1
|
Wang H, Wen L, Jiang F, Ren P, Yang Y, Song S, Yang Z, Wang Y. A comprehensive review of advances in hepatocyte microencapsulation: selecting materials and preserving cell viability. Front Immunol 2024; 15:1385022. [PMID: 38694507 PMCID: PMC11061843 DOI: 10.3389/fimmu.2024.1385022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 03/28/2024] [Indexed: 05/04/2024] Open
Abstract
Liver failure represents a critical medical condition with a traditionally grim prognosis, where treatment options have been notably limited. Historically, liver transplantation has stood as the sole definitive cure, yet the stark disparity between the limited availability of liver donations and the high demand for such organs has significantly hampered its feasibility. This discrepancy has necessitated the exploration of hepatocyte transplantation as a temporary, supportive intervention. In light of this, our review delves into the burgeoning field of hepatocyte transplantation, with a focus on the latest advancements in maintaining hepatocyte function, co-microencapsulation techniques, xenogeneic hepatocyte transplantation, and the selection of materials for microencapsulation. Our examination of hepatocyte microencapsulation research highlights that, to date, most studies have been conducted in vitro or using liver failure mouse models, with a notable paucity of experiments on larger mammals. The functionality of microencapsulated hepatocytes is primarily inferred through indirect measures such as urea and albumin production and the rate of ammonia clearance. Furthermore, research on the mechanisms underlying hepatocyte co-microencapsulation remains limited, and the practicality of xenogeneic hepatocyte transplantation requires further validation. The potential of hepatocyte microencapsulation extends beyond the current scope of application, suggesting a promising horizon for liver failure treatment modalities. Innovations in encapsulation materials and techniques aim to enhance cell viability and function, indicating a need for comprehensive studies that bridge the gap between small-scale laboratory success and clinical applicability. Moreover, the integration of bioengineering and regenerative medicine offers novel pathways to refine hepatocyte transplantation, potentially overcoming the challenges of immune rejection and ensuring the long-term functionality of transplanted cells. In conclusion, while hepatocyte microencapsulation and transplantation herald a new era in liver failure therapy, significant strides must be made to translate these experimental approaches into viable clinical solutions. Future research should aim to expand the experimental models to include larger mammals, thereby providing a clearer understanding of the clinical potential of these therapies. Additionally, a deeper exploration into the mechanisms of cell survival and function within microcapsules, alongside the development of innovative encapsulation materials, will be critical in advancing the field and offering new hope to patients with liver failure.
Collapse
Affiliation(s)
- Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Lebin Wen
- Department of Thyroid, Sichuan Second Hospital of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Fengdi Jiang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Pengyu Ren
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yixin Yang
- Department of Clinical Medicine, The First Clinical Medical College of Norman Bethune University of Medical Sciences, Jilin, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Zhengteng Yang
- Department of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Yi Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
2
|
Gómez‐Álvarez M, Bueno‐Fernandez C, Rodríguez‐Eguren A, Francés‐Herrero E, Agustina‐Hernández M, Faus A, Gisbert Roca F, Martínez‐Ramos C, Galán A, Pellicer A, Ferrero H, Cervelló I. Hybrid Endometrial-Derived Hydrogels: Human Organoid Culture Models and In Vivo Perspectives. Adv Healthc Mater 2024; 13:e2303838. [PMID: 37983675 PMCID: PMC11468130 DOI: 10.1002/adhm.202303838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Indexed: 11/22/2023]
Abstract
The endometrium plays a vital role in fertility, providing a receptive environment for embryo implantation and development. Understanding the endometrial physiology is essential for developing new strategies to improve reproductive healthcare. Human endometrial organoids (hEOs) are emerging as powerful models for translational research and personalized medicine. However, most hEOs are cultured in a 3D microenvironment that significantly differs from the human endometrium, limiting their applicability in bioengineering. This study presents a hybrid endometrial-derived hydrogel that combines the rigidity of PuraMatrix (PM) with the natural scaffold components and interactions of a porcine decellularized endometrial extracellular matrix (EndoECM) hydrogel. This hydrogel provides outstanding support for hEO culture, enhances hEO differentiation efficiency due to its biochemical similarity with the native tissue, exhibits superior in vivo stability, and demonstrates xenogeneic biocompatibility in mice over a 2-week period. Taken together, these attributes position this hybrid endometrial-derived hydrogel as a promising biomaterial for regenerative treatments in reproductive medicine.
Collapse
Affiliation(s)
- María Gómez‐Álvarez
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Clara Bueno‐Fernandez
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
- Universitat de ValènciaDepartment of PediatricsObstetrics and GynaecologyValencia46010Spain
| | - Adolfo Rodríguez‐Eguren
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Emilio Francés‐Herrero
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
- Universitat de ValènciaDepartment of PediatricsObstetrics and GynaecologyValencia46010Spain
| | - Marcos Agustina‐Hernández
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Amparo Faus
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Fernando Gisbert Roca
- Universitat Politècnica de ValènciaCentre for Biomaterials and Tissue EngineeringValencia46022Spain
| | - Cristina Martínez‐Ramos
- Universitat Politècnica de ValènciaCentre for Biomaterials and Tissue EngineeringValencia46022Spain
- Unitat Predepartamental de MedicinaUniversitat Jaume ICastellón de la Plana12071Spain
| | - Amparo Galán
- Laboratory of NeuroendocrinologyPrince Felipe Research Center (CIPF)Valencia46012Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)Instituto de Salud Carlos IIIMadrid28029Spain
| | | | - Hortensia Ferrero
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Irene Cervelló
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| |
Collapse
|
3
|
Al-Nimry SS, Daghmash RM. Three Dimensional Printing and Its Applications Focusing on Microneedles for Drug Delivery. Pharmaceutics 2023; 15:1597. [PMID: 37376046 DOI: 10.3390/pharmaceutics15061597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Microneedles (MNs) are considered to be a novel smart injection system that causes significantly low skin invasion upon puncturing, due to the micron-sized dimensions that pierce into the skin painlessly. This allows transdermal delivery of numerous therapeutic molecules, such as insulin and vaccines. The fabrication of MNs is carried out through conventional old methods such as molding, as well as through newer and more sophisticated technologies, such as three-dimensional (3D) printing, which is considered to be a superior, more accurate, and more time- and production-efficient method than conventional methods. Three-dimensional printing is becoming an innovative method that is used in education through building intricate models, as well as being employed in the synthesis of fabrics, medical devices, medical implants, and orthoses/prostheses. Moreover, it has revolutionary applications in the pharmaceutical, cosmeceutical, and medical fields. Having the capacity to design patient-tailored devices according to their dimensions, along with specified dosage forms, has allowed 3D printing to stand out in the medical field. The different techniques of 3D printing allow for the production of many types of needles with different materials, such as hollow MNs and solid MNs. This review covers the benefits and drawbacks of 3D printing, methods used in 3D printing, types of 3D-printed MNs, characterization of 3D-printed MNs, general applications of 3D printing, and transdermal delivery using 3D-printed MNs.
