1
|
Lu TY, Ji Y, Lyu C, Shen EN, Sun Y, Xiang Y, Meng-Saccoccio T, Feng GS, Chen S. Bioprinted high cell density liver model with improved hepatic metabolic functions. Biomaterials 2025; 320:123256. [PMID: 40101310 DOI: 10.1016/j.biomaterials.2025.123256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
In vitro liver tissue models are valuable for studying liver function, understanding liver diseases, and screening candidate drugs for toxicity and efficacy. While three-dimensional (3D) bioprinting shows promise in creating various types of functional tissues, current efforts to engineer a functional liver tissue face challenges in replicating native high cell density (HCD) and maintaining long-term cell viability. HCD is crucial for establishing the cell-cell interactions necessary to mimic the liver's metabolic and detoxification functions. However, HCD bioinks exacerbate light scattering in light-based 3D bioprinting. In this study, we incorporated iodixanol into our bioink formulation to minimize light scattering, enabling the fabrication of hepatic tissue constructs with an HCD of 8 × 107 cells/mL while maintaining high cell viability (∼80 %). The printed dense hepatic tissue constructs showed enhanced cell-cell interactions, as evidenced by increased expression of E-cadherin and ZO-1. Furthermore, these constructs promoted albumin secretion, urea production, and P450 metabolic activity. Additionally, HCD hepatic tissue inactivated the YAP/TAZ pathway via cell-cell interactions, preserving primary hepatocyte functions. Further screening revealed that hepatocytes in the dense model were more sensitive to drug treatments than those in a lower-density hepatic model, highlighting the importance of HCD in recapitulating the physiological drug responses. Overall, our approach represents a significant advancement in liver tissue engineering, providing a promising platform for the development of physiologically relevant in vitro liver models for drug screening and toxicity testing.
Collapse
Affiliation(s)
- Ting-Yu Lu
- Program in Materials Science and Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yichun Ji
- Department of Pathology, Department of Molecular Biology, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Cheng Lyu
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Erin Nicole Shen
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yazhi Sun
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yi Xiang
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tobias Meng-Saccoccio
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Gen-Sheng Feng
- Department of Pathology, Department of Molecular Biology, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Shaochen Chen
- Program in Materials Science and Engineering, University of California San Diego, La Jolla, CA, 92093, USA; Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
2
|
Deng W, Yang X, Yu J, Omari-Siaw E, Xu X. Recent advances of physiochemical cues on surfaces for directing cell fates. Colloids Surf B Biointerfaces 2025; 250:114550. [PMID: 39929022 DOI: 10.1016/j.colsurfb.2025.114550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/12/2025]
Abstract
Surface modification plays an essential role in dictating cell behavior and fate, as it creates a microenvironment that profoundly influences cell attachment, migration, proliferation, and differentiation. This review aims to the intricate interplay of culture surface properties, including topography, stiffness, charge, and chemical modifications, demonstrating their profound impact on cell destiny. We explore the nuanced responses of cells to varying surface topographies, from nano- to microscale features, elucidating the influence of geometric patterns and roughness. We also investigate the impact of substrate stiffness, highlighting the way cells perceive and respond to mechanical cues mimicking their native environments. The role of surface charge is examined, revealing how electrostatic interactions influence cell adhesion, signaling, and cell fate decisions. Finally, we delve into the diverse effects of chemical modifications, including the presentation of bioactive molecules, growth factors, and extracellular matrix (ECM) components, demonstrating their ability to guide cell behavior and stimulate specific cellular responses. This review offers comprehensive insights into the important role of surface properties in shaping cell fate, offering promising avenues for developing sophisticated cell culture platforms for applications in drug discovery, regenerative medicine, and fundamental research.
Collapse
Affiliation(s)
- Wenwen Deng
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Xiufen Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Emmanuel Omari-Siaw
- Department of Pharmaceutical Science, Kumasi Technical University, PO Box 854, Kumasi, Ashanti, Ghana
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| |
Collapse
|
3
|
Min K, Karuppannan SK, Tae G. The impact of matrix stiffness on hepatic cell function, liver fibrosis, and hepatocellular carcinoma-Based on quantitative data. BIOPHYSICS REVIEWS 2024; 5:021306. [PMID: 38846007 PMCID: PMC11151446 DOI: 10.1063/5.0197875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/14/2024] [Indexed: 06/09/2024]
Abstract
Over the past few decades, extensive research has explored the development of supportive scaffold materials for in vitro hepatic cell culture, to effectively mimic in vivo microenvironments. It is crucial for hepatic disease modeling, drug screening, and therapeutic evaluations, considering the ethical concerns and practical challenges associated with in vivo experiments. This review offers a comprehensive perspective on hepatic cell culture using bioscaffolds by encompassing all stages of hepatic diseases-from a healthy liver to fibrosis and hepatocellular carcinoma (HCC)-with a specific focus on matrix stiffness. This review begins by providing physiological and functional overviews of the liver. Subsequently, it explores hepatic cellular behaviors dependent on matrix stiffness from previous reports. For hepatic cell activities, softer matrices showed significant advantages over stiffer ones in terms of cell proliferation, migration, and hepatic functions. Conversely, stiffer matrices induced myofibroblastic activation of hepatic stellate cells, contributing to the further progression of fibrosis. Elevated matrix stiffness also correlates with HCC by increasing proliferation, epithelial-mesenchymal transition, metastasis, and drug resistance of HCC cells. In addition, we provide quantitative information on available data to offer valuable perspectives for refining the preparation and development of matrices for hepatic tissue engineering. We also suggest directions for further research on this topic.
Collapse
Affiliation(s)
- Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Sathish Kumar Karuppannan
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
4
|
Carpentier N, Ye S, Delemarre MD, Van der Meeren L, Skirtach AG, van der Laan LJW, Schneeberger K, Spee B, Dubruel P, Van Vlierberghe S. Gelatin-Based Hybrid Hydrogels as Matrices for Organoid Culture. Biomacromolecules 2024; 25:590-604. [PMID: 38174962 DOI: 10.1021/acs.biomac.2c01496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The application of liver organoids is very promising in the field of liver tissue engineering; however, it is still facing some limitations. One of the current major limitations is the matrix in which they are cultured. The mainly undefined and murine-originated tumor matrices derived from Engelbreth-Holm-Swarm (EHS) sarcoma, such as Matrigel, are still the standard culturing matrices for expansion and differentiation of organoids toward hepatocyte-like cells, which will obstruct its future clinical application potential. In this study, we exploited the use of newly developed highly defined hydrogels as potential matrices for the culture of liver organoids and compared them to Matrigel and two hydrogels that were already researched in the field of organoid research [i.e., polyisocyanopeptides, enriched with laminin-entactin complex (PIC-LEC) and gelatin methacryloyl (GelMA)]. The newly developed hydrogels are materials that have a physicochemical resemblance with native liver tissue. Norbornene-modified dextran cross-linked with thiolated gelatin (DexNB-GelSH) has a swelling ratio and macro- and microscale properties that highly mimic liver tissue. Norbornene-modified chondroitin sulfate cross-linked with thiolated gelatin (CSNB-GelSH) contains chondroitin sulfate, which is a glycosaminoglycan (GAG) that is present in the liver ECM. Furthermore, CSNB-GelSH hydrogels with different mechanical properties were evaluated. Bipotent intrahepatic cholangiocyte organoids (ICOs) were applied in this work and encapsulated in these materials. This research revealed that the newly developed materials outperformed Matrigel, PIC-LEC, and GelMA in the differentiation of ICOs toward hepatocyte-like cells. Furthermore, some trends indicate that an interplay of both the chemical composition and the mechanical properties has an influence on the relative expression of certain hepatocyte markers. Both DexNB-GelSH and CSNB-GelSH showed promising results for the expansion and differentiation of intrahepatic cholangiocyte organoids. The stiffest CSNB-GelSH hydrogel even significantly outperformed Matrigel based on ALB, BSEP, and CYP3A4 gene expression, being three important hepatocyte markers.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Shicheng Ye
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Maarten D Delemarre
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Louis Van der Meeren
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - André G Skirtach
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Kerstin Schneeberger
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Bart Spee
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| |
Collapse
|
5
|
Carvalho AM, Bansal R, Barrias CC, Sarmento B. The Material World of 3D-Bioprinted and Microfluidic-Chip Models of Human Liver Fibrosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307673. [PMID: 37961933 DOI: 10.1002/adma.202307673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Biomaterials are extensively used to mimic cell-matrix interactions, which are essential for cell growth, function, and differentiation. This is particularly relevant when developing in vitro disease models of organs rich in extracellular matrix, like the liver. Liver disease involves a chronic wound-healing response with formation of scar tissue known as fibrosis. At early stages, liver disease can be reverted, but as disease progresses, reversion is no longer possible, and there is no cure. Research for new therapies is hampered by the lack of adequate models that replicate the mechanical properties and biochemical stimuli present in the fibrotic liver. Fibrosis is associated with changes in the composition of the extracellular matrix that directly influence cell behavior. Biomaterials could play an essential role in better emulating the disease microenvironment. In this paper, the recent and cutting-edge biomaterials used for creating in vitro models of human liver fibrosis are revised, in combination with cells, bioprinting, and/or microfluidics. These technologies have been instrumental to replicate the intricate structure of the unhealthy tissue and promote medium perfusion that improves cell growth and function, respectively. A comprehensive analysis of the impact of material hints and cell-material interactions in a tridimensional context is provided.
