1
|
Field EH, Ratcliffe J, Johnson CJ, Binger KJ, Reynolds NP. Self-healing, 3D printed bioinks from self-assembled peptide and alginate hybrid hydrogels. BIOMATERIALS ADVANCES 2024; 169:214145. [PMID: 39675342 DOI: 10.1016/j.bioadv.2024.214145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/17/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
There is a pressing need for new cell-laden, printable, biomaterials that are rigid and highly biocompatible. These materials can mimic stiffer tissues such as cartilage, fibrotic tissue and cancer microenvironments, and thus have exciting applications in regenerative medicine, wound healing and cancer research. Self-assembled peptides (SAPs) functionalised with aromatic groups such as Fluorenyl-9-methoxycarbonyl (Fmoc) show promise as components of these biomaterials. However, the harsh basic conditions often used to solubilise SAPs leads to issues with toxicity and reproducibility. Here, we have designed a hybrid material comprised of self-assembled Fmoc-diphenylalanine (Fmoc-FF) assemblies dispersed throughout a sodium alginate matrix and investigated the influence of different organic solvents as peptide solubilising agents. Bioinks fabricated from peptides dissolved in 1,1,1,3,3,3-Hexafluoro-2-propanol (HFIP) showed improved biocompatibility compared to those made from Dimethyl Sulfoxide (DMSO) peptide stocks, due to the increased volatility and reduced surface tension of HFIP, allowing for more efficient expulsion from the system. Through optimisation of assembly and solvent conditions we can generate hybrid bioinks with stiffnesses up to 8 times greater than sodium alginate alone that remain highly printable, even when laden with high concentrations of cells. In addition, the shear-thinning nature of the self-assembled peptide assemblies gave the hybrid bioinks highly desirable self-healing capabilities. Our developed hybrid materials allow the bioprinting of materials previously considered too stiff to extrude without causing shear induced cytotoxicity with applications in tissue engineering and biosensing.
Collapse
Affiliation(s)
- Emily H Field
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Julian Ratcliffe
- La Trobe University Bioimaging platform, La Trobe University, Australia, Melbourne, Victoria 3086, Australia
| | - Chad J Johnson
- La Trobe University Bioimaging platform, La Trobe University, Australia, Melbourne, Victoria 3086, Australia
| | - Katrina J Binger
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria 3086, Australia; Centre for Cardiovascular Biology & Disease Research, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria 3086, Australia
| | - Nicholas P Reynolds
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria 3086, Australia; The Biomedical and Environmental Sensor Technology (BEST) Research Centre, Biosensors Program, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria 3086, Australia.
| |
Collapse
|
2
|
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
|
3
|
Advanced 3D Cell Culture Techniques in Micro-Bioreactors, Part II: Systems and Applications. Processes (Basel) 2020. [DOI: 10.3390/pr9010021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this second part of our systematic review on the research area of 3D cell culture in micro-bioreactors we give a detailed description of the published work with regard to the existing micro-bioreactor types and their applications, and highlight important results gathered with the respective systems. As an interesting detail, we found that micro-bioreactors have already been used in SARS-CoV research prior to the SARS-CoV2 pandemic. As our literature research revealed a variety of 3D cell culture configurations in the examined bioreactor systems, we defined in review part one “complexity levels” by means of the corresponding 3D cell culture techniques applied in the systems. The definition of the complexity is thereby based on the knowledge that the spatial distribution of cell-extracellular matrix interactions and the spatial distribution of homologous and heterologous cell–cell contacts play an important role in modulating cell functions. Because at least one of these parameters can be assigned to the 3D cell culture techniques discussed in the present review, we structured the studies according to the complexity levels applied in the MBR systems.
Collapse
|
4
|
Advanced 3D Cell Culture Techniques in Micro-Bioreactors, Part I: A Systematic Analysis of the Literature Published between 2000 and 2020. Processes (Basel) 2020. [DOI: 10.3390/pr8121656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bioreactors have proven useful for a vast amount of applications. Besides classical large-scale bioreactors and fermenters for prokaryotic and eukaryotic organisms, micro-bioreactors, as specialized bioreactor systems, have become an invaluable tool for mammalian 3D cell cultures. In this systematic review we analyze the literature in the field of eukaryotic 3D cell culture in micro-bioreactors within the last 20 years. For this, we define complexity levels with regard to the cellular 3D microenvironment concerning cell–matrix-contact, cell–cell-contact and the number of different cell types present at the same time. Moreover, we examine the data with regard to the micro-bioreactor design including mode of cell stimulation/nutrient supply and materials used for the micro-bioreactors, the corresponding 3D cell culture techniques and the related cellular microenvironment, the cell types and in vitro models used. As a data source we used the National Library of Medicine and analyzed the studies published from 2000 to 2020.
Collapse
|
5
|
Xing F, Zhou C, Hui D, Du C, Wu L, Wang L, Wang W, Pu X, Gu L, Liu L, Xiang Z, Zhang X. Hyaluronic acid as a bioactive component for bone tissue regeneration: Fabrication, modification, properties, and biological functions. NANOTECHNOLOGY REVIEWS 2020. [DOI: 10.1515/ntrev-2020-0084] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Abstract
Hyaluronic acid (HA) is widely distributed in the human body, and it is heavily involved in many physiological functions such as tissue hydration, wound repair, and cell migration. In recent years, HA and its derivatives have been widely used as advanced bioactive polymers for bone regeneration. Many medical products containing HA have been developed because this natural polymer has been proven to be nontoxic, noninflammatory, biodegradable, and biocompatible. Moreover, HA-based composite scaffolds have shown good potential for promoting osteogenesis and mineralization. Recently, many HA-based biomaterials have been fabricated for bone regeneration by combining with electrospinning and 3D printing technology. In this review, the polymer structures, processing, properties, and applications in bone tissue engineering are summarized. The challenges and prospects of HA polymers are also discussed.
Collapse
Affiliation(s)
- Fei Xing
- Department of Orthopaedics, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, Sichuan University , 610064 , Chengdu , China
- College of Biomedical Engineering, Sichuan University , 610064 , Chengdu , China
| | - Didi Hui
- Innovatus Oral Cosmetic & Surgical Institute , Norman , OK, 73069 , United States of America
| | - Colin Du
- Innovatus Oral Cosmetic & Surgical Institute , Norman , OK, 73069 , United States of America
| | - Lina Wu
- National Engineering Research Center for Biomaterials, Sichuan University , 610064 , Chengdu , China
- College of Biomedical Engineering, Sichuan University , 610064 , Chengdu , China
| | - Linnan Wang
- Department of Orthopaedics, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Wenzhao Wang
- Department of Orthopaedics, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Xiaobing Pu
- Department of Orthopedics Medical Center, West China School of Public Health and West China Fourth Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Linxia Gu
- Department of Biomedical and Chemical Engineering and Sciences, College of Engineering & Science, Florida Institute of Technology , Melbourne , FL, 32901 , United States of America
| | - Lei Liu
- Department of Orthopaedics, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Zhou Xiang
- Department of Orthopaedics, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University , 610064 , Chengdu , China
- College of Biomedical Engineering, Sichuan University , 610064 , Chengdu , China
| |
Collapse
|
6
|
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
|
7
|
Zhang Y, Lu J, Li Z, Zhu D, Yu X, Li L. Enhanced cellular functions of hepatocytes in the hyaluronate-alginate-chitosan microcapsules. Int J Artif Organs 2020; 44:340-349. [PMID: 32969286 DOI: 10.1177/0391398820959345] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The study aimed to develop a biocompatible microcapsule for hepatocytes and create a bio-mimic microenvironment for maintaining hepatic-specific functions of hepatocytes in vitro. The work is proposed for the bioartificial liver system in the treatment of liver failure. In this study, microcapsules were prepared with hyaluronate (HA)/sodium alginate (SA) as an inner core and an outer chitosan (CS) shell via one-step spraying method. C3A cells were encapsulated in microcapsules to examine the biocompatibility of HA-SA-CS microcapsules. MTT and fluorescence microscopy indicated that C3A cells had high viability in the HA-SA-CS microcapsules. The liver-specific functions, such as urea and albumin synthesis, and CYP1A2 and CYP3A4 activities from encapsulated cells were increased in the HA-SA-CS microcapsules compared to the SA-CS microcapsules. The gene expressions of CYP450 related genes were also increased by HA on day 3. The study suggests that HA-SA-CS microcapsules have good biocompatibility and can maintain a favorable environment for hepatocytes. This approach has improved the preservation of liver cells' metabolic functions and could be a candidate for the bioartificial liver system.
