1
|
Anjum MR, Subramaniam V, Higgins BR, Abrahan C, Chisolm SJ, Krishnaprasad KA, Azie O, Palmer GD, Angelini TE, Sarntinoranont M. Determining Rates of Molecular Secretion from Supernatant Concentration Measurements in a 3D-Bioprinted Human Liver Tissue Model. ACS Biomater Sci Eng 2024; 10:6711-6720. [PMID: 39259932 DOI: 10.1021/acsbiomaterials.4c01086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
The secretion rate of albumin is a key indicator of function in liver tissue models used for hepatotoxicity and pharmacokinetic testing. However, it is not generally clear how to determine molecular secretion rates from measurements of the molecular concentration in supernatant media. Here, we develop computational and analytical models of molecular transport in an experimental system that enable determination of albumin secretion rates based on measurements of albumin concentration in supernatant media. The experimental system is a 3D-bioprinted human liver tissue construct embedded in a 3D culture environment made from packed microgel particles swollen in liquid growth media. The mathematical models reveal that the range of albumin synthesis rates necessary to match experimentally measured albumin concentrations corresponds to reaction-limited conditions, where a steady state of albumin spatial distribution is rapidly reached between media exchanges. Our results show that temporally resolved synthesis rates can be inferred from serial concentration measurements of collected supernatant media. This link is critical to confidently assessing in vitro tissue performance in applications where critical quality attributes must be quantified, like in drug development and screening.
Collapse
Affiliation(s)
- M Rasheed Anjum
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Vignesh Subramaniam
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Brett R Higgins
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Carolina Abrahan
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Florida, Gainesville, Florida 32611, United States
| | - Steven J Chisolm
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - K A Krishnaprasad
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Obiora Azie
- Otomagnetics, Inc., Bethesda, Maryland 20852, United States
| | - Glyn D Palmer
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Florida, Gainesville, Florida 32611, United States
| | - Thomas E Angelini
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
- Department of Materials Science and Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Malisa Sarntinoranont
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
2
|
Leal F, Zeiringer S, Jeitler R, Costa PF, Roblegg E. A comprehensive overview of advanced dynamic in vitro intestinal and hepatic cell culture models. Tissue Barriers 2024; 12:2163820. [PMID: 36680530 PMCID: PMC10832944 DOI: 10.1080/21688370.2022.2163820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/22/2022] [Indexed: 01/22/2023] Open
Abstract
Orally administered drugs pass through the gastrointestinal tract before being absorbed in the small intestine and metabolised in the liver. To test the efficacy and toxicity of drugs, animal models are often employed; however, they are not suitable for investigating drug-tissue interactions and making reliable predictions, since the human organism differs drastically from animals in terms of absorption, distribution, metabolism and excretion of substances. Likewise, simple static in vitro cell culture systems currently used in preclinical drug screening often do not resemble the native characteristics of biological barriers. Dynamic models, on the other hand, provide in vivo-like cell phenotypes and functionalities that offer great potential for safety and efficacy prediction. Herein, current microfluidic in vitro intestinal and hepatic models are reviewed, namely single- and multi-tissue micro-bioreactors, which are associated with different methods of cell cultivation, i.e., scaffold-based versus scaffold-free.
Collapse
Affiliation(s)
- Filipa Leal
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Scarlett Zeiringer
- Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Institute of Pharmaceutical Sciences, Universitaetsplatz 1, Graz, Austria
| | - Ramona Jeitler
- Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Institute of Pharmaceutical Sciences, Universitaetsplatz 1, Graz, Austria
| | - Pedro F. Costa
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Eva Roblegg
- Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Institute of Pharmaceutical Sciences, Universitaetsplatz 1, Graz, Austria
| |
Collapse
|
3
|
Mir TA, Alzhrani A, Nakamura M, Iwanaga S, Wani SI, Altuhami A, Kazmi S, Arai K, Shamma T, Obeid DA, Assiri AM, Broering DC. Whole Liver Derived Acellular Extracellular Matrix for Bioengineering of Liver Constructs: An Updated Review. Bioengineering (Basel) 2023; 10:1126. [PMID: 37892856 PMCID: PMC10604736 DOI: 10.3390/bioengineering10101126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 10/29/2023] Open
Abstract
Biomaterial templates play a critical role in establishing and bioinstructing three-dimensional cellular growth, proliferation and spatial morphogenetic processes that culminate in the development of physiologically relevant in vitro liver models. Various natural and synthetic polymeric biomaterials are currently available to construct biomimetic cell culture environments to investigate hepatic cell-matrix interactions, drug response assessment, toxicity, and disease mechanisms. One specific class of natural biomaterials consists of the decellularized liver extracellular matrix (dECM) derived from xenogeneic or allogeneic sources, which is rich in bioconstituents essential for the ultrastructural stability, function, repair, and regeneration of tissues/organs. Considering the significance of the key design blueprints of organ-specific acellular substrates for physiologically active graft reconstruction, herein we showcased the latest updates in the field of liver decellularization-recellularization technologies. Overall, this review highlights the potential of acellular matrix as a promising biomaterial in light of recent advances in the preparation of liver-specific whole organ scaffolds. The review concludes with a discussion of the challenges and future prospects of liver-specific decellularized materials in the direction of translational research.
Collapse
Affiliation(s)
- Tanveer Ahmed Mir
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Alaa Alzhrani
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21423, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Makoto Nakamura
- Division of Biomedical System Engineering, Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan; (M.N.); (S.I.)
| | - Shintaroh Iwanaga
- Division of Biomedical System Engineering, Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan; (M.N.); (S.I.)
| | - Shadil Ibrahim Wani
- Division of Biomedical System Engineering, Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan; (M.N.); (S.I.)
| | - Abdullah Altuhami
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Shadab Kazmi
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- Department of Child Health, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Kenchi Arai
- Department of Clinical Biomaterial Applied Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Talal Shamma
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Dalia A. Obeid
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Abdullah M. Assiri
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Dieter C. Broering
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| |
Collapse
|
4
|
Grebenyuk S, Abdel Fattah AR, Kumar M, Toprakhisar B, Rustandi G, Vananroye A, Salmon I, Verfaillie C, Grillo M, Ranga A. Large-scale perfused tissues via synthetic 3D soft microfluidics. Nat Commun 2023; 14:193. [PMID: 36635264 PMCID: PMC9837048 DOI: 10.1038/s41467-022-35619-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/13/2022] [Indexed: 01/14/2023] Open
Abstract
The vascularization of engineered tissues and organoids has remained a major unresolved challenge in regenerative medicine. While multiple approaches have been developed to vascularize in vitro tissues, it has thus far not been possible to generate sufficiently dense networks of small-scale vessels to perfuse large de novo tissues. Here, we achieve the perfusion of multi-mm3 tissue constructs by generating networks of synthetic capillary-scale 3D vessels. Our 3D soft microfluidic strategy is uniquely enabled by a 3D-printable 2-photon-polymerizable hydrogel formulation, which allows for precise microvessel printing at scales below the diffusion limit of living tissues. We demonstrate that these large-scale engineered tissues are viable, proliferative and exhibit complex morphogenesis during long-term in-vitro culture, while avoiding hypoxia and necrosis. We show by scRNAseq and immunohistochemistry that neural differentiation is significantly accelerated in perfused neural constructs. Additionally, we illustrate the versatility of this platform by demonstrating long-term perfusion of developing neural and liver tissue. This fully synthetic vascularization platform opens the door to the generation of human tissue models at unprecedented scale and complexity.