Collapse
Affiliation(s)
- Ana Margarida Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ruchi Bansal
- Translational Liver Research, Department of Medical Cell Biophysics, Technical Medical Center, Faculty of Science and Technology, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Cristina C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| |
Collapse
|
6
|
Carpentier N, Van der Meeren L, Skirtach AG, Devisscher L, Van Vlierberghe H, Dubruel P, Van Vlierberghe S. Gelatin-Based Hybrid Hydrogel Scaffolds: Toward Physicochemical Liver Mimicry. Biomacromolecules 2023; 24:4333-4347. [PMID: 35914189 DOI: 10.1021/acs.biomac.2c00643] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
There exists a clear need to develop novel materials that could serve liver tissue engineering purposes. Those materials need to be researched for the development of bioengineered liver tissue as an alternative to donor livers, as well as for materials that could be applied for scaffolds to develop an in vitro model for drug-induced liver injury (DILI) detection . In this paper, the hydrogels oxidized dextran-gelatin (Dexox-Gel) and norbornene-modified dextran-thiolated gelatin (DexNB-GelSH) were developed, and their feasibility toward processing via indirect 3D-printing was investigated with the aim to develop hydrogel scaffolds that physicochemically mimic native liver tissue. Furthermore, their in vitro biocompatibility was assessed using preliminary biological tests using HepG2 cells. Both materials were thoroughly physicochemically characterized and benchmarked to the methacrylated gelatin (GelMA) reference material. Due to inferior properties, Dexox-gel was not further processed into 3D-hydrogel scaffolds. This research revealed that DexNB-GelSH exhibited physicochemical properties that were in excellent agreement with the properties of natural liver tissue in contrast to GelMA. In combination with an equally good biological evaluation of DexNB-GelSH in comparison with GelMA based on an MTS proliferation assay and an albumin quantification assay, DexNB-GelSH can be considered promising in the field of liver tissue engineering.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Louis Van der Meeren
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - André G Skirtach
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - Lindsey Devisscher
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences; Liver Research Center Ghent, Ghent University, Ghent 9000, Belgium
| | - Hans Van Vlierberghe
- Hepatology Research Unit, Dpt Internal Medicine and Pediatrics; Liver Research Center Ghent, Ghent University, Ghent 9000, Belgium
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| |
Collapse
|
7
|
Oliva-Vilarnau N, Vorrink SU, Büttner FA, Heinrich T, Sensbach J, Koscielski I, Wienke D, Petersson C, Perrin D, Lauschke VM. Comparative analysis of YAP/TEAD inhibitors in 2D and 3D cultures of primary human hepatocytes reveals a novel non-canonical mechanism of CYP induction. Biochem Pharmacol 2023; 215:115755. [PMID: 37607620 DOI: 10.1016/j.bcp.2023.115755] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/24/2023]
Abstract
Induction of cytochrome P450 (CYP) genes constitutes an important cause of drug-drug interactions and preclinical evaluation of induction liability is mandatory for novel drug candidates. YAP/TEAD signaling has emerged as an attractive target for various oncological indications and multiple chemically distinct YAP/TEAD inhibitors are rapidly progressing towards clinical stages. Here, we tested the liability for CYP induction of a diverse set of YAP/TEAD inhibitors with different modes of action and TEAD isoform selectivity profiles in monolayers and 3D spheroids of primary human hepatocytes (PHH). We found that YAP/TEAD inhibition resulted in broad induction of CYPs in 2D monolayers, whereas, if at all, only marginal induction was seen in spheroid culture. Comprehensive RNA-Seq indicated that YAP/TEAD signaling was increased in 2D culture compared to spheroids, which was paralleled by elevated activities of the interacting transcription factors LXR and ESRRA, likely at least in part due to altered mechanosensing. Inhibition of this YAP/TEAD hyperactivation resulted in an overall reduction of hepatocyte dedifferentiation marked by increased hepatic functionality, including CYPs. These results thus demonstrate that the observed induction is due to on-target effects of the compounds rather than direct activation of xenobiotic sensing nuclear receptors. Combined, the presented data link hepatocyte dedifferentiation to YAP/TEAD dysregulation, reveal a novel non-canonical pathway of CYP induction and highlight the advantage of organotypic 3D cultures to predict clinically relevant pharmacokinetic properties, particularly for atypical induction mechanisms.
Collapse
Affiliation(s)
- Nuria Oliva-Vilarnau
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Florian A Büttner
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany
| | - Timo Heinrich
- Department of Medicinal Chemistry and Drug Design, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Janike Sensbach
- Department of Chemical and Pre-Clinical Safety, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Isabel Koscielski
- Department of Chemical and Pre-Clinical Safety, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Dirk Wienke
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Carl Petersson
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Dominique Perrin
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; HepaPredict AB, Stockholm, Sweden; Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
8
|
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: 4.5] [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
|
9
|
Bate TSR, Shanahan W, Casillo JP, Grant R, Forbes SJ, Callanan A. Rat liver ECM incorporated into electrospun polycaprolactone scaffolds as a platform for hepatocyte culture. J Biomed Mater Res B Appl Biomater 2022; 110:2612-2623. [PMID: 35734943 PMCID: PMC9796056 DOI: 10.1002/jbm.b.35115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/13/2022] [Accepted: 06/08/2022] [Indexed: 12/30/2022]
Abstract
Liver disease is expanding across the globe; however, health-care systems still lack approved pharmaceutical treatment strategies to mitigate potential liver failures. Organ transplantation is the only treatment for liver failure and with increasing cases of liver disease, transplant programs increasingly cannot provide timely transplant availability for all patients. The development of pharmaceutical mitigation strategies is clearly necessary and methods to improve drug development processes are considered vital for this purpose. Herein, we present a methodology for incorporating whole organ decellularised rat liver ECM (rLECM) into polycaprolactone (PCL) electrospun scaffolds with the aim of producing biologically relevant liver tissue models. rLECM PCL scaffolds have been produced with 5 w/w% and 10 w/w% rLECM:PCL and were analyzed by SEM imaging, tensile mechanical analyses and FTIR spectroscopy. The hepatocellular carcinoma cell line, HepG2, was cultured upon the scaffolds for 14 days and were analyzed through cell viability assay, DNA quantification, albumin quantification, immunohistochemistry, and RT-qPCR gene expression analysis. Results showed significant increases in proliferative activity of HepG2 on rLECM containing scaffolds alongside maintained key gene expression. This study confirms that rLECM can be utilized to modulate the bioactivity of electrospun PCL scaffolds and has the potential to produce electrospun scaffolds suitable for enhanced hepatocyte cultures and in-vitro liver tissue models.
Collapse
Affiliation(s)
- Thomas S. R. Bate
- School of EngineeringInstitute for Bioengineering, University of EdinburghEdinburghUK
| | - William Shanahan
- School of EngineeringInstitute for Bioengineering, University of EdinburghEdinburghUK
| | - Joseph P. Casillo
- School of GeoSciencesUniversity of Edinburgh, Grant InstituteEdinburghUK
| | - Rhiannon Grant
- MERLN InstituteMaastricht UniversityMaastrichtThe Netherlands
| | - Stuart J. Forbes
- Centre for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | - Anthony Callanan
- School of EngineeringInstitute for Bioengineering, University of EdinburghEdinburghUK
| |
Collapse
|
10
|
Kaur S, Kidambi S, Ortega-Ribera M, Thuy LTT, Nieto N, Cogger VC, Xie WF, Tacke F, Gracia-Sancho J. In Vitro Models for the Study of Liver Biology and Diseases: Advances and Limitations. Cell Mol Gastroenterol Hepatol 2022; 15:559-571. [PMID: 36442812 PMCID: PMC9868680 DOI: 10.1016/j.jcmgh.2022.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
In vitro models of liver (patho)physiology, new technologies, and experimental approaches are progressing rapidly. Based on cell lines, induced pluripotent stem cells or primary cells derived from mouse or human liver as well as whole tissue (slices), such in vitro single- and multicellular models, including complex microfluidic organ-on-a-chip systems, provide tools to functionally understand mechanisms of liver health and disease. The International Society of Hepatic Sinusoidal Research (ISHSR) commissioned this working group to review the currently available in vitro liver models and describe the advantages and disadvantages of each in the context of evaluating their use for the study of liver functionality, disease modeling, therapeutic discovery, and clinical applicability.
Collapse
Affiliation(s)
- Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Srivatsan Kidambi
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Martí Ortega-Ribera
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Le Thi Thanh Thuy
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Natalia Nieto
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois
| | - Victoria C Cogger
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jordi Gracia-Sancho
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, CIBEREHD, Barcelona, Spain; Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland; Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland.
| |
Collapse
|
11
|
Li N, Zhang X, Zhou J, Li W, Shu X, Wu Y, Long M. Multiscale biomechanics and mechanotransduction from liver fibrosis to cancer. Adv Drug Deliv Rev 2022; 188:114448. [PMID: 35820602 DOI: 10.1016/j.addr.2022.114448] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/08/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023]
Abstract
A growing body of multiscale biomechanical studies has been proposed to highlight the mechanical cues in the development of hepatic fibrosis and cancer. At the cellular level, changes in mechanical microenvironment induce phenotypic and functional alterations of hepatic cells, initiating a positive feedback loop that promotes liver fibrogenesis and hepatocarcinogenesis. Tumor mechanical microenvironment of hepatocellular carcinoma facilitates tumor cell growth and metastasis, and hinders the drug delivery and immunotherapy. At the molecular level, mechanical forces are sensed and transmitted into hepatic cells via allosteric activation of mechanoreceptors on the cell membrane, leading to the activation of various mechanotransduction pathways including integrin and YAP signaling and then regulating cell function. Thus, the application of mechanomedicine concept in the treatment of liver diseases is promising for rational design and cell-specific delivery of therapeutic drugs. This review mainly discusses the correlation between biomechanical cues and liver diseases from the viewpoint of mechanobiology.
Collapse
Affiliation(s)
- Ning Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Zhou
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Wang Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyu Shu
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Wu
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mian Long
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
12
|
Dai Q, Jiang W, Huang F, Song F, Zhang J, Zhao H. Recent Advances in Liver Engineering With Decellularized Scaffold. Front Bioeng Biotechnol 2022; 10:831477. [PMID: 35223793 PMCID: PMC8866951 DOI: 10.3389/fbioe.2022.831477] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/24/2022] [Indexed: 12/02/2022] Open
Abstract
Liver transplantation is currently the only effective treatment for patients with end-stage liver disease; however, donor liver scarcity is a notable concern. As a result, extensive endeavors have been made to diversify the source of donor livers. For example, the use of a decellularized scaffold in liver engineering has gained considerable attention in recent years. The decellularized scaffold preserves the original orchestral structure and bioactive chemicals of the liver, and has the potential to create a de novo liver that is fit for transplantation after recellularization. The structure of the liver and hepatic extracellular matrix, decellularization, recellularization, and recent developments are discussed in this review. Additionally, the criteria for assessment and major obstacles in using a decellularized scaffold are covered in detail.
Collapse
Affiliation(s)
- Qingqing Dai
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Wei Jiang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Huang
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fei Song
- Department of Urology, Jena University Hospital, Jena, Germany
| | - Jiqian Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Jiqian Zhang, ; Hongchuan Zhao,
| | - Hongchuan Zhao
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Jiqian Zhang, ; Hongchuan Zhao,
| |
Collapse
|
13
|
Gwon K, Hong HJ, Gonzalez-Suarez AM, Slama MQ, Choi D, Hong J, Baskaran H, Stybayeva G, Peterson QP, Revzin A. Bioactive hydrogel microcapsules for guiding stem cell fate decisions by release and reloading of growth factors. Bioact Mater 2021; 15:1-14. [PMID: 35386345 PMCID: PMC8941170 DOI: 10.1016/j.bioactmat.2021.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/22/2021] [Accepted: 12/12/2021] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cells (hPSC) hold considerable promise as a source of adult cells for treatment of diseases ranging from diabetes to liver failure. Some of the challenges that limit the clinical/translational impact of hPSCs are high cost and difficulty in scaling-up of existing differentiation protocols. In this paper, we sought to address these challenges through the development of bioactive microcapsules. A co-axial flow focusing microfluidic device was used to encapsulate hPSCs in microcapsules comprised of an aqueous core and a hydrogel shell. Importantly, the shell contained heparin moieties for growth factor (GF) binding and release. The aqueous core enabled rapid aggregation of hPSCs into 3D spheroids while the bioactive hydrogel shell was used to load inductive cues driving pluripotency maintenance and endodermal differentiation. Specifically, we demonstrated that one-time, 1 h long loading of pluripotency signals, fibroblast growth factor (FGF)-2 and transforming growth factor (TGF)-β1, into bioactive microcapsules was sufficient to induce and maintain pluripotency of hPSCs over the course of 5 days at levels similar to or better than a standard protocol with soluble GFs. Furthermore, stem cell-carrying microcapsules that previously contained pluripotency signals could be reloaded with an endodermal cue, Nodal, resulting in higher levels of endodermal markers compared to stem cells differentiated in a standard protocol. Overall, bioactive heparin-containing core-shell microcapsules decreased GF usage five-fold while improving stem cell phenotype and are well suited for 3D cultivation of hPSCs. Heparin-containing microcapsules enable sustained release of inductive cues (growth factors) over the course of seven to nine days. Heparin-growth factor binding is reversible which means that different growth factors may be loaded in a sequential manner. Loading inductive cues into microcapsules results in better differentiation of pluripotent stem cells. Loading inductive cues into microcapsules allows to decrease the usage of growth factors by several fold.
