1
|
Wojciechowski C, Wasyłeczko M, Lewińska D, Chwojnowski A. A Comprehensive Review of Hollow-Fiber Membrane Fabrication Methods across Biomedical, Biotechnological, and Environmental Domains. Molecules 2024; 29:2637. [PMID: 38893513 PMCID: PMC11174095 DOI: 10.3390/molecules29112637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
This work presents methods of obtaining polymeric hollow-fiber membranes produced via the dry-wet phase inversion method that were published in renowned specialized membrane publications in the years 2010-2020. Obtaining hollow-fiber membranes, unlike flat membranes, requires the use of a special installation for their production, the most important component of which is the hollow fiber forming spinneret. This method is most often used in obtaining membranes made of polysulfone, polyethersulfone, polyurethane, cellulose acetate, and its derivatives. Many factors affect the properties of the membranes obtained. By changing the parameters of the spinning process, we change the thickness of the membranes' walls and the diameter of the hollow fibers, which causes changes in the membranes' structure and, as a consequence, changes in their transport/separation parameters. The type of bore fluid affects the porosity of the inner epidermal layer or causes its atrophy. Porogenic compounds such as polyvinylpyrrolidones and polyethylene glycols and other substances that additionally increase the membrane porosity are often added to the polymer solution. Another example is a blend of two- or multi-component membranes and dual-layer membranes that are obtained using a three-nozzle spinneret. In dual-layer membranes, one layer is the membrane scaffolding, and the other is the separation layer. Also, the temperature during the process, the humidity, and the composition of the solution in the coagulating bath have impact on the parameters of the membranes obtained.
Collapse
Affiliation(s)
- Cezary Wojciechowski
- Nalecz Institute of Biocybernetic and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4 Str., 02-109 Warsaw, Poland; (M.W.); (D.L.); (A.C.)
| | | | | | | |
Collapse
|
2
|
Wasyłeczko M, Wojciechowski C, Chwojnowski A. Polyethersulfone Polymer for Biomedical Applications and Biotechnology. Int J Mol Sci 2024; 25:4233. [PMID: 38673817 PMCID: PMC11049998 DOI: 10.3390/ijms25084233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Polymers stand out as promising materials extensively employed in biomedicine and biotechnology. Their versatile applications owe much to the field of tissue engineering, which seamlessly integrates materials engineering with medical science. In medicine, biomaterials serve as prototypes for organ development and as implants or scaffolds to facilitate body regeneration. With the growing demand for innovative solutions, synthetic and hybrid polymer materials, such as polyethersulfone, are gaining traction. This article offers a concise characterization of polyethersulfone followed by an exploration of its diverse applications in medical and biotechnological realms. It concludes by summarizing the significant roles of polyethersulfone in advancing both medicine and biotechnology, as outlined in the accompanying table.
Collapse
Affiliation(s)
- Monika Wasyłeczko
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ksiecia Trojdena 4, 02-109 Warsaw, Poland; (C.W.); (A.C.)
| | | | | |
Collapse
|
3
|
Huang K, Li Q, Xue Y, Wang Q, Chen Z, Gu Z. Application of colloidal photonic crystals in study of organoids. Adv Drug Deliv Rev 2023; 201:115075. [PMID: 37625595 DOI: 10.1016/j.addr.2023.115075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 07/09/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
As alternative disease models, other than 2D cell lines and patient-derived xenografts, organoids have preferable in vivo physiological relevance. However, both endogenous and exogenous limitations impede the development and clinical translation of these organoids. Fortunately, colloidal photonic crystals (PCs), which benefit from favorable biocompatibility, brilliant optical manipulation, and facile chemical decoration, have been applied to the engineering of organoids and have achieved the desirable recapitulation of the ECM niche, well-defined geometrical onsets for initial culture, in situ multiphysiological parameter monitoring, single-cell biomechanical sensing, and high-throughput drug screening with versatile functional readouts. Herein, we review the latest progress in engineering organoids fabricated from colloidal PCs and provide inputs for future research.
Collapse
Affiliation(s)
- Kai Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yufei Xue
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Qiong Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China.
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
4
|
Liu T, Ge Y, Chen Z, Wu L, Tian T, Yao W, Zhao J. Synergistic Modulation of a Tunable Microenvironment to Fabricate a Liver Fibrosis Chip for Drug Testing. ACS Biomater Sci Eng 2023; 9:4893-4906. [PMID: 37523767 DOI: 10.1021/acsbiomaterials.3c00213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Liver fibrosis is a progressive physiological change that occurs after liver injury and seriously endangers human health. The lack of reliable and physiologically relevant pathological models of liver fibrosis leads to a longer drug development period and sizeable economic investment. The fabrication of a biomimetic liver-on-a-chip is significant for liver disease treatment and drug development. Here, a sandwich chip with a microwell array structure in its bottom layer was fabricated to simulate the Disse space structure of hepatic sinusoids in vitro. By synergistic modulation of the cross-linking degree of gelatin-methacryloyl (GelMA) hydrogels and the induction of transforming growth factor-beta (TGF-β), the early and late stages of liver fibrosis were designed in the chip. Owing to its three-dimensional-mixed-culture strategy, it was possible to construct a liver sinusoid model in vitro to allow for faithful physiological emulation. The model was further subjected to drug treatment, and it presented a significant difference in treatment response in early and late fibrosis progression. Our system provides a unique method for emulating liver function through a vitro liver fibrosis-on-a-chip, potentially paving the way for investigating human liver fibrosis and related drug development.