Collapse
Affiliation(s)
- Sergei Grebenyuk
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.
| | - Abdel Rahman Abdel Fattah
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Manoj Kumar
- Stem Cell Institute Leuven and Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Burak Toprakhisar
- Stem Cell Institute Leuven and Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Gregorius Rustandi
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Anja Vananroye
- Laboratory of Soft Matter, Rheology and Technology, Department of Chemical Engineering, KU Leuven, Leuven, Belgium
| | - Idris Salmon
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Catherine Verfaillie
- Stem Cell Institute Leuven and Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Mark Grillo
- Grillo Consulting Inc., San Francisco, CA, USA
| | - Adrian Ranga
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.
- Leuven Brain Institute, KU Leuven, Leuven, Belgium.
- Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium.
| |
Collapse
|
5
|
Dupard SJ, Garcia AG, Bourgine PE. Customizable 3D printed perfusion bioreactor for the engineering of stem cell microenvironments. Front Bioeng Biotechnol 2023; 10:1081145. [PMID: 36698631 PMCID: PMC9870251 DOI: 10.3389/fbioe.2022.1081145] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Faithful modeling of tissues and organs requires the development of systems reflecting their dynamic 3D cellular architecture and organization. Current technologies suffer from a lack of design flexibility and complex prototyping, preventing their broad adoption by the scientific community. To make 3D cell culture more available and adaptable we here describe the use of the fused deposition modeling (FDM) technology to rapid-prototype 3D printed perfusion bioreactors. Our 3D printed bioreactors are made of polylactic acid resulting in reusable systems customizable in size and shape. Following design confirmation, our bioreactors were biologically validated for the culture of human mesenchymal stromal cells under perfusion for up to 2 weeks on collagen scaffolds. Microenvironments of various size/volume (6-12 mm in diameter) could be engineered, by modulating the 3D printed bioreactor design. Metabolic assay and confocal microscopy confirmed the homogenous mesenchymal cell distribution throughout the material pores. The resulting human microenvironments were further exploited for the maintenance of human hematopoietic stem cells. Following 1 week of stromal coculture, we report the recapitulation of 3D interactions between the mesenchymal and hematopoietic fractions, associated with a phenotypic expansion of the blood stem cell populations.Our data confirm that perfusion bioreactors fit for cell culture can be generated using a 3D printing technology and exploited for the 3D modeling of complex stem cell systems. Our approach opens the gates for a more faithful investigation of cellular processes in relation to a dynamic 3D microenvironment.
Collapse
Affiliation(s)
- Steven J. Dupard
- Cell, Tissue and Organ engineering laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Alejandro Garcia Garcia
- Cell, Tissue and Organ engineering laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Paul E. Bourgine
- Cell, Tissue and Organ engineering laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Poornima K, Francis AP, Hoda M, Eladl MA, Subramanian S, Veeraraghavan VP, El-Sherbiny M, Asseri SM, Hussamuldin ABA, Surapaneni KM, Mony U, Rajagopalan R. Implications of Three-Dimensional Cell Culture in Cancer Therapeutic Research. Front Oncol 2022; 12:891673. [PMID: 35646714 PMCID: PMC9133474 DOI: 10.3389/fonc.2022.891673] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Replicating the naturalistic biomechanical milieu of cells is a primary requisite to uncover the fundamental life processes. The native milieu is significantly not replicated in the two-dimensional (2D) cell cultures. Alternatively, the current three-dimensional (3D) culture techniques can replicate the properties of extracellular matrix (ECM), though the recreation of the original microenvironment is challenging. The organization of cells in a 3D manner contributes to better insight about the tumorigenesis mechanism of the in vitro cancer models. Gene expression studies are susceptible to alterations in their microenvironment. Physiological interactions among neighboring cells also contribute to gene expression, which is highly replicable with minor modifications in 3D cultures. 3D cell culture provides a useful platform for identifying the biological characteristics of tumor cells, particularly in the drug sensitivity area of translational medicine. It promises to be a bridge between traditional 2D culture and animal experiments and is of great importance for further research in tumor biology. The new imaging technology and the implementation of standard protocols can address the barriers interfering with the live cell observation in a natural 3D physiological environment.
Collapse
Affiliation(s)
- Kolluri Poornima
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - Arul Prakash Francis
- Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Muddasarul Hoda
- Department of Biological Sciences, Aliah University, Kolkata, India
| | - Mohamed Ahmed Eladl
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Srividya Subramanian
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Saad Mohamed Asseri
- Department of Clinical Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | | | - Krishna Mohan Surapaneni
- Departments of Biochemistry, Molecular Virology, Research, Clinical Skills, and Simulation, Panimalar Medical College Hospital and Research Institute, Chennai, India
| | - Ullas Mony
- Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Rukkumani Rajagopalan
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India
| |
Collapse
|
7
|
Kanabekova P, Kadyrova A, Kulsharova G. Microfluidic Organ-on-a-Chip Devices for Liver Disease Modeling In Vitro. MICROMACHINES 2022; 13:428. [PMID: 35334720 PMCID: PMC8950395 DOI: 10.3390/mi13030428] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
Mortality from liver disease conditions continues to be very high. As liver diseases manifest and progress silently, prompt measures after diagnosis are essential in the treatment of these conditions. Microfluidic organs-on-chip platforms have significant potential for the study of the pathophysiology of liver diseases in vitro. Different liver-on-a-chip microphysiological platforms have been reported to study cell-signaling pathways such as those activating stellate cells within liver diseases. Moreover, the drug efficacy for liver conditions might be evaluated on a cellular metabolic level. Here, we present a comprehensive review of microphysiological platforms used for modelling liver diseases. First, we briefly introduce the concept and importance of organs-on-a-chip in studying liver diseases in vitro, reflecting on existing reviews of healthy liver-on-a-chip platforms. Second, the techniques of cell cultures used in the microfluidic devices, including 2D, 3D, and spheroid cells, are explained. Next, the types of liver diseases (NAFLD, ALD, hepatitis infections, and drug injury) on-chip are explained for a further comprehensive overview of the design and methods of developing liver diseases in vitro. Finally, some challenges in design and existing solutions to them are reviewed.
Collapse
Affiliation(s)
- Perizat Kanabekova
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Adina Kadyrova
- Department of Biological Sciences, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Gulsim Kulsharova
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| |
Collapse
|
8
|
Sasikumar S, Chameettachal S, Kingshott P, Cromer B, Pati F. Influence of Liver Extracellular Matrix in Predicting Drug-Induced Liver Injury: An Alternate Paradigm. ACS Biomater Sci Eng 2022; 8:834-846. [PMID: 34978414 DOI: 10.1021/acsbiomaterials.1c00994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In vitro drug-induced liver injury (DILI) models are promising tools for drug development to predict adverse events during clinical usage. However, the currently available DILI models are not specific or not able to predict the injury accurately. This is believed to be mainly because of failure to conserve the hepatocyte phenotype, lack of longevity, and difficulty in maintaining the tissue-specific microenvironment. In this study, we have assessed the potential of decellularized liver extracellular matrix (DLM) in retaining the hepatic cellular phenotype and functionality in the presence of a tissue-specific microenvironment along with its role in influencing the effect of the drug on hepatic cells. We show that DLM helps maintain the phenotype of the hepatic cell line HepG2, a well-known cell line for secretion of human proteins that is easily available. Also, the DLM enhanced the expression of a metabolic marker carbamoyl phosphate synthetase I (CPS1), a regulator of urea cycle, and bile salt export pump (BSEP), a marker of hepatocyte polarity. We further validated the DLM for its influence on the sensitivity of cells toward different classes of drugs. Interestingly, the coculture model, in the presence of endothelial cells and stellate cells, exhibited a higher sensitivity for both acetaminophen and trovafloxacin, a toxic compound that does not show any toxicity on preclinical screening. Thus, our results demonstrate for the first time that a multicellular combination along with DLM can be a potential and reliable DILI model to screen multiple drugs.