Collapse
Affiliation(s)
- Kihak Gwon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55902, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55902, USA
| | | | - Michael Q. Slama
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55902, USA
| | - Daheui Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55902, USA
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55902, USA
| | - Quinn P. Peterson
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55902, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55902, USA
- Corresponding author.
| |
Collapse
|
14
|
Monckton CP, Brougham-Cook A, Kaylan KB, Underhill GH, Khetani SR. Elucidating Extracellular Matrix and Stiffness Control of Primary Human Hepatocyte Phenotype Via Cell Microarrays. ADVANCED MATERIALS INTERFACES 2021; 8:2101284. [PMID: 35111564 PMCID: PMC8803000 DOI: 10.1002/admi.202101284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Indexed: 05/30/2023]
Abstract
How the liver's extracellular matrix (ECM) protein composition and stiffness cooperatively regulate primary human hepatocyte (PHH) phenotype is unelucidated. Here, we utilize protein microarrays and high content imaging with single-cell resolution to assess PHH attachment/functions on 10 major liver ECM proteins in single and two-way combinations robotically spotted onto polyacrylamide gels of 1 kPa or 25 kPa stiffness. Albumin, cytochrome-P450 3A4 (CYP3A4), and hepatocyte nuclear factor alpha (HNF4α) positively correlate with each other and cell density on both stiffnesses. The 25 kPa stiffness supports higher average albumin and HNF4α expression after 14 days, while ECM protein composition significantly modulates PHH functions across both stiffnesses. Unlike previous rodent data, PHH functions are highest only when collagen-IV or fibronectin are mixed with specific proteins, whereas non-collagenous proteins without mixed collagens downregulate functions. Combination of collagen-IV and hyaluronic acid retains high CYP3A4 on 1 kPa, whereas collagens-IV and -V better retain HNF4α on 25 kPa over 14 days. Adapting ECM conditions to 96-well plates containing conjugated hydrogels reveals novel regulation of other functions (urea, CYP1A2/2A6/2C9) and drug-mediated CYP induction by the ECM protein composition/stiffness. This high-throughput pipeline can be adapted to elucidate ECM's role in liver diseases and facilitate optimization of engineered tissues.
Collapse
Affiliation(s)
- Chase P Monckton
- Department of Biomedical Engineering, University of Illinois at Chicago, 851 South Morgan Street, Chicago, Illinois, 60607, USA
| | - Aidan Brougham-Cook
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 2112 Everitt Laboratory, 1406 West Green Street, Urbana, Illinois, 61801, USA
| | - Kerim B Kaylan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 2112 Everitt Laboratory, 1406 West Green Street, Urbana, Illinois, 61801, USA
| | - Gregory H Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 2112 Everitt Laboratory, 1406 West Green Street, Urbana, Illinois, 61801, USA
| | - Salman R Khetani
- Department of Biomedical Engineering, University of Illinois at Chicago, 851 South Morgan Street, Chicago, Illinois, 60607, USA
| |
Collapse
|
15
|
Perspective: The Mechanobiology of Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13174275. [PMID: 34503085 PMCID: PMC8428343 DOI: 10.3390/cancers13174275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second most deadly primary cancer in the world and is thus a major global health challenge. HCC primarily develops in patients with an underlying chronic liver disease, the vast majority with advanced cirrhosis, characterized by increased matrix deposition and liver stiffness. Liver stiffness is highly associated with cancer development and poor patient outcome and is measured clinically to assess cancer risk; cirrhotic livers greatly exceed the threshold stiffness shown to alter hepatocyte cell behavior and to increase the malignancy of cancer cells. Recent studies have shown that cirrhotic liver cells have highly irregular nuclear morphologies and that nuclear deformation mediates mechanosensitive signaling. Separate research has shown that nuclear deformation can increase genetic instability and the accumulation of DNA damage in migrating cancer cells. We hypothesize that the mechanical changes associated with chronic liver disease are drivers of oncogenesis, activating mechanosensitive signaling pathways, increasing rates of DNA damage, and ultimately inducing malignant transformation.
Collapse
|
16
|
Heydari Z, Zarkesh I, Ghanian MH, Aghdaei MH, Kotova S, Zahmatkesh E, Farzaneh Z, Piryaei A, Akbarzadeh I, Shpichka A, Gramignoli R, Timashev P, Baharvand H, Vosough M. Biofabrication of size-controlled liver microtissues incorporated with ECM-derived microparticles to prolong hepatocyte function. Biodes Manuf 2021. [DOI: 10.1007/s42242-021-00137-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
17
|
Luo C, Lü D, Zheng L, Zhang F, Zhang X, Lü S, Zhang C, Jia X, Shu X, Li P, Li Z, Long M. Hepatic differentiation of human embryonic stem cells by coupling substrate stiffness and microtopography. Biomater Sci 2021; 9:3776-3790. [PMID: 33876166 DOI: 10.1039/d1bm00174d] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mechanical or physical cues are associated with the growth and differentiation of embryonic stem cells (ESCs). While the substrate stiffness or topography independently affects the differentiation of ESCs, their cooperative regulation on lineage-specific differentiation remains largely unknown. Here, four topographical configurations on stiff or soft polyacrylamide hydrogel were combined to direct hepatic differentiation of human H1 cells via a four-stage protocol, and the coupled impacts of stiffness and topography were quantified at distinct stages. Data indicated that the substrate stiffness is dominant in stemness maintenance on stiff gel and hepatic differentiation on soft gel while substrate topography assists the differentiation of hepatocyte-like cells in positive correlation with the circularity of H1 clones initially formed on the substrate. The differentiated cells exhibited liver-specific functions such as maintaining the capacities of CYP450 metabolism, glycogen synthesis, ICG engulfment, and repairing liver injury in CCl4-treated mice. These results implied that the coupling of substrate stiffness and topography, combined with the biochemical signals, is favorable to improve the efficiency and functionality of hepatic differentiation of human ESCs.
Collapse
Affiliation(s)
- Chunhua Luo
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China.
| | - Dongyuan Lü
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lu Zheng
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fan Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shouqin Lü
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China.
| | - Xiaohua Jia
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Xinyu Shu
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiwen Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China.
| | - Zhan Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China.
| | - Mian Long
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Adetunji CO, Akram M, Michael OS, Shahzad K, Ayeni AE, Hasan S, Adetunji JB, Hasan SM, Inamuddin, Olaniyan M, Muhibi MA. Polysaccharides Derived From Natural Sources: A Panacea to Health and Nutritional Challenges. POLYSACCHARIDES 2021. [DOI: 10.1002/9781119711414.ch32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
19
|
Exploring Interactions between Primary Hepatocytes and Non-Parenchymal Cells on Physiological and Pathological Liver Stiffness. BIOLOGY 2021; 10:biology10050408. [PMID: 34063016 PMCID: PMC8147966 DOI: 10.3390/biology10050408] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/19/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary Chronic liver disease is characterized by progressive hepatic fibrosis leading to the formation of cirrhosis irrespective of the etiology with no effective treatment currently available. Liver stiffness (LS) is currently the best clinical predictor of this fibrosis progression irrespective of the cause of the disease. However, it is not well understood how does LS regulate the critical hepatocytes–non parenchymal cell interactions. We here present, to the best of our knowledge, the first analyses of the impact of physiological and pathological stiffness on hepatocytes–non parenchymal cell interaction. Our findings indicate the role of stiffness in regulating the hepatocytes interactions with NPCs necessary for maintenance of hepatocytes function. Abstract Chronic liver disease is characterized by progressive hepatic fibrosis leading to the formation of cirrhosis irrespective of the etiology with no effective treatment currently available. Liver stiffness (LS) is currently the best clinical predictor of this fibrosis progression irrespective of the etiology. LS and hepatocytes-nonparenchymal cells (NPC) interactions are two variables known to be important in regulating hepatic function during liver fibrosis, but little is known about the interplay of these cues. Here, we use polydimethyl siloxane (PDMS) based substrates with tunable mechanical properties to study how cell–cell interaction and stiffness regulates hepatocytes function. Specifically, primary rat hepatocytes were cocultured with NIH-3T3 fibroblasts on soft (2 kPa) and stiff substrates that recreates physiologic (2 kPa) and cirrhotic liver stiffness (55 kPa). Urea synthesis by primary hepatocytes depended on the presence of fibroblast and was independent of the substrate stiffness. However, albumin synthesis and Cytochrome P450 enzyme activity increased in hepatocytes on soft substrates and when in coculture with a fibroblast. Western blot analysis of hepatic markers, E-cadherin, confirmed that hepatocytes on soft substrates in coculture promoted better maintenance of the hepatic phenotype. These findings indicate the role of stiffness in regulating the hepatocytes interactions with NPCs necessary for maintenance of hepatocytes function.
Collapse
|
20
|
Sullivan KM, Park CG, Ito JD, Kandel M, Popescu G, Kim YJ, Kong H. Matrix Softness-Mediated 3D Zebrafish Hepatocyte Modulates Response to Endocrine Disrupting Chemicals. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:13797-13806. [PMID: 32975940 PMCID: PMC8202163 DOI: 10.1021/acs.est.0c01988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Endocrine disrupting chemicals (EDC) include synthetic compounds that mimic the structure or function of natural hormones. While most studies utilize live embryos or primary cells from adult fish, these cells rapidly lose functionality when cultured on plastic or glass substrates coated with extracellular matrix proteins. This study hypothesizes that the softness of a matrix with adhered fish cells can regulate the intercellular organization and physiological function of engineered hepatoids during EDC exposure. We scrutinized this hypothesis by culturing zebrafish hepatocytes (ZF-L) on collagen-based hydrogels with controlled elastic moduli by examining morphology, urea production, and intracellular oxidative stress of hepatoids exposed to 17β-estradiol. Interestingly, the softer gel drove cells to form a cell sheet with a canaliculi-like structure compared to its stiffer gel counterpart. The hepatoids cultured on the softer gel exhibited more active urea production upon exposure to 17β-estradiol and displayed faster recovery of intracellular reactive oxygen species level confirmed by gradient light interference microscopy (GLIM), a live-cell imaging technique. These results are broadly useful to improve screening and understanding of potential EDC impacts on aquatic organisms and human health.