Collapse
Affiliation(s)
- Ting Liu
- Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P. R. China
- Xiangfu Laboratory, Jiashan 314102, P. R. China
| | - Yuqing Ge
- Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P. R. China
| | - Zhongping Chen
- Institute of Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, P. R. China
| | - Lei Wu
- Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P. R. China
| | - Tian Tian
- Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P. R. China
| | - Wei Yao
- Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P. R. China
| | - Jianlong Zhao
- Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China
| |
Collapse
|
5
|
Nikolopoulos VK, Augustine R, Camci-Unal G. Harnessing the potential of oxygen-generating materials and their utilization in organ-specific delivery of oxygen. Biomater Sci 2023; 11:1567-1588. [PMID: 36688522 PMCID: PMC10015602 DOI: 10.1039/d2bm01329k] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The limited availability of transplantable organs hinders the success of patient treatment through organ transplantation. In addition, there are challenges with immune rejection and the risk of disease transmission when receiving organs from other individuals. Tissue engineering aims to overcome these challenges by generating functional three-dimensional (3D) tissue constructs. When developing tissues or organs of a particular shape, structure, and size as determined by the specific needs of the therapeutic intervention, a tissue specific oxygen supply to all parts of the tissue construct is an utmost requirement. Moreover, the lack of a functional vasculature in engineered tissues decreases cell survival upon implantation in the body. Oxygen-generating materials can alleviate this challenge in engineered tissue constructs by providing oxygen in a sustained and controlled manner. Oxygen-generating materials can be incorporated into 3D scaffolds allowing the cells to receive and utilize oxygen efficiently. In this review, we present an overview of the use of oxygen-generating materials in various tissue engineering applications in an organ specific manner as well as their potential use in the clinic.
Collapse
Affiliation(s)
- Vasilios K Nikolopoulos
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
| | - Robin Augustine
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
6
|
Carpentier N, Urbani L, Dubruel P, Van Vlierberghe S. The native liver as inspiration to create superior in vitro hepatic models. Biomater Sci 2023; 11:1091-1115. [PMID: 36594602 DOI: 10.1039/d2bm01646j] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Drug induced liver injury (DILI) is one of the major reasons of drug withdrawal during the different phases of drug development. The later in the drug development a drug is discovered to be toxic, the higher the economical as well as the ethical impact will be. In vitro models for early detection of drug liver toxicity are under constant development, however to date a superior model of the liver is still lacking. Ideally, a highly reliable model should be established to maintain the different hepatic cell functionalities to the greatest extent possible, during a period of time long enough to allow for tracking of the toxicity of compounds. In the case of DILI, toxicity can appear even after months of exposure. To reach this goal, an in vitro model should be developed that mimics the in vivo liver environment, function and response to external stimuli. The different approaches for the development of liver models currently used in the field of tissue engineering will be described in this review. Combining different technologies, leading to optimal materials, cells and 3D-constructs will ultimately lead to an ideal superior model that fully recapitulates the liver.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Luca Urbani
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK.,Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| |
Collapse
|
7
|
Ray P, Chakraborty R, Banik O, Banoth E, Kumar P. Surface Engineering of a Bioartificial Membrane for Its Application in Bioengineering Devices. ACS OMEGA 2023; 8:3606-3629. [PMID: 36743049 PMCID: PMC9893455 DOI: 10.1021/acsomega.2c05983] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/04/2023] [Indexed: 06/18/2023]
Abstract
Membrane technology is playing a crucial role in cutting-edge innovations in the biomedical field. One such innovation is the surface engineering of a membrane for enhanced longevity, efficient separation, and better throughput. Hence, surface engineering is widely used while developing membranes for its use in bioartificial organ development, separation processes, extracorporeal devices, etc. Chemical-based surface modifications are usually performed by functional group/biomolecule grafting, surface moiety modification, and altercation of hydrophilic and hydrophobic properties. Further, creation of micro/nanogrooves, pillars, channel networks, and other topologies is achieved to modify physio-mechanical processes. These surface modifications facilitate improved cellular attachment, directional migration, and communication among the neighboring cells and enhanced diffusional transport of nutrients, gases, and waste across the membrane. These modifications, apart from improving functional efficiency, also help in overcoming fouling issues, biofilm formation, and infection incidences. Multiple strategies are adopted, like lysozyme enzymatic action, topographical modifications, nanomaterial coating, and antibiotic/antibacterial agent doping in the membrane to counter the challenges of biofilm formation, fouling challenges, and microbial invasion. Therefore, in the current review, we have comprehensibly discussed different types of membranes, their fabrication and surface modifications, antifouling/antibacterial strategies, and their applications in bioengineering. Thus, this review would benefit bioengineers and membrane scientists who aim to improve membranes for applications in tissue engineering, bioseparation, extra corporeal membrane devices, wound healing, and others.