Collapse
Affiliation(s)
- Shyama Sasikumar
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India.,Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Shibu Chameettachal
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Peter Kingshott
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia.,ARC Training Centre Training Centre in Surface Engineering for Advanced Materials (SEAM), School of Engineering, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Brett Cromer
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| |
Collapse
|
9
|
Fernandez-Checa JC, Bagnaninchi P, Ye H, Sancho-Bru P, Falcon-Perez JM, Royo F, Garcia-Ruiz C, Konu O, Miranda J, Lunov O, Dejneka A, Elfick A, McDonald A, Sullivan GJ, Aithal GP, Lucena MI, Andrade RJ, Fromenty B, Kranendonk M, Cubero FJ, Nelson LJ. Advanced preclinical models for evaluation of drug-induced liver injury - consensus statement by the European Drug-Induced Liver Injury Network [PRO-EURO-DILI-NET]. J Hepatol 2021; 75:935-959. [PMID: 34171436 DOI: 10.1016/j.jhep.2021.06.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023]
Abstract
Drug-induced liver injury (DILI) is a major cause of acute liver failure (ALF) and one of the leading indications for liver transplantation in Western societies. Given the wide use of both prescribed and over the counter drugs, DILI has become a major health issue for which there is a pressing need to find novel and effective therapies. Although significant progress has been made in understanding the molecular mechanisms underlying DILI, our incomplete knowledge of its pathogenesis and inability to predict DILI is largely due to both discordance between human and animal DILI in preclinical drug development and a lack of models that faithfully recapitulate complex pathophysiological features of human DILI. This is exemplified by the hepatotoxicity of acetaminophen (APAP) overdose, a major cause of ALF because of its extensive worldwide use as an analgesic. Despite intensive efforts utilising current animal and in vitro models, the mechanisms involved in the hepatotoxicity of APAP are still not fully understood. In this expert Consensus Statement, which is endorsed by the European Drug-Induced Liver Injury Network, we aim to facilitate and outline clinically impactful discoveries by detailing the requirements for more realistic human-based systems to assess hepatotoxicity and guide future drug safety testing. We present novel insights and discuss major players in APAP pathophysiology, and describe emerging in vitro and in vivo pre-clinical models, as well as advanced imaging and in silico technologies, which may improve prediction of clinical outcomes of DILI.
Collapse
Affiliation(s)
- Jose C Fernandez-Checa
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Consejo Superior Investigaciones Científicas (CSIC), Spain; Liver Unit, Hospital Clínic, Barcelona, Spain; Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; USC Research Center for ALPD, Keck School of Medicine, Los Angeles, United States, CA 90033.
| | - Pierre Bagnaninchi
- Center for Regenerative Medicine, Institute for Regenerative and Repair, The University of Edinburgh, Edinburgh, UK, EH16 4UU; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Faraday Building, Colin Maclaurin Road, EH9 3 DW, Scotland, UK
| | - Hui Ye
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Pau Sancho-Bru
- Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Juan M Falcon-Perez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, 48160, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia, 48015, Spain
| | - Felix Royo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, 48160, Spain
| | - Carmen Garcia-Ruiz
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Consejo Superior Investigaciones Científicas (CSIC), Spain; Liver Unit, Hospital Clínic, Barcelona, Spain; Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; USC Research Center for ALPD, Keck School of Medicine, Los Angeles, United States, CA 90033
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey; Interdisciplinary Neuroscience Program, Bilkent University, Ankara, Turkey; UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Joana Miranda
- Research Institute for iMedicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alistair Elfick
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh EH8 3DW, UK
| | - Alison McDonald
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh EH8 3DW, UK
| | - Gareth J Sullivan
- University of Oslo and the Oslo University Hospital, Oslo, Norway; Hybrid Technology Hub-Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Pediatric Research, Oslo University Hosptial, Oslo, Norway
| | - Guruprasad P Aithal
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospital NHS Trust and University of Nottingham, Nottingham, UK
| | - M Isabel Lucena
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, UICEC SCReN, Universidad de Málaga, Málaga, Spain
| | - Raul J Andrade
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Unidad de Gestión Clínica de Enfermedades Digestivas, Instituto de Investigación, Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Malaga, Spain
| | - Bernard Fromenty
- INSERM, Univ Rennes, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, F-35000 Rennes, France
| | - Michel Kranendonk
- Center for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculty of Medical Sciences, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Francisco Javier Cubero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Leonard J Nelson
- Center for Regenerative Medicine, Institute for Regenerative and Repair, The University of Edinburgh, Edinburgh, UK, EH16 4UU; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Faraday Building, Colin Maclaurin Road, EH9 3 DW, Scotland, UK; Institute of Biological Chemistry, Biophysics and Bioengineering (IB3), School of Engineering and Physical Sciences (EPS), Heriot-Watt University, Edinburgh EH12 2AS, Scotland, UK.
| |
Collapse
|
10
|
Kulsharova G, Kurmangaliyeva A. Liver microphysiological platforms for drug metabolism applications. Cell Prolif 2021; 54:e13099. [PMID: 34291515 PMCID: PMC8450120 DOI: 10.1111/cpr.13099] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/21/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022] Open
Abstract
Drug development is a costly and lengthy process with low success rates. To improve the efficiency of drug development, there has been an increasing need in developing alternative methods able to eliminate toxic compounds early in the drug development pipeline. Drug metabolism plays a key role in determining the efficacy of a drug and its potential side effects. Since drug metabolism occurs mainly in the liver, liver cell‐based alternative engineering platforms have been growing in the last decade. Microphysiological liver cell‐based systems called liver‐on‐a‐chip platforms can better recapitulate the environment for human liver cells in laboratory settings and have the potential to reduce the number of animal models used in drug development by predicting the response of the liver to a drug in vitro. In this review, we discuss the liver microphysiological platforms from the perspective of drug metabolism studies. We highlight the stand‐alone liver‐on‐a‐chip platforms and multi‐organ systems integrating liver‐on‐a‐chip devices used for drug metabolism mimicry in vitro and review the state‐of‐the‐art platforms reported in the last few years. With the development of more robust and reproducible liver cell‐based microphysiological platforms, the drug development field has the potential of reducing the costs and lengths associated with currently existing drug testing methods.
