1
|
Liu JS, Madruga LYC, Yuan Y, Kipper MJ, Khetani SR. Decellularized Liver Nanofibers Enhance and Stabilize the Long-Term Functions of Primary Human Hepatocytes In Vitro. Adv Healthc Mater 2023; 12:e2202302. [PMID: 36947401 PMCID: PMC11469040 DOI: 10.1002/adhm.202202302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/07/2023] [Indexed: 03/23/2023]
Abstract
Owing to significant differences across species in liver functions, in vitro human liver models are used for screening the metabolism and toxicity of compounds, modeling diseases, and cell-based therapies. However, the extracellular matrix (ECM) scaffold used for such models often does not mimic either the complex composition or the nanofibrous topography of native liver ECM. Thus, here novel methods are developed to electrospin decellularized porcine liver ECM (PLECM) and collagen I into nano- and microfibers (≈200-1000 nm) without synthetic polymer blends. Primary human hepatocytes (PHHs) on nanofibers in monoculture or in coculture with nonparenchymal cells (3T3-J2 embryonic fibroblasts or primary human liver endothelial cells) display higher albumin secretion, urea synthesis, and cytochrome-P450 1A2, 2A6, 2C9, and 3A4 enzyme activities than on conventionally adsorbed ECM controls. PHH functions are highest on the collagen/PLECM blended nanofibers (up to 34-fold higher CYP3A4 activity relative to adsorbed ECM) for nearly 7 weeks in the presence of the fibroblasts. In conclusion, it is shown for the first time that ECM composition and topography synergize to enhance and stabilize PHH functions for several weeks in vitro. The nanofiber platform can prove useful for the above applications and to elucidate cell-ECM interactions in the human liver.
Collapse
Affiliation(s)
- Jennifer S. Liu
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| | - Liszt Y. C. Madruga
- Department of Chemical & Biological EngineeringColorado State UniversityFort CollinsCO80523‐1370USA
| | - Yang Yuan
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| | - Matt J. Kipper
- Department of Chemical & Biological EngineeringColorado State UniversityFort CollinsCO80523‐1370USA
| | - Salman R. Khetani
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| |
Collapse
|
2
|
Ware BR, Liu JS, Monckton CP, Ballinger KR, Khetani SR. Micropatterned Coculture With 3T3-J2 Fibroblasts Enhances Hepatic Functions and Drug Screening Utility of HepaRG Cells. Toxicol Sci 2021; 181:90-104. [PMID: 33590212 DOI: 10.1093/toxsci/kfab018] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human liver models are useful for assessing compound metabolism/toxicity; however, primary human hepatocyte (PHH) lots are limited and highly variable in quality/viability. In contrast, cell lines, such as HepaRG, are cheaper and more reproducible surrogates for initial compound screening; however, hepatic functions and sensitivity for drug outcomes need improvement. Here, we show that HepaRGs cocultured with murine embryonic 3T3-J2 fibroblasts, previously shown to induce PHH functions, could address such limitations. We either micropatterned HepaRGs or seeded them "randomly" onto collagen-coated plates before 3T3-J2 coculture. Micropatterned cocultures (HepaRG-MPCCs) secreted 2- to 4-fold more albumin and displayed more stable cytochrome P450 activities than HepaRG conventional confluent monocultures (HepaRG-CCs) and HepaRG micropatterned hepatocytes (HepaRG-MPHs) for 4 weeks, even when excluding dimethyl sulfoxide from the medium. Furthermore, HepaRG-MPCCs had the most albumin-only positive cells (hepatic), lowest cytokeratin 19 (CK19)-only positive cells (cholangiocytic), and highest mean albumin intensity per cell than HepaRG random cocultures and monocultures; however, 80%-84% of HepaRGs remained bipotential (albumin+/CK19+) across all models. The 3T3-J2s also induced higher albumin in HepaRG spheroids than HepaRG-only spheroids. Additionally, although rifampin induced CYP3A4 in HepaRG-MPCCs and HepaRG-CCs, only HepaRG-MPCCs showed the dual omeprazole-mediated CYP1A2/3A4 induction as with PHHs. Lastly, when treated for 6 days with 47 drugs and evaluated for albumin and ATP to make binary hepatotoxicity calls, HepaRG-MPCCs displayed a sensitivity of 54% and specificity of 100% (70%/100% in PHH-MPCCs), whereas HepaRG-CCs misclassified several hepatotoxins. Ultimately, HepaRG-MPCCs could be a more cost-effective and reproducible model than PHHs for executing a tier 1 compound screen.
Collapse
Affiliation(s)
- Brenton R Ware
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Jennifer S Liu
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Chase P Monckton
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Kimberly R Ballinger
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Salman R Khetani
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA
| |
Collapse
|
3
|
Kukla DA, Khetani SR. Bioengineered Liver Models for Investigating Disease Pathogenesis and Regenerative Medicine. Semin Liver Dis 2021; 41:368-392. [PMID: 34139785 DOI: 10.1055/s-0041-1731016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Owing to species-specific differences in liver pathways, in vitro human liver models are utilized for elucidating mechanisms underlying disease pathogenesis, drug development, and regenerative medicine. To mitigate limitations with de-differentiated cultures, bioengineers have developed advanced techniques/platforms, including micropatterned cocultures, spheroids/organoids, bioprinting, and microfluidic devices, for perfusing cell cultures and liver slices. Such techniques improve mature functions and culture lifetime of primary and stem-cell human liver cells. Furthermore, bioengineered liver models display several features of liver diseases including infections with pathogens (e.g., malaria, hepatitis C/B viruses, Zika, dengue, yellow fever), alcoholic/nonalcoholic fatty liver disease, and cancer. Here, we discuss features of bioengineered human liver models, their uses for modeling aforementioned diseases, and how such models are being augmented/adapted for fabricating implantable human liver tissues for clinical therapy. Ultimately, continued advances in bioengineered human liver models have the potential to aid the development of novel, safe, and efficacious therapies for liver disease.
Collapse
Affiliation(s)
- David A Kukla
- Deparment of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R Khetani
- Deparment of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
4
|
Monckton CP, Brown GE, Khetani SR. Latest impact of engineered human liver platforms on drug development. APL Bioeng 2021; 5:031506. [PMID: 34286173 PMCID: PMC8286174 DOI: 10.1063/5.0051765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023] Open
Abstract
Drug-induced liver injury (DILI) is a leading cause of drug attrition, which is partly due to differences between preclinical animals and humans in metabolic pathways. Therefore, in vitro human liver models are utilized in biopharmaceutical practice to mitigate DILI risk and assess related mechanisms of drug transport and metabolism. However, liver cells lose phenotypic functions within 1–3 days in two-dimensional monocultures on collagen-coated polystyrene/glass, which precludes their use to model the chronic effects of drugs and disease stimuli. To mitigate such a limitation, bioengineers have adapted tools from the semiconductor industry and additive manufacturing to precisely control the microenvironment of liver cells. Such tools have led to the fabrication of advanced two-dimensional and three-dimensional human liver platforms for different throughput needs and assay endpoints (e.g., micropatterned cocultures, spheroids, organoids, bioprinted tissues, and microfluidic devices); such platforms have significantly enhanced liver functions closer to physiologic levels and improved functional lifetime to >4 weeks, which has translated to higher sensitivity for predicting drug outcomes and enabling modeling of diseased phenotypes for novel drug discovery. Here, we focus on commercialized engineered liver platforms and case studies from the biopharmaceutical industry showcasing their impact on drug development. We also discuss emerging multi-organ microfluidic devices containing a liver compartment that allow modeling of inter-tissue crosstalk following drug exposure. Finally, we end with key requirements for engineered liver platforms to become routine fixtures in the biopharmaceutical industry toward reducing animal usage and providing patients with safe and efficacious drugs with unprecedented speed and reduced cost.
Collapse
Affiliation(s)
- Chase P Monckton
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
5
|
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: 3.0] [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
|
6
|
Gao B, Sakaguchi K, Ogawa T, Kagawa Y, Kubo H, Shimizu T. Functional Analysis of Induced Human Ballooned Hepatocytes in a Cell Sheet-Based Three Dimensional Model. Tissue Eng Regen Med 2021; 18:217-224. [PMID: 33517537 DOI: 10.1007/s13770-020-00297-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Ballooned hepatocytes (BH) are a key histological hallmark of nonalcoholic steatohepatitis (NASH), yet their consequences for liver-specific functions are unknown. METHODS In our previous study, an experimental model of human induced-BHs (iBH) has been successfully developed based on cell sheet technology. This study aimed to determine the functions of iBHs in the primary human hepatocyte/normal human dermal fibroblast (PHH/NHDF) co-culture cell sheets. Normal hepatocytes in the PHH/3T3-J2 co-culture cell sheets were set as a control, since 3T3-J2 murine embryonic fibroblasts have exhibited previously long term maintenance of PHH functions. RESULTS It was found that, albumin secretion was not affected in iBHs, but urea synthesis as well as cytochrome P450 enzyme (CYP) activities including CYP1A2 and CYP3A4, were significantly reduced in iBHs. Besides, loss of bile canaliculi was observed in iBHs. These findings are consistent with clinical studies of human NASH. In addition, PHH/NHDF cell sheets demonstrated two fold higher TGF-β1 secretion compared with PHH/3T3-J2 cell sheets. Furthermore, treatment with a TGF-β inhibitor and a semi-synthetic bile acid analogue (obeticholic acid, phase 3 trial of NASH therapy) ameliorated the histological appearance of established iBHs. CONCLUSION In summary, this study demonstrates the priority of iBHs in recapitulating not only histology but also clinically relevant hepatic dysfunctions in human NASH and suggests TGF-β and bile acid related signal pathway may play important roles in the formation of iBHs.
Collapse
Affiliation(s)
- Botao Gao
- Guangdong Key Lab of Medical Electronic Instruments and Polymer Materials Products, National Engineering Research Center for Healthcare Devices, Guangdong Institute of Medical Instruments, Guangdong Academy of Sciences, Guangzhou, 510550, China.,Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Katsuhisa Sakaguchi
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Tetsuya Ogawa
- Ogino Memorial Laboratory, Nihon Kohden Corporation, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Yuki Kagawa
- Ogino Memorial Laboratory, Nihon Kohden Corporation, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Hirotsugu Kubo
- Ogino Memorial Laboratory, Nihon Kohden Corporation, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| |
Collapse
|
7
|
Chen AX, Chhabra A, Song HHG, Fleming HE, Chen CS, Bhatia SN. Controlled Apoptosis of Stromal Cells to Engineer Human Microlivers. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910442. [PMID: 33776613 PMCID: PMC7996305 DOI: 10.1002/adfm.201910442] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/29/2020] [Indexed: 05/02/2023]
Abstract
Engineered tissue models comprise a variety of multiplexed ensembles in which combinations of epithelial, stromal, and immune cells give rise to physiologic function. Engineering spatiotemporal control of cell-cell and cell-matrix interactions within these 3D multicellular tissues would represent a significant advance for tissue engineering. In this work, a new method, entitled CAMEO (Controlled Apoptosis in Multicellular tissues for Engineered Organogenesis) enables the non-invasive triggering of controlled apoptosis to eliminate genetically-engineered cells from a pre-established culture. Using this approach, the contribution of stromal cells to the phenotypic stability of primary human hepatocytes is examined. 3D hepatic microtissues, in which fibroblasts can enhance phenotypic stability and accelerate aggregation into spheroids, were found to rely only transiently on fibroblast interaction to support multiple axes of liver function, such as protein secretion and drug detoxification. Due to its modularity, CAMEO has the promise to be readily extendable to other applications that are tied to the complexity of 3D tissue biology, from understanding in vitro organoid models to building artificial tissue grafts.