Collapse
Affiliation(s)
- Kathryn M Sullivan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Chang Gyun Park
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Campus E 7.1, 66123 Saarbrücken, Germany
| | - John D Ito
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Mikhail Kandel
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Gabriel Popescu
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Young Jun Kim
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Campus E 7.1, 66123 Saarbrücken, Germany
| | - Hyunjoon Kong
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
21
|
West-Livingston LN, Park J, Lee SJ, Atala A, Yoo JJ. The Role of the Microenvironment in Controlling the Fate of Bioprinted Stem Cells. Chem Rev 2020; 120:11056-11092. [PMID: 32558555 PMCID: PMC7676498 DOI: 10.1021/acs.chemrev.0c00126] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The field of tissue engineering and regenerative medicine has made numerous advances in recent years in the arena of fabricating multifunctional, three-dimensional (3D) tissue constructs. This can be attributed to novel approaches in the bioprinting of stem cells. There are expansive options in bioprinting technology that have become more refined and specialized over the years, and stem cells address many limitations in cell source, expansion, and development of bioengineered tissue constructs. While bioprinted stem cells present an opportunity to replicate physiological microenvironments with precision, the future of this practice relies heavily on the optimization of the cellular microenvironment. To fabricate tissue constructs that are useful in replicating physiological conditions in laboratory settings, or in preparation for transplantation to a living host, the microenvironment must mimic conditions that allow bioprinted stem cells to proliferate, differentiate, and migrate. The advances of bioprinting stem cells and directing cell fate have the potential to provide feasible and translatable approach to creating complex tissues and organs. This review will examine the methods through which bioprinted stem cells are differentiated into desired cell lineages through biochemical, biological, and biomechanical techniques.
Collapse
Affiliation(s)
- Lauren N. West-Livingston
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Jihoon Park
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
22
|
Xia T, Zhao R, Feng F, Yang L. The Effect of Matrix Stiffness on Human Hepatocyte Migration and Function-An In Vitro Research. Polymers (Basel) 2020; 12:polym12091903. [PMID: 32846973 PMCID: PMC7564768 DOI: 10.3390/polym12091903] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/08/2020] [Accepted: 08/18/2020] [Indexed: 01/30/2023] Open
Abstract
The extracellular matrix (ECM) regulates cellular function through the dynamic biomechanical and biochemical interplay between the resident cells and their microenvironment. Pathologically stiff ECM promotes phenotype changes in hepatocytes during liver fibrosis. To investigate the effect of ECM stiffness on hepatocyte migration and function, we designed an easy fabricated polyvinyl alcohol (PVA) hydrogel in which stiffness can be controlled by changing the concentration of glutaraldehyde. Three stiffnesses of hydrogels corresponding to the health of liver tissue, early stage, and end stage of fibrosis were selected. These were 4.8 kPa (soft), 21 kPa (moderate), and 45 kPa (stiff). For hepatocytes attachment, the hydrogel was coated with fibronectin. To evaluate the optimal concentration of fibronectin, hydrogel was coated with 0.1 mg/mL, 0.01 mg/mL, 0.005 mg/mL, or 0.003 mg/mL fibronectin, and the migratory behavior of single hepatocyte cultured on different concentrations of fibronectin was analyzed. To further explore the effect of substrate stiffness on hepatocyte migration, we used a stiffness controllable commercial 3D collagen gel, which has similar substrate stiffness to that of PVA hydrogel. Our result confirmed the PVA hydrogel biocompatibility with high hepatocytes survival. Fibronectin (0.01 mg/mL) promoted optimal migratory behavior for single hepatocytes. However, for confluent hepatocytes, a stiff substrate promoted hepatocellular migration compared with the soft and moderate groups via enhancing the formation of actin- and tubulin-rich structures. The gene expression analysis and protein expression analysis showed that the stiff substrate altered the phenotype of hepatocytes and induced apoptosis. Hepatocytes in stiff 3D hydrogel showed a higher proportion of cell death and expression of filopodia.
Collapse
Affiliation(s)
| | | | | | - Li Yang
- Correspondence: (T.X.); (L.Y.)
| |
Collapse
|
23
|
Darakhshan S, Bidmeshki Pour A, Kowsari-Esfahan R, Vosough M, Montazeri L, Ghanian MH, Baharvand H, Piryaei A. Generation of Scalable Hepatic Micro-Tissues as a Platform for Toxicological Studies. Tissue Eng Regen Med 2020; 17:459-475. [PMID: 32666397 PMCID: PMC7392990 DOI: 10.1007/s13770-020-00272-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/02/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Currently, there is an urgent need for scalable and reliable in vitro models to assess the effects of therapeutic entities on the human liver. Hepatoma cell lines, including Huh-7, show weakly resemblance to human hepatocytes, limiting their significance in toxicity studies. Co-culture of hepatic cells with non-parenchymal cells, and the presence of extracellular matrix have been shown to influence the biological behavior of hepatocytes. The aim of this study was to generate the scalable and functional hepatic micro-tissues (HMTs). METHODS The size-controllable HMTs were generated through co-culturing of Huh-7 cells by mesenchymal stem cells and human umbilical vein endothelial cells in a composite hydrogel of liver-derived extracellular matrix and alginate, using an air-driven droplet generator. RESULTS The generated HMTs were functional throughout a culture period of 28 days, as assessed by monitoring glycogen storage, uptake of low-density lipoprotein and indocyanine green. The HMTs also showed increased secretion levels of albumin, alpha-1-antitrypsin, and fibrinogen, and production of urea. Evaluating the expression of genes involved in hepatic-specific and drug metabolism functions indicated a significant improvement in HMTs compared to two-dimensional (2D) culture of Huh-7 cells. Moreover, in drug testing assessments, HMTs showed higher sensitivity to hepatotoxins compared to 2D cultured Huh-7 cells. Furthermore, induction and inhibition potency of cytochrome P450 enzymes confirmed that the HMTs can be used for in vitro drug screening. CONCLUSION Overall, we developed a simple and scalable method for generation of liver micro-tissues, using Huh-7, with improved hepatic-specific functionality, which may represent a biologically relevant platform for drug studies.
Collapse
Affiliation(s)
- Sara Darakhshan
- Department of Biology, Faculty of Science, Razi University, Kermanshah, 6714414971, Iran
| | - Ali Bidmeshki Pour
- Department of Biology, Faculty of Science, Razi University, Kermanshah, 6714414971, Iran.
| | - Reza Kowsari-Esfahan
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, Iran
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19395-4719, Tehran, Iran.
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
van der Mark VA, Adam AAA, Chang JC, Oude Elferink RP, Chamuleau RAFM, Hoekstra R. Overexpression of the constitutive androstane receptor and shaken 3D-culturing increase biotransformation and oxidative phosphorylation and sensitivity to mitochondrial amiodarone toxicity of HepaRG cells. Toxicol Appl Pharmacol 2020; 399:115055. [PMID: 32428594 DOI: 10.1016/j.taap.2020.115055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023]
Abstract
The liver cell line HepaRG is one of the preferred sources of human hepatocytes for in vitro applications. However, mitochondrial energy metabolism is relatively low, which affects hepatic functionality and sensitivity to hepatotoxins. Culturing in a bioartificial liver (BAL) system with high oxygen, medium perfusion, low substrate stiffness, and 3D conformation increases HepaRG functionality and mitochondrial activity compared to conventional monolayer culturing. In addition, drug metabolism has been improved by overexpression of the constitutive androstane receptor (CAR), a regulator of drug and energy metabolism in the new HepaRG-CAR line. Here, we investigated the effect of BAL culturing on the HepaRG-CAR line by applying a simple and downscaled BAL culture procedure based on shaking 3D cultures, named Bal-in-a-dish (BALIAD). We compared monolayer and BALIAD cultures of HepaRG and HepaRG-CAR cells. CAR overexpression and BALIAD culturing synergistically or additively increased transcript levels of CAR and three of the seven tested CAR target genes in biotransformation. Additionally, Cytochrome P450 3A4 activity was 35-fold increased. The mitochondrial energy metabolism was enhanced; lactate production and glucose consumption switched into lactate elimination and glucose production. BALIAD culturing alone reduced glycogen content and increased oxygen consumption and mitochondrial content. Both CAR overexpression and BALIAD culturing decreased mitochondrial superoxide levels. HepaRG-CAR BALIADs were most sensitive to mitochondrial toxicity induced by the hepatotoxin amiodarone, as indicated by oxygen consumption and mitochondrial superoxide accumulation. These data show that BALIAD culturing of HepaRG-CAR cells induces high mitochondrial energy metabolism and xenobiotic metabolism, increasing its potential for drug toxicity studies.
Collapse
Affiliation(s)
- Vincent A van der Mark
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands
| | - Aziza A A Adam
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| | - Jung-Chin Chang
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| | - Ronald P Oude Elferink
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| | - Robert A F M Chamuleau
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| | - Ruurdtje Hoekstra
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AG&M, Meibergdreef 69-71, 1105 BK, Amsterdam, the Netherlands.
