1
|
Feng L, Wang Y, Fu Y, Li T, He G. Stem Cell-Based Strategies: The Future Direction of Bioartificial Liver Development. Stem Cell Rev Rep 2024; 20:601-616. [PMID: 38170319 DOI: 10.1007/s12015-023-10672-5] [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] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
Acute liver failure (ALF) results from severe liver damage or end-stage liver disease. It is extremely fatal and causes serious health and economic burdens worldwide. Once ALF occurs, liver transplantation (LT) is the only definitive and recommended treatment; however, LT is limited by the scarcity of liver grafts. Consequently, the clinical use of bioartificial liver (BAL) has been proposed as a treatment strategy for ALF. Human primary hepatocytes are an ideal cell source for these methods. However, their high demand and superior viability prevent their widespread use. Hence, finding alternatives that meet the seed cell quality and quantity requirements is imperative. Stem cells with self-renewing, immunogenic, and differentiative capacities are potential cell sources. MSCs and its secretomes encompass a spectrum of beneficial properties, such as anti-inflammatory, immunomodulatory, anti-ROS (reactive oxygen species), anti-apoptotic, pro-metabolomic, anti-fibrogenesis, and pro-regenerative attributes. This review focused on the recent status and future directions of stem cell-based strategies in BAL for ALF. Additionally, we discussed the opportunities and challenges associated with promoting such strategies for clinical applications.
Collapse
Affiliation(s)
- Lei Feng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550000, Guizhou, China.
| | - Yi Wang
- Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Yu Fu
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Ting Li
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510140, Guangdong, China.
| | - Guolin He
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
2
|
Borges MF, Maurmann N, Pranke P. Easy-to-Assembly System for Decellularization and Recellularization of Liver Grafts in a Bioreactor. MICROMACHINES 2023; 14:449. [PMID: 36838149 PMCID: PMC9962055 DOI: 10.3390/mi14020449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/31/2023] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
Decellularization of organs creates an acellular scaffold, ideal for being repopulated by cells. In this work, a low-cost perfusion system was created to be used in the process of liver decellularization and as a bioreactor after recellularization. It consists of a glass chamber to house the organ coupled to a peristaltic pump to promote liquid flow through the organ vascular tree. The rats' liver decellularization was made with a solution of sodium dodecyl sulfate. The recellularization was made with 108 mesenchymal stromal/stem cells and cultivated for seven days. The decellularized matrices showed an absence of DNA while preserving the collagen and glycosaminoglycans quantities, confirming the efficiency of the process. The functional analyses showed a rise in lactate dehydrogenase levels occurring in the first days of the cultivation, suggesting that there is cell death in this period, which stabilized on the seventh day. Histological analysis showed conservation of the collagen web and some groups of cells next to the vessels. It was possible to establish a system for decellularization and a bioreactor to use for the recellularization method. It is easy to assemble, can be ready to use in little time and be easily sterilized.
Collapse
Affiliation(s)
- Maurício Felisberto Borges
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90610-000, Brazil
| | - Natasha Maurmann
- Postgraduate Program in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90050-170, Brazil
| | - Patricia Pranke
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90610-000, Brazil
| |
Collapse
|
3
|
Chen S, Zhu J, Xue J, Wang X, Jing P, Zhou L, Cui Y, Wang T, Gong X, Lü S, Long M. Numerical simulation of flow characteristics in a permeable liver sinusoid with leukocytes. Biophys J 2022; 121:4666-4678. [PMID: 36271623 PMCID: PMC9748252 DOI: 10.1016/j.bpj.2022.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/31/2022] [Accepted: 10/17/2022] [Indexed: 02/07/2023] Open
Abstract
Double-layered channels of sinusoid lumen and Disse space separated by fenestrated liver sinusoidal endothelial cells (LSECs) endow the unique mechanical environment of the liver sinusoid network, which further guarantees its biological function. It is also known that this mechanical environment changes dramatically under liver fibrosis and cirrhosis, including the reduced plasma penetration and metabolite exchange between the two flow channels and the reduced Disse space deformability. The squeezing of leukocytes through narrow sinusoid lumen also affects the mechanical environment of liver sinusoid. To date, the detailed flow-field profile of liver sinusoid is still far from clear due to experimental limitations. It also remains elusive whether and how the varied physical properties of the pathological liver sinusoid regulate the fluid flow characteristics. Here a numerical model based on the immersed boundary method was established, and the effects of Disse space and leukocyte elasticities, endothelium permeability, and sinusoidal stenosis degree on fluid flow as well as leukocyte trafficking were specified upon a mimic liver sinusoid structure. Results showed that endothelium permeability dominantly controlled the plasma penetration velocity across the endothelium, whereas leukocyte squeezing promoted local penetration and significantly regulated wall shear stress on hepatocytes, which was strongly related to the Disse space and leukocyte deformability. Permeability and elasticity cooperatively regulated the process of leukocytes trafficking through the liver sinusoid, especially for stiffer leukocytes. This study will offer new insights into deeper understanding of the elaborate mechanical features of liver sinusoid and corresponding biological function.