Collapse
Affiliation(s)
- Pragyan Ray
- BioDesign
and Medical Devices Laboratory, Department of Biotechnology and Medical
Engineering, National Institute of Technology,
Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Ruchira Chakraborty
- BioDesign
and Medical Devices Laboratory, Department of Biotechnology and Medical
Engineering, National Institute of Technology,
Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Oindrila Banik
- BioDesign
and Medical Devices Laboratory, Department of Biotechnology and Medical
Engineering, National Institute of Technology,
Rourkela, Sector-1, Rourkela 769008, Odisha, India
- Opto-Biomedical
Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Earu Banoth
- Opto-Biomedical
Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Prasoon Kumar
- BioDesign
and Medical Devices Laboratory, Department of Biotechnology and Medical
Engineering, National Institute of Technology,
Rourkela, Sector-1, Rourkela 769008, Odisha, India
| |
Collapse
|
8
|
Camponogara F, Zanotti F, Trentini M, Tiengo E, Zanolla I, Pishavar E, Soliani E, Scatto M, Gargiulo P, Zambito Y, De Luca S, Ferroni L, Zavan B. Biomaterials for Regenerative Medicine in Italy: Brief State of the Art of the Principal Research Centers. Int J Mol Sci 2022; 23:8245. [PMID: 35897825 PMCID: PMC9368060 DOI: 10.3390/ijms23158245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/30/2022] [Accepted: 07/22/2022] [Indexed: 02/01/2023] Open
Abstract
Regenerative medicine is the branch of medicine that effectively uses stem cell therapy and tissue engineering strategies to guide the healing or replacement of damaged tissues or organs. A crucial element is undoubtedly the biomaterial that guides biological events to restore tissue continuity. The polymers, natural or synthetic, find wide application thanks to their great adaptability. In fact, they can be used as principal components, coatings or vehicles to functionalize several biomaterials. There are many leading centers for the research and development of biomaterials in Italy. The aim of this review is to provide an overview of the current state of the art on polymer research for regenerative medicine purposes. The last five years of scientific production of the main Italian research centers has been screened to analyze the current advancement in tissue engineering in order to highlight inputs for the development of novel biomaterials and strategies.
Collapse
Affiliation(s)
- Francesca Camponogara
- Translational Medicine Department, University of Ferrara, 44121 Ferrara, Italy; (F.C.); (F.Z.); (M.T.); (E.T.); (E.P.)
| | - Federica Zanotti
- Translational Medicine Department, University of Ferrara, 44121 Ferrara, Italy; (F.C.); (F.Z.); (M.T.); (E.T.); (E.P.)
| | - Martina Trentini
- Translational Medicine Department, University of Ferrara, 44121 Ferrara, Italy; (F.C.); (F.Z.); (M.T.); (E.T.); (E.P.)
| | - Elena Tiengo
- Translational Medicine Department, University of Ferrara, 44121 Ferrara, Italy; (F.C.); (F.Z.); (M.T.); (E.T.); (E.P.)
| | - Ilaria Zanolla
- Medical Sciences Department, University of Ferrara, 44121 Ferrara, Italy;
| | - Elham Pishavar
- Translational Medicine Department, University of Ferrara, 44121 Ferrara, Italy; (F.C.); (F.Z.); (M.T.); (E.T.); (E.P.)
| | - Elisa Soliani
- Bioengineering Department, Imperial College London, London SW7 2BX, UK;
| | - Marco Scatto
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Via Torino 155, 30172 Venezia, Italy;
| | - Paolo Gargiulo
- Institute for Biomedical and Neural Engineering, Reykjavík University, 101 Reykjavík, Iceland;
- Department of Science, Landspítali, 101 Reykjavík, Iceland
| | - Ylenia Zambito
- Chemical Department, University of Pisa, 56124 Pisa, Italy;
| | - Stefano De Luca
- Unit of Naples, Institute of Applied Sciences and Intelligent Systems, National Research Council, Via P. Castellino 111, 80131 Napoli, Italy;
| | - Letizia Ferroni
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy;
| | - Barbara Zavan
- Translational Medicine Department, University of Ferrara, 44121 Ferrara, Italy; (F.C.); (F.Z.); (M.T.); (E.T.); (E.P.)