| |
Collapse
|
25
|
Rahmati M, Silva EA, Reseland JE, A Heyward C, Haugen HJ. Biological responses to physicochemical properties of biomaterial surface. Chem Soc Rev 2020; 49:5178-5224. [PMID: 32642749 DOI: 10.1039/d0cs00103a] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Biomedical scientists use chemistry-driven processes found in nature as an inspiration to design biomaterials as promising diagnostic tools, therapeutic solutions, or tissue substitutes. While substantial consideration is devoted to the design and validation of biomaterials, the nature of their interactions with the surrounding biological microenvironment is commonly neglected. This gap of knowledge could be owing to our poor understanding of biochemical signaling pathways, lack of reliable techniques for designing biomaterials with optimal physicochemical properties, and/or poor stability of biomaterial properties after implantation. The success of host responses to biomaterials, known as biocompatibility, depends on chemical principles as the root of both cell signaling pathways in the body and how the biomaterial surface is designed. Most of the current review papers have discussed chemical engineering and biological principles of designing biomaterials as separate topics, which has resulted in neglecting the main role of chemistry in this field. In this review, we discuss biocompatibility in the context of chemistry, what it is and how to assess it, while describing contributions from both biochemical cues and biomaterials as well as the means of harmonizing them. We address both biochemical signal-transduction pathways and engineering principles of designing a biomaterial with an emphasis on its surface physicochemistry. As we aim to show the role of chemistry in the crosstalk between the surface physicochemical properties and body responses, we concisely highlight the main biochemical signal-transduction pathways involved in the biocompatibility complex. Finally, we discuss the progress and challenges associated with the current strategies used for improving the chemical and physical interactions between cells and biomaterial surface.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway. h.j.haugen.odont.uio.no
| | | | | | | | | |
Collapse
|
26
|
Serna-Márquez N, Rodríguez-Hernández A, Ayala-Reyes M, Martínez-Hernández LO, Peña-Rico MÁ, Carretero-Ortega J, Hautefeuille M, Vázquez-Victorio G. Fibrillar Collagen Type I Participates in the Survival and Aggregation of Primary Hepatocytes Cultured on Soft Hydrogels. Biomimetics (Basel) 2020; 5:E30. [PMID: 32630500 PMCID: PMC7345357 DOI: 10.3390/biomimetics5020030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Liver is an essential organ that carries out multiple functions such as glycogen storage, the synthesis of plasma proteins, and the detoxification of xenobiotics. Hepatocytes are the parenchyma that sustain almost all the functions supported by this organ. Hepatocytes and non-parenchymal cells respond to the mechanical alterations that occur in the extracellular matrix (ECM) caused by organogenesis and regenerating processes. Rearrangements of the ECM modify the composition and mechanical properties that result in specific dedifferentiation programs inside the hepatic cells. Quiescent hepatocytes are embedded in the soft ECM, which contains an important concentration of fibrillar collagens in combination with a basement membrane-associated matrix (BM). This work aims to evaluate the role of fibrillar collagens and BM on actin cytoskeleton organization and the function of rat primary hepatocytes cultured on soft elastic polyacrylamide hydrogels (PAA HGs). We used rat tail collagen type I and Matrigel® as references of fibrillar collagens and BM respectively and mixed different percentages of collagen type I in combination with BM. We also used peptides obtained from decellularized liver matrices (dECM). Remarkably, hepatocytes showed a poor adhesion in the absence of collagen on soft PAA HGs. We demonstrated that collagen type I inhibited apoptosis and activated extracellular signal-regulated kinases 1/2 (ERK1/2) in primary hepatocytes cultured on soft hydrogels. Epidermal growth factor (EGF) was not able to rescue cell viability in conjugated BM but affected cell aggregation in soft PAA HGs conjugated with combinations of different proportions of collagen and BM. Interestingly, actin cytoskeleton was localized and preserved close to plasma membrane (cortical actin) and proximal to intercellular ducts (canaliculi-like structures) in soft conditions; however, albumin protein expression was not preserved, even though primary hepatocytes did not remodel their actin cytoskeleton significantly in soft conditions. This investigation highlights the important role of fibrillar collagens on soft hydrogels for the maintenance of survival and aggregation of the hepatocytes. Data suggest evaluating the conditions that allow the establishment of optimal biomimetic environments for physiology and cell biology studies, where the phenotype of primary cells may be preserved for longer periods of time.
Collapse
Affiliation(s)
- Nathalia Serna-Márquez
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Adriana Rodríguez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Marisol Ayala-Reyes
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Lorena Omega Martínez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Miguel Ángel Peña-Rico
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Jorge Carretero-Ortega
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Mathieu Hautefeuille
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| | - Genaro Vázquez-Victorio
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| |
Collapse
|
27
|
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: 2.2] [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
|
28
|
Hosseini V, Maroufi NF, Saghati S, Asadi N, Darabi M, Ahmad SNS, Hosseinkhani H, Rahbarghazi R. Current progress in hepatic tissue regeneration by tissue engineering. J Transl Med 2019; 17:383. [PMID: 31752920 PMCID: PMC6873477 DOI: 10.1186/s12967-019-02137-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Liver, as a vital organ, is responsible for a wide range of biological functions to maintain homeostasis and any type of damages to hepatic tissue contributes to disease progression and death. Viral infection, trauma, carcinoma, alcohol misuse and inborn errors of metabolism are common causes of liver diseases are a severe known reason for leading to end-stage liver disease or liver failure. In either way, liver transplantation is the only treatment option which is, however, hampered by the increasing scarcity of organ donor. Over the past years, considerable efforts have been directed toward liver regeneration aiming at developing new approaches and methodologies to enhance the transplantation process. These approaches include producing decellularized scaffolds from the liver organ, 3D bio-printing system, and nano-based 3D scaffolds to simulate the native liver microenvironment. The application of small molecules and micro-RNAs and genetic manipulation in favor of hepatic differentiation of distinct stem cells could also be exploited. All of these strategies will help to facilitate the application of stem cells in human medicine. This article reviews the most recent strategies to generate a high amount of mature hepatocyte-like cells and updates current knowledge on liver regenerative medicine.
Collapse
Affiliation(s)
- Vahid Hosseini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Fathi Maroufi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Saghati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahideh Asadi
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Darabi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Nazari Soltan Ahmad
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Sarkar J, Kamble SC, Patil R, Kumar A, Gosavi SW. Gelatin interpenetration in poly N-isopropylacrylamide network reduces the compressive modulus of the scaffold: A property employed to mimic hepatic matrix stiffness. Biotechnol Bioeng 2019; 117:567-579. [PMID: 31691950 DOI: 10.1002/bit.27218] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/22/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The progression of liver disease from normal to cirrhotic state is characterized by modulation of the stiffness of the extracellular matrix (ECM). Mimicking this modulation in vitro scaffold could provide a better insight into hepatic cell behavior. In this study, interpenetrating poly(N-isopropylacrylamide-co-gelatin) cryogels were synthesized in 48 different compositions to yield scaffolds of different properties. It was observed that a high concentration of N-isopropylacrylamide (NIPAAm) leads to the formation of small pores while gelatin interpenetration on poly-NIPAAm framework renders porous structure. Swelling properties and porosity of the gels decreased with an increase in NIPAAm concentration owing to the increased compactness of the gels. Gelatin interpenetration relaxed the gels and enhanced these properties. An increase in gelatin concentration led to a reduction in compressive moduli indicating that gelatin interpenetration in the poly-NIPAAm network softens the cryogel. With the increase in NIPAAm concentration, the effect of gelatin interpenetration in reducing the compressive moduli expanded. The cytocompatibility studies indicated that the gels are cell-adherent and compatible with HepG2. Furthermore, biochemical and real-time polymerase chain reaction studies revealed that HepG2 and Huh-7 cells cultured on scaffolds mimicking the ECM stiffness of normal liver (1.5-2.5 kPa) exhibited optimum liver-specific functionalities. Increasing the stiffness to fibrotic (4-9 kPa) and cirrhotic (10-20 kPa) ECM decreases the functionality.
Collapse
Affiliation(s)
- Joyita Sarkar
- Department of Physics, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Swapnil C Kamble
- Department of Technology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Rajendra Patil
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India.,Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India.,Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Suresh W Gosavi
- Department of Physics, Savitribai Phule Pune University, Pune, Maharashtra, India
| |
Collapse
|
30
|
Vermeulen M, Del Vento F, Kanbar M, Pyr Dit Ruys S, Vertommen D, Poels J, Wyns C. Generation of Organized Porcine Testicular Organoids in Solubilized Hydrogels from Decellularized Extracellular Matrix. Int J Mol Sci 2019; 20:E5476. [PMID: 31684200 PMCID: PMC6862040 DOI: 10.3390/ijms20215476] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/25/2019] [Accepted: 10/30/2019] [Indexed: 01/15/2023] Open
Abstract
Cryopreservation of immature testicular tissue (ITT) prior to chemo/radiotherapy is now ethically accepted and is currently the only way to preserve fertility of prepubertal boys about to undergo cancer therapies. So far, three-dimensional culture of testicular cells isolated from prepubertal human testicular tissue was neither efficient nor reproducible to obtain mature spermatozoa, and ITT transplantation is not a safe option when there is a risk of cancer cell contamination of the testis. Hence, generation of testicular organoids (TOs) after cell selection is a novel strategy aimed at restoring fertility in these patients. Here, we created TOs using hydrogels developed from decellularized porcine ITT and compared cell numbers, organization and function to TOs generated in collagen only hydrogel. Organotypic culture of porcine ITT was used as a control. Rheological and mass spectrometry analyses of both hydrogels highlighted differences in terms of extracellular matrix stiffness and composition, respectively. Sertoli cells (SCs) and germ cells (GCs) assembled into seminiferous tubule-like structures delimited by a basement membrane while Leydig cells (LCs) and peritubular cells localized outside. TOs were maintained for 45 days in culture and secreted stem cell factor and testosterone demonstrating functionality of SCs and LCs, respectively. In both TOs GC numbers decreased and SC numbers increased. However, LC numbers decreased significantly in the collagen hydrogel TOs (p < 0.05) suggesting a better preservation of growth factors within TOs developed from decellularized ITT and thus a better potential to restore the reproductive capacity.
Collapse
Affiliation(s)
- Maxime Vermeulen
- Gynecology-Andrology Research Unit, Institut de Recherche Expérimentale et Clinique, Medical School, Université Catholique de Louvain, 1200 Brussels, Belgium.
| | - Federico Del Vento
- Gynecology-Andrology Research Unit, Institut de Recherche Expérimentale et Clinique, Medical School, Université Catholique de Louvain, 1200 Brussels, Belgium.
| | - Marc Kanbar
- Gynecology-Andrology Research Unit, Institut de Recherche Expérimentale et Clinique, Medical School, Université Catholique de Louvain, 1200 Brussels, Belgium.
| | - Sébastien Pyr Dit Ruys
- Phosphorylation - MassProt Unit, Institut de Duve, Université Catholique de Louvain, 1200 Brussels, Belgium.
| | - Didier Vertommen
- Phosphorylation - MassProt Unit, Institut de Duve, Université Catholique de Louvain, 1200 Brussels, Belgium.
| | - Jonathan Poels
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium.
| | - Christine Wyns
- Gynecology-Andrology Research Unit, Institut de Recherche Expérimentale et Clinique, Medical School, Université Catholique de Louvain, 1200 Brussels, Belgium.
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium.
| |
Collapse
|
31
|
Leichner C, Jelkmann M, Bernkop-Schnürch A. Thiolated polymers: Bioinspired polymers utilizing one of the most important bridging structures in nature. Adv Drug Deliv Rev 2019; 151-152:191-221. [PMID: 31028759 DOI: 10.1016/j.addr.2019.04.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022]
Abstract
Thiolated polymers designated "thiomers" are obtained by covalent attachment of thiol functionalities on the polymeric backbone of polymers. In 1998 these polymers were first described as mucoadhesive and in situ gelling compounds forming disulfide bonds with cysteine-rich substructures of mucus glycoproteins and crosslinking through inter- and intrachain disulfide bond formation. In the following, it was shown that thiomers are able to form disulfides with keratins and membrane-associated proteins exhibiting also cysteine-rich substructures. Furthermore, permeation enhancing, enzyme inhibiting and efflux pump inhibiting properties were demonstrated. Because of these capabilities thiomers are promising tools for drug delivery guaranteeing a strongly prolonged residence time as well as sustained release on mucosal membranes. Apart from that, thiomers are used as drugs per se. In particular, for treatment of dry eye syndrome various thiolated polymers are in development and a first product has already reached the market. Within this review an overview about the thiomer-technology and its potential for different applications is provided discussing especially the outcome of studies in non-rodent animal models and that of numerous clinical trials. Moreover, an overview on product developments is given.