Collapse
Affiliation(s)
- Yanhong Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zuhong Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaopeng Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Jang M, Kleber A, Ruckelshausen T, Betzholz R, Manz A. Differentiation of the human liver progenitor cell line (HepaRG) on a microfluidic-based biochip. J Tissue Eng Regen Med 2019; 13:482-494. [PMID: 30746894 DOI: 10.1002/term.2802] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/26/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022]
Abstract
HepaRG is a bipotent stem cell line that can be differentiated towards hepatocyte-like and biliary-like cells. The entire cultivation process requires 1 month and relies on the addition of 2% dimethyl sulfoxide (DMSO) to the culture. Our motivation in this research is to differentiate HepaRG cells (progenitor cells and undifferentiated cells) towards hepatocyte-like cells by minimizing the cultivation time and without using DMSO treatment by instead using a microfluidic device combined with the following strategies: (a) comparison of extracellular matrices (matrigel and collagen I), (b) types of flow (one or both sides), and (c) effects of DMSO. Our results demonstrate that matrigel promotes the differentiation of progenitor cells towards hepatocytes and biliary-like cells. Moreover, the frequent formation of HepaRG cell clusters was observed by a supply of both sides of flow, and the cell viability and liver specific functions were influenced by DMSO. Finally, differentiated HepaRG progenitor cells cultured in a microfluidic device for 14 days without DMSO treatment yielded 70% of hepatocyte-like cells with a highly polarized organization that reacted to stimulation with IL-6 to produce C-reactive protein (CRP). This culture model has high potential for investigating cell differentiation and liver pathophysiology research.
Collapse
Affiliation(s)
- Mi Jang
- Department of system engineering, Saarland University, Saarbrücken, Germany.,Microfluidics group, KIST Europe, Saarbrücken, Germany.,Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
| | - Astrid Kleber
- Rhineland Palantinate Centre of Excellence for climate Change Impacts, Trippstadt, Germany
| | - Thomas Ruckelshausen
- Dynamic Biomaterial group, INM - Leibniz-Institut für Neue Materialien GmbH, Saarbrücken, Germany.,Service and Support group, PicoQuant, Rudower Chaussee 29, Berlin, Germany
| | - Ralf Betzholz
- School of Physics, Huazhong University of Science and Technology, Wuhan, China
| | - Andreas Manz
- Department of system engineering, Saarland University, Saarbrücken, Germany.,Microfluidics group, KIST Europe, Saarbrücken, Germany
| |
Collapse
|
9
|
Gouliarmou V, Lostia AM, Coecke S, Bernasconi C, Bessems J, Dorne JL, Ferguson S, Testai E, Remy UG, Brian Houston J, Monshouwer M, Nong A, Pelkonen O, Morath S, Wetmore BA, Worth A, Zanelli U, Zorzoli MC, Whelan M. Establishing a systematic framework to characterise in vitro methods for human hepatic metabolic clearance. Toxicol In Vitro 2018; 53:233-244. [DOI: 10.1016/j.tiv.2018.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 07/17/2018] [Accepted: 08/08/2018] [Indexed: 12/26/2022]
|
10
|
Starokozhko V, Hemmingsen M, Larsen L, Mohanty S, Merema M, Pimentel RC, Wolff A, Emnéus J, Aspegren A, Groothuis G, Dufva M. Differentiation of human-induced pluripotent stem cell under flow conditions to mature hepatocytes for liver tissue engineering. J Tissue Eng Regen Med 2018; 12:1273-1284. [PMID: 29499107 PMCID: PMC5969064 DOI: 10.1002/term.2659] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 01/30/2018] [Accepted: 02/17/2018] [Indexed: 12/27/2022]
Abstract
Hepatic differentiation of human-induced pluripotent stem cells (hiPSCs) under flow conditions in a 3D scaffold is expected to be a major step forward for construction of bioartificial livers. The aims of this study were to induce hepatic differentiation of hiPSCs under perfusion conditions and to perform functional comparisons with fresh human precision-cut liver slices (hPCLS), an excellent benchmark for the human liver in vivo. The majority of the mRNA expression of CYP isoenzymes and transporters and the tested CYP activities, Phase II metabolism, and albumin, urea, and bile acid synthesis in the hiPSC-derived cells reached values that overlap those of hPCLS, which indicates a higher degree of hepatic differentiation than observed until now. Differentiation under flow compared with static conditions had a strong inducing effect on Phase II metabolism and suppressed AFP expression but resulted in slightly lower activity of some of the Phase I metabolism enzymes. Gene expression data indicate that hiPSCs differentiated into both hepatic and biliary directions. In conclusion, the hiPSC differentiated under flow conditions towards hepatocytes express a wide spectrum of liver functions at levels comparable with hPCLS indicating excellent future perspectives for the development of a bioartificial liver system for toxicity testing or as liver support device for patients.
Collapse
Affiliation(s)
- Viktoriia Starokozhko
- Groningen Research Institute for PharmacyUniversity of GroningenGroningenThe Netherlands
| | - Mette Hemmingsen
- Department of Micro‐ and NanotechnologyTechnical University of DenmarkDenmark
| | - Layla Larsen
- Department of Micro‐ and NanotechnologyTechnical University of DenmarkDenmark
| | | | - Marjolijn Merema
- Groningen Research Institute for PharmacyUniversity of GroningenGroningenThe Netherlands
| | - Rodrigo C. Pimentel
- Department of Micro‐ and NanotechnologyTechnical University of DenmarkDenmark
| | - Anders Wolff
- Department of Micro‐ and NanotechnologyTechnical University of DenmarkDenmark
| | - Jenny Emnéus
- Department of Micro‐ and NanotechnologyTechnical University of DenmarkDenmark
| | | | - Geny Groothuis
- Groningen Research Institute for PharmacyUniversity of GroningenGroningenThe Netherlands
| | - Martin Dufva
- Department of Micro‐ and NanotechnologyTechnical University of DenmarkDenmark
| |
Collapse
|
11
|
Schuerlein S, Schwarz T, Krziminski S, Gätzner S, Hoppensack A, Schwedhelm I, Schweinlin M, Walles H, Hansmann J. A versatile modular bioreactor platform for Tissue Engineering. Biotechnol J 2016; 12. [PMID: 27492568 PMCID: PMC5333457 DOI: 10.1002/biot.201600326] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 12/24/2022]
Abstract
Tissue Engineering (TE) bears potential to overcome the persistent shortage of donor organs in transplantation medicine. Additionally, TE products are applied as human test systems in pharmaceutical research to close the gap between animal testing and the administration of drugs to human subjects in clinical trials. However, generating a tissue requires complex culture conditions provided by bioreactors. Currently, the translation of TE technologies into clinical and industrial applications is limited due to a wide range of different tissue‐specific, non‐disposable bioreactor systems. To ensure a high level of standardization, a suitable cost‐effectiveness, and a safe graft production, a generic modular bioreactor platform was developed. Functional modules provide robust control of culture processes, e.g. medium transport, gas exchange, heating, or trapping of floating air bubbles. Characterization revealed improved performance of the modules in comparison to traditional cell culture equipment such as incubators, or peristaltic pumps. By combining the modules, a broad range of culture conditions can be achieved. The novel bioreactor platform allows using disposable components and facilitates tissue culture in closed fluidic systems. By sustaining native carotid arteries, engineering a blood vessel, and generating intestinal tissue models according to a previously published protocol the feasibility and performance of the bioreactor platform was demonstrated.