Collapse
Affiliation(s)
- Amanda X Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arnav Chhabra
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - H-H Greco Song
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Heather E Fleming
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
8
|
Wencel A, Ciezkowska M, Wisniewska M, Zakrzewska KE, Pijanowska DG, Pluta KD. Effects of genetically modified human skin fibroblasts, stably overexpressing hepatocyte growth factor, on hepatic functions of cocultured C3A cells. Biotechnol Bioeng 2020; 118:72-81. [PMID: 32880912 DOI: 10.1002/bit.27551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/04/2020] [Accepted: 09/01/2020] [Indexed: 01/18/2023]
Abstract
Diseases leading to terminal hepatic failure are among the most common causes of death worldwide. Transplant of the whole organ is the only effective method to cure liver failure. Unfortunately, this treatment option is not available universally due to the serious shortage of donors. Thus, alternative methods have been developed that are aimed at prolonging the life of patients, including hepatic cells transplantation and bridging therapy based on hybrid bioartificial liver devices. Parenchymal liver cells are highly differentiated and perform many complex functions, such as detoxification and protein synthesis. Unfortunately, isolated hepatocytes display a rapid decline in viability and liver-specific functions. A number of methods have been developed to maintain hepatocytes in their highly differentiated state in vitro, amongst them the most promising being 3D growth scaffolds and decellularized tissues or coculture with other cell types required for the heterotypic cell-cell interactions. Here we present a novel approach to the hepatic cells culture based on the feeder layer cells genetically modified using lentiviral vector to stably produce additional amounts of hepatocyte growth factor and show the positive influence of these coculture conditions on the preservation of the hepatic functions of the liver parenchymal cells' model-C3A cells.
Collapse
Affiliation(s)
- Agnieszka Wencel
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Ciezkowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Monika Wisniewska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Karolina E Zakrzewska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland.,Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Dorota G Pijanowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Krzysztof D Pluta
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
9
|
Ehrlich A, Duche D, Ouedraogo G, Nahmias Y. Challenges and Opportunities in the Design of Liver-on-Chip Microdevices. Annu Rev Biomed Eng 2020; 21:219-239. [PMID: 31167098 DOI: 10.1146/annurev-bioeng-060418-052305] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The liver is the central hub of xenobiotic metabolism and consequently the organ most prone to cosmetic- and drug-induced toxicity. Failure to detect liver toxicity or to assess compound clearance during product development is a major cause of postmarketing product withdrawal, with disastrous clinical and financial consequences. While small animals are still the preferred model in drug development, the recent ban on animal use in the European Union created a pressing need to develop precise and efficient tools to detect human liver toxicity during cosmetic development. This article includes a brief review of liver development, organization, and function and focuses on the state of the art of long-term cell culture, including hepatocyte cell sources, heterotypic cell-cell interactions, oxygen demands, and culture medium formulation. Finally, the article reviews emerging liver-on-chip devices and discusses the advantages and pitfalls of individual designs. The goal of this review is to provide a framework to design liver-on-chip devices and criteria with which to evaluate this emerging technology.
Collapse
Affiliation(s)
- Avner Ehrlich
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Daniel Duche
- L'Oréal Research and Innovation, Aulnay-sous-Bois 93600, France
| | | | - Yaakov Nahmias
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, Hebrew University of Jerusalem, Jerusalem 91904, Israel.,Department of Cell and Developmental Biology, Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 91904, Israel.,Tissue Dynamics Ltd., Jerusalem 91904, Israel
| |
Collapse
|
10
|
Kukla DA, Crampton AL, Wood DK, Khetani SR. Microscale Collagen and Fibroblast Interactions Enhance Primary Human Hepatocyte Functions in Three-Dimensional Models. Gene Expr 2020; 20:1-18. [PMID: 32290899 PMCID: PMC7284102 DOI: 10.3727/105221620x15868728381608] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Human liver models that are three-dimensional (3D) in architecture are indispensable for compound metabolism/toxicity screening, to model liver diseases for drug discovery, and for cell-based therapies; however, further development of such models is needed to maintain high levels of primary human hepatocyte (PHH) functions for weeks to months. Therefore, here we determined how microscale 3D collagen I presentation and fibroblast interaction affect the longevity of PHHs. High-throughput droplet microfluidics was utilized to generate reproducibly sized (∼300-μm diameter) microtissues containing PHHs encapsulated in collagen I ± supportive fibroblasts, namely, 3T3-J2 murine embryonic fibroblasts or primary human hepatic stellate cells (HSCs); self-assembled spheroids and bulk collagen gels (macrogels) containing PHHs served as controls. Hepatic functions and gene expression were subsequently measured for up to 6 weeks. We found that microtissues placed within multiwell plates rescued PHH functions at 2- to 30-fold higher levels than spheroids or macrogels. Further coating of PHH microtissues with 3T3-J2s led to higher hepatic functions than when the two cell types were either coencapsulated together or when HSCs were used for the coating instead. Importantly, the 3T3-J2-coated PHH microtissues displayed 6+ weeks of relatively stable hepatic gene expression and function at levels similar to freshly thawed PHHs. Lastly, microtissues responded in a clinically relevant manner to drug-mediated cytochrome P450 induction or hepatotoxicity. In conclusion, fibroblast-coated collagen microtissues containing PHHs display high hepatic functions for 6+ weeks and are useful for assessing drug-mediated CYP induction and hepatotoxicity. Ultimately, microtissues may find utility for modeling liver diseases and as building blocks for cell-based therapies.
Collapse
Affiliation(s)
- David A. Kukla
- *Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Alexandra L. Crampton
- †Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - David K. Wood
- †Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Salman R. Khetani
- *Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
11
|
Kukla DA, Stoppel WL, Kaplan DL, Khetani SR. Assessing the compatibility of primary human hepatocyte culture within porous silk sponges. RSC Adv 2020; 10:37662-37674. [PMID: 35515172 PMCID: PMC9057238 DOI: 10.1039/d0ra04954a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/04/2020] [Indexed: 12/24/2022] Open
Abstract
Donor organ shortages have prompted the development of alternative implantable human liver tissues for patients suffering from end-stage liver failure. Purified silk proteins provide desirable features for generating implantable tissues, including sustainable sourcing from insects/arachnids, biocompatibility, tunable mechanical properties and degradation rates, and low immunogenicity upon implantation. While different cell types were previously cultured for weeks within silk-based scaffolds, it remains unclear whether such scaffolds can be used to culture primary human hepatocytes (PHH) isolated from livers. Therefore, here we assessed the compatibility of PHH culture within porous silk scaffolds that enable diffusion of oxygen/nutrients through the pores. We found that incorporation of type I collagen during the fabrication and/or autoclaving of porous silk scaffolds, as opposed to simple adsorption of collagen onto pre-fabricated silk scaffolds, was necessary to enable robust PHH attachment/function. Scaffolds with small pores (73 ± 25 μm) promoted larger PHH spheroids and consequently higher PHH functions than large pores (235 ± 84 μm) for at least 1 month in culture. Further incorporation of supportive fibroblasts into scaffolds enhanced PHH functions up to 5-fold relative to scaffolds with PHHs alone and 2D co-cultures on plastic. Lastly, encapsulating PHHs within protein hydrogels while housed in the silk scaffold led to higher functions than protein hydrogel-only or silk-only controls. In conclusion, porous silk scaffolds containing extracellular matrix proteins can be used for the culture of PHHs ± supportive non-parenchymal cells, which can be further built on in the future to create optimized silk-based liver tissue surrogates for cell-based therapy. Porous silk scaffolds hybridized with extracellular matrix proteins are useful for culture of primary human hepatocytes ± supportive non-parenchymal cells.![]()
Collapse
Affiliation(s)
- David A. Kukla
- Department of Bioengineering
- University of Illinois at Chicago
- Chicago
- USA
| | | | - David L. Kaplan
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Salman R. Khetani
- Department of Bioengineering
- University of Illinois at Chicago
- Chicago
- USA
| |
Collapse
|
12
|
Underhill GH, Khetani SR. Emerging trends in modeling human liver disease in vitro. APL Bioeng 2019; 3:040902. [PMID: 31893256 PMCID: PMC6930139 DOI: 10.1063/1.5119090] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/29/2019] [Indexed: 12/18/2022] Open
Abstract
The liver executes 500+ functions, such as protein synthesis, xenobiotic metabolism, bile production, and metabolism of carbohydrates/fats/proteins. Such functions can be severely degraded by drug-induced liver injury, nonalcoholic fatty liver disease, hepatitis B and viral infections, and hepatocellular carcinoma. These liver diseases, which represent a significant global health burden, are the subject of novel drug discovery by the pharmaceutical industry via the use of in vitro models of the human liver, given significant species-specific differences in disease profiles and drug outcomes. Isolated primary human hepatocytes (PHHs) are a physiologically relevant cell source to construct such models; however, these cells display a rapid decline in the phenotypic function within conventional 2-dimensional monocultures. To address such a limitation, several engineered platforms have been developed such as high-throughput cellular microarrays, micropatterned cocultures, self-assembled spheroids, bioprinted tissues, and perfusion devices; many of these platforms are being used to coculture PHHs with liver nonparenchymal cells to model complex cell cross talk in liver pathophysiology. In this perspective, we focus on the utility of representative platforms for mimicking key features of liver dysfunction in the context of chronic liver diseases and liver cancer. We further discuss pending issues that will need to be addressed in this field moving forward. Collectively, these in vitro liver disease models are being increasingly applied toward the development of new therapeutics that display an optimal balance of safety and efficacy, with a focus on expediting development, reducing high costs, and preventing harm to patients.
Collapse
Affiliation(s)
- Gregory H. Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Salman R. Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
13
|
Ware BR, Brown GE, Soldatow VY, LeCluyse EL, Khetani SR. Long-Term Engineered Cultures of Primary Mouse Hepatocytes for Strain and Species Comparison Studies During Drug Development. Gene Expr 2019; 19:199-214. [PMID: 31340881 PMCID: PMC6827040 DOI: 10.3727/105221619x15638857793317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Testing drugs in isogenic rodent strains to satisfy regulatory requirements is insufficient for derisking organ toxicity in genetically diverse human populations; in contrast, advances in mouse genetics can help mitigate these limitations. Compared to the expensive and slower in vivo testing, in vitro cultures enable the testing of large compound libraries toward prioritizing lead compounds and selecting an animal model with human-like response to a compound. In the case of the liver, a leading cause of drug attrition, isolated primary mouse hepatocytes (PMHs) rapidly decline in function within current culture platforms, which restricts their use for assessing the effects of longer-term compound exposure. Here we addressed this challenge by fabricating mouse micropatterned cocultures (mMPCC) containing PMHs and 3T3-J2 murine embryonic fibroblasts that displayed 4 weeks of functions; mMPCCs created from either C57Bl/6J or CD-1 PMHs outperformed collagen/Matrigel™ sandwich-cultured hepatocyte monocultures by ∼143-fold, 413-fold, and 10-fold for albumin secretion, urea synthesis, and cytochrome P450 activities, respectively. Such functional longevity of mMPCCs enabled in vivo relevant comparisons across strains for CYP induction and hepatotoxicity following exposure to 14 compounds with subsequent comparison to responses in primary human hepatocytes (PHHs). In conclusion, mMPCCs display high levels of major liver functions for several weeks and can be used to assess strain- and species-specific compound effects when used in conjunction with responses in PHHs. Ultimately, mMPCCs can be used to leverage the power of mouse genetics for characterizing subpopulations sensitive to compounds, characterizing the degree of interindividual variability, and elucidating genetic determinants of severe hepatotoxicity in humans.