Collapse
|
32
|
Huynh C, Shih TY, Mammoo A, Samant A, Pathan S, Nelson DW, Ferran C, Mooney D, LoGerfo F, Pradhan-Nabzdyk L. Delivery of targeted gene therapies using a hybrid cryogel-coated prosthetic vascular graft. PeerJ 2019; 7:e7377. [PMID: 31497383 PMCID: PMC6707340 DOI: 10.7717/peerj.7377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/28/2019] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES The success of prosthetic vascular grafts in the management of peripheral arterial disease is frequently limited by the development of anastomotic neointimal hyperplasia (ANIH), with the host response to prosthetic grafts beginning soon after implantation. To address this, we combine a platform of polyethylene terephthalate (PET) fabric with an applied cryogel layer containing biologic agents to create a bioactive prosthetic graft system, with the ability to deliver therapeutics targeting modulators of the ANIH-associated transcriptome response, along with antithrombotic agents. METHODS Hybrid graft materials were synthesized by cryopolymerization of methacrylated alginate and heparin onto electrospun (ePET), knitted PET (kPET), or woven PET (wPET). Arg-Gly-Asp (RGD) peptides were added to increase cell adhesion. Scanning electron microscopy (SEM) was used to study the microstructure at 1 day, and 2, 4, and 8 weeks. Physical properties such as swelling ratio, pore connectivity, shape recovery, and stiffness were evaluated. Human aortic endothelial cell (HAoEC) adherence was visualized using confocal microscopy after 24 hours and proliferation was evaluated with a resazurin-based assay for 7 days. Confocal microscopy was used to assess delivery of adeno-associated virus (AAV-GFP) after incubation of hybrid grafts with HAoECs. Heparin activity of the materials was measured using an anti-Xa assay. RESULTS SEM demonstrated large interconnected pores throughout the entire structure for all graft types, with minimal degradation of the cryogel after 8 weeks. Hybrid materials showed a trend towards increased shape recovery, increased stiffness, decreased swelling ratio, and no difference in pore connectivity. HAoECs incorporated, adhered, and proliferated over 7 days on all materials. HAoECs were successfully transduced with AAV-GFP from the hybrid graft materials. Anti-Xa assay confirmed continued activity of heparin from all materials for over 7 days. CONCLUSIONS We have developed a bioactive prosthetic graft system with a cryogel coating capable of delivering biologic agents with antithrombotic activity. By applying the cryogel and selected agents onto PET prior to graft implantation, this study sets the stage for the system to be individualized and tailored to the patient, with bioengineering and targeted gene therapy strategies dovetailing to create an improved prosthetic graft adaptable to emerging knowledge and technologies.
Collapse
Affiliation(s)
- Cindy Huynh
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Department of Surgery, State University of New York (SUNY), Syracuse, NY, United States of America
| | - Ting-Yu Shih
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Alexander Mammoo
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- Division of Pharmacology, Department of Pharmaceutical Biosciences, Uppsala Universitet, Uppsala, Sweden
| | - Amruta Samant
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Saif Pathan
- BioSurfaces, Inc, Ashland, MA, United States of America
| | | | - Christiane Ferran
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - David Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Frank LoGerfo
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Leena Pradhan-Nabzdyk
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| |
Collapse
|
33
|
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: 6.2] [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
|
34
|
Mattei G, Magliaro C, Pirone A, Ahluwalia A. Bioinspired liver scaffold design criteria. Organogenesis 2018; 14:129-146. [PMID: 30156955 PMCID: PMC6300109 DOI: 10.1080/15476278.2018.1505137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/20/2018] [Accepted: 07/20/2018] [Indexed: 12/26/2022] Open
Abstract
Maintaining hepatic functional characteristics in-vitro is considered one of the main challenges in engineering liver tissue. As hepatocytes cultured ex-vivo are deprived of their native extracellular matrix (ECM) milieu, developing scaffolds that mimic the biomechanical and physicochemical properties of the native ECM is thought to be a promising approach for successful tissue engineering and regenerative medicine applications. On the basis that the decellularized liver matrix represents the ideal design template for engineering bioinspired hepatic scaffolds, to derive quantitative descriptors of liver ECM architecture, we characterised decellularised liver matrices in terms of their biochemical, viscoelastic and structural features along with porosity, permeability and wettability. Together, these data provide a unique set of quantitative design criteria which can be used to generate guidelines for fabricating biomaterial scaffolds for liver tissue engineering. As proof-of-concept, we investigated hepatic cell response to substrate viscoelasticity. On collagen hydrogels mimicking decellularised liver mechanics, cells showed superior morphology, higher viability and albumin secretion than on stiffer and less viscous substrates. Although scaffold properties are generally inspired by those of native tissues, our results indicate significant differences between the mechano-structural characteristics of untreated and decellularised hepatic tissue. Therefore, we suggest that design rules - such as mechanical properties and swelling behaviour - for engineering biomimetic scaffolds be re-examined through further studies on substrates matching the features of decellularized liver matrices.
Collapse
Affiliation(s)
- Giorgio Mattei
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Optics11 B.V, Amsterdam, The Netherlands
- Biophotonics & Medical Imaging and Laser LaB, VU University Amsterdam, Amsterdam, The Netherlands
| | - Chiara Magliaro
- Research Centre “E. Piaggio”, University of Pisa, Pisa, Italy
| | - Andrea Pirone
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | - Arti Ahluwalia
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Research Centre “E. Piaggio”, University of Pisa, Pisa, Italy
| |
Collapse
|
35
|
Zhao C, Lin JS, Choolani M, Dan YY, Pastorin G, Ho HK. Enhanced hepatic differentiation of human amniotic epithelial cells on polyethylene glycol-linked multiwalled carbon nanotube-coated hydrogels. J Tissue Eng Regen Med 2018; 12:1556-1566. [PMID: 29700978 DOI: 10.1002/term.2672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 01/16/2018] [Accepted: 04/12/2018] [Indexed: 01/24/2023]
Abstract
Polyethylene glycol-linked multiwalled carbon nanotube-coated poly-acrylamide hydrogel (CNT-PA) was customized to mimic human liver stiffness and nanostructured surface in liver cells for modulating differentiation of human amniotic epithelial cells (hAECs) into functional hepatocyte-like cells (HLCs) in vitro. This composite of CNT-PA matrix enhanced the hepatic differentiation of hAECs into HLCs with suppression of pluripotent markers and up-regulation of hepatic markers at both transcript and protein levels. Furthermore, the HLCs on CNT-PA demonstrated hepatocytic functions in terms of albumin secretion, higher uptake of indocyanine green, and comparable CYP3A4 enzymatic function and inducibility when matched against HepG2 cells. Taken together, CNT-PA provides an efficient and scalable platform for the expansion of HLCs from hAECs and could be explored further for downstream development.
Collapse
Affiliation(s)
- Chunyan Zhao
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.,NanoCore, Faculty of Engineering, National University of Singapore, Singapore
| | - Jamie Siqi Lin
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yock Young Dan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Health System, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.,NanoCore, Faculty of Engineering, National University of Singapore, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| |
Collapse
|
36
|
Ijima H, Nakamura S, Bual R, Shirakigawa N, Tanoue S. Physical Properties of the Extracellular Matrix of Decellularized Porcine Liver. Gels 2018; 4:gels4020039. [PMID: 30674815 PMCID: PMC6209282 DOI: 10.3390/gels4020039] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/19/2018] [Accepted: 04/26/2018] [Indexed: 02/08/2023] Open
Abstract
The decellularization of organs has attracted attention as a new functional methodology for regenerative medicine based on tissue engineering. In previous work we developed an L-ECM (Extracellular Matrix) as a substrate-solubilized decellularized liver and demonstrated its effectiveness as a substrate for culturing and transplantation. Importantly, the physical properties of the substrate constitute important factors that control cell behavior. In this study, we aimed to quantify the physical properties of L-ECM and L-ECM gels. L-ECM was prepared as a liver-specific matrix substrate from solubilized decellularized porcine liver. In comparison to type I collagen, L-ECM yielded a lower elasticity and exhibited an abrupt decrease in its elastic modulus at 37 °C. Its elastic modulus increased at increased temperatures, and the storage elastic modulus value never fell below the loss modulus value. An increase in the gel concentration of L-ECM resulted in a decrease in the biodegradation rate and in an increase in mechanical strength. The reported properties of L-ECM gel (10 mg/mL) were equivalent to those of collagen gel (3 mg/mL), which is commonly used in regenerative medicine and gel cultures. Based on reported findings, the physical properties of the novel functional substrate for culturing and regenerative medicine L-ECM were quantified.
Collapse
Affiliation(s)
- Hiroyuki Ijima
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, Fukuoka 819-0395, Japan.
| | - Shintaro Nakamura
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, Fukuoka 819-0395, Japan.
| | - Ronald Bual
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, Fukuoka 819-0395, Japan.
| | - Nana Shirakigawa
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, Fukuoka 819-0395, Japan.
| | - Shuichi Tanoue
- Frontier Fiber Science and Technology, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan.
| |
Collapse
|
37
|
Xia T, Zhao R, Liu W, Huang Q, Chen P, Waju YN, Al-Ani MK, Lv Y, Yang L. Effect of substrate stiffness on hepatocyte migration and cellular Young's modulus. J Cell Physiol 2018; 233:6996-7006. [PMID: 29345322 DOI: 10.1002/jcp.26491] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/16/2018] [Indexed: 01/01/2023]
Abstract
Hepatic fibrosis progress accompanied by an unbalanced extracellular matrix (ECM) degradation and deposition leads to an increased tissue stiffness. Hepatocytes interplay with all intrahepatic cell populations inside the liver. However, how hepatocytes migration and cellular Young's modulus influenced by the substrate stiffness are not well understood. Here, we established a stiffness-controllable in vitro cell culture model by using a polyvinyl alcohol (PVA) hydrogel that mimicked the same physical stiffness as a fibrotic liver. Three levels of stiffness were used in our experiment that corresponded to the stiffness levels found in normal liver tissue (4.5 kPa), the early (19 kPa) and late stages (37 kPa) of fibrotic liver tissues. Cytoskeleton of hepatocyte was influenced by substrate stiffness. Soft substrate promoted the cellular migration and directionality. The cellular Young's modulus firstly increased and then decreased with increasing substrate stiffness. Integrin-β1 and β-catenin expression on cytomembrane were up-regulated and down-regulated with the increase of substrate stiffness, respectively. Our data not only suggested that hepatocytes were sensitive to substrate stiffness, but also suggested that there may be a potential relationship among substrate stiffness, cellular Young's modulus and the dynamic balance of integrin-β1 and β-catenin pathways. These results may provide us a new insight in mechanism investigation of mechano-dependent diseases, especially like fibrosis related diseases.