Collapse
Affiliation(s)
- Shenbao Chen
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jingchen Zhu
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jian Xue
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China; State Key Laboratory of Nonlinear Mechanics (LNM), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Xiaolong Wang
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Jing
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lüwen Zhou
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yuhong Cui
- Department of Mechanics, Tianjin University, Tianjin, China
| | - Tianhao Wang
- Department of Mechanics, Tianjin University, Tianjin, China
| | - Xiaobo Gong
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Shouqin Lü
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.
| | - Mian Long
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
4
|
Cao L, Zhao H, Qian M, Shao C, Zhang Y, Yang J. Construction of polysaccharide scaffold-based perfusion bioreactor supporting liver cell aggregates for drug screening. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2249-2269. [PMID: 35848470 DOI: 10.1080/09205063.2022.2102715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Rebuilding a suitable microenvironment of liver cells is the key challenge to enhancing the expression of hepatic functions for drug screening in vitro. To improve the microenvironment by providing the specific adhesive ligands for hepatocytes in the three-dimensional dynamic culture, a perfusion bioreactor with a pectin/alginate blend porous scaffold was constructed in this study. The galactosyl component in the main chain of pectin was able to be specifically recognized by the asialoglycoprotein receptor on the surface of hepatocytes, and subsequently promoted the adhesion and aggregation of hepatocytes co-cultured with hepatic non-parenchymal cells. The bioreactor was optimized for 4 h of dynamic inoculation followed by perfusion at a flow rate of 2 mL/min, which provided adequate oxygen supply and good mass transfer to the liver cells. During dynamic cultured in the bioreactor for 14 days, more multicellular aggregates were formed and were evenly distributed in the pectin/alginate blend scaffolds. The expressions of intercellular interaction and hepatic functions of the hepatocytes in aggregates were significantly enhanced in the three-dimensional dynamic group. Furthermore, the bioreactor not only markedly upregulated the cell polarity markers expression of hepatocytes but also enhanced their metabolic capacity to acetaminophen, isoniazid, and tolbutamide, which exhibited a significant concentration-dependent manner. Therefore, the pectin/alginate blend scaffold-based perfusion bioreactor appeared to be a promising candidate in the field of drug development and liver regeneration research.
Collapse
Affiliation(s)
- Lei Cao
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China.,Biological Sample Resource Sharing Center, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Huicun Zhao
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Mengyuan Qian
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Chuxiao Shao
- Department of Hepatopancreatobiliary Surgery, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Yan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
| | - Jun Yang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| |
Collapse
|
5
|
Tuerxun K, He J, Ibrahim I, Yusupu Z, Yasheng A, Xu Q, Tang R, Aikebaier A, Wu Y, Tuerdi M, Nijiati M, Zou X, Xu T. Bioartificial livers: a review of their design and manufacture. Biofabrication 2022; 14. [PMID: 35545058 DOI: 10.1088/1758-5090/ac6e86] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
Acute liver failure (ALF) is a rapidly progressive disease with high morbidity and mortality rates. Liver transplantation and artificial liver support systems, such as artificial livers (ALs) and bioartificial livers (BALs), are the two major therapies for ALF. Compared to ALs, BALs are composed of functional hepatocytes that provide essential liver functions, including detoxification, metabolite synthesis, and biotransformation. Furthermore, BALs can potentially provide effective support as a form of bridging therapy to liver transplantation or spontaneous recovery for patients with ALF. In this review, we systematically discussed the currently available state-of-the-art designs and manufacturing processes for BAL support systems. Specifically, we classified the cell sources and bioreactors that are applied in BALs, highlighted the advanced technologies of hepatocyte culturing and bioreactor fabrication, and discussed the current challenges and future trends in developing next generation BALs for large scale clinical applications.