Collapse
Affiliation(s)
- Brenton R. Ware
- *School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- †Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Grace E. Brown
- †Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Edward L. LeCluyse
- ‡The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA
| | - Salman R. Khetani
- *School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- †Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
- §Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
14
|
Lauschke VM, Shafagh RZ, Hendriks DFG, Ingelman-Sundberg M. 3D Primary Hepatocyte Culture Systems for Analyses of Liver Diseases, Drug Metabolism, and Toxicity: Emerging Culture Paradigms and Applications. Biotechnol J 2019; 14:e1800347. [PMID: 30957976 DOI: 10.1002/biot.201800347] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/26/2019] [Indexed: 02/06/2023]
Abstract
Recent research has shown that the maintenance of relevant liver functions ex vivo requires models in which the cells exhibit an in vivo-like phenotype, often achieved by reconstitution of appropriate cellular interactions. Multiple different models have been presented that differ in the cells utilized, media, and culture conditions. Furthermore, several technologically different approaches have been presented including bioreactors, chips, and plate-based systems in fluidic or static media constituting of chemically diverse materials. Using such models, the ability to predict drug metabolism, drug toxicity, and liver functionality have increased tremendously as compared to conventional in vitro models in which cells are cultured as 2D monolayers. Here, the authors highlight important considerations for microphysiological systems for primary hepatocyte culture, review current culture paradigms, and discuss their opportunities for studies of drug metabolism, hepatotoxicity, liver biology, and disease.
Collapse
Affiliation(s)
- Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Reza Z Shafagh
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77, Stockholm, Sweden.,Department of Micro and Nanosystems, School of Electrical Engineering, Royal Institute of Technology, SE-100 44, Stockholm, Sweden
| | - Delilah F G Hendriks
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77, Stockholm, Sweden.,Present address: Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT, Utrecht, The Netherlands
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| |
Collapse
|
15
|
Brown GE, Khetani SR. Microfabrication of liver and heart tissues for drug development. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0225. [PMID: 29786560 DOI: 10.1098/rstb.2017.0225] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2017] [Indexed: 12/12/2022] Open
Abstract
Drug-induced liver- and cardiotoxicity remain among the leading causes of preclinical and clinical drug attrition, marketplace drug withdrawals and black-box warnings on marketed drugs. Unfortunately, animal testing has proven to be insufficient for accurately predicting drug-induced liver- and cardiotoxicity across many drug classes, likely due to significant differences in tissue functions across species. Thus, the field of in vitro human tissue engineering has gained increasing importance over the last 10 years. Technologies such as protein micropatterning, microfluidics, three-dimensional scaffolds and bioprinting have revolutionized in vitro platforms as well as increased the long-term phenotypic stability of both primary cells and stem cell-derived differentiated cells. Here, we discuss advances in engineering approaches for constructing in vitro human liver and heart models with utility for drug development. Design features and validation data of representative models are presented to highlight major trends followed by the discussion of pending issues. Overall, bioengineered liver and heart models have significantly advanced our understanding of organ function and injury, which will prove useful for mitigating the risk of drug-induced organ toxicity to human patients, reducing animal usage for preclinical drug testing, aiding in the discovery of novel therapeutics against human diseases, and ultimately for applications in regenerative medicine.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
16
|
Underhill GH, Khetani SR. Advances in Engineered Human Liver Platforms for Drug Metabolism Studies. Drug Metab Dispos 2018; 46:1626-1637. [PMID: 30135245 DOI: 10.1124/dmd.118.083295] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022] Open
Abstract
Metabolism in the liver often determines the overall clearance rates of many pharmaceuticals. Furthermore, induction or inhibition of the liver drug metabolism enzymes by perpetrator drugs can influence the metabolism of victim drugs (drug-drug interactions). Therefore, determining liver-drug interactions is critical during preclinical drug development. Unfortunately, studies in animals are often of limited value because of significant differences in the metabolic pathways of the liver across different species. To mitigate such limitations, the pharmaceutical industry uses a continuum of human liver models, ranging from microsomes to transfected cell lines and cultures of primary human hepatocytes (PHHs). Of these models, PHHs provide a balance of high-throughput testing capabilities together with a physiologically relevant cell type that exhibits all the characteristic enzymes, cofactors, and transporters. However, PHH monocultures display a rapid decline in metabolic capacity. Consequently, bioengineers have developed several tools, such as cellular microarrays, micropatterned cocultures, self-assembled and bioprinted spheroids, and perfusion devices, to enhance and stabilize PHH functions for ≥2 weeks. Many of these platforms have been validated for drug studies, whereas some have been adapted to include liver nonparenchymal cells that can influence hepatic drug metabolism in health and disease. Here, we focus on the design features of such platforms and their representative drug metabolism validation datasets, while discussing emerging trends. Overall, the use of engineered human liver platforms in the pharmaceutical industry has been steadily rising over the last 10 years, and we anticipate that these platforms will become an integral part of drug development with continued commercialization and validation for routine screening use.
Collapse
Affiliation(s)
- Gregory H Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; and Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; and Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
17
|
Tegge AN, Rodrigues RR, Larkin AL, Vu L, Murali TM, Rajagopalan P. Transcriptomic Analysis of Hepatic Cells in Multicellular Organotypic Liver Models. Sci Rep 2018; 8:11306. [PMID: 30054499 PMCID: PMC6063915 DOI: 10.1038/s41598-018-29455-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 07/11/2018] [Indexed: 02/08/2023] Open
Abstract
Liver homeostasis requires the presence of both parenchymal and non-parenchymal cells (NPCs). However, systems biology studies of the liver have primarily focused on hepatocytes. Using an organotypic three-dimensional (3D) hepatic culture, we report the first transcriptomic study of liver sinusoidal endothelial cells (LSECs) and Kupffer cells (KCs) cultured with hepatocytes. Through computational pathway and interaction network analyses, we demonstrate that hepatocytes, LSECs and KCs have distinct expression profiles and functional characteristics. Our results show that LSECs in the presence of KCs exhibit decreased expression of focal adhesion kinase (FAK) signaling, a pathway linked to LSEC dedifferentiation. We report the novel result that peroxisome proliferator-activated receptor alpha (PPARα) is transcribed in LSECs. The expression of downstream processes corroborates active PPARα signaling in LSECs. We uncover transcriptional evidence in LSECs for a feedback mechanism between PPARα and farnesoid X-activated receptor (FXR) that maintains bile acid homeostasis; previously, this feedback was known occur only in HepG2 cells. We demonstrate that KCs in 3D liver models display expression patterns consistent with an anti-inflammatory phenotype when compared to monocultures. These results highlight the distinct roles of LSECs and KCs in maintaining liver function and emphasize the need for additional mechanistic studies of NPCs in addition to hepatocytes in liver-mimetic microenvironments.
Collapse
Affiliation(s)
- Allison N Tegge
- Department of Computer Science, Virginia Tech, Blacksburg, USA
- Department of Statistics, Virginia Tech, Blacksburg, USA
| | - Richard R Rodrigues
- Genetics, Bioinformatics, and Computational Biology Ph.D. Program, Virginia Tech, Blacksburg, USA
| | - Adam L Larkin
- Department of Chemical Engineering, Virginia Tech, Blacksburg, USA
| | - Lucas Vu
- Department of Chemical Engineering, Virginia Tech, Blacksburg, USA
| | - T M Murali
- Department of Computer Science, Virginia Tech, Blacksburg, USA.
- ICTAS Center for Systems Biology of Engineered Tissues, Virginia Tech, Blacksburg, USA.
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, Blacksburg, USA.
- ICTAS Center for Systems Biology of Engineered Tissues, Virginia Tech, Blacksburg, USA.
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, USA.
| |
Collapse
|
18
|
Davidson MD, Kukla DA, Khetani SR. Microengineered cultures containing human hepatic stellate cells and hepatocytes for drug development. Integr Biol (Camb) 2018; 9:662-677. [PMID: 28702667 DOI: 10.1039/c7ib00027h] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In non-alcoholic steatohepatitis (NASH), hepatic stellate cells (HSC) differentiate into myofibroblast-like cells that cause fibrosis, which predisposes patients to cirrhosis and hepatocellular carcinoma. Thus, modeling interactions between activated HSCs and hepatocytes in vitro can aid in the development of anti-NASH/fibrosis therapeutics and lead to a better understanding of disease progression. Species-specific differences in drug metabolism and disease pathways now necessitate the supplementation of animal studies with data acquired using human liver models; however, current models do not adequately model the negative effects of primary human activated HSCs on the phenotype of otherwise well-differentiated primary human hepatocytes (PHHs) as in vivo. Therefore, here we first determined the long-term effects of primary human activated HSCs on PHH phenotype in a micropatterned co-culture (MPCC) platform while using 3T3-J2 murine embryonic fibroblasts as the control cell type since it has been shown previously to stabilize PHH functions for 4-6 weeks. We found that HSCs were not able to stabilize the PHH phenotype to the same magnitude and longevity as the fibroblasts, which subsequently inspired the development of a micropatterned tri-culture (MPTC) platform in which (a) micropatterned PHHs were functionally stabilized using fibroblasts, and (b) the PHH phenotype was modulated by culturing HSCs within the fibroblast monolayer at physiologically-relevant ratios with PHHs. Transwell inserts containing HSCs were placed atop MPCCs containing fibroblasts to confirm the effects of paracrine signaling between PHHs and HSCs. We found that while albumin and urea secretions were relatively similar in MPTCs and MPCCs (suggesting well-differentiated PHHs), increasing HSC numbers within MPTCs downregulated hepatic cytochrome-P450 (2A6, 3A4) and transporter activities, and caused steatosis over 2 weeks. Furthermore, MPTCs secreted higher levels of pro-inflammatory interleukin-6 (IL-6) cytokine and C-reactive protein (CRP) than MPCCs. Treatment of MPCCs with HSC-conditioned culture medium confirmed that HSC secretions mediate the altered phenotype of PHHs observed in MPTCs, partly via IL-6 signaling. Lastly, we found that NADPH oxidase (NOX) inhibition and farnesoid X receptor (FXR) activation using clinically relevant drugs alleviated hepatic dysfunctions in MPTCs. In conclusion, MPTCs recapitulate symptoms of NASH- and early fibrosis-like dysfunctions in PHHs and have utility for drug discovery in this space.