| |
Collapse
|
9
|
Hou Y, Mi K, Sun L, Zhou K, Wang L, Zhang L, Liu Z, Huang L. The Application of Hollow Fiber Cartridge in Biomedicine. Pharmaceutics 2022; 14:pharmaceutics14071485. [PMID: 35890380 PMCID: PMC9316653 DOI: 10.3390/pharmaceutics14071485] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
The hollow fiber cartridge has the advantages of good semi-permeability, high surface area to volume ratio, convenient operation, and so on. Its application in chemical analysis, drug in vitro experiment, hemodialysis, and other fields has been deeply studied. This paper introduces the basic structure of hollow fiber cartridge, compares the advantages and disadvantages of a hollow fiber infection model constructed by a hollow fiber cartridge with traditional static model and animal infection model and introduces its application in drug effects, mechanism of drug resistance, and evaluation of combined drug regimen. The principle and application of hollow fiber bioreactors for cell culture and hollow fiber dialyzer for dialysis and filtration were discussed. The hollow fiber cartridge, whether used in drug experiments, artificial liver, artificial kidney, etc., has achieved controllable experimental operation and efficient and accurate experimental results, and will provide more convenience and support for drug development and clinical research in the future.
Collapse
Affiliation(s)
- Yixuan Hou
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China; (Y.H.); (K.M.); (Z.L.)
| | - Kun Mi
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China; (Y.H.); (K.M.); (Z.L.)
| | - Lei Sun
- MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (K.Z.); (L.W.)
| | - Kaixiang Zhou
- MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (K.Z.); (L.W.)
| | - Lei Wang
- MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (K.Z.); (L.W.)
| | - Lan Zhang
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China;
| | - Zhenli Liu
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China; (Y.H.); (K.M.); (Z.L.)
- MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (K.Z.); (L.W.)
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China; (Y.H.); (K.M.); (Z.L.)
- MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (K.Z.); (L.W.)
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China;
- Correspondence:
| |
Collapse
|
10
|
Tuerxun K, He J, Ibrahim I, Yusupu Z, Yasheng A, Xu Q, Tang R, Aikebaier A, Wu Y, Tuerdi M, Nijiati M, Zou X, Xu T. Bioartificial livers: a review of their design and manufacture. Biofabrication 2022; 14. [PMID: 35545058 DOI: 10.1088/1758-5090/ac6e86] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
Acute liver failure (ALF) is a rapidly progressive disease with high morbidity and mortality rates. Liver transplantation and artificial liver support systems, such as artificial livers (ALs) and bioartificial livers (BALs), are the two major therapies for ALF. Compared to ALs, BALs are composed of functional hepatocytes that provide essential liver functions, including detoxification, metabolite synthesis, and biotransformation. Furthermore, BALs can potentially provide effective support as a form of bridging therapy to liver transplantation or spontaneous recovery for patients with ALF. In this review, we systematically discussed the currently available state-of-the-art designs and manufacturing processes for BAL support systems. Specifically, we classified the cell sources and bioreactors that are applied in BALs, highlighted the advanced technologies of hepatocyte culturing and bioreactor fabrication, and discussed the current challenges and future trends in developing next generation BALs for large scale clinical applications.
Collapse
Affiliation(s)
- Kahaer Tuerxun
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Jianyu He
- Department of Mechanical Engineering, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing, Beijing, 100084, CHINA
| | - Irxat Ibrahim
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Zainuer Yusupu
- Department of Ultrasound, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Abudoukeyimu Yasheng
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Qilin Xu
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Ronghua Tang
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Aizemaiti Aikebaier
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Yuanquan Wu
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Maimaitituerxun Tuerdi
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Mayidili Nijiati
- Medical imaging center, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Xiaoguang Zou
- Hospital Organ, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Tao Xu
- Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing, 100084, CHINA
| |
Collapse
|
11
|
Stengelin E, Thiele J, Seiffert S. Multiparametric Material Functionality of Microtissue-Based In Vitro Models as Alternatives to Animal Testing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105319. [PMID: 35043598 PMCID: PMC8981905 DOI: 10.1002/advs.202105319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Indexed: 05/12/2023]
Abstract
With the definition of the 3R principle by Russel and Burch in 1959, the search for an adequate substitute for animal testing has become one of the most important tasks and challenges of this time, not only from an ethical, but also from a scientific, economic, and legal point of view. Microtissue-based in vitro model systems offer a valuable approach to address this issue by accounting for the complexity of natural tissues in a simplified manner. To increase the functionality of these model systems and thus make their use as a substitute for animal testing more likely in the future, the fundamentals need to be continuously improved. Corresponding requirements exist in the development of multifunctional, hydrogel-based materials, whose properties are considered in this review under the aspects of processability, adaptivity, biocompatibility, and stability/degradability.
