-made scaffolds to a more bottoms up approach that starts from building the microscopic subunit of the whole tissue. In this review, we discuss briefly various biomaterials used for liver tissue engineering with respect to design parameters such as scaffold composition and chemistry, biomechanical properties, topography, cell-cell interactions and oxygenation.
Collapse
Affiliation(s)
- Era Jain
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India.,Biomedical Engineering Department, St. Louis University, St. Louis, MO, USA
| | - Apeksha Damania
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India.
| |
Collapse
|
33
|
Tamai M, Adachi E, Tagawa YI. Characterization of a liver organoid tissue composed of hepatocytes and fibroblasts in dense collagen fibrils. Tissue Eng Part A 2013; 19:2527-35. [PMID: 23815236 DOI: 10.1089/ten.tea.2012.0704] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The adult liver is wrapped in a connective tissue sheet called the liver capsule, which consists of collagen fibrils and fibroblasts. In this study, we set out to construct a liver organoid tissue that would be comparable to the endogenous liver, using a bioreactor. In vitro liver organoid tissue was generated by combining collagen fibrils, fibroblasts, and primary murine hepatocytes or Hep G2 on a mesh of poly-lactic acid fabric using a bioreactor. Then, the suitability of this liver organoid tissue for transplantation was tested by implanting the constructs into partially hepatectomized BALB/cA-nu/nu mice. As determined by using scanning and transmission electron microscopes, the liver organoid tissues were composed of densely packed collagen fibrils with fibroblasts and aggregates of oval or spherical hepatocytes. Angiogenesis was induced after the transplantation, and blood vessels connected the liver organoid tissue with the surrounding tissue. Thus, a novel approach was applied to generate transplantable liver organoid tissue within a condensed collagen fibril matrix. These results suggested that a dense collagen network populated with fibroblasts can hold a layer of concentrated hepatocytes, providing a three-dimensional microenvrionment suitable for the reestablishment of cell-cell and cell-extracellular matrix (ECM) interactions, and resulting in the maintenance of their liver-specific functions. This liver organoid tissue may be useful for the study of intrahepatic functions of various cells, cytokines, and ECMs, and may fulfill the fundamental requirements of a donor tissue.
Collapse
Affiliation(s)
- Miho Tamai
- 1 Department of Biomolecular Engineering, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology , Kanagawa, Japan
| | | | | |
Collapse
|
34
|
Yang N, Yang MK, Bi SX, Chen L, Zhu ZY, Gao YT, Du Z. Cells behaviors and genotoxicity on topological surface. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2013; 33:3465-3473. [PMID: 23706235 DOI: 10.1016/j.msec.2013.04.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 02/20/2013] [Accepted: 04/16/2013] [Indexed: 12/11/2022]
Abstract
To investigate different cells behaviors and genotoxicity, which were driven by specific microenvironments, three patterned surfaces (pillars, wide grooves and narrow grooves) and one smooth surface were prepared by template-based technique. Vinculin is a membrane-cytoskeletal protein in focal adhesion plaques and associates with cell-cell and cell-matrix junctions, which can promote cell adhesion and spreading. The immunofluorescence staining of vinculin revealed that the narrow grooves patterned substrate was favorable for L929 cell adhesion. For cell multiplication, the narrow grooves surface was fitted for the proliferation of L929, L02 and MSC cells, the pillars surface was only in favor of L929 cells to proliferate during 7 days of cell cultivation. Cell genetic toxicity was evaluated by cellular micronuclei test (MNT). The results indicated that topological surfaces were more suitable for L929 cells to proliferate and maintain the stability of genome. On the contrary, the narrow grooves surface induced higher micronuclei ratio of L02 and MSC cells than other surfaces. With the comprehensive results of cell multiplication and MNT, it was concluded that the wide grooves surface was best fitted for L02 cells to proliferate and have less DNA damages, and the smooth surface was optimum for the research of MSC cells in vitro.