Collapse
Affiliation(s)
- Sebastian Schuerlein
- University Hospital Wuerzburg; Department Tissue Engineering and Regenerative Medicine (TERM); Wuerzburg Germany
| | - Thomas Schwarz
- Translational Center Wuerzburg of the Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB); Wuerzburg Germany
| | - Steffan Krziminski
- Translational Center Wuerzburg of the Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB); Wuerzburg Germany
| | - Sabine Gätzner
- University Hospital Wuerzburg; Department Tissue Engineering and Regenerative Medicine (TERM); Wuerzburg Germany
| | - Anke Hoppensack
- University Hospital Wuerzburg; Department Tissue Engineering and Regenerative Medicine (TERM); Wuerzburg Germany
| | - Ivo Schwedhelm
- University Hospital Wuerzburg; Department Tissue Engineering and Regenerative Medicine (TERM); Wuerzburg Germany
| | - Matthias Schweinlin
- University Hospital Wuerzburg; Department Tissue Engineering and Regenerative Medicine (TERM); Wuerzburg Germany
| | - Heike Walles
- University Hospital Wuerzburg; Department Tissue Engineering and Regenerative Medicine (TERM); Wuerzburg Germany
- Translational Center Wuerzburg of the Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB); Wuerzburg Germany
| | - Jan Hansmann
- University Hospital Wuerzburg; Department Tissue Engineering and Regenerative Medicine (TERM); Wuerzburg Germany
- Translational Center Wuerzburg of the Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB); Wuerzburg Germany
| |
Collapse
|
12
|
Open-Porous Hydroxyapatite Scaffolds for Three-Dimensional Culture of Human Adult Liver Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6040146. [PMID: 27403430 PMCID: PMC4925947 DOI: 10.1155/2016/6040146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/25/2016] [Indexed: 11/17/2022]
Abstract
Liver cell culture within three-dimensional structures provides an improved culture system for various applications in basic research, pharmacological screening, and implantable or extracorporeal liver support. Biodegradable calcium-based scaffolds in such systems could enhance liver cell functionality by providing endothelial and hepatic cell support through locally elevated calcium levels, increased surface area for cell attachment, and allowing three-dimensional tissue restructuring. Open-porous hydroxyapatite scaffolds were fabricated and seeded with primary adult human liver cells, which were embedded within or without gels of extracellular matrix protein collagen-1 or hyaluronan. Metabolic functions were assessed after 5, 15, and 28 days. Longer-term cultures exhibited highest cell numbers and liver specific gene expression when cultured on hydroxyapatite scaffolds in collagen-1. Endothelial gene expression was induced in cells cultured on scaffolds without extracellular matrix proteins. Hydroxyapatite induced gene expression for cytokeratin-19 when cells were cultured in collagen-1 gel while culture in hyaluronan increased cytokeratin-19 gene expression independent of the use of scaffold in long-term culture. The implementation of hydroxyapatite composites with extracellular matrices affected liver cell cultures and cell differentiation depending on the type of matrix protein and the presence of a scaffold. The hydroxyapatite scaffolds enable scale-up of hepatic three-dimensional culture models for regenerative medicine applications.
Collapse
|
13
|
Nishii K, Reese G, Moran EC, Sparks JL. Multiscale computational model of fluid flow and matrix deformation in decellularized liver. J Mech Behav Biomed Mater 2016; 57:201-14. [PMID: 26722987 PMCID: PMC4831654 DOI: 10.1016/j.jmbbm.2015.11.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/23/2015] [Accepted: 11/30/2015] [Indexed: 01/26/2023]
Abstract
Currently little is known about the biomechanical environment in decellularized tissue. The goal of this research is to quantify the mechanical microenvironment in decellularized liver, for varying organ-scale perfusion conditions, using a combined experimental/computational approach. Needle-guided ultra-miniature pressure sensors were inserted into liver tissue to measure parenchymal fluid pressure ex-situ in portal vein-perfused native (n=5) and decellularized (n=7) ferret liver, for flow rates from 3-12mL/min. Pressures were also recorded at the inlet near the portal vein cannula to estimate total vascular resistance of the specimens. Experimental results were fit to a multiscale computational model to simulate perfusion conditions inside native versus decellularized livers for four experimental flow rates. The multiscale model consists of two parts: an organ-scale electrical analog model of liver hemodynamics and a tissue-scale model that predicts pore fluid pressure, pore fluid velocity, and solid matrix stress and deformation throughout the 3D hepatic lobule. Distinct models were created for native versus decellularized liver. Results show that vascular resistance decreases by 82% as a result of decellularization. The hydraulic conductivity of the decellularized liver lobule, a measure of tissue permeability, was 5.6 times that of native liver. For the four flow rates studied, mean fluid pressures in the decellularized lobule were 0.6-2.4mmHg, mean fluid velocities were 211-767μm/s, and average solid matrix principal strains were 1.7-6.1%. In the future this modeling platform can be used to guide the optimization of perfusion seeding and conditioning strategies for decellularized scaffolds in liver bioengineering.
Collapse
Affiliation(s)
- Kenichiro Nishii
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, United States
| | - Greg Reese
- Research Computing Support Group, Miami University, Oxford, OH, United States
| | - Emma C Moran
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, United States
| | - Jessica L Sparks
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, United States.
| |
Collapse
|
14
|
Schmelzer E, Gerlach JC. Multicompartmental Hollow-Fiber-Based Bioreactors for Dynamic Three-Dimensional Perfusion Culture. Methods Mol Biol 2016; 1502:1-19. [PMID: 27075977 DOI: 10.1007/7651_2016_335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The creation of larger-scale three-dimensional tissue constructs depends on proper medium mass and gas exchange, as well as removal of metabolites, which cannot be achieved in conventional static two-dimensional petri dish culture. In cultures of tissue-density this problem can be addressed by decentral perfusion through artificial micro-capillaries. While the static medium exchange in petri dishes leads to metabolite peaks, perfusion culture provides a dynamic medium supply, thereby preventing non-physiological peaks. To overcome the limitations of conventional static two-dimensional culture, a three-dimensional perfusion bioreactor technology has been developed, providing decentral and high-performance mass exchange as well as integral oxygenation. Similar to organ systems in vivo, the perfusion with medium provides nutrition and removes waste metabolites, and the perfusion with gas delivers oxygen and carbon dioxide for pH regulation. Such bioreactors are available at various dimensions ranging from 0.2 to 800 mL cell compartment volumes (manufactured by StemCell Systems, Berlin, Germany). Here, we describe in detail the setup and maintenance of a small-scale 4-chamber bioreactor with its tubing circuit and perfusion system.
Collapse
Affiliation(s)
- Eva Schmelzer
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, 3025 E. Carson St., Pittsburgh, PA, 15203, USA.
| | - Jörg C Gerlach
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, 3025 E. Carson St., Pittsburgh, PA, 15203, USA
| |
Collapse
|
15
|
Gerlach JC, Over P, Foka HG, Turner ME, Thompson RL, Gridelli B, Schmelzer E. Role of transcription factor CCAAT/enhancer-binding protein alpha in human fetal liver cell types in vitro. Hepatol Res 2015; 45:919-32. [PMID: 25195540 DOI: 10.1111/hepr.12420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 09/02/2014] [Accepted: 09/03/2014] [Indexed: 12/15/2022]
Abstract
AIM The transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) has been shown to play an important role in liver development, cell proliferation and differentiation. It is, however, largely unknown if C/EBPα regulates cell differentiation and proliferation differently in the diverse cell types of the human liver. We investigated the role of C/EBPα in primary human fetal liver cells and liver cell subpopulations in vitro using a 3-D perfusion bioreactor as an advanced in vivo-like human organ culture model. METHODS Human fetal liver cells were investigated in vitro. C/EBPα gene expression was knocked down using siRNA or overexpressed by plasmid transfection. Cell type-specific gene expression was studied, cell populations and their proliferation were investigated, and metabolic parameters were analyzed. RESULTS When C/EBPα gene expression was knocked down, we observed a significantly reduced expression of typical endothelial, hematopoietic and mesenchymal genes such as CD31, vWF, CD90, CD45 and α-smooth muscle actin in fetal cells. The intracellular expression of hepatic proteins and genes for liver-specific serum proteins α-fetoprotein and albumin were reduced, their protein secretion was increased. Fetal endothelial cell numbers were reduced and hepatoblast numbers were increased. C/EBPα overexpression in fetal cells resulted in increased endothelial numbers, but did not affect mesenchymal cell types or hepatoblasts. CONCLUSION We demonstrated that the effects of C/EBPα are specific for the different human fetal liver cell types, using an advanced 3-D perfusion bioreactor as a human in vivo-like model.