Collapse
Affiliation(s)
- Elena Stengelin
- Department of ChemistryJohannes Gutenberg‐University MainzD‐55128MainzGermany
| | - Julian Thiele
- Leibniz‐Institut für Polymerforschung Dresden e.V.Hohe Straße 6D‐01069DresdenGermany
| | - Sebastian Seiffert
- Department of ChemistryJohannes Gutenberg‐University MainzD‐55128MainzGermany
| |
Collapse
|
12
|
de Hoyos-Vega JM, Hong HJ, Stybayeva G, Revzin A. Hepatocyte cultures: From collagen gel sandwiches to microfluidic devices with integrated biosensors. APL Bioeng 2021; 5:041504. [PMID: 34703968 PMCID: PMC8519630 DOI: 10.1063/5.0058798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocytes are parenchymal cells of the liver responsible for drug detoxification, urea and bile production, serum protein synthesis, and glucose homeostasis. Hepatocytes are widely used for drug toxicity studies in bioartificial liver devices and for cell-based liver therapies. Because hepatocytes are highly differentiated cells residing in a complex microenvironment in vivo, they tend to lose hepatic phenotype and function in vitro. This paper first reviews traditional culture approaches used to rescue hepatic function in vitro and then discusses the benefits of emerging microfluidic-based culture approaches. We conclude by reviewing integration of hepatocyte cultures with bioanalytical or sensing approaches.
Collapse
Affiliation(s)
- Jose M. de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| |
Collapse
|
13
|
Rizki-Safitri A, Tokito F, Nishikawa M, Tanaka M, Maeda K, Kusuhara H, Sakai Y. Prospect of in vitro Bile Fluids Collection in Improving Cell-Based Assay of Liver Function. FRONTIERS IN TOXICOLOGY 2021; 3:657432. [PMID: 35295147 PMCID: PMC8915818 DOI: 10.3389/ftox.2021.657432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
The liver plays a pivotal role in the clearance of drugs. Reliable assays for liver function are crucial for various metabolism investigation, including toxicity, disease, and pre-clinical testing for drug development. Bile is an aqueous secretion of a functioning liver. Analyses of bile are used to explain drug clearance and related effects and are thus important for toxicology and pharmacokinetic research. Bile fluids collection is extensively performed in vivo, whereas this process is rarely reproduced as in the in vitro studies. The key to success is the technology involved, which needs to satisfy multiple criteria. To ensure the accuracy of subsequent chemical analyses, certain amounts of bile are needed. Additionally, non-invasive and continuous collections are preferable in view of cell culture. In this review, we summarize recent progress and limitations in the field. We highlight attempts to develop advanced liver cultures for bile fluids collection, including methods to stimulate the secretion of bile in vitro. With these strategies, researchers have used a variety of cell sources, extracellular matrix proteins, and growth factors to investigate different cell-culture environments, including three-dimensional spheroids, cocultures, and microfluidic devices. Effective combinations of expertise and technology have the potential to overcome these obstacles to achieve reliable in vitro bile assay systems.
Collapse
Affiliation(s)
- Astia Rizki-Safitri
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Fumiya Tokito
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Masaki Nishikawa
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Minoru Tanaka
- Laboratory of Stem Cell Regulation, Institute for Quantitative Biosciences (IQB), The University of Tokyo, Tokyo, Japan
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
14
|
Mansouri M, Leipzig ND. Advances in removing mass transport limitations for more physiologically relevant in vitro 3D cell constructs. BIOPHYSICS REVIEWS 2021; 2:021305. [PMID: 38505119 PMCID: PMC10903443 DOI: 10.1063/5.0048837] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/31/2021] [Indexed: 03/21/2024]
Abstract
Spheroids and organoids are promising models for biomedical applications ranging from human disease modeling to drug discovery. A main goal of these 3D cell-based platforms is to recapitulate important physiological parameters of their in vivo organ counterparts. One way to achieve improved biomimetic architectures and functions is to culture cells at higher density and larger total numbers. However, poor nutrient and waste transport lead to low stability, survival, and functionality over extended periods of time, presenting outstanding challenges in this field. Fortunately, important improvements in culture strategies have enhanced the survival and function of cells within engineered microtissues/organs. Here, we first discuss the challenges of growing large spheroids/organoids with a focus on mass transport limitations, then highlight recent tools and methodologies that are available for producing and sustaining functional 3D in vitro models. This information points toward the fact that there is a critical need for the continued development of novel cell culture strategies that address mass transport in a physiologically relevant human setting to generate long-lasting and large-sized spheroids/organoids.
Collapse
Affiliation(s)
- Mona Mansouri
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| | - Nic D. Leipzig
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| |
Collapse
|
15
|
Morelli S, Piscioneri A, Salerno S, De Bartolo L. Hollow Fiber and Nanofiber Membranes in Bioartificial Liver and Neuronal Tissue Engineering. Cells Tissues Organs 2021; 211:447-476. [PMID: 33849029 DOI: 10.1159/000511680] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 09/16/2020] [Indexed: 11/19/2022] Open
Abstract
To date, the creation of biomimetic devices for the regeneration and repair of injured or diseased tissues and organs remains a crucial challenge in tissue engineering. Membrane technology offers advanced approaches to realize multifunctional tools with permissive environments well-controlled at molecular level for the development of functional tissues and organs. Membranes in fiber configuration with precisely controlled, tunable topography, and physical, biochemical, and mechanical cues, can direct and control the function of different kinds of cells toward the recovery from disorders and injuries. At the same time, fiber tools also provide the potential to model diseases in vitro for investigating specific biological phenomena as well as for drug testing. The purpose of this review is to present an overview of the literature concerning the development of hollow fibers and electrospun fiber membranes used in bioartificial organs, tissue engineered constructs, and in vitro bioreactors. With the aim to highlight the main biomedical applications of fiber-based systems, the first part reviews the fibers for bioartificial liver and liver tissue engineering with special attention to their multifunctional role in the long-term maintenance of specific liver functions and in driving hepatocyte differentiation. The second part reports the fiber-based systems used for neuronal tissue applications including advanced approaches for the creation of novel nerve conduits and in vitro models of brain tissue. Besides presenting recent advances and achievements, this work also delineates existing limitations and highlights emerging possibilities and future prospects in this field.