Collapse
Affiliation(s)
- Kahaer Tuerxun
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Jianyu He
- Department of Mechanical Engineering, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing, Beijing, 100084, CHINA
| | - Irxat Ibrahim
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Zainuer Yusupu
- Department of Ultrasound, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Abudoukeyimu Yasheng
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Qilin Xu
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Ronghua Tang
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Aizemaiti Aikebaier
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Yuanquan Wu
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Maimaitituerxun Tuerdi
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Mayidili Nijiati
- Medical imaging center, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Xiaoguang Zou
- Hospital Organ, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Tao Xu
- Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing, 100084, CHINA
| |
Collapse
|
6
|
The adverse effects of hypoxia on hiHep functions via HIF-1α/PGC-1α axis are alleviated by PFDC emulsion. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2021.108152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
7
|
Wang J, Ren H, Liu Y, Sun L, Zhang Z, Zhao Y, Shi X. Bioinspired Artificial Liver System with hiPSC-Derived Hepatocytes for Acute Liver Failure Treatment. Adv Healthc Mater 2021; 10:e2101580. [PMID: 34599859 DOI: 10.1002/adhm.202101580] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/17/2021] [Indexed: 12/21/2022]
Abstract
Bioartificial liver (BAL) system has become a promising alternative to traditional liver transplantation in rescuing acute liver failure (ALF) patients. Herein, inspired by natural microstructure of hepatic lobules, a novel biomimetic bioartificial liver system (BBALS) is developed by integrating human induced pluripotent stem cell-derived hepatocytes (hiPSC-Heps) -laden microparticles and semipermeable microtubes into a microfluidic platform. As the working units are hepatic lobules-like semipermeable microtubes surrounding with serum-free suspension differentiated hiPSC-Heps microcarriers, the BBALS is endowed with functional cell aggregates and effective circulation system. Thus, the BBALS possesses high cell viability, favorable function regeneration, and effective substances exchange. Based on these features, a 3D liver chip with multiple parallel BBALS units is created for filtering the plasma of ALF rabbits, which validates the research significance and application potential of the proposed BBALS. Moreover, the novel integrated BBALS is applied to treat ALF rabbits and shows great advantages in increasing survival, generating serum proteins, and decreasing inflammation. These properties point to the broad prospects of BBALS in treating related diseases and improving traditional clinical methods.
Collapse
Affiliation(s)
- Jinglin Wang
- Department of Hepatobiliary Surgery the Affiliated Drum Tower Hospital of Nanjing University Medical School Hepatobiliary Institute of Nanjing University Nanjing 210008 China
| | - Haozhen Ren
- Department of Hepatobiliary Surgery the Affiliated Drum Tower Hospital of Nanjing University Medical School Hepatobiliary Institute of Nanjing University Nanjing 210008 China
| | - Yuxiao Liu
- Department of Hepatobiliary Surgery the Affiliated Drum Tower Hospital of Nanjing University Medical School Hepatobiliary Institute of Nanjing University Nanjing 210008 China
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
| | - Lingyu Sun
- Department of Hepatobiliary Surgery the Affiliated Drum Tower Hospital of Nanjing University Medical School Hepatobiliary Institute of Nanjing University Nanjing 210008 China
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
| | - Zhuohao Zhang
- Department of Hepatobiliary Surgery the Affiliated Drum Tower Hospital of Nanjing University Medical School Hepatobiliary Institute of Nanjing University Nanjing 210008 China
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
| | - Yuanjin Zhao
- Department of Hepatobiliary Surgery the Affiliated Drum Tower Hospital of Nanjing University Medical School Hepatobiliary Institute of Nanjing University Nanjing 210008 China
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
| | - Xiaolei Shi
- Department of Hepatobiliary Surgery the Affiliated Drum Tower Hospital of Nanjing University Medical School Hepatobiliary Institute of Nanjing University Nanjing 210008 China
| |
Collapse
|
8
|
Li W, Li P, Li N, Du Y, Lü S, Elad D, Long M. Matrix stiffness and shear stresses modulate hepatocyte functions in a fibrotic liver sinusoidal model. Am J Physiol Gastrointest Liver Physiol 2021; 320:G272-G282. [PMID: 33296275 PMCID: PMC8609567 DOI: 10.1152/ajpgi.00379.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Extracellular matrix (ECM) rigidity has important effects on cell behaviors and increases sharply in liver fibrosis and cirrhosis. Hepatic blood flow is essential in maintaining hepatocytes' (HCs) functions. However, it is still unclear how matrix stiffness and shear stresses orchestrate HC phenotype in concert. A fibrotic three-dimensional (3-D) liver sinusoidal model is constructed using a porous membrane sandwiched between two polydimethylsiloxane (PDMS) layers with respective flow channels. The HCs are cultured in collagen gels of various stiffnesses in the lower channel, whereas the upper channel is pre-seeded with liver sinusoidal endothelial cells (LSECs) and accessible to shear flow. The results reveal that HCs cultured within stiffer matrices exhibit reduced albumin production and cytochrome P450 (CYP450) reductase expression. Low shear stresses enhance synthetic and metabolic functions of HC, whereas high shear stresses lead to the loss of HC phenotype. Furthermore, both mechanical factors regulate HC functions by complementing each other. These observations are likely attributed to mechanically induced mass transport or key signaling molecule of hepatocyte nuclear factor 4α (HNF4α). The present study results provide an insight into understanding the mechanisms of HC dysfunction in liver fibrosis and cirrhosis, especially from the viewpoint of matrix stiffness and blood flow.NEW & NOTEWORTHY A fibrotic three-dimensional (3-D) liver sinusoidal model was constructed to mimic different stages of liver fibrosis in vivo and to explore the cooperative effects of matrix stiffness and shear stresses on hepatocyte (HC) functions. Mechanically induced alterations of mass transport mainly contributed to HC functions via typical mechanosensitive signaling.