Collapse
Affiliation(s)
- Gulsim Kulsharova
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan, Kazakhstan
| | | |
Collapse
|
11
|
Yadav J, El Hassani M, Sodhi J, Lauschke VM, Hartman JH, Russell LE. Recent developments in in vitro and in vivo models for improved translation of preclinical pharmacokinetics and pharmacodynamics data. Drug Metab Rev 2021; 53:207-233. [PMID: 33989099 DOI: 10.1080/03602532.2021.1922435] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Improved pharmacokinetics/pharmacodynamics (PK/PD) prediction in the early stages of drug development is essential to inform lead optimization strategies and reduce attrition rates. Recently, there have been significant advancements in the development of new in vitro and in vivo strategies to better characterize pharmacokinetic properties and efficacy of drug leads. Herein, we review advances in experimental and mathematical models for clearance predictions, advancements in developing novel tools to capture slowly metabolized drugs, in vivo model developments to capture human etiology for supporting drug development, limitations and gaps in these efforts, and a perspective on the future in the field.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Inc., Boston, MA, USA
| | | | - Jasleen Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jessica H Hartman
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
12
|
Lee-Montiel FT, Laemmle A, Charwat V, Dumont L, Lee CS, Huebsch N, Okochi H, Hancock MJ, Siemons B, Boggess SC, Goswami I, Miller EW, Willenbring H, Healy KE. Integrated Isogenic Human Induced Pluripotent Stem Cell-Based Liver and Heart Microphysiological Systems Predict Unsafe Drug-Drug Interaction. Front Pharmacol 2021; 12:667010. [PMID: 34025426 PMCID: PMC8138446 DOI: 10.3389/fphar.2021.667010] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
Three-dimensional (3D) microphysiological systems (MPSs) mimicking human organ function in vitro are an emerging alternative to conventional monolayer cell culture and animal models for drug development. Human induced pluripotent stem cells (hiPSCs) have the potential to capture the diversity of human genetics and provide an unlimited supply of cells. Combining hiPSCs with microfluidics technology in MPSs offers new perspectives for drug development. Here, the integration of a newly developed liver MPS with a cardiac MPS—both created with the same hiPSC line—to study drug–drug interaction (DDI) is reported. As a prominent example of clinically relevant DDI, the interaction of the arrhythmogenic gastroprokinetic cisapride with the fungicide ketoconazole was investigated. As seen in patients, metabolic conversion of cisapride to non-arrhythmogenic norcisapride in the liver MPS by the cytochrome P450 enzyme CYP3A4 was inhibited by ketoconazole, leading to arrhythmia in the cardiac MPS. These results establish integration of hiPSC-based liver and cardiac MPSs to facilitate screening for DDI, and thus drug efficacy and toxicity, isogenic in the same genetic background.
Collapse
Affiliation(s)
- Felipe T Lee-Montiel
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Alexander Laemmle
- Department of Surgery, Division of Transplant Surgery, Liver Center and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States.,Institute of Clinical Chemistry and Department of Pediatrics, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Verena Charwat
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Laure Dumont
- Department of Surgery, Division of Transplant Surgery, Liver Center and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States
| | - Caleb S Lee
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Nathaniel Huebsch
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Hideaki Okochi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, United States
| | | | - Brian Siemons
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Steven C Boggess
- Department of Chemistry, University of California Berkeley, Berkeley, CA, United States
| | - Ishan Goswami
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Evan W Miller
- Departments of Chemistry and Molecular & Cell Biology, and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, United States
| | - Holger Willenbring
- Department of Surgery, Division of Transplant Surgery, Liver Center and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States
| | - Kevin E Healy
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
13
|
Li W, Li P, Li N, Du Y, Lü S, Elad D, Long M. Matrix stiffness and shear stresses modulate hepatocyte functions in a fibrotic liver sinusoidal model. Am J Physiol Gastrointest Liver Physiol 2021; 320:G272-G282. [PMID: 33296275 PMCID: PMC8609567 DOI: 10.1152/ajpgi.00379.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Extracellular matrix (ECM) rigidity has important effects on cell behaviors and increases sharply in liver fibrosis and cirrhosis. Hepatic blood flow is essential in maintaining hepatocytes' (HCs) functions. However, it is still unclear how matrix stiffness and shear stresses orchestrate HC phenotype in concert. A fibrotic three-dimensional (3-D) liver sinusoidal model is constructed using a porous membrane sandwiched between two polydimethylsiloxane (PDMS) layers with respective flow channels. The HCs are cultured in collagen gels of various stiffnesses in the lower channel, whereas the upper channel is pre-seeded with liver sinusoidal endothelial cells (LSECs) and accessible to shear flow. The results reveal that HCs cultured within stiffer matrices exhibit reduced albumin production and cytochrome P450 (CYP450) reductase expression. Low shear stresses enhance synthetic and metabolic functions of HC, whereas high shear stresses lead to the loss of HC phenotype. Furthermore, both mechanical factors regulate HC functions by complementing each other. These observations are likely attributed to mechanically induced mass transport or key signaling molecule of hepatocyte nuclear factor 4α (HNF4α). The present study results provide an insight into understanding the mechanisms of HC dysfunction in liver fibrosis and cirrhosis, especially from the viewpoint of matrix stiffness and blood flow.NEW & NOTEWORTHY A fibrotic three-dimensional (3-D) liver sinusoidal model was constructed to mimic different stages of liver fibrosis in vivo and to explore the cooperative effects of matrix stiffness and shear stresses on hepatocyte (HC) functions. Mechanically induced alterations of mass transport mainly contributed to HC functions via typical mechanosensitive signaling.
Collapse
Affiliation(s)
- Wang Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Peiwen Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Ning Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yu Du
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Shouqin Lü
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - David Elad
- 5Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Mian Long
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
14
|
Serras AS, Rodrigues JS, Cipriano M, Rodrigues AV, Oliveira NG, Miranda JP. A Critical Perspective on 3D Liver Models for Drug Metabolism and Toxicology Studies. Front Cell Dev Biol 2021; 9:626805. [PMID: 33732695 PMCID: PMC7957963 DOI: 10.3389/fcell.2021.626805] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
The poor predictability of human liver toxicity is still causing high attrition rates of drug candidates in the pharmaceutical industry at the non-clinical, clinical, and post-marketing authorization stages. This is in part caused by animal models that fail to predict various human adverse drug reactions (ADRs), resulting in undetected hepatotoxicity at the non-clinical phase of drug development. In an effort to increase the prediction of human hepatotoxicity, different approaches to enhance the physiological relevance of hepatic in vitro systems are being pursued. Three-dimensional (3D) or microfluidic technologies allow to better recapitulate hepatocyte organization and cell-matrix contacts, to include additional cell types, to incorporate fluid flow and to create gradients of oxygen and nutrients, which have led to improved differentiated cell phenotype and functionality. This comprehensive review addresses the drug-induced hepatotoxicity mechanisms and the currently available 3D liver in vitro models, their characteristics, as well as their advantages and limitations for human hepatotoxicity assessment. In addition, since toxic responses are greatly dependent on the culture model, a comparative analysis of the toxicity studies performed using two-dimensional (2D) and 3D in vitro strategies with recognized hepatotoxic compounds, such as paracetamol, diclofenac, and troglitazone is performed, further highlighting the need for harmonization of the respective characterization methods. Finally, taking a step forward, we propose a roadmap for the assessment of drugs hepatotoxicity based on fully characterized fit-for-purpose in vitro models, taking advantage of the best of each model, which will ultimately contribute to more informed decision-making in the drug development and risk assessment fields.
Collapse
Affiliation(s)
- Ana S. Serras
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Madalena Cipriano
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Armanda V. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Nuno G. Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
15
|
Son J, Kim HH, Lee JH, Jeong WI, Park JK. Assembly and Disassembly of the Micropatterned Collagen Sheets Containing Cells for Location-Based Cellular Function Analysis. BIOCHIP JOURNAL 2021. [DOI: 10.1007/s13206-021-00007-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Ballard TE, Kratochwil N, Cox LM, Moen MA, Klammers F, Ekiciler A, Goetschi A, Walter I. Simplifying the Execution of HepatoPac MetID Experiments: Metabolite Profile and Intrinsic Clearance Comparisons. Drug Metab Dispos 2020; 48:804-810. [PMID: 32623369 DOI: 10.1124/dmd.120.000013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/12/2020] [Indexed: 02/02/2023] Open
Abstract
The HepatoPac micropatterned coculture (MPCC) hepatocyte system has been shown to be an effective tool to investigate the qualitative human and preclinical species' metabolite profiles of new drug candidates. However, additional improvements to the overall study conditions and execution, layout, and human-donor count could be made. To that end, we have evaluated several ways to increase the amount of data one can generate per MPCC plate and how to more efficiently execute a MPCC study for the purpose of metabolite generation. Herein, we compare a set of compounds using single- and 10-donor pooled human MPCC hepatocytes. Intrinsic clearance and mean metabolic activities assessed by diverse enzyme markers were comparable between the single- and 10-donor pool. We have confirmed that the generated metabolite profiles were indistinguishable between the single- and 10-donor pool and also that rat MPCC can be performed at 400 µl media volume, which greatly simplifies study execution. Additional tips for successful study execution are also described. SIGNIFICANCE STATEMENT: When using the HepatoPac micropatterned coculture (MPCC) system, sometimes simple experimental condition variables or problematic plate designs can hamper productive study execution. We evaluated conditions to increase the amount of data one can generate per MPCC plate and, perhaps more importantly, execute that study more efficiently with less likelihood of error. We describe some of our key learnings, provide an examination of enzyme activity levels and clearance values, and provide some recommendations to simplify the execution of a HepatoPac experiment.