Collapse
Affiliation(s)
- Suhair S Al-Nimry
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Rawand M Daghmash
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| |
Collapse
|
4
|
Blackford SJI, Yu TTL, Norman MDA, Syanda AM, Manolakakis M, Lachowski D, Yan Z, Guo Y, Garitta E, Riccio F, Jowett GM, Ng SS, Vernia S, Del Río Hernández AE, Gentleman E, Rashid ST. RGD density along with substrate stiffness regulate hPSC hepatocyte functionality through YAP signalling. Biomaterials 2023; 293:121982. [PMID: 36640555 DOI: 10.1016/j.biomaterials.2022.121982] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Human pluripotent stem cell-derived hepatocytes (hPSC-Heps) may be suitable for treating liver diseases, but differentiation protocols often fail to yield adult-like cells. We hypothesised that replicating healthy liver niche biochemical and biophysical cues would produce hepatocytes with desired metabolic functionality. Using 2D synthetic hydrogels which independently control mechanical properties and biochemical cues, we found that culturing hPSC-Heps on surfaces matching the stiffness of fibrotic liver tissue upregulated expression of genes for RGD-binding integrins, and increased expression of YAP/TAZ and their transcriptional targets. Alternatively, culture on soft, healthy liver-like substrates drove increases in cytochrome p450 activity and ureagenesis. Knockdown of ITGB1 or reducing RGD-motif-containing peptide concentration in stiff hydrogels reduced YAP activity and improved metabolic functionality; however, on soft substrates, reducing RGD concentration had the opposite effect. Furthermore, targeting YAP activity with verteporfin or forskolin increased cytochrome p450 activity, with forskolin dramatically enhancing urea synthesis. hPSC-Heps could also be successfully encapsulated within RGD peptide-containing hydrogels without negatively impacting hepatic functionality, and compared to 2D cultures, 3D cultured hPSC-Heps secreted significantly less fetal liver-associated alpha-fetoprotein, suggesting furthered differentiation. Our platform overcomes technical hurdles in replicating the liver niche, and allowed us to identify a role for YAP/TAZ-mediated mechanosensing in hPSC-Hep differentiation.
Collapse
Affiliation(s)
- Samuel J I Blackford
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Centre for Craniofacial & Regenerative Biology, King's College London, UK; Centre for Gene Therapy & Regenerative Medicine, King's College London, UK; NIHR Imperial BRC iPSC and Organoid Core Facility, Imperial College London, UK.
| | - Tracy T L Yu
- Centre for Craniofacial & Regenerative Biology, King's College London, UK
| | - Michael D A Norman
- Centre for Craniofacial & Regenerative Biology, King's College London, UK
| | - Adam M Syanda
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; NIHR Imperial BRC iPSC and Organoid Core Facility, Imperial College London, UK
| | - Michail Manolakakis
- MRC London Institute of Medical Sciences, UK; Institute of Clinical Sciences, Imperial College London, UK
| | - Dariusz Lachowski
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, UK
| | - Ziqian Yan
- Centre for Craniofacial & Regenerative Biology, King's College London, UK
| | - Yunzhe Guo
- Centre for Craniofacial & Regenerative Biology, King's College London, UK
| | - Elena Garitta
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; NIHR Imperial BRC iPSC and Organoid Core Facility, Imperial College London, UK
| | - Federica Riccio
- Centre for Gene Therapy & Regenerative Medicine, King's College London, UK
| | - Geraldine M Jowett
- Centre for Craniofacial & Regenerative Biology, King's College London, UK; Centre for Gene Therapy & Regenerative Medicine, King's College London, UK
| | - Soon Seng Ng
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; NIHR Imperial BRC iPSC and Organoid Core Facility, Imperial College London, UK
| | - Santiago Vernia
- MRC London Institute of Medical Sciences, UK; Institute of Clinical Sciences, Imperial College London, UK
| | | | - Eileen Gentleman
- Centre for Craniofacial & Regenerative Biology, King's College London, UK.
| | - S Tamir Rashid
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; NIHR Imperial BRC iPSC and Organoid Core Facility, Imperial College London, UK.
| |
Collapse
|
5
|
Le Guilcher C, Merlen G, Dellaquila A, Labour MN, Aid R, Tordjmann T, Letourneur D, Simon-Yarza T. Engineered human liver based on pullulan-dextran hydrogel promotes mice survival after liver failure. Mater Today Bio 2023; 19:100554. [PMID: 36756209 PMCID: PMC9900439 DOI: 10.1016/j.mtbio.2023.100554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Liver tissue engineering approaches aim to support drug testing, assistance devices, or transplantation. However, their suitability for clinical application remains unsatisfactory. Herein, we demonstrate the beneficial and biocompatible use of porous pullulan-dextran hydrogel for the self-assembly of hepatocytes and biliary-like cells into functional 3D microtissues. Using HepaRG cells, we obtained 21 days maintenance of engineered liver polarity, functional detoxification and excretion systems, as well as glycogen storage in hydrogel. Implantation on two liver lobes in mice of hydrogels containing 3800 HepaRG 3D structures of 100 μm in diameter, indicated successful engraftment and no signs of liver toxicity after one month. Finally, after acetaminophen-induced liver failure, when mice were transplanted with engineered livers on left lobe and peritoneal cavity, the survival rate at 7 days significantly increased by 31.8% compared with mice without cell therapy. These findings support the clinical potential of pullulan-dextran hydrogel for liver failure management.
Collapse
Affiliation(s)
- Camille Le Guilcher
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,Corresponding author.
| | - Grégory Merlen
- Université Paris-Saclay, INSERM U1193, F- 94800 Villejuif, France
| | - Alessandra Dellaquila
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France
| | - Marie-Noëlle Labour
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,ICGM, Université de Montpellier, CNRS, ENSCM, F- 34293 Montpellier, France,École Pratique des Hautes Études, Université Paris Sciences et Lettres, F-75014 Paris, France
| | - Rachida Aid
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France
| | | | - Didier Letourneur
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,Corresponding author.
| | - Teresa Simon-Yarza
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,Corresponding author.
| |
Collapse
|
6
|
Abbott A, Gravina ME, Vandadi M, Rahbar N, Coburn JM. Influence of lyophilization primary drying time and temperature on porous silk scaffold fabrication for biomedical applications. J Biomed Mater Res A 2023; 111:118-131. [PMID: 36205385 DOI: 10.1002/jbm.a.37451] [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: 10/14/2021] [Revised: 05/20/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
Lyophilization of protein solutions, such as silk fibroin (silk), produces porous scaffolds useful for tissue engineering (TE). The impact of modifying lyophilization primary drying parameters on scaffold properties has not yet been explored previously. In this work, changes to primary drying duration and temperature were investigated using 3%, 6%, 9%, and 12% (w/v) silk solutions, via protocols labeled as Long Hold, Slow Ramp, and Standard. The 9% and 12% scaffolds were not successfully fabricated using the Standard protocol, while the Long Hold and Slow Ramp protocols resulted in scaffolds from all silk solution concentrations. Scaffolds fabricated using the Long Hold protocol had higher Young's moduli, smaller pore Feret diameters, and faster degradation. To investigate the utility of the different lyophilized scaffolds for in vitro cell culturing, the HepaRG liver cell line was cultured in the 3% to 12% scaffolds fabricated using the Long Hold protocol. The HepaRG cells grown in 3% scaffolds initially had greater lipid accumulation and metabolic activity than the other groups, although these differences were no longer apparent by Day 28. The deoxyribonucleic acid content of the HepaRG cells grown in 3% scaffold group was also initially significantly higher than the other groups. Significant differences in gene expression by 9% scaffolded HepaRG cells (CK19, HNFα) were seen on Day 14 while significant differences by 12% scaffolded HepaRG cells (ALB, APOA4) were seen on Day 28. Overall, modifying the primary drying parameters and silk concentration resulted in lyophilized scaffolds with tunable properties useful for TE applications.