Collapse
Affiliation(s)
- Matthew D Davidson
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | | | | |
Collapse
|
19
|
Ware BR, Durham MJ, Monckton CP, Khetani SR. A Cell Culture Platform to Maintain Long-term Phenotype of Primary Human Hepatocytes and Endothelial Cells. Cell Mol Gastroenterol Hepatol 2018; 5:187-207. [PMID: 29379855 PMCID: PMC5782488 DOI: 10.1016/j.jcmgh.2017.11.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/17/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS Modeling interactions between primary human hepatocytes (PHHs) and primary human liver sinusoidal endothelial cells (LSECs) in vitro can help elucidate human-specific mechanisms underlying liver physiology/disease and drug responses; however, existing hepatocyte/endothelial coculture models are suboptimal because of their use of rodent cells, cancerous cell lines, and/or nonliver endothelial cells. Hence, we sought to develop a platform that could maintain the long-term phenotype of PHHs and primary human LSECs. METHODS Primary human LSECs or human umbilical vein endothelial cells as the nonliver control were cocultivated with micropatterned PHH colonies (to control homotypic interactions) followed by an assessment of PHH morphology and functions (albumin and urea secretion, and cytochrome P-450 2A6 and 3A4 enzyme activities) over 3 weeks. Endothelial phenotype was assessed via gene expression patterns and scanning electron microscopy to visualize fenestrations. Hepatic responses in PHH/endothelial cocultures were benchmarked against responses in previously developed PHH/3T3-J2 fibroblast cocultures. Finally, PHH/fibroblast/endothelial cell tricultures were created and characterized as described previously. RESULTS LSECs, but not human umbilical vein endothelial cells, induced PHH albumin secretion for ∼11 days; however, neither endothelial cell type could maintain PHH morphology and functions to the same magnitude/longevity as the fibroblasts. In contrast, both PHHs and endothelial cells displayed stable phenotype for 3 weeks in PHH/fibroblast/endothelial cell tricultures; furthermore, layered tricultures in which PHHs and endothelial cells were separated by a protein gel to mimic the space of Disse displayed similar functional levels as the coplanar tricultures. CONCLUSIONS PHH/fibroblast/endothelial tricultures constitute a robust platform to elucidate reciprocal interactions between PHHs and endothelial cells in physiology, disease, and after drug exposure.
Collapse
Key Words
- 3T3-J2 Fibroblasts
- CD31, cluster of differentiation 31
- CD54, cluster of differentiation 54
- CYP450, cytochrome P-450
- ECM, extracellular matrix
- F8, factor VIII
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- HUVECs
- HUVECs, human umbilical vein endothelial cells
- LSECs
- LSECs, liver sinusoidal endothelial cells
- Micropatterned Cocultures
- NPCs, nonparenchymal cells
- PHHs, primary human hepatocytes
- SEM, scanning electron microscope
- Tricultures
- cDNA, complementary DNA
- vWF, von Willebrand factor
Collapse
Affiliation(s)
- Brenton R. Ware
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Mitchell J. Durham
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado
| | - Chase P. Monckton
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R. Khetani
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
- Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
20
|
Underhill GH, Khetani SR. Bioengineered Liver Models for Drug Testing and Cell Differentiation Studies. Cell Mol Gastroenterol Hepatol 2018; 5:426-439.e1. [PMID: 29675458 PMCID: PMC5904032 DOI: 10.1016/j.jcmgh.2017.11.012] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/21/2017] [Indexed: 12/19/2022]
Abstract
In vitro models of the human liver are important for the following: (1) mitigating the risk of drug-induced liver injury to human beings, (2) modeling human liver diseases, (3) elucidating the role of single and combinatorial microenvironmental cues on liver cell function, and (4) enabling cell-based therapies in the clinic. Methods to isolate and culture primary human hepatocytes (PHHs), the gold standard for building human liver models, were developed several decades ago; however, PHHs show a precipitous decline in phenotypic functions in 2-dimensional extracellular matrix-coated conventional culture formats, which does not allow chronic treatment with drugs and other stimuli. The development of several engineering tools, such as cellular microarrays, protein micropatterning, microfluidics, biomaterial scaffolds, and bioprinting, now allow precise control over the cellular microenvironment for enhancing the function of both PHHs and induced pluripotent stem cell-derived human hepatocyte-like cells; long-term (4+ weeks) stabilization of hepatocellular function typically requires co-cultivation with liver-derived or non-liver-derived nonparenchymal cell types. In addition, the recent development of liver organoid culture systems can provide a strategy for the enhanced expansion of therapeutically relevant cell types. Here, we discuss advances in engineering approaches for constructing in vitro human liver models that have utility in drug screening and for determining microenvironmental determinants of liver cell differentiation/function. Design features and validation data of representative models are presented to highlight major trends followed by the discussion of pending issues that need to be addressed. Overall, bioengineered liver models have significantly advanced our understanding of liver function and injury, which will prove useful for drug development and ultimately cell-based therapies.
Collapse
Key Words
- 3D, 3-dimensional
- BAL, bioartificial liver
- Bioprinting
- CRP, C-reactive protein
- CYP450, cytochrome P450
- Cellular Microarrays
- DILI, drug-induced liver injury
- ECM, extracellular matrix
- HSC, hepatic stellate cell
- Hepatocytes
- IL, interleukin
- KC, Kupffer cell
- LSEC, liver sinusoidal endothelial cell
- MPCC, micropatterned co-culture
- Microfluidics
- Micropatterned Co-Cultures
- NPC, nonparenchymal cell
- PEG, polyethylene glycol
- PHH, primary human hepatocyte
- Spheroids
- iHep, induced pluripotent stem cell-derived human hepatocyte-like cell
- iPS, induced pluripotent stem
Collapse
Affiliation(s)
- Gregory H. Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Salman R. Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
21
|
Elbaz A, Gao B, He Z, Gu Z. Hepatocyte Aggregate Formation on Chitin-Based Anisotropic Microstructures of Butterfly Wings. Biomimetics (Basel) 2018; 3:E2. [PMID: 31105224 PMCID: PMC6352657 DOI: 10.3390/biomimetics3010002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/09/2018] [Accepted: 01/13/2018] [Indexed: 01/03/2023] Open
Abstract
Scaffold nanotopography plays the most significant role in the mimicry of the in vivo microenvironment of the hepatocytes. Several attempts have been made to develop methods and substrates suited to growing hepatocytes into aggregates. Functional biomaterials, particularly biodegradable polymers, have been used in several studies aimed to develop improved scaffolds with ordered geometry and nanofibrous architecture for tissue engineering. However, there are still some limitation in their fabrication: it is not cost-efficient, is time-consuming, and exhibits some technological complications. The synthetic scaffolds are usually non-biodegradable and can be non-biocompatible compared to the naturally derived biomaterials. Here, we utilized a simple, cost-effective, and green method with two-step chemical treatment to get more selected hydrophilic butterfly wings from Morpho menelaus, Papilio ulysses telegonus, and Ornithoptera croesus lydius as a chitin-based natural scaffolds to growing hepatocyte aggregates. We established a three-dimensional (3D) in vitro model for culture of HepG2 cells and aggregate formation that maintained the hepatocytes function on these natural anisotropic microstructures. Cells cultured on these substrates show higher viability than those cultured on a two-dimensional (2D) culture plate. Methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay results revealed excellent viability of HepG2 cells on P. u. telegonus wings (fibrous area). The results also demonstrated appropriate cell activity, cell retention, and stable and functional expression in terms of albumin secretion and urea synthesis activity compared to the 2D monolayer culture of hepatocytes on the culture dish surface. With a slightly different degree, the other substrates also shown similar results. We anticipate that these natural anisotropic, biodegradable, and biocompatible substrates can maintain long-term hepatic culture as an in vitro 3D model for potential therapeutic applications and regenerative tissue applications. The model presented here provides a feasible alternative to the synthetic scaffolds and is expected to be more reliable for 3D organotypic liver culture models based on such scaffolds.
Collapse
Affiliation(s)
- Abdelrahman Elbaz
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China.
| | - Bingbing Gao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China.
| | - Zhenzhu He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China.
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China.
- Laboratory of Environment and Biosafety Research Institute of Southeast University in Suzhou, Suzhou 215123, China.
| |
Collapse
|
22
|
Meseguer-Ripolles J, Khetani SR, Blanco JG, Iredale M, Hay DC. Pluripotent Stem Cell-Derived Human Tissue: Platforms to Evaluate Drug Metabolism and Safety. AAPS J 2017; 20:20. [PMID: 29270863 PMCID: PMC5804345 DOI: 10.1208/s12248-017-0171-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/16/2017] [Indexed: 12/15/2022] Open
Abstract
Despite the improvements in drug screening, high levels of drug attrition persist. Although high-throughput screening platforms permit the testing of compound libraries, poor compound efficacy or unexpected organ toxicity are major causes of attrition. Part of the reason for drug failure resides in the models employed, most of which are not representative of normal organ biology. This same problem affects all the major organs during drug development. Hepatotoxicity and cardiotoxicity are two interesting examples of organ disease and can present in the late stages of drug development, resulting in major cost and increased risk to the patient. Currently, cell-based systems used within industry rely on immortalized or primary cell lines from donated tissue. These models possess significant advantages and disadvantages, but in general display limited relevance to the organ of interest. Recently, stem cell technology has shown promise in drug development and has been proposed as an alternative to current industrial systems. These offerings will provide the field with exciting new models to study human organ biology at scale and in detail. We believe that the recent advances in production of stem cell-derived hepatocytes and cardiomyocytes combined with cutting-edge engineering technologies make them an attractive alternative to current screening models for drug discovery. This will lead to fast failing of poor drugs earlier in the process, delivering safer and more efficacious medicines for the patient.
Collapse
Affiliation(s)
| | - Salman R Khetani
- University of Illinois at Chicago, Bioengineering (MC 063) 851 S Morgan St, 218 SEO, Chicago, Illinois, 60607, USA
| | - Javier G Blanco
- School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Mairi Iredale
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - David C Hay
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
23
|
Lin C, Romero R, Sorokina LV, Ballinger KR, Place LW, Kipper MJ, Khetani SR. A polyelectrolyte multilayer platform for investigating growth factor delivery modes in human liver cultures. J Biomed Mater Res A 2017; 106:971-984. [PMID: 29139224 DOI: 10.1002/jbm.a.36293] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/19/2017] [Accepted: 10/26/2017] [Indexed: 01/19/2023]
Abstract
Polyelectrolyte multilayers (PEMs) of chitosan and heparin are useful for mimicking growth factor (GF) binding to extracellular matrix (ECM) as in vivo. Here, we developed a PEM platform for delivering bound/adsorbed GFs to monocultures of primary human hepatocytes (PHHs) and PHH/non-parenchymal cell (NPC) co-cultures, which are useful for drug development and regenerative medicine. The effects of ECM protein coating (collagen I, fibronectin, and Matrigel®) and terminal PEM layer on PHH attachment/functions were determined. Then, heparin-terminated/fibronectin-coated PEMs were used to deliver varying concentrations of an adsorbed model GF, transforming growth factor β (TGFβ), to PHH monocultures while using soluble TGFβ delivery via culture medium as the conventional control. Soluble TGFβ delivery caused a severe, monotonic, and sustained downregulation of all PHH functions measured (albumin and urea secretions, cytochrome-P450 2A6 and 3A4 enzyme activities), whereas adsorbed TGFβ delivery caused transient upregulation of 3 out of 4 functions. Finally, functionally stable co-cultures of PHHs and 3T3-J2 murine embryonic fibroblasts were created on the heparin-terminated/fibronectin-coated PEMs modified with adsorbed TGFβ to elucidate similarities and differences in functional response relative to the monocultures. In conclusion, chitosan-heparin PEMs constitute a robust platform for investigating the effects of GF delivery modes on PHH monocultures and PHH/NPC co-cultures. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 971-984, 2018.