Collapse
Affiliation(s)
- N Yang
- Tianjin Key Laboratory of Fiber Modification and Functional Fiber, School of Materials Science and Engineering, Tianjin Polytechnic University, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Palakkan AA, Hay DC, Anil Kumar PR, Kumary TV, Ross JA. Liver tissue engineering and cell sources: issues and challenges. Liver Int 2013; 33:666-76. [PMID: 23490085 DOI: 10.1111/liv.12134] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 01/27/2013] [Indexed: 02/13/2023]
Abstract
Liver diseases are of major concern as they now account for millions of deaths annually. As a result of the increased incidence of liver disease, many patients die on the transplant waiting list, before a donor organ becomes available. To meet the huge demand for donor liver, alternative approaches using liver tissue engineering principles are being actively pursued. Even though adult hepatocytes, the primary cells of the liver are most preferred for tissue engineering of liver, their limited availability, isolation from diseased organs, lack of in vitro propagation and deterioration of function acts as a major drawback to their use. Various approaches have been taken to prevent the functional deterioration of hepatocytes including the provision of an adequate extracellular matrix and co-culture with non-parenchymal cells of liver. Great progress has also been made to differentiate human stem cells to hepatocytes and to use them for liver tissue engineering applications. This review provides an overview of recent challenges, issues and cell sources with regard to liver tissue engineering.
Collapse
Affiliation(s)
- Anwar A Palakkan
- Tissue Injury and Repair Group, University of Edinburgh - MRC Centre for Regenerative Medicine, Edinburgh, UK
| | | | | | | | | |
Collapse
|
36
|
Wang T, Feng ZQ, Leach MK, Wu J, Jiang Q. Nanoporous fibers of type-I collagen coated poly(l-lactic acid) for enhancing primary hepatocyte growth and function. J Mater Chem B 2013; 1:339-346. [DOI: 10.1039/c2tb00195k] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
37
|
Fraczek J, Bolleyn J, Vanhaecke T, Rogiers V, Vinken M. Primary hepatocyte cultures for pharmaco-toxicological studies: at the busy crossroad of various anti-dedifferentiation strategies. Arch Toxicol 2012; 87:577-610. [PMID: 23242478 DOI: 10.1007/s00204-012-0983-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 11/19/2012] [Indexed: 01/24/2023]
Abstract
Continuously increasing understanding of the molecular triggers responsible for the onset of diseases, paralleled by an equally dynamic evolution of chemical synthesis and screening methods, offers an abundance of pharmacological agents with a potential to become new successful drugs. However, before patients can benefit of newly developed pharmaceuticals, stringent safety filters need to be applied to weed out unfavourable drug candidates. Cost effectiveness and the need to identify compound liabilities, without exposing humans to unnecessary risks, has stimulated the shift of the safety studies to the earliest stages of drug discovery and development. In this regard, in vivo relevant organotypic in vitro models have high potential to revolutionize the preclinical safety testing. They can enable automation of the process, to match the requirements of high-throughput screening approaches, while satisfying ethical considerations. Cultures of primary hepatocytes became already an inherent part of the preclinical pharmaco-toxicological testing battery, yet their routine use, particularly for long-term assays, is limited by the progressive deterioration of liver-specific features. The availability of suitable hepatic and other organ-specific in vitro models is, however, of paramount importance in the light of changing European legal regulations in the field of chemical compounds of different origin, which gradually restrict the use of animal studies for safety assessment, as currently witnessed in cosmetic industry. Fortunately, research groups worldwide spare no effort to establish hepatic in vitro systems. In the present review, both classical and innovative methodologies to stabilize the in vivo-like hepatocyte phenotype in culture of primary hepatocytes are presented and discussed.
Collapse
Affiliation(s)
- J Fraczek
- Department of Toxicology, Faculty of Medicine and Pharmacy, Centre for Pharmaceutical Research, Vrije Universiteit Brussel, Belgium.
| | | | | | | | | |
Collapse
|
38
|
Skardal A, Smith L, Bharadwaj S, Atala A, Soker S, Zhang Y. Tissue specific synthetic ECM hydrogels for 3-D in vitro maintenance of hepatocyte function. Biomaterials 2012; 33:4565-75. [PMID: 22475531 DOI: 10.1016/j.biomaterials.2012.03.034] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 03/10/2012] [Indexed: 01/19/2023]
Abstract
Despite recent advances in biomaterial science, there is yet no culture system that supports long-term culture expansion of human adult hepatocytes, while preserving continued function. Previous studies suggested that acellular liver extracellular matrix (ECM), employed as a substrate, improved proliferation and function of liver cells. Here we investigated whether extracts prepared from acellular liver ECM (liver ECM extract, LEE), or from whole (fresh) liver tissue (liver tissue extract, LTE), could be combined with collagen Type I, hyaluronic acid (HA), or heparin-conjugated HA (HP) hydrogels to enhance survival and functional output of primary human hepatocytes. The liver-specific semi-synthetic ECMs (sECMs) were prepared by incorporating LEE or LTE into the gel matrices. Subsequently, primary human hepatocytes were maintained in sandwich-style hydrogel cultures for 4 weeks. Progressive increase in hepatocyte metabolism was observed in all HA and HP groups. Hepatocytes cultured in HA and HP hydrogels containing LEE or LTE synthesized and secreted steady levels of albumin and urea and sustained cytochrome p450-dependent drug metabolism of ethoxycoumarin. Collectively, these results indicate that customized HA hydrogels with liver-specific ECM components may be an efficient method for expansion human hepatocytes in vitro for cell therapy and drug and toxicology screening purposes.