Collapse
Affiliation(s)
- Alycia Abbott
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Mattea E Gravina
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Mobin Vandadi
- Department of Civil and Environmental Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Nima Rahbar
- Department of Civil and Environmental Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Jeannine M Coburn
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
7
|
Budharaju H, Zennifer A, Sethuraman S, Paul A, Sundaramurthi D. Designer DNA biomolecules as a defined biomaterial for 3D bioprinting applications. MATERIALS HORIZONS 2022; 9:1141-1166. [PMID: 35006214 DOI: 10.1039/d1mh01632f] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
DNA has excellent features such as the presence of functional and targeted molecular recognition motifs, tailorability, multifunctionality, high-precision molecular self-assembly, hydrophilicity, and outstanding biocompatibility. Due to these remarkable features, DNA has emerged as a leading next-generation biomaterial of choice to make hydrogels by self-assembly. In recent times, novel routes for the chemical synthesis of DNA, advances in tailorable designs, and affordable production ways have made DNA as a building block material for various applications. These advanced features have made researchers continuously explore the interesting properties of pure and hybrid DNA for 3D bioprinting and other biomedical applications. This review article highlights the topical advancements in the use of DNA as an ideal bioink for the bioprinting of cell-laden three-dimensional tissue constructs for regenerative medicine applications. Various bioprinting techniques and emerging design approaches such as self-assembly, nucleotide sequence, enzymes, and production cost to use DNA as a bioink for bioprinting applications are described. In addition, various types and properties of DNA hydrogels such as stimuli responsiveness and mechanical properties are discussed. Further, recent progress in the applications of DNA in 3D bioprinting are emphasized. Finally, the current challenges and future perspectives of DNA hydrogels in 3D bioprinting and other biomedical applications are discussed.
Collapse
Affiliation(s)
- Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India.
| | - Allen Zennifer
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India.
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India.
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
- Department of Chemistry, The University of Western Ontario, London, ON N6A 5B9, Canada
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India.
| |
Collapse
|
8
|
Abstract
Organoids-cellular aggregates derived from stem or progenitor cells that recapitulate organ function in miniature-are of growing interest in developmental biology and medicine. Organoids have been developed for organs and tissues such as the liver, gut, brain, and pancreas; they are used as organ surrogates to study a wide range of questions in basic and developmental biology, genetic disorders, and therapies. However, many organoids reported to date have been cultured in Matrigel, which is prepared from the secretion of Engelbreth-Holm-Swarm mouse sarcoma cells; Matrigel is complex and poorly defined. This complexity makes it difficult to elucidate Matrigel-specific factors governing organoid development. In this review, we discuss promising Matrigel-free methods for the generation and maintenance of organoids that use decellularized extracellular matrix (ECM), synthetic hydrogels, or gel-forming recombinant proteins.
Collapse
Affiliation(s)
- Mark T Kozlowski
- DEVCOM US Army Research Laboratory, Weapons and Materials Research Directorate, Science of Extreme Materials Division, Polymers Branch, 6300 Rodman Rd. Building 4600, Aberdeen Proving Ground, Aberdeen, MD, 21005, USA.
| | - Christiana J Crook
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Hsun Teresa Ku
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 Duarte Rd., Duarte, CA, 91010, USA
| |
Collapse
|
9
|
Rose S, Cuvellier M, Ezan F, Carteret J, Bruyère A, Legagneux V, Nesslany F, Baffet G, Langouët S. DMSO-free highly differentiated HepaRG spheroids for chronic toxicity, liver functions and genotoxicity studies. Arch Toxicol 2021; 96:243-258. [PMID: 34762139 DOI: 10.1007/s00204-021-03178-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/06/2021] [Indexed: 12/23/2022]
Abstract
The liver is essential in the elimination of environmental and food contaminants. Given the interspecies differences between rodents and humans, the development of relevant in vitro human models is crucial to investigate liver functions and toxicity in cells that better reflect pathophysiological processes. Classically, the differentiation of the hepatic HepaRG cell line requires high concentration of dimethyl sulfoxide (DMSO), which restricts its usefulness for drug-metabolism studies. Herein, we describe undifferentiated HepaRG cells embedded in a collagen matrix in DMSO-free conditions that rapidly organize into polarized hollow spheroids of differentiated hepatocyte-like cells (Hepoid-HepaRG). Our conditions allow concomitant proliferation with high levels of liver-specific functions and xenobiotic metabolism enzymes expression and activities after a few days of culture and for at least 4 weeks. By studying the toxicity of well-known injury-inducing drugs by treating cells with 1- to 100-fold of their plasmatic concentrations, we showed appropriate responses and demonstrate the sensitivity to drugs known to induce various degrees of liver injury. Our results also demonstrated that the model is well suited to estimate cholestasis and steatosis effects of drugs following chronic treatment. Additionally, DNA alterations caused by four genotoxic compounds (Aflatoxin B1 (AFB1), Benzo[a]Pyrene (B[a]P), Cyclophosphamide (CPA) and Methyl methanesulfonate (MMS)) were quantified in a dose-dependent manner by the comet and micronucleus assays. Their genotoxic effects were significantly increased after either an acute 24 h treatment (AFB1: 1.5-6 μM, CPA: 2.5-10 μM, B[a]P: 12.5-50 μM, MMS: 90-450 μM) or after a 14-day treatment at much lower concentrations (AFB1: 0.05-0.2 μM, CPA: 0.125-0.5 μM, B[a]P: 0.125-0.5 μM) representative to human exposure. Altogether, the DMSO-free 3D culture of Hepoid-HepaRG provides highly differentiated and proliferating cells relevant for various toxicological in vitro assays, especially for drug-preclinical studies and environmental chemicals risk assessment.
Collapse
Affiliation(s)
- Sophie Rose
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Marie Cuvellier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Frédéric Ezan
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Jennifer Carteret
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Vincent Legagneux
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Fabrice Nesslany
- Genotoxicology Department, Institut Pasteur de Lille, 1, Rue du Professeur Calmette, 59000, Lille, France
| | - Georges Baffet
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France.
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France.
| |
Collapse
|
10
|
Brovold M, Keller D, Devarasetty M, Dominijanni A, Shirwaiker R, Soker S. Biofabricated 3D in vitro model of fibrosis-induced abnormal hepatoblast/biliary progenitors' expansion of the developing liver. Bioeng Transl Med 2021; 6:e10207. [PMID: 34589593 PMCID: PMC8459590 DOI: 10.1002/btm2.10207] [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: 09/16/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 11/19/2022] Open
Abstract
Congenital disorders of the biliary tract are the primary reason for pediatric liver failure and ultimately for pediatric liver transplant needs. Not all causes of these disorders are well understood, but it is known that liver fibrosis occurs in many of those afflicted. The goal of this study is to develop a simple yet robust model that recapitulates physico-mechanical and cellular aspects of fibrosis mediated via hepatic stellate cells (HSCs) and their effects on biliary progenitor cells. Liver organoids were fabricated by embedding various HSCs, with distinctive abilities to generate mild to severe fibrotic environments, together with undifferentiated liver progenitor cell line, HepaRG, within a collagen I hydrogel. The fibrotic state of each organoid was characterized by examination of extracellular matrix (ECM) remodeling through quantitative image analysis, rheometry, and qPCR. In tandem, the phenotype of the liver progenitor cell and cluster formation was assessed through histology. Activated HSCs (aHSCs) created a more severe fibrotic state, exemplified by a more highly contracted and rigid ECM, as well higher relative expression of TGF-β, TIMP-1, LOXL2, and COL1A2 as compared to immortalized HSCs (LX-2). Within the more severe fibrotic environment, generated by the aHSCs, higher Notch signaling was associated with an expansion of CK19+ cells as well as the formation of larger, more densely populated cell biliary like-clusters as compared to mild and non-fibrotic controls. The expansion of CK19+ cells, coupled with a severely fibrotic environment, are phenomena found within patients suffering from a variety of congenital liver disorders of the biliary tract. Thus, the model presented here can be utilized as a novel in vitro testing platform to test drugs and identify new targets that could benefit pediatric patients that suffer from the biliary dysgenesis associated with a multitude of congenital liver diseases.