Collapse
Affiliation(s)
- Jörg C Gerlach
- Department of Surgery and Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick Over
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hubert G Foka
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Morris E Turner
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Robert L Thompson
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Bruno Gridelli
- Department of Surgery, ISMETT - Mediterranean Institute for Transplantation and Advanced Specialized Therapies, Palermo, Italy
| | - Eva Schmelzer
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Minteer DM, Young MT, Lin YC, Over PJ, Rubin JP, Gerlach JC, Marra KG. Analysis of type II diabetes mellitus adipose-derived stem cells for tissue engineering applications. J Tissue Eng 2015; 6:2041731415579215. [PMID: 26090087 PMCID: PMC4456321 DOI: 10.1177/2041731415579215] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 02/26/2015] [Indexed: 11/28/2022] Open
Abstract
To address the functionality of diabetic adipose-derived stem cells in tissue engineering applications, adipose-derived stem cells isolated from patients with and without type II diabetes mellitus were cultured in bioreactor culture systems. The adipose-derived stem cells were differentiated into adipocytes and maintained as functional adipocytes. The bioreactor system utilizes a hollow fiber–based technology for three-dimensional perfusion of tissues in vitro, creating a model in which long-term culture of adipocytes is feasible, and providing a potential tool useful for drug discovery. Daily metabolic activity of the adipose-derived stem cells was analyzed within the medium recirculating throughout the bioreactor system. At experiment termination, tissues were extracted from bioreactors for immunohistological analyses in addition to gene and protein expression. Type II diabetic adipose-derived stem cells did not exhibit significantly different glucose consumption compared to adipose-derived stem cells from patients without type II diabetes (p > 0.05, N = 3). Expression of mature adipocyte genes was not significantly different between diabetic/non-diabetic groups (p > 0.05, N = 3). Protein expression of adipose tissue grown within all bioreactors was verified by Western blotting.The results from this small-scale study reveal adipose-derived stem cells from patients with type II diabetes when removed from diabetic environments behave metabolically similar to the same cells of non-diabetic patients when cultured in a three-dimensional perfusion bioreactor, suggesting that glucose transport across the adipocyte cell membrane, the hindrance of which being characteristic of type II diabetes, is dependent on environment. The presented observation describes a tissue-engineered tool for long-term cell culture and, following future adjustments to the culture environment and increased sample sizes, potentially for anti-diabetic drug testing.
Collapse
Affiliation(s)
| | - Matthew T Young
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yen-Chih Lin
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA ; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick J Over
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Peter Rubin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA ; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jorg C Gerlach
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA ; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA ; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
17
|
Schmelzer E, Finoli A, Nettleship I, Gerlach JC. Long-term three-dimensional perfusion culture of human adult bone marrow mononuclear cells in bioreactors. Biotechnol Bioeng 2015; 112:801-10. [PMID: 25335987 DOI: 10.1002/bit.25485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 08/22/2014] [Accepted: 10/13/2014] [Indexed: 12/14/2022]
Abstract
The construction and long-term maintenance of three-dimensional in vitro bone marrow models is of great interest but still quite challenging. Here we describe the use of a multi-compartment hollow-fiber membrane based three-dimensional perfusion bioreactor for long-term culture of whole human bone marrow mononuclear cells. We also investigated bioreactors with incorporated open-porous foamed hydroxyapatite scaffolds, mimicking the in vivo bone matrix. Cells in bioreactors with and without scaffolds were cultured to 6 weeks and compared to Petri dish controls. Cells were analyzed for gene expression, surface markers by flow cytometry, metabolic activity, hematopoietic potential, viability, and attachment by immunocytochemistry. Cells in bioreactors were metabolic active during long-term culture. The percentages of hematopoietic stem cell and mature endothelial cell fractions were maintained in bioreactors. The expression of most of the analyzed genes stabilized and increased after long-term culture of 6 weeks. Compared to Petri dish culture controls, bioreactor perfusion culture improved in both the short and long-term, the colony formation unit capacity of hematopoietic progenitors. Cells attached to the ample surface area provided by hydroxyapatite scaffolds. The implementation of a hydroxyapatite scaffold did not influence colony formation capacity, percentages of cell type specific fractions, gene expression, cell viability or metabolic turnover when compared to control cells cultured in bioreactors without scaffolds. In conclusion, three-dimensional perfusion bioreactor culture enables long-term maintenance of primary human bone marrow cells, with hydroxyapatite scaffolds providing an in vivo-like scaffold for three-dimensional culture.
Collapse
Affiliation(s)
- Eva Schmelzer
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, 3025 E. Carson St., Pittsburgh, Pennsylvania, 15203.
| | | | | | | |
Collapse
|
18
|
Bachmann A, Moll M, Gottwald E, Nies C, Zantl R, Wagner H, Burkhardt B, Sánchez JJM, Ladurner R, Thasler W, Damm G, Nussler AK. 3D Cultivation Techniques for Primary Human Hepatocytes. MICROARRAYS (BASEL, SWITZERLAND) 2015; 4:64-83. [PMID: 27600213 PMCID: PMC4996383 DOI: 10.3390/microarrays4010064] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/08/2015] [Accepted: 02/03/2015] [Indexed: 01/27/2023]
Abstract
One of the main challenges in drug development is the prediction of in vivo toxicity based on in vitro data. The standard cultivation system for primary human hepatocytes is based on monolayer cultures, even if it is known that these conditions result in a loss of hepatocyte morphology and of liver-specific functions, such as drug-metabolizing enzymes and transporters. As it has been demonstrated that hepatocytes embedded between two sheets of collagen maintain their function, various hydrogels and scaffolds for the 3D cultivation of hepatocytes have been developed. To further improve or maintain hepatic functions, 3D cultivation has been combined with perfusion. In this manuscript, we discuss the benefits and drawbacks of different 3D microfluidic devices. For most systems that are currently available, the main issues are the requirement of large cell numbers, the low throughput, and expensive equipment, which render these devices unattractive for research and the drug-developing industry. A higher acceptance of these devices could be achieved by their simplification and their compatibility with high-throughput, as both aspects are of major importance for a user-friendly device.