Collapse
Affiliation(s)
- Wang Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Peiwen Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Ning Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yu Du
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Shouqin Lü
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - David Elad
- 5Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Mian Long
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
9
|
Huang D, Gibeley SB, Xu C, Xiao Y, Celik O, Ginsberg HN, Leong KW. Engineering liver microtissues for disease modeling and regenerative medicine. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909553. [PMID: 33390875 PMCID: PMC7774671 DOI: 10.1002/adfm.201909553] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Indexed: 05/08/2023]
Abstract
The burden of liver diseases is increasing worldwide, accounting for two million deaths annually. In the past decade, tremendous progress has been made in the basic and translational research of liver tissue engineering. Liver microtissues are small, three-dimensional hepatocyte cultures that recapitulate liver physiology and have been used in biomedical research and regenerative medicine. This review summarizes recent advances, challenges, and future directions in liver microtissue research. Cellular engineering approaches are used to sustain primary hepatocytes or produce hepatocytes derived from pluripotent stem cells and other adult tissues. Three-dimensional microtissues are generated by scaffold-free assembly or scaffold-assisted methods such as macroencapsulation, droplet microfluidics, and bioprinting. Optimization of the hepatic microenvironment entails incorporating the appropriate cell composition for enhanced cell-cell interactions and niche-specific signals, and creating scaffolds with desired chemical, mechanical and physical properties. Perfusion-based culture systems such as bioreactors and microfluidic systems are used to achieve efficient exchange of nutrients and soluble factors. Taken together, systematic optimization of liver microtissues is a multidisciplinary effort focused on creating liver cultures and on-chip models with greater structural complexity and physiological relevance for use in liver disease research, therapeutic development, and regenerative medicine.
Collapse
Affiliation(s)
- Dantong Huang
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Sarah B. Gibeley
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Ozgenur Celik
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Henry N. Ginsberg
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
10
|
Liu W, Zhang M, Xiao Y, Ye Z, Zhou Y, Lang M, Tan WS. Fabrication and in vitro evaluation of a packed-bed bioreactor based on galactosylated poly(ethylene terephthalate) microfibrous scaffolds. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
11
|
Yadav A, Seth B, Chaturvedi RK. Brain Organoids: Tiny Mirrors of Human Neurodevelopment and Neurological Disorders. Neuroscientist 2020; 27:388-426. [PMID: 32723210 DOI: 10.1177/1073858420943192] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Unravelling the complexity of the human brain is a challenging task. Nowadays, modern neurobiologists have developed 3D model systems called "brain organoids" to overcome the technical challenges in understanding human brain development and the limitations of animal models to study neurological diseases. Certainly like most model systems in neuroscience, brain organoids too have limitations, as these minuscule brains lack the complex neuronal circuitry required to begin the operational tasks of human brain. However, researchers are hopeful that future endeavors with these 3D brain tissues could provide mechanistic insights into the generation of circuit complexity as well as reproducible creation of different regions of the human brain. Herein, we have presented the contemporary state of brain organoids with special emphasis on their mode of generation and their utility in modelling neurological disorders, drug discovery, and clinical trials.
Collapse
Affiliation(s)
- Anuradha Yadav
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Brashket Seth
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
12
|
da Silva Morais A, Vieira S, Zhao X, Mao Z, Gao C, Oliveira JM, Reis RL. Advanced Biomaterials and Processing Methods for Liver Regeneration: State-of-the-Art and Future Trends. Adv Healthc Mater 2020; 9:e1901435. [PMID: 31977159 DOI: 10.1002/adhm.201901435] [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: 10/09/2019] [Revised: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Liver diseases contribute markedly to the global burden of mortality and disease. The limited organ disposal for orthotopic liver transplantation results in a continuing need for alternative strategies. Over the past years, important progress has been made in the field of tissue engineering (TE). Many of the early trials to improve the development of an engineered tissue construct are based on seeding cells onto biomaterial scaffolds. Nowadays, several TE approaches have been developed and are applied to one vital organ: the liver. Essential elements must be considered in liver TE-cells and culturing systems, bioactive agents or growth factors (GF), and biomaterials and processing methods. The potential of hepatocytes, mesenchymal stem cells, and others as cell sources is demonstrated. They need engineered biomaterial-based scaffolds with perfect biocompatibility and bioactivity to support cell proliferation and hepatic differentiation as well as allowing extracellular matrix deposition and vascularization. Moreover, they require a microenvironment provided using conventional or advanced processing technologies in order to supply oxygen, nutrients, and GF. Herein the biomaterials and the conventional and advanced processing technologies, including cell-sheets process, 3D bioprinting, and microfluidic systems, as well as the future trends in these major fields are discussed.