Collapse
Affiliation(s)
- Matthew Brovold
- Wake Forest Institute for Regenerative MedicineWake Forest Baptist Medical Center, Medical Center BoulevardWinston‐SalemNorth CarolinaUSA
| | - Dale Keller
- Wake Forest Institute for Regenerative MedicineWake Forest Baptist Medical Center, Medical Center BoulevardWinston‐SalemNorth CarolinaUSA
| | - Mahesh Devarasetty
- Wake Forest Institute for Regenerative MedicineWake Forest Baptist Medical Center, Medical Center BoulevardWinston‐SalemNorth CarolinaUSA
| | - Anthony Dominijanni
- Wake Forest Institute for Regenerative MedicineWake Forest Baptist Medical Center, Medical Center BoulevardWinston‐SalemNorth CarolinaUSA
| | - Rohan Shirwaiker
- Department of Industrial and Systems EngineeringNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Shay Soker
- Wake Forest Institute for Regenerative MedicineWake Forest Baptist Medical Center, Medical Center BoulevardWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
11
|
Pasqua M, Di Gesù R, Chinnici CM, Conaldi PG, Francipane MG. Generation of Hepatobiliary Cell Lineages from Human Induced Pluripotent Stem Cells: Applications in Disease Modeling and Drug Screening. Int J Mol Sci 2021; 22:8227. [PMID: 34360991 PMCID: PMC8348238 DOI: 10.3390/ijms22158227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
The possibility to reproduce key tissue functions in vitro from induced pluripotent stem cells (iPSCs) is offering an incredible opportunity to gain better insight into biological mechanisms underlying development and disease, and a tool for the rapid screening of drug candidates. This review attempts to summarize recent strategies for specification of iPSCs towards hepatobiliary lineages -hepatocytes and cholangiocytes-and their use as platforms for disease modeling and drug testing. The application of different tissue-engineering methods to promote accurate and reliable readouts is discussed. Space is given to open questions, including to what extent these novel systems can be informative. Potential pathways for improvement are finally suggested.
Collapse
Affiliation(s)
- Mattia Pasqua
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
| | - Roberto Di Gesù
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
| | - Cinzia Maria Chinnici
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
- Dipartimento della Ricerca, IRCCS ISMETT, 90127 Palermo, Italy;
| | | | - Maria Giovanna Francipane
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
12
|
Ma P, Chen Y, Lai X, Zheng J, Ye E, Loh XJ, Zhao Y, Parikh BH, Su X, You M, Wu YL, Li Z. The Translational Application of Hydrogel for Organoid Technology: Challenges and Future Perspectives. Macromol Biosci 2021; 21:e2100191. [PMID: 34263547 DOI: 10.1002/mabi.202100191] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/17/2021] [Indexed: 12/16/2022]
Abstract
Human organoids mimic the physiology and tissue architecture of organs and are of great significance for promoting the study of human diseases. Traditionally, organoid cultures rely predominantly on animal or tumor-derived extracellular matrix (ECM), resulting in poor reproducibility. This limits their utility in for large-scale drug screening and application for regenerative medicine. Recently, synthetic polymeric hydrogels, with high biocompatibility and biodegradability, stability, uniformity of compositions, and high throughput properties, have emerged as potential materials for achieving 3D architectures for organoid cultures. Compared to conventional animal or tumor-derived organoids, these newly engineered hydrogel-based organoids more closely resemble human organs, as they are able to mimic native structural and functional properties observed in-situ. In this review, recent developments in hydrogel-based organoid culture will be summarized, emergent hydrogel technology will be highlighted, and future challenges in applying them to organoid culture will be discussed.
Collapse
Affiliation(s)
- Panqin Ma
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Ying Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xiyu Lai
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Jie Zheng
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Enyi Ye
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Yi Zhao
- BayRay Innovation Center, Shenzhen Bay Laboratory (SZBL), Shenzhen, 518132, China
| | - Bhav Harshad Parikh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis, Drive, Proteos, Singapore, 138673, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore
| | - Xinyi Su
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis, Drive, Proteos, Singapore, 138673, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.,Singapore Eye Research Institute (SERI), The Academia, 20 College Road Discovery Tower Level 6, Singapore, 169856, Singapore.,Department of Ophthalmology, National University Hospital, Singapore, 119074, Singapore
| | - Mingliang You
- Hangzhou Cancer Institute, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Zibiao Li
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore.,Department of Materials Science and Engineering, National University of Singapore, Singapore, 117574, Singapore
| |
Collapse
|
13
|
Jiang T, Guo H, Xia YN, Liu Y, Chen D, Pang G, Feng Y, Yu H, Wu Y, Zhang S, Wang Y, Wang Y, Wen H, Zhang LW. Hepatotoxicity of copper sulfide nanoparticles towards hepatocyte spheroids using a novel multi-concave agarose chip method. Nanomedicine (Lond) 2021; 16:1487-1504. [PMID: 34184559 DOI: 10.2217/nnm-2021-0011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: To explore the hepatotoxicity of copper sulfide nanoparticles (CuSNPs) toward hepatocyte spheroids. Materials & methods: Other than the traditional agarose method to generate hepatocyte spheroids, we developed a multi-concave agarose chip (MCAC) method to investigate changes in hepatocyte viability, morphology, mitochondrial membrane potential, reactive oxygen species and hepatobiliary transporter by CuSNPs. Results: The MCAC method allowed a large number of spheroids to be obtained per sample. CuSNPs showed hepatotoxicity in vitro through a decrease in spheroid viability, albumin/urea production and glycogen deposition. CuSNPs also introduced hepatocyte spheroid injury through alteration of mitochondrial membrane potential and reactive oxygen species, that could be reversed by N-acetyl-l-cysteine. CuSNPs significantly decreased the activity of BSEP transporter by downregulating its mRNA and protein levels. Activity of the MRP2 transporter remained unchanged. Conclusion: We observed the hepatotoxicity of CuSNPs in vitro with associated mechanisms in an advanced 3D culture system.
Collapse
Affiliation(s)
- Tianyan Jiang
- School of Radiation Medicine & Protection, State Key Laboratory of Radiation Medicine & Protection, School for Radiological & Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, PR China
| | - Haoxiang Guo
- School of Radiation Medicine & Protection, State Key Laboratory of Radiation Medicine & Protection, School for Radiological & Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, PR China
| | - Ya-Nan Xia
- School of Radiation Medicine & Protection, State Key Laboratory of Radiation Medicine & Protection, School for Radiological & Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, PR China
| | - Yun Liu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China.,Shanghai R&D Centre for Standardization of Chinese Medicines, Shanghai, 201210, PR China
| | - Dandan Chen
- School of Radiation Medicine & Protection, State Key Laboratory of Radiation Medicine & Protection, School for Radiological & Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, PR China
| | - Guibin Pang
- State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology, Shanghai, PR China
| | - Yahui Feng
- Department of Science & Technology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, PR China
| | - Huan Yu
- School of Radiation Medicine & Protection, State Key Laboratory of Radiation Medicine & Protection, School for Radiological & Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, PR China
| | - Yanxian Wu
- School of Radiation Medicine & Protection, State Key Laboratory of Radiation Medicine & Protection, School for Radiological & Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, PR China
| | - Shaodian Zhang
- The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, PR China
| | - Yangyun Wang
- School of Radiation Medicine & Protection, State Key Laboratory of Radiation Medicine & Protection, School for Radiological & Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, PR China
| | - Yong Wang
- School of Radiation Medicine & Protection, State Key Laboratory of Radiation Medicine & Protection, School for Radiological & Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, PR China
| | - Hairuo Wen
- Beijing Key Laboratory, National Center for Safety Evaluation of Drugs, National Institutes for Food & Drug Control, Beijing, 100176, PR China
| | - Leshuai W Zhang
- School of Radiation Medicine & Protection, State Key Laboratory of Radiation Medicine & Protection, School for Radiological & Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, PR China
| |
Collapse
|
14
|
Abbott A, Coburn JM. HepaRG Maturation in Silk Fibroin Scaffolds: Toward Developing a 3D In Vitro Liver Model. ACS Biomater Sci Eng 2021. [PMID: 34105934 DOI: 10.1021/acsbiomaterials.0c01584] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In vitro liver models are necessary tools for the development of new therapeutics. HepaRG cells are a commonly used cell line to produce hepatic progenitor cells and hepatocytes. This study demonstrates for the first time the suitability of 3% silk scaffolds to support HepaRG growth and differentiation. The modulus and pore size of 3% silk scaffolds were shown to be within the desired range for liver cell growth. The optimal seeding density for HepaRG cells on silk scaffolds was determined. The growth and maturation of scaffolded HepaRG cells was evaluated for 28 days, where the first 14 days of culture were a proliferation period and the last 14 days of culture were a differentiation period using dimethyl sulfoxide (DMSO) treatment. After the first 14 days of culture, the scaffolded HepaRG cells exhibited increased metabolic activity and albumin secretion compared to monolayer cultured controls and preserved these attributes through the duration of culture. Additionally, after the first 14 days of culture, the scaffolded HepaRG cells displayed a significantly reduced expression of genes associated with hepatocyte maturation. This difference in expression was no longer apparent after 28 days of culture, suggesting that the cells underwent rapid differentiation within the scaffold. The functionalization of silk scaffolds with extracellular matrix (ECM) components (type I collagen and/or an arginylglycylaspartic acid (RGD)-containing peptide) was investigated to determine the impact on HepaRG cell attachment and maturation. The inclusion of ECM components had no noticeable impact on cell attachment but did significantly influence CYP3A4 expression and albumin secretion. Finally, the matrix support provided by the 3% silk scaffolds could prime the HepaRG cells for steatosis liver model applications, as evidenced by lipid droplet accumulation and expression of steatosis-related genes after 24 h of exposure to oleic acid. Overall, our work demonstrates the utility of silk scaffolds in providing a modifiable platform for liver cell growth.