Collapse
Affiliation(s)
- T Eric Ballard
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - N Kratochwil
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - Loretta M Cox
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - Mark A Moen
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - F Klammers
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - A Ekiciler
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - A Goetschi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - I Walter
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| |
Collapse
|
17
|
Nudischer R, Renggli K, Hierlemann A, Roth AB, Bertinetti-Lapatki C. Characterization of a long-term mouse primary liver 3D tissue model recapitulating innate-immune responses and drug-induced liver toxicity. PLoS One 2020; 15:e0235745. [PMID: 32645073 PMCID: PMC7347206 DOI: 10.1371/journal.pone.0235745] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional liver in vitro systems have recently attracted a lot of attention in drug development. These systems help to gain unprecedented insights into drug-induced liver injury (DILI), as they more closely reproduce liver biology, and as drug effects can be studied in isolated and controllable microenvironments. Many groups established human-based in vitro models but so far neglected the animal equivalent, although the availability of both models would be desirable. Animal in vitro models enable back- and forward translation of in vitro and in vivo findings, bridge the gap between rodent in vivo and human in vitro scenarios, and ultimately support the interpretation of data generated with preclinical species and humans. Since mice are often used in drug development and physiologically relevant in vitro systems are lacking, we established, for the first time, a mouse liver model that encompasses primary parenchymal and non-parenchymal cells with preserved viability and functionality over three weeks. Using our three-dimensional liver spheroids, we were able to predict the toxicity of known DILI compounds, demonstrated the interaction cascades between the different cell types and showed evidence of drug-induced steatosis and cholestasis. In summary, our mouse liver spheroids represent a valuable in vitro model that can be applied to study DILI findings, reported from mouse studies, and offers the potential to detect immune-mediated drug-induced liver toxicity.
Collapse
Affiliation(s)
- Ramona Nudischer
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
- * E-mail:
| | - Kasper Renggli
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Zurich, Switzerland
| | - Andreas Hierlemann
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Zurich, Switzerland
| | - Adrian B. Roth
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Cristina Bertinetti-Lapatki
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
18
|
Self-Organized Liver Microtissue on a Bio-Functional Surface: The Role of Human Adipose-Derived Stromal Cells in Hepatic Function. Int J Mol Sci 2020; 21:ijms21134605. [PMID: 32610471 PMCID: PMC7369942 DOI: 10.3390/ijms21134605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 01/02/2023] Open
Abstract
The maintenance of hepatocyte function is a critical research topic in liver tissue engineering. Although an increasing number of strategies have been developed, liver tissue engineering using hepatocytes as a therapeutic alternative remains challenging owing to its poor efficacy. In this study, we developed a multicellular hepatic microtissue to enhance the function of induced hepatic precursor cells. Mouse induced hepatic precursor cells (miHeps) were self-organized in 3D with human adipose-derived stem cells (hASCs) on a bio-functional matrix. We found that hepatic phenotypes, such as levels of albumin, asialoglycoprotein receptor-1, and cytochrome P450, were enhanced in miHeps-hASC microtissue comprising miHeps and hASCs relative to two-dimensional-cultured miHeps-hASCs. Additionally, the secretome of 3D-cultured hASCs increased the hepatic function of mature miHeps. Furthermore, hepatic gene expression was reduced in mature miHeps treated with conditioned media of hypoxia-inducible factor 1α (HIF1α)-depleted hASCs relative to that with conditioned media of control hASCs. Our results suggested that the hepatic function of 3D-co-cultured miHeps could be enhanced by HIF1α-dependent factors secreted from stromal cells. This study provides an insight into the factors regulating hepatic function and shows that self-organized hepatic microtissue could act as liver spheroids for liver regenerative medicine and liver toxicity tests.
Collapse
|
19
|
Agarwal T, Borrelli MR, Makvandi P, Ashrafizadeh M, Maiti TK. Paper-Based Cell Culture: Paving the Pathway for Liver Tissue Model Development on a Cellulose Paper Chip. ACS APPLIED BIO MATERIALS 2020; 3:3956-3974. [DOI: 10.1021/acsabm.0c00558] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Mimi R. Borrelli
- Department of Surgery, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Pooyan Makvandi
- Institute for Polymers, Composites and Biomaterials (IPCB), National Research Council (CNR), Naples 80078, Italy
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 51666-16471, Iran
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| |
Collapse
|
20
|
Salerno S, Tasselli F, Drioli E, De Bartolo L. Poly(ε-Caprolactone) Hollow Fiber Membranes for the Biofabrication of a Vascularized Human Liver Tissue. MEMBRANES 2020; 10:E112. [PMID: 32471264 PMCID: PMC7344547 DOI: 10.3390/membranes10060112] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/23/2020] [Accepted: 05/26/2020] [Indexed: 01/01/2023]
Abstract
The creation of a liver tissue that recapitulates the micro-architecture and functional complexity of a human organ is still one of the main challenges of liver tissue engineering. Here we report on the development of a 3D vascularized hepatic tissue based on biodegradable hollow fiber (HF) membranes of poly(ε-caprolactone) (PCL) that compartmentalize human hepatocytes on the external surface and between the fibers, and endothelial cells into the fiber lumen. To this purpose, PCL HF membranes were prepared by a dry-jet wet phase inversion spinning technique tailoring the operational parameters in order to obtain fibers with suitable properties. After characterization, the fibers were applied to generate a human vascularized hepatic unit by loading endothelial cells in their inner surface and hepatocytes on the external surface. The unit was connected to a perfusion system, and the morpho-functional behavior was evaluated. The results demonstrated the large integration of endothelial cells with the internal surface of individual PCL fibers forming vascular-like structures, and hepatocytes covered completely the external surface and the space between fibers. The perfused 3D hepatic unit retained its functional activity at high levels up to 18 days. This bottom-up tissue engineering approach represents a rational strategy to create relatively 3D vascularized tissues and organs.