Collapse
Affiliation(s)
- Anastasia Bachmann
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, Schnarrenbergstr. 95, 72076 Tü̈bingen, Germany.
| | - Matthias Moll
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, Schnarrenbergstr. 95, 72076 Tü̈bingen, Germany.
| | - Eric Gottwald
- Institute for Biological Interfaces, Karlsruhe Institute of Technology, POB 3640, 76021 Karlsruhe, Germany.
| | - Cordula Nies
- Institute for Biological Interfaces, Karlsruhe Institute of Technology, POB 3640, 76021 Karlsruhe, Germany.
| | - Roman Zantl
- GmbH, Am Klopferspitz 19, 82152 Martinsried, Germany.
| | - Helga Wagner
- GmbH, Am Klopferspitz 19, 82152 Martinsried, Germany.
| | - Britta Burkhardt
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, Schnarrenbergstr. 95, 72076 Tü̈bingen, Germany.
| | - Juan J Martínez Sánchez
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, Schnarrenbergstr. 95, 72076 Tü̈bingen, Germany.
| | - Ruth Ladurner
- Clinic for General, Visceral and Transplantation Surgery, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany.
| | - Wolfgang Thasler
- Department of Surgery, Ludwig-Maximilians-University of Munich Hospital Grosshadern, 81377 Munich, Germany.
| | - Georg Damm
- Department for General, Visceral and Transplantation Surgery, Charité Medical University Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Andreas K Nussler
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, Schnarrenbergstr. 95, 72076 Tü̈bingen, Germany.
| |
Collapse
|
19
|
Pekor C, Gerlach JC, Nettleship I, Schmelzer E. Induction of Hepatic and Endothelial Differentiation by Perfusion in a Three-Dimensional Cell Culture Model of Human Fetal Liver. Tissue Eng Part C Methods 2015; 21:705-15. [PMID: 25559936 DOI: 10.1089/ten.tec.2014.0453] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The development of functional engineered tissue constructs depends on high cell densities and appropriate vascularization. In this study we implemented a four-compartment three-dimensional perfusion bioreactor culture model for studying the effects of medium perfusion on endothelial, hepatic, and hematopoietic cell populations of primary human fetal liver in an in vivo-like environment. Human fetal liver cells were cultured in bioreactors configured to provide either perfusion or diffusion conditions. Metabolic activities of the cultures were monitored daily by measuring glucose consumption and lactate production. Cell viability during culture was analyzed by lactate dehydrogenase activity. Hepatic functionality was determined by the release of albumin and alpha-fetoprotein (AFP) in culture medium samples. After 4 days of culture, cells were analyzed for the expression of a variety of endothelial, hepatic, and hematopoietic genes, as well as the surface marker expression of CD31 and CD34 in flow cytometry. We found that medium perfusion increased the gene expression of endothelial markers such as CD31, von Willebrand factor (vWF), CD140b, CD309, and CD144 while decreasing the gene expression of the erythrocyte-surface marker CD235a. Hepatic differentiation was promoted under perfusion conditions as demonstrated by lower AFP and higher albumin secretion compared with cultures not exposed to medium perfusion. Additionally, cultures exposed to medium perfusion gave higher rates of glucose consumption and lactate production, indicating increased metabolic activity. In conclusion, high-density bioreactors configured to provide constant medium perfusion significantly induced hepatic and endothelial cell differentiation and provided improved conditions for the culture of human fetal liver cells compared with cultures without perfusion.
Collapse
Affiliation(s)
- Christopher Pekor
- 1 Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Jörg C Gerlach
- 1 Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania.,2 Department of Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Ian Nettleship
- 3 Department of Mechanical Engineering and Materials Science, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Eva Schmelzer
- 1 Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
20
|
Chan TM, Harn HJ, Lin HP, Chou PW, Chen JYR, Ho TJ, Chiou TW, Chuang HM, Chiu SC, Chen YC, Yen SY, Huang MH, Liang BC, Lin SZ. Improved human mesenchymal stem cell isolation. Cell Transplant 2015; 23:399-406. [PMID: 24816441 DOI: 10.3727/096368914x678292] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are currently available for a range of applications and benefits and have become a good material for regenerative medicine, tissue engineering, and disease therapy. Before ex vivo expansion, isolation and characterization of primary hMSCs from peripheral tissues are key steps for obtaining adequate materials for clinical application. The proportion of peripheral stem cells is very low in surrounding tissues and organs; thus the recovery ratio will be a limiting factor. In this review, we summarized current common methods used to isolate peripheral stem cells, as well as the new insights revealed to improve the quantity of stem cells and their stemness. These strategies offer alternative ways to acquire hMSCs in a convenient and/or effective manner, which is important for clinical treatments. Improved isolation and mass amplification of the hMSCs while ensuring their stemness and quantity will be an important step for clinical use. Enlarged suitable hMSCs are more clinically applicable for therapeutic transplants and may help people live longer and better.
Collapse
Affiliation(s)
- Tzu-Min Chan
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
The concept of bioreactors in biochemical engineering is a well-established process; however, the idea of applying bioreactor technology to biomedical and tissue engineering issues is relatively novel and has been rapidly accepted as a culture model. Tissue engineers have developed and adapted various types of bioreactors in which to culture many different cell types and therapies addressing several diseases, including diabetes mellitus types 1 and 2. With a rising world of bioreactor development and an ever increasing diagnosis rate of diabetes, this review aims to highlight bioreactor history and emerging bioreactor technologies used for diabetes-related cell culture and therapies.
Collapse
Affiliation(s)
- Danielle M Minteer
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jorg C Gerlach
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Gómez-Lechón MJ, Tolosa L, Conde I, Donato MT. Competency of different cell models to predict human hepatotoxic drugs. Expert Opin Drug Metab Toxicol 2014; 10:1553-68. [PMID: 25297626 DOI: 10.1517/17425255.2014.967680] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The liver is the most important target for drug-induced toxicity. This vulnerability results from functional liver features and its role in the metabolic elimination of most drugs. Drug-induced liver injury is a significant leading cause of acute, chronic liver disease and an important safety issue when developing new drugs. AREAS COVERED This review describes the advantages and limitations of hepatic cell-based models for early safety risk assessment during drug development. These models include hepatocytes cultured as monolayer, collagen-sandwich; emerging complex 3D configuration; liver-derived cell lines; stem cell-derived hepatocytes. EXPERT OPINION In vitro toxicity assays performed in hepatocytes or hepatoma cell lines can potentially provide rapid and cost-effective early feedback to identify toxic candidates for compound prioritization. However, their capacity to predict hepatotoxicity depends critically on cells' functional performance. In an attempt to improve and prolong functional properties of cultured cells, different strategies to recreate the in vivo hepatocyte environment have been explored. 3D cultures, co-cultures of hepatocytes with other cell types and microfluidic devices seem highly promising for toxicological studies. Moreover, hepatocytes derived from human pluripotent stem cells are emerging cell-based systems that may provide a stable source of hepatocytes to reliably screen metabolism and toxicity of candidate compounds.
Collapse
Affiliation(s)
- M José Gómez-Lechón
- Unidad de Hepatología Experimental Instituto de Investigación Sanitaria La Fe (IIS LA Fe) , Torre A Avda. Fernando Abril Martorell 106, 46026 Valencia , Spain +34 961246619 ;
| | | | | | | |
Collapse
|
23
|
Caralt M, Velasco E, Lanas A, Baptista PM. Liver bioengineering: from the stage of liver decellularized matrix to the multiple cellular actors and bioreactor special effects. Organogenesis 2014; 10:250-9. [PMID: 25102189 DOI: 10.4161/org.29892] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Liver bioengineering has been a field of intense research and popular excitement in the past decades. It experiences great interest since the introduction of whole liver acellular scaffolds generated by perfusion decellularization (1-3). Nevertheless, the different strategies developed so far have failed to generate hepatic tissue in vitro bioequivalent to native liver tissue. Even notable novel strategies that rely on iPSC-derived liver progenitor cells potential to self-organize in association with endothelial cells in hepatic organoids are lacking critical components of the native tissue (e.g., bile ducts, functional vascular network, hepatic microarchitecture, etc) (4). Hence, it is vital to understand the strengths and short comes of our current strategies in this quest to re-create liver organogenesis in vitro. To shed some light into these issues, this review describes the different actors that play crucial roles in liver organogenesis and highlights the steps still missing to successfully generate whole livers and hepatic organoids in vitro for multiple applications.