Collapse
Affiliation(s)
- Christine Lin
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Raimundo Romero
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Lioudmila V Sorokina
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Kimberly R Ballinger
- Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado
| | - Laura W Place
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Matt J Kipper
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado.,Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado
| | - Salman R Khetani
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
24
|
Ogoke O, Oluwole J, Parashurama N. Bioengineering considerations in liver regenerative medicine. J Biol Eng 2017; 11:46. [PMID: 29204185 PMCID: PMC5702480 DOI: 10.1186/s13036-017-0081-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/25/2017] [Indexed: 12/19/2022] Open
Abstract
Background Liver disease contributes significantly to global disease burden and is associated with rising incidence and escalating costs. It is likely that innovative approaches, arising from the emerging field of liver regenerative medicine, will counter these trends. Main body Liver regenerative medicine is a rapidly expanding field based on a rich history of basic investigations into the nature of liver structure, physiology, development, regeneration, and function. With a bioengineering perspective, we discuss all major subfields within liver regenerative medicine, focusing on the history, seminal publications, recent progress within these fields, and commercialization efforts. The areas reviewed include fundamental aspects of liver transplantation, liver regeneration, primary hepatocyte cell culture, bioartificial liver, hepatocyte transplantation and liver cell therapies, mouse liver repopulation, adult liver stem cell/progenitor cells, pluripotent stem cells, hepatic microdevices, and decellularized liver grafts. Conclusion These studies highlight the creative directions of liver regenerative medicine, the collective efforts of scientists, engineers, and doctors, and the bright outlook for a wide range of approaches and applications which will impact patients with liver disease.
Collapse
Affiliation(s)
- Ogechi Ogoke
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Furnas Hall, Buffalo, NY 14260 USA.,Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), 875 Ellicott St., Buffalo, NY 14203 USA
| | - Janet Oluwole
- Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), 875 Ellicott St., Buffalo, NY 14203 USA.,Department of Biomedical Engineering, University at Buffalo (State University of New York), Furnas Hall, 907 Furnas Hall, Buffalo, NY 14260 USA
| | - Natesh Parashurama
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Furnas Hall, Buffalo, NY 14260 USA.,Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), 875 Ellicott St., Buffalo, NY 14203 USA.,Department of Biomedical Engineering, University at Buffalo (State University of New York), Furnas Hall, 907 Furnas Hall, Buffalo, NY 14260 USA
| |
Collapse
|
25
|
Chinnici CM, Miceli V, Pampalone M, Lo Nigro A, Amico G, Conaldi PG. In vitro evidences of epithelial to mesenchymal transition in low cell-density cultured human fetal hepatocytes. Biochem Biophys Res Commun 2017. [PMID: 28624456 DOI: 10.1016/j.bbrc.2017.06.065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Culturing fetal hepatocytes in high cell-density allowed stabilization of the hepatocyte phenotype up to 8 weeks, including the maintenance of liver-specific functions. On the other hand, when cultured at low cell-density, fetal hepatocytes underwent morphological modifications and acquired fibroblastic morphology. Since a switch from E-cadherin to vimentin expression accompanied these changes, we hypothesized the occurrence of epithelial-to-mesenchymal transition when fetal hepatocytes were cultured at low cell-density. Changes in gene expressionsuch as up-regulation of fibrosis-related geneswere also observed, suggesting that the low cell-density culture system promoted the acquisition of a profibrotic phenotype in cultured hepatocytes. The origin of fibrogenic cells in the liver is not well known, and the role of hepatocytes as a source of fibrogenic cells is controversial. Therefore, we hypothesized that hepatocytes undergoing epithelial-to-mesenchymal transition could have a central role in liver fibrosis as a source of fibrogenic cells. To conclude, the high cell-density culture system could be a useful model for in vitro studies requiring long-term cultures of hepatocytes, such as the development of pharmaceutical drugs and mechanisms of viral infections. The low cell-density culture system may provide additional insights into the origin of fibrogenic cells in the liver, thus contributing to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Cinzia Maria Chinnici
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Palermo, Italy; Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneano per I Trapianti e Terapie Ad Alta Specializzazione), Palermo, Italy
| | - Vitale Miceli
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneano per I Trapianti e Terapie Ad Alta Specializzazione), Palermo, Italy.
| | - Mariangela Pampalone
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Palermo, Italy; Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneano per I Trapianti e Terapie Ad Alta Specializzazione), Palermo, Italy
| | - Antonio Lo Nigro
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Palermo, Italy; Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneano per I Trapianti e Terapie Ad Alta Specializzazione), Palermo, Italy
| | - Giandomenico Amico
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Palermo, Italy; Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneano per I Trapianti e Terapie Ad Alta Specializzazione), Palermo, Italy
| | - Pier Giulio Conaldi
- Fondazione Ri.MED, Regenerative Medicine and Biomedical Technologies Unit, Palermo, Italy; Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneano per I Trapianti e Terapie Ad Alta Specializzazione), Palermo, Italy
| |
Collapse
|
26
|
Long-term hepatitis B infection in a scalable hepatic co-culture system. Nat Commun 2017; 8:125. [PMID: 28743900 PMCID: PMC5527081 DOI: 10.1038/s41467-017-00200-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus causes chronic infections in 250 million people worldwide. Chronic hepatitis B virus carriers are at risk of developing fibrosis, cirrhosis, and hepatocellular carcinoma. A prophylactic vaccine exists and currently available antivirals can suppress but rarely cure chronic infections. The study of hepatitis B virus and development of curative antivirals are hampered by a scarcity of models that mimic infection in a physiologically relevant, cellular context. Here, we show that cell-culture and patient-derived hepatitis B virus can establish persistent infection for over 30 days in a self-assembling, primary hepatocyte co-culture system. Importantly, infection can be established without antiviral immune suppression, and susceptibility is not donor dependent. The platform is scalable to microwell formats, and we provide proof-of-concept for its use in testing entry inhibitors and antiviral compounds. The lack of models that mimic hepatitis B virus (HBV) infection in a physiologically relevant context has hampered drug development. Here, Winer et al. establish a self-assembling, primary hepatocyte co-culture system that can be infected with patient-derived HBV without further modifications.
Collapse
|
27
|
Fontenete S, Peña-Jimenez D, Perez-Moreno M. Heterocellular cadherin connections: coordinating adhesive cues in homeostasis and cancer. F1000Res 2017; 6:1010. [PMID: 28721207 PMCID: PMC5497824 DOI: 10.12688/f1000research.11357.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2017] [Indexed: 01/06/2023] Open
Abstract
This short insight covers some of the recent topics relevant to the field of cadherin-catenin adhesion in mediating connections between different cell types, so-called heterotypic or heterocellular connections, in both homeostasis and cancer. These scientific discoveries are increasing our understanding of how multiple cells residing in complex tissues can be instructed by cadherin adhesion receptors to regulate tissue architecture and function and how these cadherin-mediated heterocellular connections spur tumor growth and the acquisition of malignant characteristics in tumor cells. Overall, the findings that have emerged over the past few years are elucidating the complexity of the functional roles of the cadherin-catenin complexes. Future exciting research lies ahead in order to understand the physical basis of these heterotypic interactions and their influence on the behavior of heterogeneous cellular populations as well as their roles in mediating phenotypic and genetic changes as cells evolve through complex environments during morphogenesis and cancer.
Collapse
Affiliation(s)
- Silvia Fontenete
- Epithelial Cell Biology Group, Cancer Cell Biology Programme, Spanish Cancer Research Centre (CNIO), Madrid, Spain
| | - Daniel Peña-Jimenez
- Epithelial Cell Biology Group, Cancer Cell Biology Programme, Spanish Cancer Research Centre (CNIO), Madrid, Spain
| | - Mirna Perez-Moreno
- Epithelial Cell Biology Group, Cancer Cell Biology Programme, Spanish Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
28
|
Vu LT, Orbach SM, Ray WK, Cassin ME, Rajagopalan P, Helm RF. The hepatocyte proteome in organotypic rat liver models and the influence of the local microenvironment. Proteome Sci 2017; 15:12. [PMID: 28649179 PMCID: PMC5480101 DOI: 10.1186/s12953-017-0120-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/15/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Liver models that closely mimic the in vivo microenvironment are useful for understanding liver functions, capabilities, and intercellular communication processes. Three-dimensional (3D) liver models assembled using hepatocytes and liver sinusoidal endothelial cells (LSECs) separated by a polyelectrolyte multilayer (PEM) provide a functional system while also permitting isolation of individual cell types for proteomic analyses. METHODS To better understand the mechanisms and processes that underlie liver model function, hepatocytes were maintained as monolayers and 3D PEM-based formats in the presence or absence of primary LSECs. The resulting hepatocyte proteomes, the proteins in the PEM, and extracellular levels of urea, albumin and glucose after three days of culture were compared. RESULTS All systems were ketogenic and found to release glucose. The presence of the PEM led to increases in proteins associated with both mitochondrial and peroxisomal-based β-oxidation. The PEMs also limited production of structural and migratory proteins associated with dedifferentiation. The presence of LSECs increased levels of Phase I and Phase II biotransformation enzymes as well as several proteins associated with the endoplasmic reticulum and extracellular matrix remodeling. The proteomic analysis of the PEMs indicated that there was no significant change after three days of culture. These results are discussed in relation to liver model function. CONCLUSIONS Heterotypic cell-cell and cell-ECM interactions exert different effects on hepatocyte functions and phenotypes.