Collapse
Affiliation(s)
- Alain da Silva Morais
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Sílvia Vieira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Xinlian Zhao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Joaquim M. Oliveira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| | - Rui L. Reis
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
13
|
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]
|
14
|
Fabrication and evaluation of modified poly(ethylene terephthalate) microfibrous scaffolds for hepatocyte growth and functionality maintenance. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 109:110523. [PMID: 32228959 DOI: 10.1016/j.msec.2019.110523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/15/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022]
Abstract
For hepatocyte culture in vitro, the surface feature of utilized scaffolds exerts a direct impact on cell adhesion, growth and differentiated functionality. Herein, to regulate hepatocyte growth and differentiated functionality, modified microfibrous scaffolds were fabricated by surface grafting monoamine terminated lactobionic lactone (L-NH2) and gelatin onto non-woven poly(ethylene terephthalate) (PET) fibrous substrate (PET-Gal and PET-Gel), respectively. The physicochemical properties of PET scaffolds before and after modification were characterized. Upon 15-day culture, the effects of modified PET scaffolds on growth and differentiated functionality of human induced hepatocytes (hiHeps) were evaluated, compared with that of control without modification. Results demonstrated that both L-NH2 and gelatin modifications improved scaffold properties including hydrophilicity, water uptake ratio, stiffness and roughness, resulting in efficient cell adhesion, ~20-fold cell expansion and enhanced differentiated functionality. After culture for 15 days, PET-Gal cultured cells formed aggregates, displaying better cell viability and significantly higher differentiated functionality regarding albumin secretion, urea synthesis, phases I (cytochrome P450, CYP1A1/2 and CYP3A4) and II (uridine 5'-diphosphate glucuronosyltransferases, UGT) enzyme activity, biliary excretion and detoxification ability (ammonia elimination and bilirubin conjugation), compared with PET and PET-Gel cultured ones. Hence, as a three-dimensional (3D) microfibrous scaffold, PET-Gal promotes hiHeps growth and differentiated functionality maintenance, which is promisingly utilized in bioartificial liver (BAL) bioreactors.
Collapse
|
15
|
Liu W, Hu D, Gu C, Zhou Y, Tan WS. Fabrication and in vitro evaluation of a packed-bed bioreactor based on an optimum two-stage culture strategy. J Biosci Bioeng 2018; 127:506-514. [PMID: 30322683 DOI: 10.1016/j.jbiosc.2018.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 12/14/2022]
Abstract
A packed-bed (PB) bioreactor for bioartificial liver (BAL) was fabricated based on an optimum two-stage culture strategy and evaluated in vitro in this research. Human induced hepatocytes (hiHeps) were first expanded using Cytodex 3 microcarriers and the choice of microcarrier concentration and fetal bovine serum (FBS) content was optimized. Then, the cells expanded under the optimum expansion condition were perfused into a perfusion system containing Fibra-Cel (FC) disks to fabricate a PB bioreactor. Operating parameters including flow rate and seeding density for perfusion culture were optimized, respectively. Results indicated that during suspension culture, rapid cell proliferation and favorable amino acid metabolism were achieved at 3 mg/mL microcarriers combined with 1% FBS. While for the perfusion culture, the most effective flow rate and seeding density were 2 mL/min and 1 × 106 cells/mL, respectively. Under this optimum perfusion condition, hiHeps showed good proliferation ability, high viability, homogeneous distribution, high metabolism activities and efficient albumin secretion as well as high liver-specific genes expression. Therefore, the two-stage culture strategy based on operating parameters optimization provides a new method for the development of PB bioreactors.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Dan Hu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Ce Gu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Yan Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| |
Collapse
|
16
|
Robertson MJ, Soibam B, O’Leary JG, Sampaio LC, Taylor DA. Recellularization of rat liver: An in vitro model for assessing human drug metabolism and liver biology. PLoS One 2018; 13:e0191892. [PMID: 29377912 PMCID: PMC5788381 DOI: 10.1371/journal.pone.0191892] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/12/2018] [Indexed: 02/07/2023] Open
Abstract
Liver-like organoids that recapitulate the complex functions of the whole liver by combining cells, scaffolds, and mechanical or chemical cues are becoming important models for studying liver biology and drug metabolism. The advantages of growing cells in three-dimensional constructs include enhanced cell-cell and cell-extracellular matrix interactions and preserved cellular phenotype including, prevention of de-differentiation. In the current study, biomimetic liver constructs were made via perfusion decellularization of rat liver, with the goal of maintaining the native composition and structure of the extracellular matrix. We optimized our decellularization process to produce liver scaffolds in which immunogenic residual DNA was removed but glycosaminoglycans were maintained. When the constructs were recellularized with rat or human liver cells, the cells remained viable, capable of proliferation, and functional for 28 days. Specifically, the cells continued to express cytochrome P450 genes and maintained their ability to metabolize a model drug, midazolam. Microarray analysis showed an upregulation of genes involved in liver regeneration and fibrosis. In conclusion, these liver constructs have the potential to be used as test beds for studying liver biology and drug metabolism.