Collapse
Affiliation(s)
- Sabrina Morelli
- Institute on Membrane Technology, National Research Council of Italy, CNR-ITM, Rende, Italy
| | - Antonella Piscioneri
- Institute on Membrane Technology, National Research Council of Italy, CNR-ITM, Rende, Italy
| | - Simona Salerno
- Institute on Membrane Technology, National Research Council of Italy, CNR-ITM, Rende, Italy
| | - Loredana De Bartolo
- Institute on Membrane Technology, National Research Council of Italy, CNR-ITM, Rende, Italy
| |
Collapse
|
16
|
Agarwal T, Kazemi S, Costantini M, Perfeito F, Correia CR, Gaspar V, Montazeri L, De Maria C, Mano JF, Vosough M, Makvandi P, Maiti TK. Oxygen releasing materials: Towards addressing the hypoxia-related issues in tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111896. [PMID: 33641899 DOI: 10.1016/j.msec.2021.111896] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
Manufacturing macroscale cell-laden architectures is one of the biggest challenges faced nowadays in the domain of tissue engineering. Such living constructs, in fact, pose strict requirements for nutrients and oxygen supply that can hardly be addressed through simple diffusion in vitro or without a functional vasculature in vivo. In this context, in the last two decades, a substantial amount of work has been carried out to develop smart materials that could actively provide oxygen-release to contrast local hypoxia in large-size constructs. This review provides an overview of the currently available oxygen-releasing materials and their synthesis and mechanism of action, highlighting their capacities under in vitro tissue cultures and in vivo contexts. Additionally, we also showcase an emerging concept, herein termed as "living materials as releasing systems", which relies on the combination of biomaterials with photosynthetic microorganisms, namely algae, in an "unconventional" attempt to supply the damaged or re-growing tissue with the necessary supply of oxygen. We envision that future advances focusing on tissue microenvironment regulated oxygen-supplying materials would unlock an untapped potential for generating a repertoire of anatomic scale, living constructs with improved cell survival, guided differentiation, and tissue-specific biofunctionality.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Sara Kazemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marco Costantini
- Institute of Physical Chemistry - Polish Academy of Sciences, Warsaw, Poland
| | - Francisca Perfeito
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Clara R Correia
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - Vítor Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Carmelo De Maria
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Pooyan Makvandi
- Center for MicroBioRobotics (CMBR), Istituto Italiano di Tecnologia, Pisa, Italy
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| |
Collapse
|
17
|
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: 4.3] [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
|
18
|
Jung DJ, Byeon JH, Jeong GS. Flow enhances phenotypic and maturation of adult rat liver organoids. Biofabrication 2020; 12:045035. [PMID: 33000764 DOI: 10.1088/1758-5090/abb538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A biologically relevant in vitro model of hepatic microtissue would be a valuable tool for the preclinical study of pharmacokinetics and metabolism. Although considerable advances have been made in recent years in the establishment of alternative in vitro culture systems that mimic liver tissue, generating an effective liver model remains challenging. Specifically, existing model systems still exhibit limited functions for hepatocellular differentiation potential and cellular complexity. It is essential to improve the in vitro differentiation of liver progenitor cells (LPCs) for disease modeling and preclinical pharmatoxicological research. Here, we describe a rat liver organoid culture system under in vivo-like steady-state flow conditions; this system is capable of controlling the expansion and differentiation of rat liver organoids over 10-15 d. LPCs cultured in medium flow conditions become self-assembled liver organoids that exhibit phenotypic and functional hepato-biliary modeling. In addition, hepatocytes that are differentiated using liver organoids produced albumin and maintained polygonal morphology, which is characteristic of mature hepatocytes.