Collapse
Affiliation(s)
- Alycia Abbott
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| | - Jeannine M Coburn
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| |
Collapse
|
15
|
Yao T, Zhang Y, Lv M, Zang G, Ng SS, Chen X. Advances in 3D cell culture for liver preclinical studies. Acta Biochim Biophys Sin (Shanghai) 2021; 53:643-651. [PMID: 33973620 DOI: 10.1093/abbs/gmab046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Indexed: 11/13/2022] Open
Abstract
The 3D cell culture model is an indispensable tool in the study of liver biology in the field of health and disease and the development of clinically relevant products for liver therapies. The 3D culture model captures critical factors of the microenvironmental niche required by hepatocytes for exhibiting optimal phenotypes, thus enabling the pursuit of a range of preclinical studies that are not entirely feasible in conventional 2D cell models. In this review, we highlight the major attributes associated with and the components needed for the development of a functional 3D liver culture model for a range of applications.
Collapse
Affiliation(s)
- Ting Yao
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Yi Zhang
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Mengjiao Lv
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Guoqing Zang
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Soon Seng Ng
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W2 1PG, UK
| | - Xiaohua Chen
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| |
Collapse
|
16
|
Roy HS, Singh R, Ghosh D. SARS-CoV-2 and tissue damage: current insights and biomaterial-based therapeutic strategies. Biomater Sci 2021; 9:2804-2824. [PMID: 33666206 DOI: 10.1039/d0bm02077j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The effect of SARS-CoV-2 infection on humanity has gained worldwide attention and importance due to the rapid transmission, lack of treatment options and high mortality rate of the virus. While scientists across the world are searching for vaccines/drugs that can control the spread of the virus and/or reduce the risks associated with infection, patients infected with SARS-CoV-2 have been reported to have tissue/organ damage. With most tissues/organs having limited regenerative potential, interventions that prevent further damage or facilitate healing would be helpful. In the past few decades, biomaterials have gained prominence in the field of tissue engineering, in view of their major role in the regenerative process. Here we describe the effect of SARS-CoV-2 on multiple tissues/organs, and provide evidence for the positive role of biomaterials in aiding tissue repair. These findings are further extrapolated to explore their prospects as a therapeutic platform to address the tissue/organ damage that is frequently observed during this viral outbreak. This study suggests that the biomaterial-based approach could be an effective strategy for regenerating tissues/organs damaged by SARS-CoV-2.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Rupali Singh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Deepa Ghosh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| |
Collapse
|
17
|
Ali M, Payne SL. Biomaterial-based cell delivery strategies to promote liver regeneration. Biomater Res 2021; 25:5. [PMID: 33632335 PMCID: PMC7905561 DOI: 10.1186/s40824-021-00206-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/05/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic liver disease and cirrhosis is a widespread and untreatable condition that leads to lifelong impairment and eventual death. The scarcity of liver transplantation options requires the development of new strategies to attenuate disease progression and reestablish liver function by promoting regeneration. Biomaterials are becoming an increasingly promising option to both culture and deliver cells to support in vivo viability and long-term function. There is a wide variety of both natural and synthetic biomaterials that are becoming established as delivery vehicles with their own unique advantages and disadvantages for liver regeneration. We review the latest developments in cell transplantation strategies to promote liver regeneration, with a focus on the use of both natural and synthetic biomaterials for cell culture and delivery. We conclude that future work will need to refine the use of these biomaterials and combine them with novel strategies that recapitulate liver organization and function in order to translate this strategy to clinical use.
Collapse
Affiliation(s)
- Maqsood Ali
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Samantha L Payne
- Department of Biomedical Engineering, School of Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
18
|
Rose S, Ezan F, Cuvellier M, Bruyère A, Legagneux V, Langouët S, Baffet G. Generation of proliferating human adult hepatocytes using optimized 3D culture conditions. Sci Rep 2021; 11:515. [PMID: 33436872 PMCID: PMC7804446 DOI: 10.1038/s41598-020-80019-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/10/2020] [Indexed: 02/08/2023] Open
Abstract
Generating the proliferation of differentiated normal adult human hepatocytes is a major challenge and an expected central step in understanding the microenvironmental conditions that regulate the phenotype of human hepatocytes in vitro. In this work, we described optimized 3D culture conditions of primary human hepatocytes (PHH) to trigger two waves of proliferation and we identified matrix stiffness and cell-cell interactions as the main actors necessary for this proliferation. We demonstrated that DNA replication and overexpression of cell cycle markers are modulate by the matrix stiffness while PHH cultured in 3D without prior cellular interactions did not proliferate. Besides, we showed that PHH carry out an additional cell cycle after transient inhibition of MAPK MER1/2-ERK1/2 signaling pathway. Collagen cultured hepatocytes are organized as characteristic hollow spheroids able to maintain survival, cell polarity and hepatic differentiation for long-term culture periods of at least 28 days. Remarkably, we demonstrated by transcriptomic analysis and functional experiments that proliferating cells are mature hepatocytes with high detoxication capacities. In conclusion, the advanced 3D model described here, named Hepoid, is particularly relevant for obtaining normal human proliferating hepatocytes. By allowing concomitant proliferation and differentiation, it constitutes a promising tool for many pharmacological and biotechnological applications.