Collapse
Affiliation(s)
- Matthew J. Robertson
- Scientific Stem Cell, Texas Heart Institute, Houston, Texas, United States of America
| | - Benjamin Soibam
- Department of Computer Science and Engineering Technology, University of Houston-Downtown, Houston, Texas, United States of America
| | - Jacqueline G. O’Leary
- Annette C. & Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, Texas, United States of America
| | - Luiz C. Sampaio
- Regenerative Medicine Research Laboratories, Texas Heart Institute, Houston, Texas, United States of America
| | - Doris A. Taylor
- Regenerative Medicine Research Laboratories, Texas Heart Institute, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
17
|
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
|
18
|
Perez RA, Jung CR, Kim HW. Biomaterials and Culture Technologies for Regenerative Therapy of Liver Tissue. Adv Healthc Mater 2017; 6. [PMID: 27860372 DOI: 10.1002/adhm.201600791] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/10/2016] [Indexed: 12/18/2022]
Abstract
Regenerative approach has emerged to substitute the current extracorporeal technologies for the treatment of diseased and damaged liver tissue. This is based on the use of biomaterials that modulate the responses of hepatic cells through the unique matrix properties tuned to recapitulate regenerative functions. Cells in liver preserve their phenotype or differentiate through the interactions with extracellular matrix molecules. Therefore, the intrinsic properties of the engineered biomaterials, such as stiffness and surface topography, need to be tailored to induce appropriate cellular functions. The matrix physical stimuli can be combined with biochemical cues, such as immobilized functional groups or the delivered actions of signaling molecules. Furthermore, the external modulation of cells, through cocultures with nonparenchymal cells (e.g., endothelial cells) that can signal bioactive molecules, is another promising avenue to regenerate liver tissue. This review disseminates the recent approaches of regenerating liver tissue, with a focus on the development of biomaterials and the related culture technologies.
Collapse
Affiliation(s)
- Roman A. Perez
- Institute of Tissue Regeneration Engineering (ITREN); Dankook University; Cheonan 330-714 Republic of Korea
- Regenerative Medicine Research Institute; Universitat Internacional de Catalunya; Barcelona 08017 Spain
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine; Dankook University; Cheonan 330-714 Republic of Korea
| | - Cho-Rok Jung
- Gene Therapy Research Unit; KRIBB; 125 Gwahak-ro Yuseong-gu, Daejeon 34141 Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN); Dankook University; Cheonan 330-714 Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine; Dankook University; Cheonan 330-714 Republic of Korea
- Department of Biomaterials Science; Dankook University Dental College; Cheonan 330-714 Republic of Korea
| |
Collapse
|
19
|
Pang Y, Horimoto Y, Sutoko S, Montagne K, Shinohara M, Mathiue D, Komori K, Anzai M, Niino T, Sakai Y. Novel integrative methodology for engineering large liver tissue equivalents based on three-dimensional scaffold fabrication and cellular aggregate assembly. Biofabrication 2016; 8:035016. [PMID: 27579855 DOI: 10.1088/1758-5090/8/3/035016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A novel engineering methodology for organizing a large liver tissue equivalent was established by intergrating both 'top down' and 'bottom up' approaches. A three-dimensional (3D) scaffold was engineered comprising 43 culture chambers (volume: 11.63 cm(3)) assembled in a symmetrical pattern on 3 layers, a design which enables further scaling up of the device to a clinically significant size (volume: 500 cm(3)). In addition, an inter-connected flow channel network was designed and proved to homogenously deliver culture medium to each chamber with the same pressure drop. After fabrication using nylon-12 and a selective laser sintering process, co-cultured cellular aggregates of human hepatoma Hep G2 and TMNK-1 cells were loosely packed into the culture chambers with biodegradable poly-L-lactic acid fibre pieces for 9 days of perfusion culture. The device enabled increased hepatic function and well-maintained cell viability, demonstrating the importance of an independent medium flow supply for cell growth and function provided by the current 3D scaffold. This integrative methodology from the macro- to the micro-scale provides an efficient way of arranging engineered liver tissue with improved mass transfer, making it possible to further scale up to a construct with clinically relevant size while maintaining high per-volume-based physiological function in the near future.