Collapse
Affiliation(s)
- Tingting Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Runze Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Wanqian Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Qiping Huang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Peixing Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Yasinta N Waju
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Mohanad K Al-Ani
- Department of microbiology, College of Medicine, Tikrit University, Tikrit, Iraq
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| |
Collapse
|
38
|
Xia T, Zhao R, Feng F, Song Y, Zhang Y, Dong L, Lv Y, Yang L. Gene expression profiling of human hepatocytes grown on differing substrate stiffness. Biotechnol Lett 2018; 40:809-818. [PMID: 29605939 DOI: 10.1007/s10529-018-2536-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/05/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To study the effects of different substrate stiffness on human hepatocytes using RNA sequencing (RNA-Seq) technology. The stiffness was corresponding to physiology and pathology stiffness of liver tissues. RESULTS With the aid of RNA-Seq technology, our study characterizes the transcriptome of hepatocytes cultured on soft, moderate, stiff and plastic substrates. Compared to soft substrate, our RNA-Seq results revealed 1131 genes that were up-regulated and 2534 that were down-regulated on moderate substrate, 1370 genes that were up-regulated and 2677 down-regulated genes on stiff substrate. Functional enrichment analysis indicated that differentially expressed genes were associated with the regulation of actin cytoskeleton, focal adhesion, tight junction, adherens junction as well as antigen processing and presentation. RNA-Seq results were further verified by a quantitative real-time reverse transcriptase polymerase chain reaction. CONCLUSION Our study provides a comprehensive picture of the gene expression landscape in hepatocytes grown on different substrate stiffness, offering insights into the role of substrate stiffness in hepatic pathology.
Collapse
Affiliation(s)
- Tingting Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Runze Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Fan Feng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Yijiang Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Yu Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Lili Dong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| |
Collapse
|
39
|
3D Co-Culture with Vascular Cells Supports Long-Term Hepatocyte Phenotype and Function In Vitro. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018. [DOI: 10.1007/s40883-018-0046-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
40
|
Li Q, Niu Y, Xing P, Wang C. Bioactive polysaccharides from natural resources including Chinese medicinal herbs on tissue repair. Chin Med 2018; 13:7. [PMID: 29445417 PMCID: PMC5802060 DOI: 10.1186/s13020-018-0166-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 01/30/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Functional polysaccharides can be derived from plants (including herbs), animals and microorganisms. They have been widely used in a broad of biomedical applications, such as immunoregulatory agents or drug delivery vehicles. In the past few years, increasing studies have started to develop natural polysaccharides-based biomaterials for various applications in tissue engineering and regenerative medicine. MAIN BODY We discuss in this article the emerging applications of natural polysaccharides-particularly those derived from Chinese medicine-for wound healing. First, we introduce natural polysaccharides of three natural sources and their biological activities. Then, we focus on certain natural polysaccharides with growth factor-binding affinities and their inspired polymeric tools, with an emphasis on how these polysaccharides could possibly benefit wound healing. Finally, we report the latest progress in the discovery of polysaccharides from Chinese medicinal herbs with identified activities favouring tissue repair. CONCLUSION Natural polysaccharides with clearly elucidated compositions/structures, identified cellular activities, as well as desirable physical properties have shown the potential to serve as therapeutic tools for tissue regeneration.
Collapse
Affiliation(s)
- Qiu Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau SAR, China
| | - Yiming Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau SAR, China
| | - Panfei Xing
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau SAR, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau SAR, China
| |
Collapse
|
41
|
Liaw CY, Ji S, Guvendiren M. Engineering 3D Hydrogels for Personalized In Vitro Human Tissue Models. Adv Healthc Mater 2018; 7. [PMID: 29345429 DOI: 10.1002/adhm.201701165] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/13/2017] [Indexed: 01/17/2023]
Abstract
There is a growing interest in engineering hydrogels for 3D tissue and disease models. The major motivation is to better mimic the physiological microenvironment of the disease and human condition. 3D tissue models derived from patients' own cells can potentially revolutionize the way treatment and diagnostic alternatives are developed. This requires development of tissue mimetic hydrogels with user defined and tunable properties. In this review article, a recent summary of 3D hydrogel platforms for in vitro tissue and disease modeling is given. Hydrogel design considerations and available hydrogel systems are summarized, followed by the types of currently available hydrogel models, such as bulk hydrogels, porous scaffolds, fibrous scaffolds, hydrogel microspheres, hydrogel sandwich systems, microwells, and 3D bioprinted constructs. Although hydrogels are utilized for a wide range of tissue models, this article focuses on liver and cancer models. This article also provides a detailed section on current challenges and future perspectives of hydrogel-based tissue models.
Collapse
Affiliation(s)
- Chya-Yan Liaw
- Instructive Biomaterials and Additive Manufacturing Laboratory; Otto H. York Chemical; Biological and Pharmaceutical Engineering; Newark College of Engineering; New Jersey Institute of Technology; University Heights; 138 York Center Newark NJ 07102 USA
| | - Shen Ji
- Instructive Biomaterials and Additive Manufacturing Laboratory; Otto H. York Chemical; Biological and Pharmaceutical Engineering; Newark College of Engineering; New Jersey Institute of Technology; University Heights; 138 York Center Newark NJ 07102 USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing Laboratory; Otto H. York Chemical; Biological and Pharmaceutical Engineering; Newark College of Engineering; New Jersey Institute of Technology; University Heights; 138 York Center Newark NJ 07102 USA
| |
Collapse
|
42
|
Beckwitt CH, Clark AM, Wheeler S, Taylor DL, Stolz DB, Griffith L, Wells A. Liver 'organ on a chip'. Exp Cell Res 2018; 363:15-25. [PMID: 29291400 PMCID: PMC5944300 DOI: 10.1016/j.yexcr.2017.12.023] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
The liver plays critical roles in both homeostasis and pathology. It is the major site of drug metabolism in the body and, as such, a common target for drug-induced toxicity and is susceptible to a wide range of diseases. In contrast to other solid organs, the liver possesses the unique ability to regenerate. The physiological importance and plasticity of this organ make it a crucial system of study to better understand human physiology, disease, and response to exogenous compounds. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems.
Collapse
Affiliation(s)
- Colin H Beckwitt
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA
| | - Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sarah Wheeler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - D Lansing Taylor
- Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Donna B Stolz
- Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Linda Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA.
| |
Collapse
|
43
|
Elasticity-based development of functionally enhanced multicellular 3D liver encapsulated in hybrid hydrogel. Acta Biomater 2017; 64:67-79. [PMID: 28966094 DOI: 10.1016/j.actbio.2017.09.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/30/2017] [Accepted: 09/27/2017] [Indexed: 12/23/2022]
Abstract
Current in vitro liver models provide three-dimensional (3-D) microenvironments in combination with tissue engineering technology and can perform more accurate in vivo mimicry than two-dimensional models. However, a human cell-based, functionally mature liver model is still desired, which would provide an alternative to animal experiments and resolve low-prediction issues on species differences. Here, we prepared hybrid hydrogels of varying elasticity and compared them with a normal liver, to develop a more mature liver model that preserves liver properties in vitro. We encapsulated HepaRG cells, either alone or with supporting cells, in a biodegradable hybrid hydrogel. The elastic modulus of the 3D liver dynamically changed during culture due to the combined effects of prolonged degradation of hydrogel and extracellular matrix formation provided by the supporting cells. As a result, when the elastic modulus of the 3D liver model converges close to that of the in vivo liver (≅ 2.3 to 5.9 kPa), both phenotypic and functional maturation of the 3D liver were realized, while hepatic gene expression, albumin secretion, cytochrome p450-3A4 activity, and drug metabolism were enhanced. Finally, the 3D liver model was expanded to applications with embryonic stem cell-derived hepatocytes and primary human hepatocytes, and it supported prolonged hepatocyte survival and functionality in long-term culture. Our model represents critical progress in developing a biomimetic liver system to simulate liver tissue remodeling, and provides a versatile platform in drug development and disease modeling, ranging from physiology to pathology. STATEMENT OF SIGNIFICANCE We provide a functionally improved 3D liver model that recapitulates in vivo liver stiffness. We have experimentally addressed the issues of orchestrated effects of mechanical compliance, controlled matrix formation by stromal cells in conjunction with hepatic differentiation, and functional maturation of hepatocytes in a dynamic 3D microenvironment. Our model represents critical progress in developing a biomimetic liver system to simulate liver tissue remodeling, and provides a versatile platform in drug development and disease modeling, ranging from physiology to pathology. Additionally, recent advances in the stem-cell technologies have made the development of 3D organoid possible, and thus, our study also provides further contribution to the development of physiologically relevant stem-cell-based 3D tissues that provide an elasticity-based predefined biomimetic 3D microenvironment.
Collapse
|
44
|
Heparin/Collagen 3D Scaffold Accelerates Hepatocyte Differentiation of Wharton's Jelly-Derived Mesenchymal Stem Cells. Tissue Eng Regen Med 2017; 14:443-452. [PMID: 30603500 DOI: 10.1007/s13770-017-0048-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 01/20/2017] [Accepted: 02/02/2017] [Indexed: 12/26/2022] Open
Abstract
Both mature and stem cell-derived hepatocytes lost their phenotype and functionality under conventional culture conditions. However, the 3D scaffolds containing the main extracellular matrix constitutions, such as heparin, may provide appropriate microenvironment for hepatocytes to be functional. The current study aimed to investigate the efficacy of the differentiation capability of hepatocytes derived from human Wharton's jelly mesenchymal stem cells (WJ-MSCs) in 3D heparinized scaffold. In this case, the human WJ-MSCs were cultured on the heparinized and non-heparinized 2D collagen gels or within 3D scaffolds in the presence of hepatogenic medium. Immunostaining was performed for anti-alpha fetoprotein, cytokeratin-18 and -19 antibodies. RT-PCR was performed for detection of hepatic nuclear factor-4 (HNF-4), albumin, cytokeratin-18 and -19, glucose-6-phosphatase (G6P), c-met and Cyp2B. The results indicated that hepatogenic media induced the cells to express early liver-specific markers including HNF4, albumin, cytokeratin-18 and 19 in all conditions. The cells cultured on both heparinized culture conditions expressed late liver-specific markers such as G6P and Cyp2B as well. Besides, the hepatocytes differentiated in 3D heparinized scaffolds stored more glycogen that indicated they were more functional. Non-heparinized 2D gel was the superior condition for cholangiocyte differentiation as indicated by higher levels of cytokeratin 19 expression. In conclusion, the heparinized 3D scaffolds provided a microenvironment to mimic Disse space. Therefore, 3D heparinized collagen scaffold can be suggested as a good vehicle for hepatocyte differentiation.