Collapse
Affiliation(s)
- Lucas T. Vu
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061 USA
| | - Sophia M. Orbach
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061 USA
| | - W. Keith Ray
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia 24061 USA
| | - Margaret E. Cassin
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061 USA
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061 USA
- School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia 24061 USA
- ICTAS Center for Systems Biology and Engineered Tissues, Virginia Tech, Blacksburg, Virginia 24061 USA
| | - Richard F. Helm
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia 24061 USA
- ICTAS Center for Systems Biology and Engineered Tissues, Virginia Tech, Blacksburg, Virginia 24061 USA
| |
Collapse
|
29
|
Lin C, Khetani SR. Micropatterned Co-Cultures of Human Hepatocytes and Stromal Cells for the Assessment of Drug Clearance and Drug-Drug Interactions. ACTA ACUST UNITED AC 2017; 72:14.17.1-14.17.23. [PMID: 28463419 DOI: 10.1002/cptx.23] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Drug clearance rates from the body can determine drug exposure that can affect efficacy or toxicity. Thus, accurate prediction of drug clearance during preclinical development can help guide dose selection in humans, but animal testing is not always predictive of human outcomes. Because hepatic drug metabolism is a rate-limiting step in the overall clearance of many drugs, primary human hepatocytes (PHHs) in suspension cultures or monolayers are used for drug clearance predictions. Yet, the precipitous decline in drug metabolism capacity can lead to significant underestimation of clearance rates, particularly for low turnover compounds that have desirable one-pill-a-day dosing regimens. In contrast, micropatterned co-cultures (MPCCs) of PHHs and fibroblasts display phenotypic stability for several weeks and can help mitigate the limitations of conventional cultures. Here, we describe protocols to create and use MPCCs for drug clearance predictions, and for modeling clinically-relevant drug-drug interactions that can affect drug clearance. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Christine Lin
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
30
|
Rajendran D, Hussain A, Yip D, Parekh A, Shrirao A, Cho CH. Long-term liver-specific functions of hepatocytes in electrospun chitosan nanofiber scaffolds coated with fibronectin. J Biomed Mater Res A 2017; 105:2119-2128. [PMID: 28371246 DOI: 10.1002/jbm.a.36072] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/16/2017] [Accepted: 03/22/2017] [Indexed: 12/22/2022]
Abstract
In this study, a new 3D liver model was developed using biomimetic nanofiber scaffolds and co-culture system consisting of hepatocytes and fibroblasts for the maintenance of long-term liver functions. The chitosan nanofiber scaffolds were fabricated by the electrospinning technique. To enhance cellular adhesion and spreading, the surfaces of the chitosan scaffolds were coated with fibronectin (FN) by adsorption and evaluated for various cell types. Cellular phenotype, protein expression, and liver-specific functions were extensively characterized by immunofluorescent and histochemical stainings, albumin enzyme-linked immunosorbent assay and Cytochrome p450 detoxification assays, and scanning electron microscopy. The electrospun chitosan scaffolds exhibited a highly porous and randomly oriented nanofibrous structure. The FN coating on the surface of the chitosan nanofibers significantly enhanced cell attachment and spreading, as expected, as surface modification with this cell adhesion molecule on the chitosan surface is important for focal adhesion formation and integrin binding. Comparison of hepatocyte mono-cultures and co-cultures in 3D culture systems indicated that the hepatocytes in co-cultures formed colonies and maintained their morphologies and functions for prolonged periods of time. The 3D liver tissue model developed in this study will provide useful tools toward the development of engineered liver tissues for drug screening and tissue engineering applications. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2119-2128, 2017.
Collapse
Affiliation(s)
- Divya Rajendran
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, 07102
| | - Ali Hussain
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, 07102
| | - Derek Yip
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, 07102
| | - Amit Parekh
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, 07102
| | - Anil Shrirao
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, 08854
| | - Cheul H Cho
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, 07102
| |
Collapse
|
31
|
Lauschke VM, Hendriks DFG, Bell CC, Andersson TB, Ingelman-Sundberg M. Novel 3D Culture Systems for Studies of Human Liver Function and Assessments of the Hepatotoxicity of Drugs and Drug Candidates. Chem Res Toxicol 2016; 29:1936-1955. [DOI: 10.1021/acs.chemrestox.6b00150] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Volker M. Lauschke
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Delilah F. G. Hendriks
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Catherine C. Bell
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Tommy B. Andersson
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Cardiovascular
and Metabolic Diseases, Innovative Medicines and Early Development
Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - Magnus Ingelman-Sundberg
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| |
Collapse
|
32
|
Shan J, Logan DJ, Root DE, Carpenter AE, Bhatia SN. High-Throughput Platform for Identifying Molecular Factors Involved in Phenotypic Stabilization of Primary Human Hepatocytes In Vitro. ACTA ACUST UNITED AC 2016; 21:897-911. [PMID: 27650791 DOI: 10.1177/1087057116660277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Liver disease is a leading cause of morbidity worldwide and treatment options are limited, with organ transplantation being the only form of definitive management. Cell-based therapies have long held promise as alternatives to whole-organ transplantation but have been hindered by the rapid loss of liver-specific functions over a period of days in cultured hepatocytes. Hypothesis-driven studies have identified a handful of factors that modulate hepatocyte functions in vitro, but our understanding of the mechanisms involved remains incomplete. We thus report here the development of a high-throughput platform to enable systematic interrogation of liver biology in vitro. The platform is currently configured to enable genetic knockdown screens and includes an enzyme-linked immunosorbent assay-based functional assay to quantify albumin output as a surrogate marker for hepatocyte synthetic functions as well as an image-based viability assay that counts hepatocyte nuclei. Using this platform, we identified 12 gene products that may be important for hepatocyte viability and/or liver identity in vitro. These results represent important first steps in the elucidation of mechanisms instrumental to the phenotypic maintenance of hepatocytes in vitro, and we hope that the tools reported here will empower additional studies in various fields of liver research.
Collapse
Affiliation(s)
- Jing Shan
- Harvard-MIT Division of Health Sciences and Technology, MIT, Cambridge, MA, USA
| | - David J Logan
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA Pfizer Neuroscience and Pain Research Unit, Cambridge, MA, USA
| | - David E Root
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Sangeeta N Bhatia
- Harvard-MIT Division of Health Sciences and Technology, MIT, Cambridge, MA, USA The Broad Institute of MIT and Harvard, Cambridge, MA, USA Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
33
|
Schepers A, Li C, Chhabra A, Seney BT, Bhatia S. Engineering a perfusable 3D human liver platform from iPS cells. LAB ON A CHIP 2016; 16:2644-53. [PMID: 27296616 PMCID: PMC5318999 DOI: 10.1039/c6lc00598e] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In vitro models of human tissue are crucial to our ability to study human disease as well as develop safe and effective drug therapies. Models of single organs in static and microfluidic culture have been established and shown utility for modeling some aspects of health and disease; however, these systems lack multi-organ interactions that are critical to some aspects of drug metabolism and toxicity. Thus, as part of a consortium of researchers, we have developed a liver chip that meets the following criteria: (1) employs human iPS cells from a patient of interest, (2) cultures cells in perfusable 3D organoids, and (3) is robust to variations in perfusion rate so as to be compatible in series with other specialized tissue chips (e.g. heart, lung). In order to achieve this, we describe methods to form hepatocyte aggregates from primary and iPS-derived cells, alone and in co-culture with support cells. This necessitated a novel culture protocol for the interrupted differentiation of iPS cells that permits their removal from a plated surface and aggregation while maintaining phenotypic hepatic functions. In order to incorporate these 3D aggregates in a perfusable platform, we next encapsulated the cells in a PEG hydrogel to prevent aggregation and overgrowth once on chip. We adapted a C-trap chip architecture from the literature that enabled robust loading with encapsulated organoids and culture over a range of flow rates. Finally, we characterize the liver functions of this iHep organoid chip under perfusion and demonstrate a lifetime of at least 28 days. We envision that such this strategy can be generalized to other microfluidic tissue models and provides an opportunity to query patient-specific liver responses in vitro.
Collapse
Affiliation(s)
- Arnout Schepers
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Cheri Li
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arnav Chhabra
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Benjamin Tschudy Seney
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sangeeta Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA and Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA and Howard Hughes Medical Institute, Cambridge, MA 02139, USA
| |
Collapse
|
34
|
Arisaka Y, Kobayashi J, Ohashi K, Tatsumi K, Kim K, Akiyama Y, Yamato M, Okano T. A heparin-modified thermoresponsive surface with heparin-binding epidermal growth factor-like growth factor for maintaining hepatic functions in vitro and harvesting hepatocyte sheets. Regen Ther 2016; 3:97-106. [PMID: 31245479 PMCID: PMC6581876 DOI: 10.1016/j.reth.2016.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/08/2016] [Accepted: 03/08/2016] [Indexed: 01/21/2023] Open
Abstract
A heparin-modified thermoresponsive surface bound with heparin-binding epidermal growth factor-like growth factor (HB-EGF) was designed to allow creation of transferrable and functional hepatocyte sheets. A heparin-modified thermoresponsive surface was prepared by covalently tethering heparin onto poly(N-isopropylacrylamide-co-2-carboxyisopropylacrylamide)-grafted tissue culture polystyrene surfaces (Heparin-IC). HB-EGFs were able to stably bind to heparin-IC via affinity interaction. The survival of primary rat hepatocytes was maintained through HB-EGF-bound heparin-IC (HB-EGF/heparin-IC). Moreover, cultured rat primary hepatocytes on HB-EGF/heparin-IC exhibited higher albumin-secretion than hepatocytes cultured on PIPAAm-grafted and collagen-coated surfaces with soluble HB-EGF in the culture medium, regardless of whether soluble EGF was added. These results suggested that HB-EGF/heparin-IC is able to effectively maintain hepatic function via continuous signaling of HB-EGF. After a 4-day cultivation, the cultured hepatocytes on HB-EGF/heparin-IC detached as a cell sheet with fibronectin and HB-EGF only after the temperature was lowered to 20 °C. In addition, higher expression of hepatocyte-specific genes (albumin, hepatocyte nuclear factor 4 alpha, coagulation factor VII, and coagulation factor IX) in hepatocyte sheets was detected on HB-EGF/heparin-IC than on a PIPAAm surface with soluble HB-EGF, indicating that HB-EGF/heparin-IC suppressed the dedifferentiation of cultured hepatocytes. Hence, heparin-modified thermoresponsive surfaces bound with HB-EGF facilitate the fabrication of transferrable hepatocyte sheets with intact hepatic functions and have the potential to provide an in vitro culture system using functional hepatocyte sheet tissues, which may serve as an effective hepatocyte-based tissue engineering platform for liver disease treatments.
Collapse
Key Words
- Alb, albumin
- CIPAAm, 2-carboxyisopropylacrylamide
- DMEM, Dulbecco's modified Eagle's medium
- ECM, extracellular matrix
- EDC, 1-ethyl-3-(3-dimetylaminopropyl)-carbodiimide hydrochloride
- EDTA, trypsin/ethylenediaminetetraacetic acid
- EGF, epidermal growth factor
- ELISA, enzyme-linked immunosorbent assay
- F7, coagulation factor VII
- F9, coagulation factor IX
- FBS, fetal bovine serum
- HB-EGF, heparin-binding EGF-like growth factor
- HB-EGFX/heparin-IC, HB-EGF-bound heparin-IC
- Heparin
- Heparin-binding EGF-like growth factor
- Hepatocyte sheet
- Hnf4α, hepatocyte nuclear factor 4 alpha
- IC, poly(N-isopropylacrylamide-co-2-carboxyisopropylacrylamide) on TCPS
- IPAAm, N-isopropylacrylamide
- MES, morpholinoethanesulfonic acid monohydrate
- NHS, N-hydroxysuccinimide
- PBS, Dulbecco's phosphate buffered saline
- PIPAAm, poly(N-isopropylacrylamide) on TCPS
- PIPAAm + HB-EGFY, PIPAAm with soluble HB-EGF
- Poly(N-isopropylacrylamide)
- RT-PCR, reverse transcription polymerase chain reaction
- TCPS, tissue culture polystyrene dishe
- Thermoresponsive cell culture surface
- bFGF, basic fibroblast growth factor
- heparin-IC, heparin-modified IC
Collapse
Affiliation(s)
| | - Jun Kobayashi
- Institute of Advanced Biomedical Engineering and Science and Global Center of Excellence (COE) Program, Tokyo Women's Medical University (TWIns), 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan
| | | | | | | | | | | | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science and Global Center of Excellence (COE) Program, Tokyo Women's Medical University (TWIns), 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
35
|
Jeong D, Han C, Kang I, Park HT, Kim J, Ryu H, Gho YS, Park J. Effect of Concentrated Fibroblast-Conditioned Media on In Vitro Maintenance of Rat Primary Hepatocyte. PLoS One 2016; 11:e0148846. [PMID: 26863621 PMCID: PMC4749383 DOI: 10.1371/journal.pone.0148846] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 01/25/2016] [Indexed: 01/20/2023] Open
Abstract
The effects of concentrated fibroblast-conditioned media were tested to determine whether hepatocyte function can be maintained without direct contact between hepatocytes and fibroblasts. Primary rat hepatocytes cultured with a concentrated conditioned media of NIH-3T3 J2 cell line (final concentration of 55 mg/ml) showed significantly improved survival and functions (albumin and urea) compared to those of control groups. They also showed higher expression levels of mRNA, albumin and tyrosine aminotransferase compared to hepatocyte monoculture. The results suggest that culture with concentrated fibroblast-conditioned media could be an easy method for in vitro maintenance of primary hepatocytes. They also could be contribute to understand and analyze co-culture condition of hepatocyte with stroma cells.