Collapse
Affiliation(s)
- Sophie Rose
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Frédéric Ezan
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Marie Cuvellier
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Vincent Legagneux
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France.
| | - Georges Baffet
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France.
| |
Collapse
|
19
|
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
|
20
|
Liu Y, Hsu YH, Huang APH, Hsu SH. Semi-Interpenetrating Polymer Network of Hyaluronan and Chitosan Self-Healing Hydrogels for Central Nervous System Repair. ACS APPLIED MATERIALS & INTERFACES 2020; 12:40108-40120. [PMID: 32808527 DOI: 10.1021/acsami.0c11433] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The repair of the central nervous system (CNS) is a major challenge because of the difficulty for neurons or axons to regenerate after damages. Injectable hydrogels have been developed to deliver drugs or cells for neural repair, but these hydrogels usually require conditional stimuli or additional catalysts to control the gelling process. Self-healing hydrogels, which can be injected locally to fill tissue defects after stable gelation, are attractive candidates for CNS treatment. In the current study, the self-healing hydrogel with a semi-interpenetrating polymer network (SIPN) was prepared by incorporation of hyaluronan (HA) into the chitosan-based self-healing hydrogel. The addition of HA allowed the hydrogel to pass through a narrow needle much more easily. As the HA content increased, the hydrogel showed a more packed nanostructure and a more porous microstructure verified by coherent small-angle X-ray scattering and scanning electron microscopy. The unique structure of SIPN hydrogel enhanced the spreading, migration, proliferation, and differentiation of encapsulated neural stem cells in vitro. Compared to the pristine chitosan-based self-healing hydrogel, the SIPN hydrogel showed better biocompatibility, CNS injury repair, and functional recovery evaluated by the traumatic brain injury zebrafish model and intracerebral hemorrhage rat model. We proposed that the SIPN of HA and chitosan self-healing hydrogel allowed an adaptable environment for cell spreading and migration and had the potential as an injectable defect support for CNS repair.
Collapse
Affiliation(s)
- Yi Liu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10617, Taiwan, Republic of China
| | - Yi-Hua Hsu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei 10617, Taiwan, Republic of China
| | - Abel Po-Hao Huang
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10617, Taiwan, Republic of China
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei 10617, Taiwan, Republic of China
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10617, Taiwan, Republic of China
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan, Republic of China
| |
Collapse
|
21
|
Zhao X, Zhu Y, Laslett AL, Chan HF. Hepatic Differentiation of Stem Cells in 2D and 3D Biomaterial Systems. Bioengineering (Basel) 2020; 7:E47. [PMID: 32466173 PMCID: PMC7356247 DOI: 10.3390/bioengineering7020047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023] Open
Abstract
A critical shortage of donor livers for treating end-stage liver failure signifies the urgent need for alternative treatment options. Hepatocyte-like cells (HLC) derived from various stem cells represent a promising cell source for hepatocyte transplantation, liver tissue engineering, and development of a bioartificial liver assist device. At present, the protocols of hepatic differentiation of stem cells are optimized based on soluble chemical signals introduced in the culture medium and the HLC produced typically retain an immature phenotype. To promote further hepatic differentiation and maturation, biomaterials can be designed to recapitulate cell-extracellular matrix (ECM) interactions in both 2D and 3D configurations. In this review, we will summarize and compare various 2D and 3D biomaterial systems that have been applied to hepatic differentiation, and highlight their roles in presenting biochemical and physical cues to different stem cell sources.
Collapse
Affiliation(s)
- Xiaoyu Zhao
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; (X.Z.); (Y.Z.)
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yanlun Zhu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; (X.Z.); (Y.Z.)
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Andrew L. Laslett
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia;
- Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; (X.Z.); (Y.Z.)
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
22
|
Zhang X, Jiang T, Chen D, Wang Q, Zhang LW. Three-dimensional liver models: state of the art and their application for hepatotoxicity evaluation. Crit Rev Toxicol 2020; 50:279-309. [DOI: 10.1080/10408444.2020.1756219] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Xihui Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, P. R. China
| | - Tianyan Jiang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, P. R. China
| | - Dandan Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, P. R. China
| | - Qi Wang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control (NIFDC), China Food and Drug Administration (CFDA), Beijing, P. R. China
| | - Leshuai W. Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, P. R. China
| |
Collapse
|
23
|
Lee H, Ahn J, Jung C, Jeung Y, Cho H, Son MJ, Chung K. Optimization of 3D hydrogel microenvironment for enhanced hepatic functionality of primary human hepatocytes. Biotechnol Bioeng 2020; 117:1864-1876. [DOI: 10.1002/bit.27328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/03/2020] [Accepted: 03/07/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Ho‐Joon Lee
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| | - Jiwon Ahn
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| | - Cho‐Rock Jung
- Gene Therapy UnitKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
- Department of Functional GenomicsKorea University of Science and Technology (UST) Daejeon Republic of Korea
| | - Yun‐Ji Jeung
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| | - Hyun‐Soo Cho
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
- Department of Functional GenomicsKorea University of Science and Technology (UST) Daejeon Republic of Korea
| | - Kyung‐Sook Chung
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
- Department of Functional GenomicsKorea University of Science and Technology (UST) Daejeon Republic of Korea
- Biomedical Translational Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| |
Collapse
|
24
|
Fan D, Li Y, Wang X, Zhu T, Wang Q, Cai H, Li W, Tian Y, Liu Z. Progressive 3D Printing Technology and Its Application in Medical Materials. Front Pharmacol 2020; 11:122. [PMID: 32265689 PMCID: PMC7100535 DOI: 10.3389/fphar.2020.00122] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/28/2020] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional (3D) printing enables patient-specific anatomical level productions with high adjustability and resolution in microstructures. With cost-effective manufacturing for high productivity, 3D printing has become a leading healthcare and pharmaceutical manufacturing technology, which is suitable for variety of applications including tissue engineering models, anatomical models, pharmacological design and validation model, medical apparatus and instruments. Today, 3D printing is offering clinical available medical products and platforms suitable for emerging research fields, including tissue and organ printing. In this review, our goal is to discuss progressive 3D printing technology and its application in medical materials. The additive overview also provides manufacturing techniques and printable materials.
Collapse
Affiliation(s)
- Daoyang Fan
- Department of Orthopedic, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Yan Li
- Department of Orthopedic, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Tengjiao Zhu
- Department of Orthopedic, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Qi Wang
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Hong Cai
- Department of Orthopedic, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Weishi Li
- Department of Orthopedic, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Yun Tian
- Department of Orthopedic, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Zhongjun Liu
- Department of Orthopedic, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| |
Collapse
|
25
|
Abalymov A, Parakhonskiy B, Skirtach AG. Polymer- and Hybrid-Based Biomaterials for Interstitial, Connective, Vascular, Nerve, Visceral and Musculoskeletal Tissue Engineering. Polymers (Basel) 2020; 12:E620. [PMID: 32182751 PMCID: PMC7182904 DOI: 10.3390/polym12030620] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/19/2020] [Accepted: 03/03/2020] [Indexed: 12/11/2022] Open
Abstract
In this review, materials based on polymers and hybrids possessing both organic and inorganic contents for repairing or facilitating cell growth in tissue engineering are discussed. Pure polymer based biomaterials are predominantly used to target soft tissues. Stipulated by possibilities of tuning the composition and concentration of their inorganic content, hybrid materials allow to mimic properties of various types of harder tissues. That leads to the concept of "one-matches-all" referring to materials possessing the same polymeric base, but different inorganic content to enable tissue growth and repair, proliferation of cells, and the formation of the ECM (extra cellular matrix). Furthermore, adding drug delivery carriers to coatings and scaffolds designed with such materials brings additional functionality by encapsulating active molecules, antibacterial agents, and growth factors. We discuss here materials and methods of their assembly from a general perspective together with their applications in various tissue engineering sub-areas: interstitial, connective, vascular, nervous, visceral and musculoskeletal tissues. The overall aims of this review are two-fold: (a) to describe the needs and opportunities in the field of bio-medicine, which should be useful for material scientists, and (b) to present capabilities and resources available in the area of materials, which should be of interest for biologists and medical doctors.