Collapse
Affiliation(s)
- Mireia Caralt
- Vall d'Hebron University Hospital; Universitat Autònoma de Barcelona; Barcelona, Spain
| | | | - Angel Lanas
- University of Zaragoza; Zaragoza, Spain; IIS Aragón; CIBERehd; Zaragoza, Spain; Aragon Health Sciences Institute (IACS); Zaragoza, Spain
| | - Pedro M Baptista
- University of Zaragoza; Zaragoza, Spain; IIS Aragón; CIBERehd; Zaragoza, Spain; Aragon Health Sciences Institute (IACS); Zaragoza, Spain
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
|
27
|
Vosough M, Omidinia E, Kadivar M, Shokrgozar MA, Pournasr B, Aghdami N, Baharvand H. Generation of functional hepatocyte-like cells from human pluripotent stem cells in a scalable suspension culture. Stem Cells Dev 2013; 22:2693-705. [PMID: 23731381 DOI: 10.1089/scd.2013.0088] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Recent advances in human embryonic and induced pluripotent stem cell-based therapies in animal models of hepatic failure have led to an increased appreciation of the need to translate the proof-of-principle concepts into more practical and feasible protocols for scale up and manufacturing of functional hepatocytes. In this study, we describe a scalable stirred-suspension bioreactor culture of functional hepatocyte-like cells (HLCs) from the human pluripotent stem cells (hPSCs). To promote the initial differentiation of hPSCs in a carrier-free suspension stirred bioreactor into definitive endoderm, we used rapamycin for "priming" phase and activin A for induction. The cells were further differentiated into HLCs in the same system. HLCs were characterized and then purified based on their physiological function, the uptake of DiI-acetylated low-density lipoprotein (LDL) by flow cytometry without genetic manipulation or antibody labeling. The sorted cells were transplanted into the spleens of mice with acute liver injury from carbon tetrachloride. The differentiated HLCs had multiple features of primary hepatocytes, for example, the expression patterns of liver-specific marker genes, albumin secretion, urea production, collagen synthesis, indocyanin green and LDL uptake, glycogen storage, and inducible cytochrome P450 activity. They increased the survival rate, engrafted successfully into the liver, and continued to present hepatic function (i.e., albumin secretion after implantation). This amenable scaling up and outlined enrichment strategy provides a new platform for generating functional HLCs. This integrated approach may facilitate biomedical applications of the hPSC-derived hepatocytes.
Collapse
Affiliation(s)
- Massoud Vosough
- 1 Department of Biochemistry, Pasteur Institute of Iran , Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
28
|
Erro E, Bundy J, Massie I, Chalmers SA, Gautier A, Gerontas S, Hoare M, Sharratt P, Choudhury S, Lubowiecki M, Llewellyn I, Legallais C, Fuller B, Hodgson H, Selden C. Bioengineering the liver: scale-up and cool chain delivery of the liver cell biomass for clinical targeting in a bioartificial liver support system. Biores Open Access 2013; 2:1-11. [PMID: 23514704 PMCID: PMC3569957 DOI: 10.1089/biores.2012.0286] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Acute liver failure has a high mortality unless patients receive a liver transplant; however, there are insufficient donor organs to meet the clinical need. The liver may rapidly recover from acute injury by hepatic cell regeneration given time. A bioartificial liver machine can provide temporary liver support to enable such regeneration to occur. We developed a bioartificial liver machine using human-derived liver cells encapsulated in alginate, cultured in a fluidized bed bioreactor to a level of function suitable for clinical use (performance competence). HepG2 cells were encapsulated in alginate using a JetCutter to produce ∼500 μm spherical beads containing cells at ∼1.75 million cells/mL beads. Within the beads, encapsulated cells proliferated to form compact cell spheroids (AELS) with good cell-to-cell contact and cell function, that were analyzed functionally and by gene expression at mRNA and protein levels. We established a methodology to enable a ∼34-fold increase in cell density within the AELS over 11-13 days, maintaining cell viability. Optimized nutrient and oxygen provision were numerically modeled and tested experimentally, achieving a cell density at harvest of >45 million cells/mL beads; >5×10(10) cells were produced in 1100 mL of beads. This process is scalable to human size ([0.7-1]×10(11)). A short-term storage protocol at ambient temperature was established, enabling transport from laboratory to bedside over 48 h, appropriate for clinical translation of a manufactured bioartificial liver machine.
Collapse
Affiliation(s)
- Eloy Erro
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| | - James Bundy
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| | - Isobel Massie
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| | - Sherri-Ann Chalmers
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| | - Aude Gautier
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| | - Spyridon Gerontas
- The Advanced Center for Biochemical Engineering, Department of Biochemical Engineering; University College London, London, United Kingdom
| | - Mike Hoare
- The Advanced Center for Biochemical Engineering, Department of Biochemical Engineering; University College London, London, United Kingdom
| | - Peter Sharratt
- PNAC Facility, Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Choudhury
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| | - Marcin Lubowiecki
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| | - Ian Llewellyn
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| | - Cécile Legallais
- CNRS UMR 6600 Biomechanics and Bioengineering, University of Technology of Compiègne, Compiègne, France
| | - Barry Fuller
- Cell, Tissue & Organ Preservation Unit, University Department of Surgery, UCL Medical School, Royal Free Hospital Campus, London, United Kingdom
| | - Humphrey Hodgson
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| | - Clare Selden
- Liver Group, UCL Institute of Liver & Digestive Health, London, United Kingdom
| |
Collapse
|
29
|
Mandenius CF. Biomechatronics for designing bioprocess monitoring and control systems: Application to stem cell production. J Biotechnol 2012; 162:430-40. [DOI: 10.1016/j.jbiotec.2012.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 07/23/2012] [Accepted: 09/03/2012] [Indexed: 12/26/2022]
|
30
|
Soldatow VY, Lecluyse EL, Griffith LG, Rusyn I. In vitro models for liver toxicity testing. Toxicol Res (Camb) 2012; 2:23-39. [PMID: 23495363 DOI: 10.1039/c2tx20051a] [Citation(s) in RCA: 320] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Over the years, various liver-derived in vitro model systems have been developed to enable investigation of the potential adverse effects of chemicals and drugs. Liver tissue slices, isolated microsomes, perfused liver, immortalized cell lines, and primary hepatocytes have been used extensively. Immortalized cell lines and primary isolated liver cells are currently most widely used in vitro models for liver toxicity testing. Limited throughput, loss of viability, and decreases in liver-specific functionality and gene expression are common shortcomings of these models. Recent developments in the field of in vitro hepatotoxicity include three-dimensional tissue constructs and bioartificial livers, co-cultures of various cell types with hepatocytes, and differentiation of stem cells into hepatic lineage-like cells. In an attempt to provide a more physiological environment for cultured liver cells, some of the novel cell culture systems incorporate fluid flow, micro-circulation, and other forms of organotypic microenvironments. Co-cultures aim to preserve liver-specific morphology and functionality beyond those provided by cultures of pure parenchymal cells. Stem cells, both embryonic- and adult tissue-derived, may provide a limitless supply of hepatocytes from multiple individuals to improve reproducibility and enable testing of the individual-specific toxicity. This review describes various traditional and novel in vitro liver models and provides a perspective on the challenges and opportunities afforded by each individual test system.