Collapse
Affiliation(s)
- Y Pang
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, University of Tokyo, Komaba 4-6-1, Meguro-ku, Tokyo 153-8505, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Loneker AE, Faulk DM, Hussey GS, D'Amore A, Badylak SF. Solubilized liver extracellular matrix maintains primary rat hepatocyte phenotype in-vitro. J Biomed Mater Res A 2016; 104:957-65. [PMID: 26704367 DOI: 10.1002/jbm.a.35636] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 12/11/2015] [Accepted: 12/21/2015] [Indexed: 01/12/2023]
Abstract
Whole organ engineering and cell-based regenerative medicine approaches are being investigated as potential therapeutic options for end-stage liver failure. However, a major challenge of these strategies is the loss of hepatic specific function after hepatocytes are removed from their native microenvironment. The objective of the present study was to determine if solubilized liver extracellular matrix (ECM), when used as a media supplement, can better maintain hepatocyte phenotype compared to type I collagen alone or solubilized ECM harvested from a non-liver tissue source. Liver extracellular matrix (LECM) from four different species was isolated via liver tissue decellularization, solubilized, and then used as a media supplement for primary rat hepatocytes (PRH). The four species of LECM investigated were human, porcine, canine and rat. Cell morphology, albumin secretion, and ammonia metabolism were used to assess maintenance of hepatocyte phenotype. Biochemical and mechanical characterization of each LECM were also conducted. Results showed that PRH's supplemented with canine and porcine LECM maintained their phenotype to a greater extent compared to all other groups. PRH's supplemented with canine and porcine LECM showed increased bile production, increased albumin production, and the formation of multinucleate cells. The findings of the present study suggest that solubilized liver ECM can support in-vitro hepatocyte culture and should be considered for therapeutic and diagnostic techniques that utilize hepatocytes.
Collapse
Affiliation(s)
- Abigail E Loneker
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Denver M Faulk
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George S Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Antonio D'Amore
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,RiMED Foundation, Palermo, 90133, Italy
| | - Stephen F Badylak
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
21
|
Gao Y, Liang J, Luo Y, Gong Y. Structural optimization design of perfusion bioreactors with multilayer circular parallel plates scaffold. BIOTECHNOL BIOTEC EQ 2015. [DOI: 10.1080/13102818.2015.1021279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
22
|
Gao YB, Liang JX, Luo YX, Yan J. A tracer liquid image velocimetry for multi-layer radial flow in bioreactors. Biomed Eng Online 2015; 14:10. [PMID: 25888748 PMCID: PMC4339657 DOI: 10.1186/s12938-015-0002-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/21/2015] [Indexed: 01/05/2023] Open
Abstract
Background This paper presents a Tracer Liquid Image Velocimetry (TLIV) for multi-layer radial flow in bioreactors used for cells cultivation of tissue engineering. The goal of this approach is to use simple devices to get good measuring precision, specialized for the case in which the uniform level of fluid shear stress was required while fluid velocity varied smoothly. Methods Compared to the widely used Particles Image Velocimetry (PIV), this method adopted a bit of liquid as tracer, without the need of laser source. Sub-pixel positioning algorithm was used to overcome the adverse effects of the tracer liquid deformation. In addition, a neighborhood smoothing algorithm was used to restrict the measurement perturbation caused by diffusion. Experiments were carried out in a parallel plates flow chamber. And mathematical models of the flow chamber and Computational Fluid Dynamics (CFD) simulation were separately employed to validate the measurement precision of TLIV. Results The mean relative error between the simulated and measured data can be less than 2%, while in similar validations using PIV, the error was around 8.8%. Conclusions TLIV avoided the contradiction between the particles’ visibility and following performance with tested fluid, which is difficult to overcome in PIV. And TLIV is easier to popularize for its simple experimental condition and low cost.