Collapse
|
45
|
Tetsuka K, Ohbuchi M, Tabata K. Recent Progress in Hepatocyte Culture Models and Their Application to the Assessment of Drug Metabolism, Transport, and Toxicity in Drug Discovery: The Value of Tissue Engineering for the Successful Development of a Microphysiological System. J Pharm Sci 2017; 106:2302-2311. [PMID: 28533121 DOI: 10.1016/j.xphs.2017.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/23/2017] [Accepted: 05/05/2017] [Indexed: 12/14/2022]
Abstract
Tissue engineering technology has provided many useful culture models. This article reviews the merits of this technology in a hepatocyte culture system and describes the applications of the sandwich-cultured hepatocyte model in drug discovery. In addition, we also review recent investigations of the utility of the 3-dimensional bioprinted human liver tissue model and spheroid model. Finally, we present the future direction and developmental challenges of a hepatocyte culture model for the successful establishment of a microphysiological system, represented as an organ-on-a-chip and even as a human-on-a-chip. A merit of advanced culture models is their potential use for detecting hepatotoxicity through repeated exposure to chemicals as they allow long-term culture while maintaining hepatocyte functionality. As a future direction, such advanced hepatocyte culture systems can be connected to other tissue models for evaluating tissue-to-tissue interaction beyond cell-to-cell interaction. This combination of culture models could represent parts of the human body in a microphysiological system.
Collapse
Affiliation(s)
- Kazuhiro Tetsuka
- Analysis & Pharmacokinetics Research Labs., Astellas Pharma Inc., 21 Miyukigaoka Tsukuba-shi, Ibaraki, Japan.
| | - Masato Ohbuchi
- Analysis & Pharmacokinetics Research Labs., Astellas Pharma Inc., 21 Miyukigaoka Tsukuba-shi, Ibaraki, Japan
| | - Kenji Tabata
- Analysis & Pharmacokinetics Research Labs., Astellas Pharma Inc., 21 Miyukigaoka Tsukuba-shi, Ibaraki, Japan
| |
Collapse
|
46
|
Domian IJ, Yu H, Mittal N. On Materials for Cardiac Tissue Engineering. Adv Healthc Mater 2017; 6. [PMID: 27774763 DOI: 10.1002/adhm.201600768] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Indexed: 11/08/2022]
Abstract
In this essay the authors argue that chamber pressure dominates the biomechanics of the contraction cycle of the heart, while tissue stiffness dominates the relaxation cycle. This appears to be an under-recognized challenge in cardiac tissue engineering. Optimal approaches will involve constructing chambers or modulating the stiffness of the scaffold/substrate in synchrony with the beating cycle.
Collapse
Affiliation(s)
- Ibrahim J. Domian
- Cardiovascular Research Center; Massachusetts General Hospital; Boston MA 02114 USA
- Harvard Medical School; Boston MA 02115 USA
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology; Singapore 138669
- Singapore-MIT Alliance for Research and Technology; 1 CREATE Way, #10-01 CREATE Tower Singapore 138602
- Department of Physiology; Yong Loo Lin School of Medicine; National University Health System; Singapore 117597
| | - Nikhil Mittal
- Cardiovascular Research Center; Massachusetts General Hospital; Boston MA 02114 USA
- Institute of Bioengineering and Nanotechnology; Singapore 138669
| |
Collapse
|
47
|
Perez RA, Jung CR, Kim HW. Biomaterials and Culture Technologies for Regenerative Therapy of Liver Tissue. Adv Healthc Mater 2017; 6. [PMID: 27860372 DOI: 10.1002/adhm.201600791] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/10/2016] [Indexed: 12/18/2022]
Abstract
Regenerative approach has emerged to substitute the current extracorporeal technologies for the treatment of diseased and damaged liver tissue. This is based on the use of biomaterials that modulate the responses of hepatic cells through the unique matrix properties tuned to recapitulate regenerative functions. Cells in liver preserve their phenotype or differentiate through the interactions with extracellular matrix molecules. Therefore, the intrinsic properties of the engineered biomaterials, such as stiffness and surface topography, need to be tailored to induce appropriate cellular functions. The matrix physical stimuli can be combined with biochemical cues, such as immobilized functional groups or the delivered actions of signaling molecules. Furthermore, the external modulation of cells, through cocultures with nonparenchymal cells (e.g., endothelial cells) that can signal bioactive molecules, is another promising avenue to regenerate liver tissue. This review disseminates the recent approaches of regenerating liver tissue, with a focus on the development of biomaterials and the related culture technologies.
Collapse
Affiliation(s)
- Roman A. Perez
- Institute of Tissue Regeneration Engineering (ITREN); Dankook University; Cheonan 330-714 Republic of Korea
- Regenerative Medicine Research Institute; Universitat Internacional de Catalunya; Barcelona 08017 Spain
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine; Dankook University; Cheonan 330-714 Republic of Korea
| | - Cho-Rok Jung
- Gene Therapy Research Unit; KRIBB; 125 Gwahak-ro Yuseong-gu, Daejeon 34141 Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN); Dankook University; Cheonan 330-714 Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine; Dankook University; Cheonan 330-714 Republic of Korea
- Department of Biomaterials Science; Dankook University Dental College; Cheonan 330-714 Republic of Korea
| |
Collapse
|
48
|
Haque A, Gheibi P, Gao Y, Foster E, Son KJ, You J, Stybayeva G, Patel D, Revzin A. Cell biology is different in small volumes: endogenous signals shape phenotype of primary hepatocytes cultured in microfluidic channels. Sci Rep 2016; 6:33980. [PMID: 27681582 PMCID: PMC5041105 DOI: 10.1038/srep33980] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022] Open
Abstract
The approaches for maintaining hepatocytes in vitro are aimed at recapitulating aspects of the native liver microenvironment through the use of co-cultures, surface coatings and 3D spheroids. This study highlights the effects of spatial confinement-a less studied component of the in vivo microenvironment. We demonstrate that hepatocytes cultured in low-volume microfluidic channels (microchambers) retain differentiated hepatic phenotype for 21 days whereas cells cultured in regular culture plates under identical conditions de-differentiate after 7 days. Careful consideration of nutrient delivery and oxygen tension suggested that these factors could not solely account for enhanced cell function in microchambers. Through a series of experiments involving microfluidic chambers of various heights and inhibition of key molecular pathways, we confirmed that phenotype of hepatocytes in small volumes was shaped by endogenous signals, both hepato-inductive growth factors (GFs) such as hepatocyte growth factor (HGF) and hepato-disruptive GFs such as transforming growth factor (TGF)-β1. Hepatocytes are not generally thought of as significant producers of GFs–this role is typically assigned to nonparenchymal cells of the liver. Our study demonstrates that, in an appropriate microenvironment, hepatocytes produce hepato-inductive and pro-fibrogenic signals at the levels sufficient to shape their phenotype and function.
Collapse
Affiliation(s)
- Amranul Haque
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Pantea Gheibi
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Yandong Gao
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Elena Foster
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Kyung Jin Son
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Jungmok You
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA.,Department of Plant and Environmental New Resources, Kyung Hee University, Youngin-si, Gyeonggi-do, South Korea
| | - Gulnaz Stybayeva
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Dipali Patel
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Alexander Revzin
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| |
Collapse
|
49
|
Mittal N, Tasnim F, Yue C, Qu Y, Phan D, Choudhury Y, Tan MH, Yu H. Substrate Stiffness Modulates the Maturation of Human Pluripotent Stem-Cell-Derived Hepatocytes. ACS Biomater Sci Eng 2016; 2:1649-1657. [PMID: 33440598 DOI: 10.1021/acsbiomaterials.6b00475] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obtaining functional hepatocytes from human pluripotent stem cells (hPSCs) holds great potential for applications in drug safety testing, as well in the field of regenerative medicine. However, developing functionally mature hPSC-derived hepatocytes (hPSC-Heps) remains a challenge. We hypothesized that the cellular microenvironment plays a vital role in the maturation of immature hepatocytes. In this study, we examined the role of mechanical stiffness, a key component of the cellular microenvironment, in the maturation of hPSC-Heps. We cultured hPSC-Heps on collagen-coated polyacrylamide hydrogels with varying elastic moduli. On softer substrates the hPSC-Heps formed compact colonies while on stiffer substrates they formed a diffuse monolayer. We observed an inverse correlation between albumin production and substrate stiffness. The expression of key cytochrome enzymes, which are expressed at higher levels in the adult liver compared to the fetal liver, also correlated inversely with substrate stiffness, whereas fetal markers such as Cyp3A7 and AFP showed no correlation with stiffness. Culture of hPSC-Heps on soft substrates for 12 days led to 10-30 fold increases in the expression of drug-metabolizing enzymes. These results demonstrate that substrate stiffness similar to that of the liver enables aspects of the maturation of hPSC-Heps.
Collapse
Affiliation(s)
- Nikhil Mittal
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Farah Tasnim
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Cao Yue
- Institute of Bioengineering and Nanotechnology, Singapore 138669.,Mechanobiology Institute, 10-01 T-Lab, National University of Singapore, 5A Engineering Drive, Singapore 117411
| | - Yinghua Qu
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Derek Phan
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Yukti Choudhury
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Min-Han Tan
- Institute of Bioengineering and Nanotechnology, Singapore 138669.,National Cancer Centre Singapore, Singapore 169610
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, Singapore 138669.,Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, Singapore 117597.,Mechanobiology Institute, 10-01 T-Lab, National University of Singapore, 5A Engineering Drive, Singapore 117411.,Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, #10-01 CREATE Tower, Singapore 138602
| |
Collapse
|
50
|
Deterministically patterned biomimetic human iPSC-derived hepatic model via rapid 3D bioprinting. Proc Natl Acad Sci U S A 2016; 113:2206-11. [PMID: 26858399 DOI: 10.1073/pnas.1524510113] [Citation(s) in RCA: 553] [Impact Index Per Article: 61.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The functional maturation and preservation of hepatic cells derived from human induced pluripotent stem cells (hiPSCs) are essential to personalized in vitro drug screening and disease study. Major liver functions are tightly linked to the 3D assembly of hepatocytes, with the supporting cell types from both endodermal and mesodermal origins in a hexagonal lobule unit. Although there are many reports on functional 2D cell differentiation, few studies have demonstrated the in vitro maturation of hiPSC-derived hepatic progenitor cells (hiPSC-HPCs) in a 3D environment that depicts the physiologically relevant cell combination and microarchitecture. The application of rapid, digital 3D bioprinting to tissue engineering has allowed 3D patterning of multiple cell types in a predefined biomimetic manner. Here we present a 3D hydrogel-based triculture model that embeds hiPSC-HPCs with human umbilical vein endothelial cells and adipose-derived stem cells in a microscale hexagonal architecture. In comparison with 2D monolayer culture and a 3D HPC-only model, our 3D triculture model shows both phenotypic and functional enhancements in the hiPSC-HPCs over weeks of in vitro culture. Specifically, we find improved morphological organization, higher liver-specific gene expression levels, increased metabolic product secretion, and enhanced cytochrome P450 induction. The application of bioprinting technology in tissue engineering enables the development of a 3D biomimetic liver model that recapitulates the native liver module architecture and could be used for various applications such as early drug screening and disease modeling.
Collapse
|