Collapse
Affiliation(s)
- Simona Salerno
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
| | - Franco Tasselli
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
| | - Enrico Drioli
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
- Department of Energy, Engineering, Hanyang University, Seoul 04763, Korea
| | - Loredana De Bartolo
- CNR-ITM, Institute on Membrane Technology, National Research Council of Italy, via P. Bucci, cubo 17/C, I-87036 Rende, Italy; (S.S.); (F.T.); (E.D.)
| |
Collapse
|
21
|
Bell CC, Chouhan B, Andersson LC, Andersson H, Dear JW, Williams DP, Söderberg M. Functionality of primary hepatic non-parenchymal cells in a 3D spheroid model and contribution to acetaminophen hepatotoxicity. Arch Toxicol 2020; 94:1251-1263. [PMID: 32112222 PMCID: PMC7225187 DOI: 10.1007/s00204-020-02682-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/20/2020] [Indexed: 12/12/2022]
Abstract
In addition to hepatocytes, the liver comprises a host of specialised non-parenchymal cells which are important to consider in the development of in vitro models which are both physiologically and toxicologically relevant. We have characterized a 3D co-culture system comprising primary human hepatocytes (PHH) and non-parenchymal cells (NPC) and applied it to the investigation of acetaminophen-induced toxicity. Firstly, we titrated ratios of PHH:NPC and confirmed the presence of functional NPCs via both immunohistochemistry and activation with both LPS and TGF-β. Based on these data we selected a ratio of 2:1 PHH:NPC for further studies. We observed that spheroids supplemented with NPCs were protected against acetaminophen (APAP) toxicity as determined by ATP (up to threefold difference in EC50 at day 14 compared to hepatocytes alone) and glutathione depletion, as well as miR-122 release. APAP metabolism was also altered in the presence of NPCs, with significantly lower levels of APAP-GSH detected. Expression of several CYP450 enzymes involved in the bioactivation of APAP was also lower in NPC-containing spheroids. Spheroids containing NPCs also expressed higher levels of miRNAs which have been implicated in APAP-induced hepatotoxicity, including miR-382 and miR-155 which have potential roles in liver regeneration and inflammation, respectively. These data indicate that the interaction between hepatocytes and NPCs can have significant metabolic and toxicological consequences important for the correct elucidation of hepatic safety mechanisms.
Collapse
Affiliation(s)
- Catherine C Bell
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Bhavik Chouhan
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Linda C Andersson
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Håkan Andersson
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - James W Dear
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Dominic P Williams
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Magnus Söderberg
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
22
|
Abstract
High-throughput in vitro models lack human-relevant complexity, which undermines their ability to accurately mimic in vivo biologic and pathologic responses. The emergence of microphysiological systems (MPS) presents an opportunity to revolutionize in vitro modeling for both basic biomedical research and applied drug discovery. The MPS platform has been an area of interdisciplinary collaboration to develop new, predictive, and reliable in vitro methods for regulatory acceptance. The current MPS models have been developed to recapitulate an organ or tissue on a smaller scale. However, the complexity of these models (ie, including all cell types present in the in vivo tissue) with appropriate structural, functional, and biochemical attributes are often not fully characterized. Here, we provide an overview of the capabilities and limitations of the microfluidic MPS model (aka organs-on-chips) within the scope of drug development. We recommend the engagement of pathologists early in the MPS design, characterization, and validation phases, because this will enable development of more robust and comprehensive MPS models that can accurately replicate normal biology and pathophysiology and hence be more predictive of human responses.
Collapse
Affiliation(s)
| | - Terry Van Vleet
- Global Preclinical Safety, AbbVie Inc, North Chicago, IL, USA
| | - Brian R Berridge
- National Toxicology Program, The National Institute of Environmental Health Sciences, Durham, NC, USA
| |
Collapse
|
23
|
Hosseini V, Maroufi NF, Saghati S, Asadi N, Darabi M, Ahmad SNS, Hosseinkhani H, Rahbarghazi R. Current progress in hepatic tissue regeneration by tissue engineering. J Transl Med 2019; 17:383. [PMID: 31752920 PMCID: PMC6873477 DOI: 10.1186/s12967-019-02137-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Liver, as a vital organ, is responsible for a wide range of biological functions to maintain homeostasis and any type of damages to hepatic tissue contributes to disease progression and death. Viral infection, trauma, carcinoma, alcohol misuse and inborn errors of metabolism are common causes of liver diseases are a severe known reason for leading to end-stage liver disease or liver failure. In either way, liver transplantation is the only treatment option which is, however, hampered by the increasing scarcity of organ donor. Over the past years, considerable efforts have been directed toward liver regeneration aiming at developing new approaches and methodologies to enhance the transplantation process. These approaches include producing decellularized scaffolds from the liver organ, 3D bio-printing system, and nano-based 3D scaffolds to simulate the native liver microenvironment. The application of small molecules and micro-RNAs and genetic manipulation in favor of hepatic differentiation of distinct stem cells could also be exploited. All of these strategies will help to facilitate the application of stem cells in human medicine. This article reviews the most recent strategies to generate a high amount of mature hepatocyte-like cells and updates current knowledge on liver regenerative medicine.
Collapse
Affiliation(s)
- Vahid Hosseini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Fathi Maroufi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Saghati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahideh Asadi
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Darabi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Nazari Soltan Ahmad
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
24
|
Geetha Bai R, Muthoosamy K, Manickam S, Hilal-Alnaqbi A. Graphene-based 3D scaffolds in tissue engineering: fabrication, applications, and future scope in liver tissue engineering. Int J Nanomedicine 2019; 14:5753-5783. [PMID: 31413573 PMCID: PMC6662516 DOI: 10.2147/ijn.s192779] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
Tissue engineering embraces the potential of recreating and replacing defective body parts by advancements in the medical field. Being a biocompatible nanomaterial with outstanding physical, chemical, optical, and biological properties, graphene-based materials were successfully employed in creating the perfect scaffold for a range of organs, starting from the skin through to the brain. Investigations on 2D and 3D tissue culture scaffolds incorporated with graphene or its derivatives have revealed the capability of this carbon material in mimicking in vivo environment. The porous morphology, great surface area, selective permeability of gases, excellent mechanical strength, good thermal and electrical conductivity, good optical properties, and biodegradability enable graphene materials to be the best component for scaffold engineering. Along with the apt microenvironment, this material was found to be efficient in differentiating stem cells into specific cell types. Furthermore, the scope of graphene nanomaterials in liver tissue engineering as a promising biomaterial is also discussed. This review critically looks into the unlimited potential of graphene-based nanomaterials in future tissue engineering and regenerative therapy.
Collapse
Affiliation(s)
- Renu Geetha Bai
- Nanotechnology and Advanced Materials (NATAM), Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Selangor, 43500, Malaysia
| | - Kasturi Muthoosamy
- Nanotechnology and Advanced Materials (NATAM), Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Selangor, 43500, Malaysia
| | - Sivakumar Manickam
- Nanotechnology and Advanced Materials (NATAM), Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Selangor, 43500, Malaysia
| | - Ali Hilal-Alnaqbi
- Electromechanical Technology, Abu Dhabi Polytechnic, Abu Dhabi, United Arab Emirates
| |
Collapse
|
25
|
Ye S, Boeter JWB, Penning LC, Spee B, Schneeberger K. Hydrogels for Liver Tissue Engineering. Bioengineering (Basel) 2019; 6:E59. [PMID: 31284412 PMCID: PMC6784004 DOI: 10.3390/bioengineering6030059] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
Bioengineered livers are promising in vitro models for drug testing, toxicological studies, and as disease models, and might in the future be an alternative for donor organs to treat end-stage liver diseases. Liver tissue engineering (LTE) aims to construct liver models that are physiologically relevant. To make bioengineered livers, the two most important ingredients are hepatic cells and supportive materials such as hydrogels. In the past decades, dozens of hydrogels have been developed to act as supportive materials, and some have been used for in vitro models and formed functional liver constructs. However, currently none of the used hydrogels are suitable for in vivo transplantation. Here, the histology of the human liver and its relationship with LTE is introduced. After that, significant characteristics of hydrogels are described focusing on LTE. Then, both natural and synthetic materials utilized in hydrogels for LTE are reviewed individually. Finally, a conclusion is drawn on a comparison of the different hydrogels and their characteristics and ideal hydrogels are proposed to promote LTE.