Collapse
Affiliation(s)
- Valerie Y Soldatow
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
31
|
Wang Y, Zhang Y, Zhang S, Peng G, Liu T, Li Y, Xiang D, Wassler MJ, Shelat HS, Geng Y. Rotating Microgravity-Bioreactor Cultivation Enhances the Hepatic Differentiation of Mouse Embryonic Stem Cells on Biodegradable Polymer Scaffolds. Tissue Eng Part A 2012; 18:2376-85. [DOI: 10.1089/ten.tea.2012.0097] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Yingjie Wang
- The Artificial Liver Lab., Southwest Hospital, The Third Military Medical University, Chongqing, China
- The University of Texas Health Science Center and Texas Heart Institute, Houston, Texas
| | - Yunping Zhang
- The University of Texas Health Science Center and Texas Heart Institute, Houston, Texas
- Department of Emergency Medicine, JaoTong University, Shanghai, China
| | - Shichang Zhang
- The Artificial Liver Lab., Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Guangyong Peng
- Division of Immunobiology, Department of Internal Medicine, Saint Louis University School of Medicine, Edward A Doisy Research Center, St. Louis, Missouri
| | - Tao Liu
- The Artificial Liver Lab., Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Yangxin Li
- The University of Texas Health Science Center and Texas Heart Institute, Houston, Texas
| | - Dedong Xiang
- The Artificial Liver Lab., Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Michael J. Wassler
- The University of Texas Health Science Center and Texas Heart Institute, Houston, Texas
| | - Harnath S. Shelat
- The University of Texas Health Science Center and Texas Heart Institute, Houston, Texas
| | - Yongjian Geng
- The University of Texas Health Science Center and Texas Heart Institute, Houston, Texas
| |
Collapse
|
32
|
Mueller D, Tascher G, Damm G, Nüssler AK, Heinzle E, Noor F. Real-time in situ viability assessment in a 3D bioreactor with liver cells using resazurin assay. Cytotechnology 2012; 65:297-305. [PMID: 22828753 DOI: 10.1007/s10616-012-9486-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 07/10/2012] [Indexed: 01/18/2023] Open
Abstract
Three-dimensional cultivation of human cells is promising especially for long-term maintenance of specific functions and mimicking the in vivo tissue environment. However, direct viability assessment is very difficult in such systems. Commonly applied indirect methods such as glucose consumption, albumin or urea production are greatly affected by culture conditions, stress and time of cultivation and do not reflect the real time viability of the cells. In this study we established a real-time in situ viability assay namely; resazurin assay, in a 3D hollow-fiber bioreactor using human liver cells. Resazurin assay is based on the conversion of resazurin to a fluorescent dye by cytoplasmatic and mitochondrial enzymes. We show that the resazurin reagent in concentrations used in this study is non-toxic and could be rapidly removed out of the system. Moreover, we observed that dead cells do not affect the results of the assay. We optimized the assay on HepG2 cells and tested it with primary human hepatocytes. Moreover, we maintained primary human hepatocytes in the 3D bioreactor system in serum-free conditions and also assessed viability before and after the exposure to amiodarone using the resazurin assay. We show that this approach is applicable during long-term cultivation of cells in bioreactors under different conditions and can moreover be applied to pharmacological studies, e.g. investigation of chronic drug effects in such 3D bioreactors.
Collapse
Affiliation(s)
- Daniel Mueller
- Biochemical Engineering Institute, Saarland University, Geb. A1 5, 66123, Saarbruecken, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Nibourg GAA, Chamuleau RAFM, van Gulik TM, Hoekstra R. Proliferative human cell sources applied as biocomponent in bioartificial livers: a review. Expert Opin Biol Ther 2012; 12:905-21. [DOI: 10.1517/14712598.2012.685714] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
34
|
Mandenius CF, Björkman M. Scale-up of cell culture bioreactors using biomechatronic design. Biotechnol J 2012; 7:1026-39. [DOI: 10.1002/biot.201100463] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Revised: 02/19/2012] [Accepted: 03/01/2012] [Indexed: 01/23/2023]
|
35
|
Gridelli B, Vizzini G, Pietrosi G, Luca A, Spada M, Gruttadauria S, Cintorino D, Amico G, Chinnici C, Miki T, Schmelzer E, Conaldi PG, Triolo F, Gerlach JC. Efficient human fetal liver cell isolation protocol based on vascular perfusion for liver cell-based therapy and case report on cell transplantation. Liver Transpl 2012; 18:226-37. [PMID: 22034152 DOI: 10.1002/lt.22322] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although hepatic cell transplantation (CT) holds the promise of bridging patients with end-stage chronic liver failure to whole liver transplantation, suitable cell populations are under debate. In addition to hepatic cells, mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) are being considered as alternative cell sources for initial clinical cell work. Fetal liver (FL) tissue contains potential progenitors for all these cell lineages. Based on the collagenase incubation of tissue fragments, traditional isolation techniques yield only a fraction of the number of available cells. We report a 5-step method in which a portal vein in situ perfusion technique is used for tissue from the late second trimester. This method results in the high viabilities known for adult liver vascular perfusion, addresses the low cell yields of conventional digestion methods, and reduces the exposure of the tissue to collagenase 4-fold. We used donated tissue from gestational weeks 18 to 22, which yielded 1.8 ± 0.7 × 10(9) cells with an average viability of 78%. Because HSC transplantation and MSC transplantation are of interest for the treatment of hepatic failure, we phenotypically confirmed that in addition to hepatic progenitors, the resulting cell preparation contained cells expressing typical MSC and HSC markers. The percentage of FL cells expressing proliferation markers was 45 times greater than the percentage of adult hepatocytes expressing these markers and was comparable to the percentage of immortalized HepG2 liver hepatocellular carcinoma cells; this indicated the strong proliferative capacity of fetal cells. We report a case of human FL CT with the described liver cell population for clinical end-stage chronic liver failure. The patient's Model for End-Stage Liver Disease (MELD) score improved from 15 to 10 within the first 18 months of observation. In conclusion, this human FL cell isolation protocol may be of interest for further clinical translation work on the development of liver cell-based therapies.
Collapse
Affiliation(s)
- Bruno Gridelli
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15203, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhang M, Zhong Y, Chen J. Model systems and clinical applications of hepatic stem cells for liver regeneration. Hepatol Int 2011. [DOI: 10.1007/s12072-011-9323-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
Toni R, Tampieri A, Zini N, Strusi V, Sandri M, Dallatana D, Spaletta G, Bassoli E, Gatto A, Ferrari A, Martin I. Ex situ bioengineering of bioartificial endocrine glands: A new frontier in regenerative medicine of soft tissue organs. Ann Anat 2011; 193:381-94. [DOI: 10.1016/j.aanat.2011.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 06/14/2011] [Accepted: 06/17/2011] [Indexed: 01/14/2023]
|
38
|
Zhang SC, Liu T, Wang YJ. Porous and single-skinned polyethersulfone membranes support the growth of HepG2 cells: A potential biomaterial for bioartificial liver systems. J Biomater Appl 2011; 27:359-66. [PMID: 21750186 DOI: 10.1177/0885328211406299] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In this study, we evaluated a porous and single-layer skin polyethersulfone (PES) membrane as a material for use in hybrid bioartificial liver support systems. The PES membrane has been characterized as a single-layer skin structure, with a rough porous surface. Specifically, we studied the ability of the human hepatoblastoma cell lines (HepG2) to adhere, grow, and spread on the PES membrane. Furthermore, we examined albumin secretion, low-density lipoprotein uptake, and CYP450 activity of HepG2 cells that grew on the membrane. HepG2 cells readily adhered onto the outer surfaces of PES membranes. Over time, HepG2 cells proliferated actively, and confluent monolayer of cells covered the available surface area of the membrane, eventually forming cell clusters and three-dimensional aggregates. Furthermore, HepG2 cells grown on PES membranes maintained highly specific functions, including uptake capability, biosynthesis and biotransformation. These results indicate that PES membranes are potential substrates for the growth of human liver cells and may be useful in the construction of hollow fiber bioreactors. Porous and single-layer skin PES membranes and HepG2 cells may be potential biomaterials for the development of biohybrid liver devices.