Collapse
Affiliation(s)
- Dayeong Jeong
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeong-buk, Republic of Korea
| | - Chungmin Han
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeong-buk, Republic of Korea
| | - Inhye Kang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeong-buk, Republic of Korea
| | - Hyun Taek Park
- Department of Life Science and Division of Molecular and Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeong-buk, Republic of Korea
| | - Jiyoon Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeong-buk, Republic of Korea
| | - Hayoung Ryu
- Chadwick International School, Songdo, Incheon, Republic of Korea
| | - Yong Song Gho
- Department of Life Science and Division of Molecular and Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeong-buk, Republic of Korea
| | - Jaesung Park
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeong-buk, Republic of Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeong-buk, Republic of Korea
- * E-mail:
| |
Collapse
|
36
|
March S, Ramanan V, Trehan K, Ng S, Galstian A, Gural N, Scull MA, Shlomai A, Mota MM, Fleming HE, Khetani SR, Rice CM, Bhatia SN. Micropatterned coculture of primary human hepatocytes and supportive cells for the study of hepatotropic pathogens. Nat Protoc 2015; 10:2027-53. [PMID: 26584444 PMCID: PMC5867906 DOI: 10.1038/nprot.2015.128] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The development of therapies and vaccines for human hepatropic pathogens requires robust model systems that enable the study of host-pathogen interactions. However, in vitro liver models of infection typically use either hepatoma cell lines that exhibit aberrant physiology or primary human hepatocytes in culture conditions in which they rapidly lose their hepatic phenotype. To achieve stable and robust in vitro primary human hepatocyte models, we developed micropatterned cocultures (MPCCs), which consist of primary human hepatocytes organized into 2D islands that are surrounded by supportive fibroblast cells. By using this system, which can be established over a period of days, and maintained over multiple weeks, we demonstrate how to recapitulate in vitro hepatic life cycles for the hepatitis B and C viruses and the Plasmodium pathogens P. falciparum and P. vivax. The MPCC platform can be used to uncover aspects of host-pathogen interactions, and it has the potential to be used for drug and vaccine development.
Collapse
Affiliation(s)
- Sandra March
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Vyas Ramanan
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Kartik Trehan
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Shengyong Ng
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ani Galstian
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Nil Gural
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Margaret A Scull
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, New York, USA
| | - Amir Shlomai
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, New York, USA
| | - Maria M Mota
- Unidade de Malaria, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Heather E Fleming
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Salman R Khetani
- Department of Mechanical Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, New York, USA
| | - Sangeeta N Bhatia
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
37
|
Ryabinin VE. [Problems and prospects of creation of extracorporal systems for support of functional livers status]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2015; 61:545-59. [PMID: 26539863 DOI: 10.18097/pbmc20156105545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review considers features of efferent therapy employing extracorporeal systems, the devices known as "artificial liver" and "bioartificial liver" in the treatment of liver insufficiency. Analysis of literature data shows the need for further development of these biomedical studies and the search for optimal solutions in the selection of the source of hepatocytes, the development of bioreactors and biomaterials forming the basis of devices like "bioartificial liver". Taking into consideration certain advantages and disadvantages typical for various methods of extracorporeal support of the functional state of the liver one can evaluate prior experience in the treatment of liver diseases and approaches to the development of new, more effective medical technologies.
Collapse
Affiliation(s)
- V E Ryabinin
- South-Ural State Medical University, Chelyabinsk, Russia
| |
Collapse
|
38
|
Stevens KR, Miller JS, Blakely BL, Chen CS, Bhatia SN. Degradable hydrogels derived from PEG-diacrylamide for hepatic tissue engineering. J Biomed Mater Res A 2015; 103:3331-8. [PMID: 25851120 PMCID: PMC4890565 DOI: 10.1002/jbm.a.35478] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/02/2015] [Accepted: 04/02/2015] [Indexed: 01/19/2023]
Abstract
Engineered tissue constructs have the potential to augment or replace whole organ transplantation for the treatment of liver failure. Poly(ethylene glycol) (PEG)‐based systems are particularly promising for the construction of engineered liver tissue due to their biocompatibility and amenability to modular addition of bioactive factors. To date, primary hepatocytes have been successfully encapsulated in non‐degradable hydrogels based on PEG‐diacrylate (PEGDA). In this study, we describe a hydrogel system based on PEG‐diacrylamide (PEGDAAm) containing matrix‐metalloproteinase sensitive (MMP‐sensitive) peptide in the hydrogel backbone that is suitable for hepatocyte culture both in vitro and after implantation. By replacing hydrolytically unstable esters in PEGDA with amides in PEGDAAm, resultant hydrogels resisted non‐specific hydrolysis, while still allowing for MMP‐mediated hydrogel degradation. Optimization of polymerization conditions, hepatocellular density, and multicellular tissue composition modulated both the magnitude and longevity of hepatic function in vitro. Importantly, hepatic PEGDAAm‐based tissues survived and functioned for over 3 weeks after implantation ectopically in the intraperitoneal (IP) space of nude mice. Together, these studies suggest that MMP‐sensitive PEGDAAm‐based hydrogels may be a useful material system for applications in tissue engineering and regenerative medicine. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 103A: 3331–3338, 2015.
Collapse
Affiliation(s)
- Kelly R Stevens
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| | - Jordan S Miller
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Brandon L Blakely
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Christopher S Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Sangeeta N Bhatia
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139.,Howard Hughes Medical Institute, Cambridge, Massachusetts, 02139.,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| |
Collapse
|
39
|
Berger DR, Ware BR, Davidson MD, Allsup SR, Khetani SR. Enhancing the functional maturity of induced pluripotent stem cell-derived human hepatocytes by controlled presentation of cell-cell interactions in vitro. Hepatology 2015; 61:1370-81. [PMID: 25421237 DOI: 10.1002/hep.27621] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 11/19/2014] [Indexed: 12/26/2022]
Abstract
UNLABELLED Induced pluripotent stem cell-derived human hepatocyte-like cells (iHeps) could provide a powerful tool for studying the mechanisms underlying human liver development and disease, testing the efficacy and safety of pharmaceuticals across different patients (i.e., personalized medicine), and enabling cell-based therapies in the clinic. However, current in vitro protocols that rely upon growth factors and extracellular matrices (ECMs) alone yield iHeps with low levels of liver functions relative to adult primary human hepatocytes (PHHs). Moreover, these low hepatic functions in iHeps are difficult to maintain for prolonged times (weeks to months) in culture. Here, we engineered a micropatterned coculture (iMPCC) platform in a multiwell format that, in contrast to conventional confluent cultures, significantly enhanced the functional maturation and longevity of iHeps in culture for at least 4 weeks in vitro when benchmarked against multiple donors of PHHs. In particular, iHeps were micropatterned onto collagen-coated domains of empirically optimized dimensions, surrounded by 3T3-J2 murine embryonic fibroblasts, and then sandwiched with a thin layer of ECM gel (Matrigel). We assessed iHep maturity by global gene expression profiles, hepatic polarity, secretion of albumin and urea, basal cytochrome P450 (CYP450) activities, phase II conjugation, drug-mediated CYP450 induction, and drug-induced hepatotoxicity. CONCLUSION Controlling both homotypic interactions between iHeps and heterotypic interactions with stromal fibroblasts significantly matures iHep functions and maintains them for several weeks in culture. In the future, iMPCCs could prove useful for drug screening, studying molecular mechanisms underlying iHep differentiation, modeling liver diseases, and integration into human-on-a-chip systems being designed to assess multiorgan responses to compounds.
Collapse
Affiliation(s)
- Dustin R Berger
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO
| | | | | | | | | |
Collapse
|
40
|
Usta OB, McCarty WJ, Bale S, Hegde M, Jindal R, Bhushan A, Golberg I, Yarmush ML. Microengineered cell and tissue systems for drug screening and toxicology applications: Evolution of in-vitro liver technologies. TECHNOLOGY 2015; 3:1-26. [PMID: 26167518 PMCID: PMC4494128 DOI: 10.1142/s2339547815300012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The liver performs many key functions, the most prominent of which is serving as the metabolic hub of the body. For this reason, the liver is the focal point of many investigations aimed at understanding an organism's toxicological response to endogenous and exogenous challenges. Because so many drug failures have involved direct liver toxicity or other organ toxicity from liver generated metabolites, the pharmaceutical industry has constantly sought superior, predictive in-vitro models that can more quickly and efficiently identify problematic drug candidates before they incur major development costs, and certainly before they are released to the public. In this broad review, we present a survey and critical comparison of in-vitro liver technologies along a broad spectrum, but focus on the current renewed push to develop "organs-on-a-chip". One prominent set of conclusions from this review is that while a large body of recent work has steered the field towards an ever more comprehensive understanding of what is needed, the field remains in great need of several key advances, including establishment of standard characterization methods, enhanced technologies that mimic the in-vivo cellular environment, and better computational approaches to bridge the gap between the in-vitro and in-vivo results.
Collapse
Affiliation(s)
- O B Usta
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - W J McCarty
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - S Bale
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - M Hegde
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - R Jindal
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - A Bhushan
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - I Golberg
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - M L Yarmush
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA ; Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd., Piscataway, NJ 08854, USA
| |
Collapse
|
41
|
Khetani SR, Berger DR, Ballinger KR, Davidson MD, Lin C, Ware BR. Microengineered liver tissues for drug testing. ACTA ACUST UNITED AC 2015; 20:216-50. [PMID: 25617027 DOI: 10.1177/2211068214566939] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Indexed: 01/09/2023]
Abstract
Drug-induced liver injury (DILI) is a leading cause of drug attrition. Significant and well-documented differences between animals and humans in liver pathways now necessitate the use of human-relevant in vitro liver models for testing new chemical entities during preclinical drug development. Consequently, several human liver models with various levels of in vivo-like complexity have been developed for assessment of drug metabolism, toxicity, and efficacy on liver diseases. Recent trends leverage engineering tools, such as those adapted from the semiconductor industry, to enable precise control over the microenvironment of liver cells and to allow for miniaturization into formats amenable for higher throughput drug screening. Integration of liver models into organs-on-a-chip devices, permitting crosstalk between tissue types, is actively being pursued to obtain a systems-level understanding of drug effects. Here, we review the major trends, challenges, and opportunities associated with development and implementation of engineered liver models created from primary cells, cell lines, and stem cell-derived hepatocyte-like cells. We also present key applications where such models are currently making an impact and highlight areas for improvement. In the future, engineered liver models will prove useful for selecting drugs that are efficacious, safer, and, in some cases, personalized for specific patient populations.