Collapse
Affiliation(s)
- Shicheng Ye
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Jochem W B Boeter
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Louis C Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Kerstin Schneeberger
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
26
|
Young AT, Rivera KR, Erb PD, Daniele MA. Monitoring of Microphysiological Systems: Integrating Sensors and Real-Time Data Analysis toward Autonomous Decision-Making. ACS Sens 2019; 4:1454-1464. [PMID: 30964652 DOI: 10.1021/acssensors.8b01549] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Microphysiological systems replicate human organ function and are promising technologies for discovery of translatable biomarkers, pharmaceuticals, and regenerative therapies. Because microphysiological systems require complex microscale anatomical structures and heterogeneous cell populations, a major challenge remains to manufacture and operate these products with reproducible and standardized function. In this Perspective, three stages of microphysiological system monitoring, including process, development, and function, are assessed. The unique features and remaining technical challenges for the required sensors are discussed. Monitoring of microphysiological systems requires nondestructive, continuous biosensors and imaging techniques. With such tools, the extent of cellular and tissue development, as well as function, can be autonomously determined and optimized by correlating physical and chemical sensor outputs with markers of physiological performance. Ultimately, data fusion and analyses across process, development, and function monitors can be implemented to adopt microphysiological systems for broad research and commercial applications.
Collapse
Affiliation(s)
- Ashlyn T. Young
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Drive, Raleigh, North Carolina 27695, United States
| | - Kristina R. Rivera
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Drive, Raleigh, North Carolina 27695, United States
| | - Patrick D. Erb
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Drive, Raleigh, North Carolina 27695, United States
| | - Michael A. Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Drive, Raleigh, North Carolina 27695, United States
- Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Drive, Raleigh, North Carolina 27695, United States
| |
Collapse
|
27
|
Prantil-Baun R, Novak R, Das D, Somayaji MR, Przekwas A, Ingber DE. Physiologically Based Pharmacokinetic and Pharmacodynamic Analysis Enabled by Microfluidically Linked Organs-on-Chips. Annu Rev Pharmacol Toxicol 2019; 58:37-64. [PMID: 29309256 DOI: 10.1146/annurev-pharmtox-010716-104748] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling and simulation approaches are beginning to be integrated into drug development and approval processes because they enable key pharmacokinetic (PK) parameters to be predicted from in vitro data. However, these approaches are hampered by many limitations, including an inability to incorporate organ-specific differentials in drug clearance, distribution, and absorption that result from differences in cell uptake, transport, and metabolism. Moreover, such approaches are generally unable to provide insight into pharmacodynamic (PD) parameters. Recent development of microfluidic Organ-on-a-Chip (Organ Chip) cell culture devices that recapitulate tissue-tissue interfaces, vascular perfusion, and organ-level functionality offer the ability to overcome these limitations when multiple Organ Chips are linked via their endothelium-lined vascular channels. Here, we discuss successes and challenges in the use of existing culture models and vascularized Organ Chips for PBPK and PD modeling of human drug responses, as well as in vitro to in vivo extrapolation (IVIVE) of these results, and how these approaches might advance drug development and regulatory review processes in the future.
Collapse
Affiliation(s)
- Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA;
| | - Richard Novak
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA;
| | - Debarun Das
- CFD Research Corporation, Huntsville, Alabama 35806, USA
| | | | | | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA; .,Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
28
|
Baze A, Parmentier C, Hendriks DFG, Hurrell T, Heyd B, Bachellier P, Schuster C, Ingelman-Sundberg M, Richert L. Three-Dimensional Spheroid Primary Human Hepatocytes in Monoculture and Coculture with Nonparenchymal Cells. Tissue Eng Part C Methods 2019; 24:534-545. [PMID: 30101670 DOI: 10.1089/ten.tec.2018.0134] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent advances in the development of various culture platforms are promising for achieving more physiologically relevant in vitro hepatic models using primary human hepatocytes (PHHs). Previous studies have shown the value of PHHs three-dimensional (3D) spheroid models, cultured in low cell number (1330-2000 cells/3D spheroid), to study long-term liver function as well as pharmacological drug effects and toxicity. In this study, we report that only plateable PHHs aggregate and form compact 3D spheroids with a success rate of 79%, and 96% reproducibility. Out of 3D spheroid forming PHH lots, 65% were considered stable (<50% ATP decrease) over the subsequent 14 days of culture, with reproducibility of a given PHH lot being 82%. We also report successful coculturing of PHHs with human liver nonparenchymal cells (NPCs). Crude P1c-NPC fractions were obtained by low centrifugation of the PHH supernatant fraction followed by a few days of culture before harvesting and cryopreservation. At aggregation of PHHs/P1c-NPCs (2:1 ratio 3D spheroids), liver sinusoidal endothelial cells, Kupffer cells, and hepatic stellate cells were successfully integrated and remained present throughout the subsequent 14-day culture period as revealed by mRNA expression markers and immunostaining. Increased mRNA expression of albumin (ALB), apolipoprotein B (APOB), cytochrome P450 3A4 (CYP3A4), and increased albumin secretion compared to PHH 3D spheroid monocultures highlighted that in a 3D spheroid coculture, configuration with NPCs, PHH functionality is increased. We thus achieved the development of a more integrated coculture model system requiring low cell numbers, of particular interest due to the scarcity of human liver NPCs.
Collapse
Affiliation(s)
- Audrey Baze
- 1 KaLy-Cell, Plobsheim , France .,2 Université de Strasbourg , Strasbourg, France
| | | | - Delilah F G Hendriks
- 3 Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet , Stockholm, Sweden
| | - Tracey Hurrell
- 3 Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet , Stockholm, Sweden
| | - Bruno Heyd
- 4 Hôpital Jean Minjoz , Besançon, France .,5 Université de Bourgogne Franche-Comté , Besançon, France
| | - Philippe Bachellier
- 2 Université de Strasbourg , Strasbourg, France .,6 Hôpital de Hautepierre , Strasbourg, France
| | - Catherine Schuster
- 2 Université de Strasbourg , Strasbourg, France .,7 INSERM, UMR_S1110, Institut de Recherche sur les Maladies Virales et Hépatiques , Strasbourg, France
| | - Magnus Ingelman-Sundberg
- 3 Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet , Stockholm, Sweden
| | - Lysiane Richert
- 1 KaLy-Cell, Plobsheim , France .,5 Université de Bourgogne Franche-Comté , Besançon, France
| |
Collapse
|
29
|
Rivera KR, Yokus MA, Erb PD, Pozdin VA, Daniele M. Measuring and regulating oxygen levels in microphysiological systems: design, material, and sensor considerations. Analyst 2019; 144:3190-3215. [PMID: 30968094 PMCID: PMC6564678 DOI: 10.1039/c8an02201a] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
As microfabrication techniques and tissue engineering methods improve, microphysiological systems (MPS) are being engineered that recapitulate complex physiological and pathophysiological states to supplement and challenge traditional animal models. Although MPS provide unique microenvironments that transcend common 2D cell culture, without proper regulation of oxygen content, MPS often fail to provide the biomimetic environment necessary to activate and investigate fundamental pathways of cellular metabolism and sub-cellular level. Oxygen exists in the human body in various concentrations and partial pressures; moreover, it fluctuates dramatically depending on fasting, exercise, and sleep patterns. Regulating oxygen content inside MPS necessitates a sensitive biological sensor to quantify oxygen content in real-time. Measuring oxygen in a microdevice is a non-trivial requirement for studies focused on understanding how oxygen impacts cellular processes, including angiogenesis and tumorigenesis. Quantifying oxygen inside a microdevice can be achieved via an array of technologies, with each method having benefits and limitations in terms of sensitivity, limits of detection, and invasiveness that must be considered and optimized. This article will review oxygen physiology in organ systems and offer comparisons of organ-specific MPS that do and do not consider oxygen microenvironments. Materials used in microphysiological models will also be analyzed in terms of their ability to control oxygen. Finally, oxygen sensor technologies are critically compared and evaluated for use in MPS.