Collapse
Affiliation(s)
- Shi-Chang Zhang
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Tao Liu
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ying-Jie Wang
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
39
|
Prestwich GD. Hyaluronic acid-based clinical biomaterials derived for cell and molecule delivery in regenerative medicine. J Control Release 2011; 155:193-9. [PMID: 21513749 DOI: 10.1016/j.jconrel.2011.04.007] [Citation(s) in RCA: 270] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Revised: 04/04/2011] [Accepted: 04/04/2011] [Indexed: 02/01/2023]
Abstract
The development of injectable and biocompatible vehicles for delivery, retention, growth, and differentiation of stem cells is of paramount importance for regenerative medicine. For cell therapy and the development of clinical combination products, we created a hyaluronan (HA)-based synthetic extracellular matrix (sECM) that provides highly reproducible, manufacturable, approvable, and affordable biomaterials. The composition of the sECM can be customized for use with progenitor and mature cell populations obtained from skin, fat, liver, heart, muscle, bone, cartilage, nerves, and other tissues. This overview describes the design criteria for "living" HA derivatives, and the many uses of this in situ crosslinkable HA-based sECM hydrogel for three-dimensional (3-D) culture of cells in vitro and translational use in vivo. Recent advances allow rapid expansion and recovery of cells in 3-D, and the bioprinting of engineered tissue constructs. The uses of HA-derived sECMs for cell and molecule delivery in vivo will be reviewed, including applications in cancer biology and tumor imaging.
Collapse
Affiliation(s)
- Glenn D Prestwich
- Center for Therapeutic Biomaterials and Department of Medicinal Chemistry, University of Utah, 419 Wakara Way #205, Salt Lake City, UT 84108-1257, USA.
| |
Collapse
|
40
|
Burdick JA, Prestwich GD. Hyaluronic acid hydrogels for biomedical applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H41-56. [PMID: 21394792 PMCID: PMC3730855 DOI: 10.1002/adma.201003963] [Citation(s) in RCA: 1371] [Impact Index Per Article: 97.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 01/03/2011] [Indexed: 05/10/2023]
Abstract
Hyaluronic acid (HA), an immunoneutral polysaccharide that is ubiquitous in the human body, is crucial for many cellular and tissue functions and has been in clinical use for over thirty years. When chemically modified, HA can be transformed into many physical forms-viscoelastic solutions, soft or stiff hydrogels, electrospun fibers, non-woven meshes, macroporous and fibrillar sponges, flexible sheets, and nanoparticulate fluids-for use in a range of preclinical and clinical settings. Many of these forms are derived from the chemical crosslinking of pendant reactive groups by addition/condensation chemistry or by radical polymerization. Clinical products for cell therapy and regenerative medicine require crosslinking chemistry that is compatible with the encapsulation of cells and injection into tissues. Moreover, an injectable clinical biomaterial must meet marketing, regulatory, and financial constraints to provide affordable products that can be approved, deployed to the clinic, and used by physicians. Many HA-derived hydrogels meet these criteria, and can deliver cells and therapeutic agents for tissue repair and regeneration. This progress report covers both basic concepts and recent advances in the development of HA-based hydrogels for biomedical applications.
Collapse
Affiliation(s)
- Jason A. Burdick
- Prof. J.A. Burdick, Department of Bioengineering, University of Pennsylvania, 210 S 33th Street, Philadelphia, PA 19104 (USA),
| | - Glenn D. Prestwich
- Prof. G.D. Prestwich, Department of Medicinal Chemistry and Center for Therapeutic Biomaterials, University of Utah, 419 Wakara Way, Suite 205, Salt Lake City, UT 84108 (USA),
| |
Collapse
|
41
|
Abstract
A bioreactor is defined as a specifically designed vessel to facilitate the growth of organisms and cells through application of physical and/or electrical stimulus. When cells with therapeutic potential were first discovered, they were initially cultured and expanded in two-dimensional (2-D) culture vessels such as plates or T-flasks. However, it was soon discovered that bioreactors could be used to expand and maintain cultures more easily and efficiently. Since then, bioreactors have come to be accepted as an indispensable tool to advance cell and tissue culture further. A wide array of bioreactors has been developed to date, and in recent years businesses have started supplying bioreactors commercially. Bioreactors in the research arena range from stirred tank bioreactors for suspension culture to those with various mechanical actuators that can apply different fluidic and mechanical stresses to tissues and three-dimensional (3-D) scaffolds. As regenerative medicine gains more traction in the clinic, bioreactors for use with cellular therapies are being developed and marketed. While many of the simpler bioreactors are fit for purpose, others fail to satisfy the complex requirements of tissues in culture. We have examined the use of different types of bioreactors in regenerative medicine and evaluated the application of bioreactors in the realization of emerging cellular therapies.
Collapse
Affiliation(s)
- M W Naing
- Healthcare Engineering Research Group, Centre for Biological Engineering, Loughborough University, Loughborough, UK
| | | |
Collapse
|
42
|
Wobus AM, Löser P. Present state and future perspectives of using pluripotent stem cells in toxicology research. Arch Toxicol 2011; 85:79-117. [PMID: 21225242 PMCID: PMC3026927 DOI: 10.1007/s00204-010-0641-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 12/21/2010] [Indexed: 02/08/2023]
Abstract
The use of novel drugs and chemicals requires reliable data on their potential toxic effects on humans. Current test systems are mainly based on animals or in vitro–cultured animal-derived cells and do not or not sufficiently mirror the situation in humans. Therefore, in vitro models based on human pluripotent stem cells (hPSCs) have become an attractive alternative. The article summarizes the characteristics of pluripotent stem cells, including embryonic carcinoma and embryonic germ cells, and discusses the potential of pluripotent stem cells for safety pharmacology and toxicology. Special attention is directed to the potential application of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) for the assessment of developmental toxicology as well as cardio- and hepatotoxicology. With respect to embryotoxicology, recent achievements of the embryonic stem cell test (EST) are described and current limitations as well as prospects of embryotoxicity studies using pluripotent stem cells are discussed. Furthermore, recent efforts to establish hPSC-based cell models for testing cardio- and hepatotoxicity are presented. In this context, methods for differentiation and selection of cardiac and hepatic cells from hPSCs are summarized, requirements and implications with respect to the use of these cells in safety pharmacology and toxicology are presented, and future challenges and perspectives of using hPSCs are discussed.
Collapse
Affiliation(s)
- Anna M Wobus
- In Vitro Differentiation Group, Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Corrensstr. 3, 06466 Gatersleben, Germany.
| | | |
Collapse
|
43
|
Katsuda T, Teratani T, Ochiya T, Sakai Y. Transplantation of a fetal liver cell-loaded hyaluronic acid sponge onto the mesentery recovers a Wilson's disease model rat. J Biochem 2010; 148:281-8. [PMID: 20562412 DOI: 10.1093/jb/mvq063] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
An auxiliary liver represents a promising alternative for liver transplantation. The use of a large amount of mature hepatocytes, however, despite their high function, is limited in a clinical setting. Here, we propose a novel transplantation system that dramatically improved a diseased animal by incorporating fetal liver cells (FLCs) as a cell source, the mesentery as a transplantation site and a hyaluronic acid (HA) sponge as a cell scaffold. We transplanted wild-type Long Evans Agouti rat FLCs embedded in HA sponges onto the mesentery of Long Evans Cinnamon (LEC) rats, an animal model for Wilson's disease. The FLC-loaded HA sponges successfully grafted and consequently prevented jaundice. Accordingly, the treated animals showed a significant reduction in blood copper concentration, which consequently led to significant decreases in serum total bilirubin and direct bilirubin, and to a significant increase in albumin productivity. Furthermore, haematoxylin and eosin staining of the host livers demonstrated that fibrosis at the periportal area was moderated in the treated animals. In conclusion, we transplanted FLC-loaded HA sponges onto the mesenteric blood vessels, leading to thick, liver-like tissue possessing blood vessels, and the liver tissue engineered thus exhibited a remarkable therapeutic effect on the copper metabolism deficiency of LEC rats.
Collapse
Affiliation(s)
- Takeshi Katsuda
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan.
| | | | | | | |
Collapse
|
44
|
Mechatronics design principles for biotechnology product development. Trends Biotechnol 2010; 28:230-6. [DOI: 10.1016/j.tibtech.2010.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 02/10/2010] [Accepted: 02/17/2010] [Indexed: 11/19/2022]
|