Collapse
Affiliation(s)
- Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC 27101, USA
| | | | | | | | | | | |
Collapse
|
39
|
NeuZ̆il P, Xu Y, Manz A. Trends and Perspectives. MICRODROPLET TECHNOLOGY 2012. [PMCID: PMC7122354 DOI: 10.1007/978-1-4614-3265-4_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Throughout the book chapters, researchers have highlighted the recent advancement in microfluidic areas, particularly those involving microdroplets.
Collapse
|
40
|
Kim SJ, Ise H, Goto M, Akaike T. Interactions of vimentin- or desmin-expressing liver cells with N-acetylglucosamine-bearing polymers. Biomaterials 2011; 33:2154-64. [PMID: 22177839 DOI: 10.1016/j.biomaterials.2011.11.084] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 11/28/2011] [Indexed: 10/14/2022]
Abstract
It is necessary to develop highly functionalized liver cell culture systems for liver tissue engineering such as bioartificial livers and liver cell chips. To maintain a high level of hepatocyte function, well-organized patterning culture systems of hepatocytes and nonparenchymal cells would be advantageous. To design the patterning culture system using these cells, cell-recognizable polymers should be useful to regulate not only the hepatocytes, but also the nonparenchymal cells. Here, we report that N-acetylglucosamine (GlcNAc)-bearing polymers are useful as nonparenchymal cell-recognizable polymers. It has previously been reported that mesenchymal cells adhered to GlcNAc-bearing polymer-coated dishes through surface vimentin. It was also observed that nonparenchymal cells expressing vimentin or desmin specifically adhered to GlcNAc-bearing polymer-coated dishes. Especially, in hepatic stellate cells (HSCs) cultured on GlcNAc-bearing polymer-coated dishes, the expression of α-smooth muscle actin as an activated HSCs marker was suppressed in long-term. Therefore, HSCs were shown to maintain a quiescent state on PVGlcNAc-coated dishes during a long-term culture. These results demonstrated that GlcNAc-bearing polymers could be beneficial to culture nonparenchymal cells such as HSCs. Our findings suggest that galactose- and GlcNAc-bearing polymers can regulate the culture of all liver cells and may be useful tools for the establishment of liver tissue engineering.
Collapse
Affiliation(s)
- Sun-Jung Kim
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | | | | | | |
Collapse
|
41
|
Delgado-Coello B, Briones-Orta MA, Macías-Silva M, Mas-Oliva J. Cholesterol: recapitulation of its active role during liver regeneration. Liver Int 2011; 31:1271-84. [PMID: 21745289 DOI: 10.1111/j.1478-3231.2011.02542.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Liver regeneration is a compensatory hyperplasia produced by several stimuli that promotes proliferation in order to provide recovery of the liver mass and architecture. This process involves complex signalling cascades that receive feedback from autocrine and paracrine pathways, recognized by parenchymal as well as non-parenchymal cells. Nowadays the dynamic role of lipids in biological processes is widely recognized; however, a systematic analysis of their importance during liver regeneration is still missing. Therefore, in this review we address the role of lipids including the bioactive ones such as sphingolipids, but with special emphasis on cholesterol. Cholesterol is not only considered as a structural component but also as a relevant lipid involved in the control of the intermediate metabolism of different liver cell types such as hepatocytes, hepatic stellate cells and Kupffer cells. Cholesterol plays a significant role at the level of specific membrane domains, as well as modulating the expression of sterol-dependent proteins. Moreover, several enzymes related to the catabolism of cholesterol and whose activity is down regulated are related to the protection of liver tissue from toxicity during the process of regeneration. This review puts in perspective the necessity to study and understand the basic mechanisms involving lipids during the process of liver regeneration. On the other hand, the knowledge acquired in this area in the past years, can be considered invaluable in order to provide further insights into processes such as general organogenesis and several liver-related pathologies, including steatosis and fibrosis.
Collapse
Affiliation(s)
- Blanca Delgado-Coello
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, DF Mexico
| | | | | | | |
Collapse
|