Collapse
Affiliation(s)
- Anatolii Abalymov
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | | | - Andre G. Skirtach
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
26
|
Spontaneously and reversibly forming phospholipid polymer hydrogels as a matrix for cell engineering. Biomaterials 2020; 230:119628. [DOI: 10.1016/j.biomaterials.2019.119628] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 12/16/2022]
|
27
|
Generation of expandable human pluripotent stem cell-derived hepatocyte-like liver organoids. J Hepatol 2019; 71:970-985. [PMID: 31299272 DOI: 10.1016/j.jhep.2019.06.030] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS The development of hepatic models capable of long-term expansion with competent liver functionality is technically challenging in a personalized setting. Stem cell-based organoid technologies can provide an alternative source of patient-derived primary hepatocytes. However, self-renewing and functionally competent human pluripotent stem cell (PSC)-derived hepatic organoids have not been developed. METHODS We developed a novel method to efficiently and reproducibly generate functionally mature human hepatic organoids derived from PSCs, including human embryonic stem cells and induced PSCs. The maturity of the organoids was validated by a detailed transcriptome analysis and functional performance assays. The organoids were applied to screening platforms for the prediction of toxicity and the evaluation of drugs that target hepatic steatosis through real-time monitoring of cellular bioenergetics and high-content analyses. RESULTS Our organoids were morphologically indistinguishable from adult liver tissue-derived epithelial organoids and exhibited self-renewal. With further maturation, their molecular features approximated those of liver tissue, although these features were lacking in 2D differentiated hepatocytes. Our organoids preserved mature liver properties, including serum protein production, drug metabolism and detoxifying functions, active mitochondrial bioenergetics, and regenerative and inflammatory responses. The organoids exhibited significant toxic responses to clinically relevant concentrations of drugs that had been withdrawn from the market due to hepatotoxicity and recapitulated human disease phenotypes such as hepatic steatosis. CONCLUSIONS Our organoids exhibit self-renewal (expandable and further able to differentiate) while maintaining their mature hepatic characteristics over long-term culture. These organoids may provide a versatile and valuable platform for physiologically and pathologically relevant hepatic models in the context of personalized medicine. LAY SUMMARY A functionally mature, human cell-based liver model exhibiting human responses in toxicity prediction and drug evaluation is urgently needed for pre-clinical drug development. Here, we develop a novel human pluripotent stem cell-derived hepatocyte-like liver organoid that is critically advanced in terms of its generation method, functional performance, and application technologies. Our organoids can contribute to the better understanding of liver development and regeneration, and provide insights for metabolic studies and disease modeling, as well as toxicity assessments and drug screening for personalized medicine.
Collapse
|
28
|
Agarwal T, Subramanian B, Maiti TK. Liver Tissue Engineering: Challenges and Opportunities. ACS Biomater Sci Eng 2019; 5:4167-4182. [PMID: 33417776 DOI: 10.1021/acsbiomaterials.9b00745] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver tissue engineering aims at the possibility of reproducing a fully functional organ for the treatment of acute and chronic liver disorders. Approaches in this field endeavor to replace organ transplantation (gold standard treatment for liver diseases in a clinical setting) with in vitro developed liver tissue constructs. However, the complexity of the liver microarchitecture and functionality along with the limited supply of cellular components of the liver pose numerous challenges. This review provides a comprehensive outlook onto how the physicochemical, mechanobiological, and spatiotemporal aspects of the substrates could be tuned to address current challenges in the field. We also highlight the strategic advancements made in the field so far for the development of artificial liver tissue. We further showcase the currently available prototypes in research and clinical trials, which shows the hope for the future of liver tissue engineering.
Collapse
|
29
|
Ryu JS, Lee M, Mun SJ, Hong SH, Lee HJ, Ahn HS, Chung KS, Kim GH, Son MJ. Targeting CYP4A attenuates hepatic steatosis in a novel multicellular organotypic liver model. J Biol Eng 2019; 13:69. [PMID: 31406506 PMCID: PMC6686528 DOI: 10.1186/s13036-019-0198-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) begins as simple hepatic steatosis, but further progress to chronic liver diseases results in severe liver damage and hepatic failure. However, therapeutic options are scarce due to the lack of reliable human in vitro liver models for understanding disease progression mechanisms and developing therapies. Results We describe here a novel method for generating 3D hepatic spheroids using HepaRG cells, vascular endothelial cells, and mesenchymal stem cells cultured on a thick layer of soft matrix in a narrow conical tube; this method improved self-organization efficiency and functional competence. We further developed a 3D hepatic steatosis model with excess glucose and palmitate, accurately recapitulating steatosis phenotypes such as neutral lipid accumulation, enhanced expression of lipogenesis and gluconeogenesis markers, increased intracellular triglyceride content, and reduced glucose uptake. The expression and activity of cytochrome P450 4A (CYP4A), a hepatic glucose and lipid homeostasis enzyme, that is highly expressed in liver tissues from NAFLD patients, was induced in our in vitro steatosis model, and inhibiting CYP4A with the selective inhibitor HET0016 or a specific siRNA ameliorated steatosis-related pathology through reduced ER stress and improved insulin signaling. Conclusions We provide here a novel 3D human cell-based hepatic model that can be easily generated and reliably simulate hepatic steatosis pathology. We have experimentally validated its potential for target validation and drug evaluation by focusing on CYP4A, which may serve as a translational platform for drug development.
Collapse
Affiliation(s)
- Jae-Sung Ryu
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Minji Lee
- 2Drug and Disease Target Team, Division of Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Chungcheong 28119 Republic of Korea.,3Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon, 34113 Republic of Korea
| | - Seon Ju Mun
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea.,4Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-gu, Daejeon, 34113 Republic of Korea
| | - Sin-Hyoung Hong
- 2Drug and Disease Target Team, Division of Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Chungcheong 28119 Republic of Korea.,3Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon, 34113 Republic of Korea
| | - Ho-Joon Lee
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Hyo-Suk Ahn
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Kyung-Sook Chung
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea.,4Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-gu, Daejeon, 34113 Republic of Korea.,5Biomedical Translational Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Gun-Hwa Kim
- 2Drug and Disease Target Team, Division of Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Chungcheong 28119 Republic of Korea.,3Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon, 34113 Republic of Korea.,6Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, 34134 Republic of Korea
| | - Myung Jin Son
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea.,4Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-gu, Daejeon, 34113 Republic of Korea
| |
Collapse
|
30
|
Natale A, Vanmol K, Arslan A, Van Vlierberghe S, Dubruel P, Van Erps J, Thienpont H, Buzgo M, Boeckmans J, De Kock J, Vanhaecke T, Rogiers V, Rodrigues RM. Technological advancements for the development of stem cell-based models for hepatotoxicity testing. Arch Toxicol 2019; 93:1789-1805. [PMID: 31037322 DOI: 10.1007/s00204-019-02465-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/18/2019] [Indexed: 02/07/2023]
Abstract
Stem cells are characterized by their self-renewal capacity and their ability to differentiate into multiple cell types of the human body. Using directed differentiation strategies, stem cells can now be converted into hepatocyte-like cells (HLCs) and therefore, represent a unique cell source for toxicological applications in vitro. However, the acquired hepatic functionality of stem cell-derived HLCs is still significantly inferior to primary human hepatocytes. One of the main reasons for this is that most in vitro models use traditional two-dimensional (2D) setups where the flat substrata cannot properly mimic the physiology of the human liver. Therefore, 2D-setups are progressively being replaced by more advanced culture systems, which attempt to replicate the natural liver microenvironment, in which stem cells can better differentiate towards HLCs. This review highlights the most recent cell culture systems, including scaffold-free and scaffold-based three-dimensional (3D) technologies and microfluidics that can be employed for culture and hepatic differentiation of stem cells intended for hepatotoxicity testing. These methodologies have shown to improve in vitro liver cell functionality according to the in vivo liver physiology and allow to establish stem cell-based hepatic in vitro platforms for the accurate evaluation of xenobiotics.