Collapse
Affiliation(s)
- Da Jung Jung
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| | | | | |
Collapse
|
19
|
Salerno S, Tasselli F, Drioli E, De Bartolo L. Poly(ε-Caprolactone) Hollow Fiber Membranes for the Biofabrication of a Vascularized Human Liver Tissue. MEMBRANES 2020; 10:E112. [PMID: 32471264 PMCID: PMC7344547 DOI: 10.3390/membranes10060112] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/23/2020] [Accepted: 05/26/2020] [Indexed: 01/01/2023]
Abstract
The creation of a liver tissue that recapitulates the micro-architecture and functional complexity of a human organ is still one of the main challenges of liver tissue engineering. Here we report on the development of a 3D vascularized hepatic tissue based on biodegradable hollow fiber (HF) membranes of poly(ε-caprolactone) (PCL) that compartmentalize human hepatocytes on the external surface and between the fibers, and endothelial cells into the fiber lumen. To this purpose, PCL HF membranes were prepared by a dry-jet wet phase inversion spinning technique tailoring the operational parameters in order to obtain fibers with suitable properties. After characterization, the fibers were applied to generate a human vascularized hepatic unit by loading endothelial cells in their inner surface and hepatocytes on the external surface. The unit was connected to a perfusion system, and the morpho-functional behavior was evaluated. The results demonstrated the large integration of endothelial cells with the internal surface of individual PCL fibers forming vascular-like structures, and hepatocytes covered completely the external surface and the space between fibers. The perfused 3D hepatic unit retained its functional activity at high levels up to 18 days. This bottom-up tissue engineering approach represents a rational strategy to create relatively 3D vascularized tissues and organs.
Collapse
Affiliation(s)
- Simona Salerno
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
| | - Franco Tasselli
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
| | - Enrico Drioli
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
- Department of Energy, Engineering, Hanyang University, Seoul 04763, Korea
| | - Loredana De Bartolo
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
| |
Collapse
|
20
|
Sharifi F, Firoozabadi B, Firoozbakhsh K. Numerical Investigations of Hepatic Spheroids Metabolic Reactions in a Perfusion Bioreactor. Front Bioeng Biotechnol 2019; 7:221. [PMID: 31572719 PMCID: PMC6751279 DOI: 10.3389/fbioe.2019.00221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022] Open
Abstract
Miniaturized culture systems of hepatic cells are emerging as a strong tool facilitating studies related to liver diseases and drug discovery. However, the experimental optimization of various parameters involved in the operation of these systems is time-consuming and expensive. Hence, developing numerical tools predicting the function of such systems can significantly reduce the associated cost. In this paper, a perfusion-based three dimensional (3D) bioreactor comprising encapsulated human liver hepatocellular carcinoma (HepG2) spheroids are analyzed. The flow and mass transfer equations for oxygen as well as different metabolites such as albumin, glucose, glutamine, ammonia, and urea were solved in three different domains, i.e., free flow, hydrogel, and spheroid porous media sections. Since the spheroids were encapsulated inside the hydrogel, shear stress imposed on them were found to be less than tolerable thresholds. The predicted cumulative albumin concentration over the 7 days of culture period showed a good agreement with the experimental data. Based on the critical role of oxygen supply to the hepatocytes, a parametric study was performed and the effect of various parameters was investigated. Results illustrated that convection mechanism was the dominant transport mechanism in the main-stream section contrary to the intra spheroids parts where the diffusion was the prevailing transport mechanism. In the hydrogel parts, the rate of diffusion and convection mechanisms were almost identical. As expected, higher perfusion rate would provide high oxygen level for the cells and, smaller spheroids with a diameter of 100 μm were at the low risk of hypoxic conditions due to short diffusive oxygen penetration depth. Numerical results evidenced that spheroids with diameter size >200 μm at low porosities (ε = 0.2-0.3) were at risk of oxygen depletion, especially at locations near the core center. Therefore, these results could be beneficial in preventing hypoxic conditions during in vitro experiments. The presented numerical model provides a numerical platform which can help researchers to design and optimize complex bioreactors and obtain numerical indexes of the main metabolites in a very short time prior to any fabrications. Such numerical indexes can be helpful in certifying the outcomes of forensic investigations.
Collapse
Affiliation(s)
| | - Bahar Firoozabadi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | | |
Collapse
|
21
|
Li K, Yang X, Gao X. Probing tumor microtissue formation and epithelial-mesenchymal transition on a well-mesh microchip. BIOMICROFLUIDICS 2019; 13:014102. [PMID: 30867873 PMCID: PMC6404933 DOI: 10.1063/1.5064838] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/21/2018] [Indexed: 05/09/2023]
Abstract
Three-dimensional cultures of tumor microtissues and biomimetic simulation of tumor microenvironments are of great significance in the study of tumorigenesis and development processes. In this study, a well-mesh microchip was developed to realize the formation and culture of tumor microtissues in vitro. Human lung adenocarcinoma HCC827 cells and large-cell lung cancer NCI-H460 cells were used. The size and morphology of the microtissues have been observed. In addition, we constructed an in situ three-dimensional co-culture model with tumor cell microtissues (HCC827 or NCI-H460 cells), extracellular matrix (Matrigel), and human umbilical vein endothelial cells. HCC827 microtissue epithelial-mesenchymal transition (EMT) in the established well-mesh microchip also was investigated, and the results showed that recombinant transforming growth factor could activate the Snail and Akt gene and promote migration and EMT with the decrease of E-cadherin expression for HCC827. This well-mesh microchip features simple operation and easy observation, and could provide a new method for the study of tumor cells and tumor microenvironments in vitro. Therefore, this model has potential application value in organ-on-chip technology, tissue engineering, and drug evaluation.