Collapse
Affiliation(s)
- Salman R Khetani
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Dustin R Berger
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Kimberly R Ballinger
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Matthew D Davidson
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Christine Lin
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Brenton R Ware
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
42
|
Abstract
The liver is the largest internal organ in mammals, serving a wide spectrum of vital functions. Loss of liver function due to drug toxicity, progressive fatty liver disease, or viral infection is a major cause of death in the United States of America. Pharmaceutical and cosmetic toxicity screening, basic research and the development of bioartificial liver devices require long-term hepatocyte culture techniques that sustain hepatocyte morphology and function. In recent years, several techniques have been developed that can support high levels of liver-specific gene expression, metabolic function, and synthetic activity for several weeks in culture. These include the collagen double gel configuration, hepatocyte spheroids, coculture with nonparenchymal cells, and micropatterned cocultures. This chapter will cover the current status of hepatocyte culture techniques, including media formulation, oxygen supply, and heterotypic cell-cell interactions.
Collapse
Affiliation(s)
- Merav Cohen
- Department of Cell and Developmental Biology, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | |
Collapse
|
43
|
Hegde M, Jindal R, Bhushan A, Bale SS, McCarty WJ, Golberg I, Usta OB, Yarmush ML. Dynamic interplay of flow and collagen stabilizes primary hepatocytes culture in a microfluidic platform. LAB ON A CHIP 2014; 14:2033-9. [PMID: 24770663 PMCID: PMC4036071 DOI: 10.1039/c4lc00071d] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The creation of stable flow cultures of hepatocytes is highly desirable for the development of platforms for drug toxicity screening, bio-artificial liver support devices, and models for investigating liver physiology and pathophysiology. Given that hepatocytes cultured using the collagen overlay or in 'sandwich' configuration maintain a wide range of differentiated functions, we describe a simple method for adapting this culture configuration within a microfluidic device. The device design consists of a porous membrane sandwiched between two layers of PDMS resulting in a two-chambered device. In the bottom chamber, hepatocytes are cultured in the collagen sandwich configuration, while the top chamber is accessible for flow. We demonstrate that hepatocytes cultured under flow exhibit higher albumin and urea secretions and induce cytochrome P450 1A1 activity in comparison to static cultures. Furthermore, over two weeks, hepatocytes cultured under flow show a well-connected cellular network with bile canaliculi formation, whereas static cultures show formation of gaps in the cellular network that progressively increase over time. Although enhanced functional response of hepatocytes cultured under flow has been observed in multiple prior studies, the exact mechanism for this flow induced effect remains unknown. In our work, we identified that hepatocytes secrete a higher level of collagen in the flow cultures; inhibiting collagen secretion within the flow cultures reduced albumin secretion and restored the appearance of gaps in the cellular network similar to the static cultures. These results demonstrate the importance of the increased collagen secretion by hepatocytes cultured under flow as a mechanism to maintain a well-connected cellular network and a differentiated function.
Collapse
Affiliation(s)
| | - Rohit Jindal
- corresponding authors: Dr. Rohit Jindal, Center for Engineering in Medicine (CEM), Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, 51 Blossom Street, Boston, MA 02114, . Dr. Martin L. Yarmush, Center for Engineering in Medicine (CEM), Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, 51 Blossom Street, Boston, MA 02114,
| | | | | | | | | | | | - Martin L. Yarmush
- corresponding authors: Dr. Rohit Jindal, Center for Engineering in Medicine (CEM), Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, 51 Blossom Street, Boston, MA 02114, . Dr. Martin L. Yarmush, Center for Engineering in Medicine (CEM), Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, 51 Blossom Street, Boston, MA 02114,
| |
Collapse
|
44
|
Highly robust protein production by co-culture of CHO spheroids layered on feeder cells in serum-free medium. Colloid Polym Sci 2014. [DOI: 10.1007/s00396-013-3093-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
45
|
Otsuka H, Sasaki K, Okimura S, Nagamura M, Watanabe R, Kawabe M. Contribution of Fibroblasts Cultured on 3D Silica Nonwoven Fabrics to Cocultured Hepatocytes Function. CHEM LETT 2014. [DOI: 10.1246/cl.130955] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Hidenori Otsuka
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science
- Department of Chemical Sciences and Technology, Graduate School of Chemical Science and Technology, Tokyo University of Science
| | - Kohei Sasaki
- Department of Chemical Sciences and Technology, Graduate School of Chemical Science and Technology, Tokyo University of Science
| | - Saya Okimura
- Department of Chemical Sciences and Technology, Graduate School of Chemical Science and Technology, Tokyo University of Science
| | - Masako Nagamura
- Department of Chemical Sciences and Technology, Graduate School of Chemical Science and Technology, Tokyo University of Science
| | | | | |
Collapse
|
46
|
Handa K, Matsubara K, Fukumitsu K, Guzman-Lepe J, Watson A, Soto-Gutierrez A. Assembly of human organs from stem cells to study liver disease. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:348-57. [PMID: 24333262 DOI: 10.1016/j.ajpath.2013.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/04/2013] [Accepted: 11/18/2013] [Indexed: 01/01/2023]
Abstract
Recently, significant developments in the field of liver tissue engineering have raised new possibilities for the study of complex physiological and pathophysiological processes in vitro, as well as the potential to assemble entire organs for transplantation. Human-induced pluripotent stem cells have been differentiated into relatively functional populations of hepatic cells, and novel techniques to generate whole organ acellular three-dimensional scaffolds have been developed. In this review, we highlight the most recent advances in organ assembly regarding the development of liver tissue in vitro. We emphasize applications that involve multiple types of cells with a biomimetic spatial organization for which three-dimensional configurations could be used for drug development or to explain mechanisms of disease. We also discuss applications of liver organotypic surrogates and the challenges of translating the highly promising new field of tissue engineering into a proven platform for predicting drug metabolism and toxicity.
Collapse
Affiliation(s)
- Kan Handa
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Transplantation Section, Children's Hospital of Pittsburgh, Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kentaro Matsubara
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Transplantation Section, Children's Hospital of Pittsburgh, Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ken Fukumitsu
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jorge Guzman-Lepe
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Transplantation Section, Children's Hospital of Pittsburgh, Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alicia Watson
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; Transplantation Section, Children's Hospital of Pittsburgh, Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
47
|
Larkin AL, Rodrigues RR, Murali TM, Rajagopalan P. Designing a multicellular organotypic 3D liver model with a detachable, nanoscale polymeric Space of Disse. Tissue Eng Part C Methods 2013; 19:875-84. [PMID: 23556413 DOI: 10.1089/ten.tec.2012.0700] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The design of in vitro models that mimic the stratified multicellular hepatic microenvironment continues to be challenging. Although several in vitro hepatic cultures have been shown to exhibit liver functions, their physiological relevance is limited due to significant deviation from in vivo cellular composition. We report the assembly of a novel three-dimensional (3D) organotypic liver model incorporating three different cell types (hepatocytes, liver sinusoidal endothelial cells, and Kupffer cells) and a polymeric interface that mimics the Space of Disse. The nanoscale interface is detachable, optically transparent, derived from self-assembled polyelectrolyte multilayers, and exhibits a Young's modulus similar to in vivo values for liver tissue. Only the 3D liver models simultaneously maintain hepatic phenotype and elicit proliferation, while achieving cellular ratios found in vivo. The nanoscale detachable polymeric interfaces can be modulated to mimic basement membranes that exhibit a wide range of physical properties. This facile approach offers a versatile new avenue in the assembly of engineered tissues. These results demonstrate the ability of the tri-cellular 3D cultures to serve as an organotypic hepatic model that elicits proliferation and maintenance of phenotype and in vivo-like cellular ratios.
Collapse
Affiliation(s)
- Adam L Larkin
- 1 Department of Chemical Engineering, Virginia Tech , Blacksburg, Virginia
| | | | | | | |
Collapse
|
48
|
Rosen MB, Das KP, Wood CR, Wolf CJ, Abbott BD, Lau C. Evaluation of perfluoroalkyl acid activity using primary mouse and human hepatocytes. Toxicology 2013; 308:129-37. [DOI: 10.1016/j.tox.2013.03.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 03/22/2013] [Accepted: 03/25/2013] [Indexed: 01/09/2023]
|
49
|
Tandon N, Marolt D, Cimetta E, Vunjak-Novakovic G. Bioreactor engineering of stem cell environments. Biotechnol Adv 2013; 31:1020-31. [PMID: 23531529 DOI: 10.1016/j.biotechadv.2013.03.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 12/02/2012] [Accepted: 03/11/2013] [Indexed: 12/31/2022]
Abstract
Stem cells hold promise to revolutionize modern medicine by the development of new therapies, disease models and drug screening systems. Standard cell culture systems have limited biological relevance because they do not recapitulate the complex 3-dimensional interactions and biophysical cues that characterize the in vivo environment. In this review, we discuss the current advances in engineering stem cell environments using novel biomaterials and bioreactor technologies. We also reflect on the challenges the field is currently facing with regard to the translation of stem cell based therapies into the clinic.
Collapse
Affiliation(s)
- Nina Tandon
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | | | | | | |
Collapse
|
50
|
Ukairo O, Kanchagar C, Moore A, Shi J, Gaffney J, Aoyama S, Rose K, Krzyzewski S, McGeehan J, Andersen ME, Khetani SR, LeCluyse EL. Long-Term Stability of Primary Rat Hepatocytes in Micropatterned Cocultures. J Biochem Mol Toxicol 2013; 27:204-12. [DOI: 10.1002/jbt.21469] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/17/2012] [Accepted: 11/19/2012] [Indexed: 11/12/2022]
Affiliation(s)
| | | | - Amanda Moore
- Hepregen Corporation, Suite 1500; Medford; MA; 2155; USA
| | - Julianne Shi
- Hepregen Corporation, Suite 1500; Medford; MA; 2155; USA
| | | | - Simon Aoyama
- Hepregen Corporation, Suite 1500; Medford; MA; 2155; USA
| | - Kelly Rose
- The Hamner Institutes for Health Sciences; Research Triangle Park; NC; 27709; USA
| | | | - Jack McGeehan
- Hepregen Corporation, Suite 1500; Medford; MA; 2155; USA
| | - Melvin E. Andersen
- The Hamner Institutes for Health Sciences; Research Triangle Park; NC; 27709; USA
| | - Salman R. Khetani
- Colorado State University; Mechanical and Biomedical Engineering; Fort Collins; CO; 80523-1374; USA
| | - Edward L. LeCluyse
- The Hamner Institutes for Health Sciences; Research Triangle Park; NC; 27709; USA
| |
Collapse
|