Collapse
Affiliation(s)
- Alessandra Natale
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Koen Vanmol
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | - Aysu Arslan
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Jürgen Van Erps
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | - Hugo Thienpont
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | | | - Joost Boeckmans
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Joery De Kock
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Vera Rogiers
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Robim M Rodrigues
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
31
|
Ahn J, Lee HJ, Oh SJ, Kim W, Mun SJ, Lee JH, Jung CR, Cho HS, Kim DS, Son MJ, Chung KS. Developing scalable cultivation systems of hepatic spheroids for drug metabolism via genomic and functional analyses. Biotechnol Bioeng 2019; 116:1496-1508. [PMID: 30737956 DOI: 10.1002/bit.26954] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 01/18/2019] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
Spheroids, a widely used three-dimensional (3D) culture model, are standard in hepatocyte culture as they preserve long-term hepatocyte functionality and enhance survivability. In this study, we investigated the effects of three operation modes in 3D culture - static, orbital shaking, and under vertical bidirectional flow using spheroid forming units (SFUs) on hepatic differentiation and drug metabolism to propose the best for mass production of functionally enhanced spheroids. Spheroids in SFUs exhibited increased hepatic gene expression, albumin secretion, and cytochrome P450 3A4 (CYP3A4) activity during the differentiation period (12 days). SFUs advantages include facilitated mass production and a relatively earlier peak of CYP3A4 activity. However, CYP3A4 activity was not well maintained under dimethyl sulfoxide (DMSO)-free conditions (13-18 days), dramatically reducing drug metabolism capability. Continued shear stimulation without differentiation stimuli in assay conditions markedly attenuated CYP3A4 activity, which was less severe in static conditions. In this condition, SFU spheroids exhibited dedifferentiation characteristics, such as increased proliferation and Notch signaling genes. We found that the dedifferentiation could be overcome by using the serum-free medium formulation. Therefore, we suggest that SFUs represent the best option for the mass production of functionally improved spheroids and so the serum-free conditions should be maintained during drug metabolism analysis.
Collapse
Affiliation(s)
- Jiwon Ahn
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ho-Joon Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Soo Jin Oh
- New Drug Development Center, Asan Medical Center and Convergence Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Wantae Kim
- Biomedical Translational Research Center, KRIBB, Daejeon, Republic of Korea
| | - Seon Ju Mun
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jae-Hye Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Cho-Rock Jung
- Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.,Gene Therapy Unit, KRIBB, Daejeon, Republic of Korea
| | - Hyun-Soo Cho
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Dae-Soo Kim
- Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.,Genome Research Center, KRIBB, Daejeon, Republic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kyung-Sook Chung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Biomedical Translational Research Center, KRIBB, Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
32
|
Lü WD, Sun RF, Hu YR, Lu JR, Gu L, Liu ZG, Lei GY, Qiang Z, Cai L. Photooxidatively crosslinked acellular tumor extracellular matrices as potential tumor engineering scaffolds. Acta Biomater 2018; 71:460-473. [PMID: 29555461 DOI: 10.1016/j.actbio.2018.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/08/2018] [Accepted: 03/08/2018] [Indexed: 12/12/2022]
Abstract
Acellular tumor extracellular matrices (ECMs) have limitations when employed as three-dimensional (3D) scaffolds for tumor engineering. In this work, methylene blue-mediated photooxidation was used to crosslink acellular tumor ECMs. Photooxidative crosslinking greatly increased the stiffness of acellular tumor ECM scaffolds but barely altered the Amide III band of the secondary structure of polypeptides and proteins. MCF-7, HepG2 and A549 cells cultured on photooxidatively crosslinked acellular tumor ECM scaffolds exhibited greater cell number per scaffold, more IL-8 and VEGF secretion, and increase migration and invasion abilities than cells cultured on uncrosslinked acellular tumor ECM scaffolds. The three tumor cell lines cultured on the stiffer photooxidatively crosslinked acellular matrices acquire mesenchymal properties (mesenchymal shift) and dedifferentiated phenotypes. Furthermore, the malignant phenotypes induced in vitro when cultured on the crosslinked scaffold promoted the in vivo tumor growth of BALB/c nude mice. Finally, the dedifferentiated cancer cells, including MCF-7, HepG2 and A549 cells, were less sensitive to chemotherapeutics. Thus, photooxidatively crosslinked acellular tumor ECMs have potentials as 3D tumor engineering scaffolds for cancer research. STATEMENT OF SIGNIFICANCE Natural material scaffolds have been successfully used as 3D matrices to study the in vitro tumor cell growth and mimic the in vivo tumor microenvironment. Acellular tumor ECMs are developed as 3D scaffolds for tumor engineering but have limitations in terms of elastic modulus and cell spheroid formation. Here we use methylene blue-mediated photooxidation to crosslink acellular tumor ECMs and investigate the influence of photooxidative crosslinking on structural, mechanical and biological characteristics of acellular tumor ECM scaffolds. It is the first study to evaluate the feasibility of photooxidatively crosslinked acellular tumor ECMs as 3D scaffolds for cancer research and the results are encouraging. Moreover, this study provides new research areas in regard to photodynamic therapy (PDT) for Cancer.
Collapse
Affiliation(s)
- Wei-Dong Lü
- Department of Thoracic Surgery, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Rui-Fang Sun
- Department of Pathology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Ye-Rong Hu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| | - Jan-Rong Lu
- Department of Pathology, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Lu Gu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Zhi-Gang Liu
- Department of Thoracic Surgery, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Guang-Yan Lei
- Department of Thoracic Surgery, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zhun Qiang
- Department of Thoracic Surgery, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Lin Cai
- Department of Pathology, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| |
Collapse
|
33
|
Grix T, Ruppelt A, Thomas A, Amler AK, Noichl BP, Lauster R, Kloke L. Bioprinting Perfusion-Enabled Liver Equivalents for Advanced Organ-on-a-Chip Applications. Genes (Basel) 2018; 9:genes9040176. [PMID: 29565814 PMCID: PMC5924518 DOI: 10.3390/genes9040176] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/08/2018] [Accepted: 03/19/2018] [Indexed: 12/30/2022] Open
Abstract
Many tissue models have been developed to mimic liver-specific functions for metabolic and toxin conversion in in vitro assays. Most models represent a 2D environment rather than a complex 3D structure similar to native tissue. To overcome this issue, spheroid cultures have become the gold standard in tissue engineering. Unfortunately, spheroids are limited in size due to diffusion barriers in their dense structures, limiting nutrient and oxygen supply. Recent developments in bioprinting techniques have enabled us to engineer complex 3D structures with perfusion-enabled channel systems to ensure nutritional supply within larger, densely-populated tissue models. In this study, we present a proof-of-concept for the feasibility of bioprinting a liver organoid by combining HepaRG and human stellate cells in a stereolithographic printing approach, and show basic characterization under static cultivation conditions. Using standard tissue engineering analytics, such as immunohistology and qPCR, we found higher albumin and cytochrome P450 3A4 (CYP3A4) expression in bioprinted liver tissues compared to monolayer controls over a two-week cultivation period. In addition, the expression of tight junctions, liver-specific bile transporter multidrug resistance-associated protein 2 (MRP2), and overall metabolism (glucose, lactate, lactate dehydrogenase (LDH)) were found to be stable. Furthermore, we provide evidence for the perfusability of the organoids’ intrinsic channel system. These results motivate new approaches and further development in liver tissue engineering for advanced organ-on-a-chip applications and pharmaceutical developments.
Collapse
Affiliation(s)
| | - Alicia Ruppelt
- Fachgebiet für Medizinische Biotechnologie, Technische Universität Berlin, 13355 Berlin, Germany.
| | | | - Anna-Klara Amler
- Fachgebiet für Medizinische Biotechnologie, Technische Universität Berlin, 13355 Berlin, Germany.
| | - Benjamin P Noichl
- Fachgebiet für Medizinische Biotechnologie, Technische Universität Berlin, 13355 Berlin, Germany.
| | - Roland Lauster
- Fachgebiet für Medizinische Biotechnologie, Technische Universität Berlin, 13355 Berlin, Germany.
| | | |
Collapse
|