Collapse
Affiliation(s)
| | | | - Xinghua Gao
- Author to whom correspondence should be addressed: . Tel.: +86-69982223
| |
Collapse
|
22
|
Legallais C, Kim D, Mihaila SM, Mihajlovic M, Figliuzzi M, Bonandrini B, Salerno S, Yousef Yengej FA, Rookmaaker MB, Sanchez Romero N, Sainz-Arnal P, Pereira U, Pasqua M, Gerritsen KGF, Verhaar MC, Remuzzi A, Baptista PM, De Bartolo L, Masereeuw R, Stamatialis D. Bioengineering Organs for Blood Detoxification. Adv Healthc Mater 2018; 7:e1800430. [PMID: 30230709 DOI: 10.1002/adhm.201800430] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 08/23/2018] [Indexed: 12/11/2022]
Abstract
For patients with severe kidney or liver failure the best solution is currently organ transplantation. However, not all patients are eligible for transplantation and due to limited organ availability, most patients are currently treated with therapies using artificial kidney and artificial liver devices. These therapies, despite their relative success in preserving the patients' life, have important limitations since they can only replace part of the natural kidney or liver functions. As blood detoxification (and other functions) in these highly perfused organs is achieved by specialized cells, it seems relevant to review the approaches leading to bioengineered organs fulfilling most of the native organ functions. There, the culture of cells of specific phenotypes on adapted scaffolds that can be perfused takes place. In this review paper, first the functions of kidney and liver organs are briefly described. Then artificial kidney/liver devices, bioartificial kidney devices, and bioartificial liver devices are focused on, as well as biohybrid constructs obtained by decellularization and recellularization of animal organs. For all organs, a thorough overview of the literature is given and the perspectives for their application in the clinic are discussed.
Collapse
Affiliation(s)
- Cécile Legallais
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Dooli Kim
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| | - Sylvia M. Mihaila
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Milos Mihajlovic
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Marina Figliuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
| | - Barbara Bonandrini
- Department of Chemistry; Materials and Chemical Engineering “Giulio Natta”; Politecnico di Milano; Piazza Leonardo da Vinci 32 20133 Milan Italy
| | - Simona Salerno
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Fjodor A. Yousef Yengej
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Maarten B. Rookmaaker
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | | | - Pilar Sainz-Arnal
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Instituto Aragonés de Ciencias de la Salud (IACS); 50009 Zaragoza Spain
| | - Ulysse Pereira
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Mattia Pasqua
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Karin G. F. Gerritsen
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Andrea Remuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
| | - Pedro M. Baptista
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd); 28029 Barcelona Spain
- Fundación ARAID; 50009 Zaragoza Spain
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz; 28040 Madrid Spain. Department of Biomedical and Aerospace Engineering; Universidad Carlos III de Madrid; 28911 Madrid Spain
| | - Loredana De Bartolo
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Rosalinde Masereeuw
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Dimitrios Stamatialis
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| |
Collapse
|
23
|
Salerno S, Curcio E, Bader A, Giorno L, Drioli E, De Bartolo L. Gas permeable membrane bioreactor for the co-culture of human skin derived mesenchymal stem cells with hepatocytes and endothelial cells. J Memb Sci 2018. [DOI: 10.1016/j.memsci.2018.06.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
24
|
Piscioneri A, Ahmed HMM, Morelli S, Khakpour S, Giorno L, Drioli E, De Bartolo L. Membrane bioreactor to guide hepatic differentiation of human mesenchymal stem cells. J Memb Sci 2018. [DOI: 10.1016/j.memsci.2018.07.083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
25
|
Yajima Y, Lee CN, Yamada M, Utoh R, Seki M. Development of a perfusable 3D liver cell cultivation system via bundling-up assembly of cell-laden microfibers. J Biosci Bioeng 2018; 126:111-118. [PMID: 29502942 DOI: 10.1016/j.jbiosc.2018.01.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/10/2018] [Accepted: 01/31/2018] [Indexed: 01/20/2023]
Abstract
Although the reconstruction of functional 3D liver tissue models in vitro presents numerous challenges, it is in great demand for drug development, regenerative medicine, and physiological studies. Here we propose a new approach to perform perfusion cultivation of liver cells by assembling cell-laden hydrogel microfibers. HepG2 cells were densely packed into the core of sandwich-type anisotropic microfibers, which were produced using microfluidic devices. The obtained microfibers were bundled up and packed into a perfusion chamber, and perfusion cultivation was performed. We evaluated cell viability and functions, and also monitored the oxygen consumption. Furthermore, fibers covered with vascular endothelial cells were united during the perfusion culture, to form vascular network-like conduits between fibers. The presented technique can structurally mimic the hepatic lobule in vivo and could prove to be a useful model for various biomedical research applications.
Collapse
Affiliation(s)
- Yuya Yajima
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Chu Ning Lee
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Masumi Yamada
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Rie Utoh
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Minoru Seki
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|