Collapse
Affiliation(s)
- Yu-Bao Gao
- School of Engineering, Sun Yat-sen University, Guangdong, China.
| | - Jiu-Xing Liang
- School of Engineering, Sun Yat-sen University, Guangdong, China.
| | - Yu-Xi Luo
- School of Engineering, Sun Yat-sen University, Guangdong, China.
| | - Jia Yan
- School of Engineering, Sun Yat-sen University, Guangdong, China.
| |
Collapse
|
23
|
Sarika PR, Sidhy Viha CV, Sajin Raj RG, Nirmala RJ, Anil Kumar PR. A non-adhesive hybrid scaffold from gelatin and gum Arabic as packed bed matrix for hepatocyte perfusion culture. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 46:341-7. [PMID: 25491996 DOI: 10.1016/j.msec.2014.10.044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 09/27/2014] [Accepted: 10/21/2014] [Indexed: 10/24/2022]
Abstract
Development of liver support systems has become one of the most investigated areas for the last 50 years because of the shortage of donor organs for orthotopic liver transplantations. Bioartificial liver (BAL) device is one of the alternatives for liver failure which provides a curing method and support patients to recover from certain liver failure diseases. The biological compartment of BAL is called the bioreactor where functionally active hepatocytes are maintained to support the liver specific functions. We have developed a packed bed bioreactor with a cytocompatible, polysaccharide-protein hybrid scaffold. The scaffold prepared from gelatin and gum Arabic acts as a packed bed matrix for hepatocyte culture. Quantitative evaluation of the hepatocytes cultured using packed bed bioreactor demonstrated that cells maintained liver specific functions like albumin and urea synthesis for seven days. These results indicated that the system can be scaled up to form the biological component of a bioartificial liver.
Collapse
Affiliation(s)
- P R Sarika
- Department of Chemistry, Indian Institute of Space Science and Technology, Valiamala, Thiruvananthapuram, Kerala 695 547, India
| | - C V Sidhy Viha
- Tissue Culture Laboratory, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, Kerala 695 012, India
| | - R G Sajin Raj
- Device Testing Laboratory, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, Kerala 695 012, India
| | - Rachel James Nirmala
- Department of Chemistry, Indian Institute of Space Science and Technology, Valiamala, Thiruvananthapuram, Kerala 695 547, India
| | - P R Anil Kumar
- Tissue Culture Laboratory, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, Kerala 695 012, India.
| |
Collapse
|
24
|
Hynds RE, Giangreco A. Concise review: the relevance of human stem cell-derived organoid models for epithelial translational medicine. Stem Cells 2014. [PMID: 23203919 DOI: 10.1002/stem.1290] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Epithelial organ remodeling is a major contributing factor to worldwide death and disease, costing healthcare systems billions of dollars every year. Despite this, most fundamental epithelial organ research fails to produce new therapies and mortality rates for epithelial organ diseases remain unacceptably high. In large part, this failure in translating basic epithelial research into clinical therapy is due to a lack of relevance in existing preclinical models. To correct this, new models are required that improve preclinical target identification, pharmacological lead validation, and compound optimization. In this review, we discuss the relevance of human stem cell-derived, three-dimensional organoid models for addressing each of these challenges. We highlight the advantages of stem cell-derived organoid models over existing culture systems, discuss recent advances in epithelial tissue-specific organoids, and present a paradigm for using organoid models in human translational medicine.
Collapse
Affiliation(s)
- Robert E Hynds
- Lungs for Living Research Centre, Division of Medicine, University College London, London, UK
| | | |
Collapse
|
25
|
|
26
|
Kang CW, Wang Y, Tania M, Zhou H, Gao Y, Ba T, Tan GDS, Kim S, Leo HL. Computational fluid modeling and performance analysis of a bidirectional rotating perfusion culture system. Biotechnol Prog 2013; 29:1002-12. [DOI: 10.1002/btpr.1736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 03/11/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Chang-Wei Kang
- Fluid Dynamic Group; Institute of High Performance Computing; Singapore 138632 Singapore
| | - Yan Wang
- Dept. of Hepatobiliary Surgery; Southern Medical University Zhujiang Hospital; Guangzhou 510280 P.R. China
| | - Marshella Tania
- Dept. of Bioengineering; National University of Singapore; Singapore 117576 Singapore
| | - Huancheng Zhou
- Dept. of Hepatobiliary Surgery; Southern Medical University Zhujiang Hospital; Guangzhou 510280 P.R. China
| | - Yi Gao
- Dept. of Hepatobiliary Surgery; Southern Medical University Zhujiang Hospital; Guangzhou 510280 P.R. China
| | - Te Ba
- Fluid Dynamic Group; Institute of High Performance Computing; Singapore 138632 Singapore
| | - Guo-Dong Sean Tan
- Dept. of Bioengineering; National University of Singapore; Singapore 117576 Singapore
| | - Sangho Kim
- Dept. of Bioengineering; National University of Singapore; Singapore 117576 Singapore
| | - Hwa Liang Leo
- Dept. of Bioengineering; National University of Singapore; Singapore 117576 Singapore
| |
Collapse
|