Collapse
Affiliation(s)
- Kristina R Rivera
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA.
| | | | | | | | | |
Collapse
|
30
|
Marx U, Walles H, Hoffmann S, Lindner G, Horland R, Sonntag F, Klotzbach U, Sakharov D, Tonevitsky A, Lauster R. ‘Human-on-a-chip’ Developments: A Translational Cutting-edge Alternative to Systemic Safety Assessment and Efficiency Evaluation of Substances in Laboratory Animals and Man? Altern Lab Anim 2019; 40:235-57. [DOI: 10.1177/026119291204000504] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Uwe Marx
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | | | - Silke Hoffmann
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | - Gerd Lindner
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | - Reyk Horland
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | - Frank Sonntag
- Fraunhofer-Institut für Werkstoff- und Strahltechnik IWS, Dresden, Germany
| | - Udo Klotzbach
- Fraunhofer-Institut für Werkstoff- und Strahltechnik IWS, Dresden, Germany
| | | | | | - Roland Lauster
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| |
Collapse
|
31
|
Tomlinson L, Hyndman L, Firman JW, Bentley R, Kyffin JA, Webb SD, McGinty S, Sharma P. In vitro Liver Zonation of Primary Rat Hepatocytes. Front Bioeng Biotechnol 2019; 7:17. [PMID: 30834246 PMCID: PMC6387900 DOI: 10.3389/fbioe.2019.00017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/23/2019] [Indexed: 01/05/2023] Open
Abstract
The ability of the liver to simultaneously carry out multiple functions is dependent on the metabolic heterogeneity of hepatocytes spatially located within a liver lobule spanning from the portal triad to the central vein. This complex zonal architecture of the liver, however, makes accurate in vitro modeling a challenge and often standard culture systems assume a homogenous model which may lead to inaccurate translatability of results. Here, we use a combination of mathematical modeling and experimental data to demonstrate a readily constructible in vitro flow system capable of liver zonation in primary rat hepatocytes. We show the differential expression of zonation markers, enhanced functionality when compared to standard static cultures and zone-specific metabolism and cell damage in the presence of paracetamol, a known zone-specific toxin. This type of advanced system provides a more in-depth and essential understanding of liver physiology and pathophysiology as well as the accurate evaluation of pharmacological interventions at a zone-specific level.
Collapse
Affiliation(s)
- Lauren Tomlinson
- MRC Centre for Drug Safety Science, Department of Clinical and Molecular Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Lauren Hyndman
- Division of Biomedical Engineering, University of Glasgow, Glasgow, United Kingdom
| | - James W. Firman
- MRC Centre for Drug Safety Science, Department of Clinical and Molecular Pharmacology, University of Liverpool, Liverpool, United Kingdom
- Department of Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Robert Bentley
- MRC Centre for Drug Safety Science, Department of Clinical and Molecular Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Jonathan A. Kyffin
- Department of Applied Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
| | - Steven D. Webb
- Department of Applied Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
- EPSRC Liverpool Centre for Mathematics in Healthcare, Department of Mathematical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Sean McGinty
- Division of Biomedical Engineering, University of Glasgow, Glasgow, United Kingdom
| | - Parveen Sharma
- MRC Centre for Drug Safety Science, Department of Clinical and Molecular Pharmacology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
32
|
Underhill GH, Khetani SR. Advances in Engineered Human Liver Platforms for Drug Metabolism Studies. Drug Metab Dispos 2018; 46:1626-1637. [PMID: 30135245 PMCID: PMC6199629 DOI: 10.1124/dmd.118.083295] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
33
|
Kuna L, Bozic I, Kizivat T, Bojanic K, Mrso M, Kralj E, Smolic R, Wu GY, Smolic M. Models of Drug Induced Liver Injury (DILI) - Current Issues and Future Perspectives. Curr Drug Metab 2018; 19:830-838. [PMID: 29788883 PMCID: PMC6174638 DOI: 10.2174/1389200219666180523095355] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/20/2018] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
Abstract
Background: Drug-induced Liver Injury (DILI) is an important cause of acute liver failure cases in the United States, and remains a common cause of withdrawal of drugs in both preclinical and clinical phases. Methods: A structured search of bibliographic databases – Web of Science Core Collection, Scopus and Medline for peer-reviewed articles on models of DILI was performed. The reference lists of relevant studies was prepared and a citation search for the included studies was carried out. In addition, the characteristics of screened studies were described. Results: One hundred and six articles about the existing knowledge of appropriate models to study DILI in vitro and in vivo with special focus on hepatic cell models, variations of 3D co-cultures, animal models, databases and predictive modeling and translational biomarkers developed to understand the mechanisms and pathophysiology of DILI are described. Conclusion: Besides descriptions of current applications of existing modeling systems, associated advantages and limitations of each modeling system and future directions for research development are discussed as well.
Collapse
Affiliation(s)
- Lucija Kuna
- Department of Chemistry and Biochemistry, Faculty of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| | - Ivana Bozic
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Tomislav Kizivat
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Kristina Bojanic
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Margareta Mrso
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Edgar Kralj
- Inspecto, LLC, Martina Divalta 193, 31000 Osijek, Croatia
| | - Robert Smolic
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - George Y Wu
- Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, United States
| | - Martina Smolic
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia.,Department of Pharmacology, Faculty Of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| |
Collapse
|
34
|
Edington CD, Chen WLK, Geishecker E, Kassis T, Soenksen LR, Bhushan BM, Freake D, Kirschner J, Maass C, Tsamandouras N, Valdez J, Cook CD, Parent T, Snyder S, Yu J, Suter E, Shockley M, Velazquez J, Velazquez JJ, Stockdale L, Papps JP, Lee I, Vann N, Gamboa M, LaBarge ME, Zhong Z, Wang X, Boyer LA, Lauffenburger DA, Carrier RL, Communal C, Tannenbaum SR, Stokes CL, Hughes DJ, Rohatgi G, Trumper DL, Cirit M, Griffith LG. Interconnected Microphysiological Systems for Quantitative Biology and Pharmacology Studies. Sci Rep 2018. [PMID: 29540740 PMCID: PMC5852083 DOI: 10.1038/s41598-018-22749-0] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Microphysiological systems (MPSs) are in vitro models that capture facets of in vivo organ function through use of specialized culture microenvironments, including 3D matrices and microperfusion. Here, we report an approach to co-culture multiple different MPSs linked together physiologically on re-useable, open-system microfluidic platforms that are compatible with the quantitative study of a range of compounds, including lipophilic drugs. We describe three different platform designs – “4-way”, “7-way”, and “10-way” – each accommodating a mixing chamber and up to 4, 7, or 10 MPSs. Platforms accommodate multiple different MPS flow configurations, each with internal re-circulation to enhance molecular exchange, and feature on-board pneumatically-driven pumps with independently programmable flow rates to provide precise control over both intra- and inter-MPS flow partitioning and drug distribution. We first developed a 4-MPS system, showing accurate prediction of secreted liver protein distribution and 2-week maintenance of phenotypic markers. We then developed 7-MPS and 10-MPS platforms, demonstrating reliable, robust operation and maintenance of MPS phenotypic function for 3 weeks (7-way) and 4 weeks (10-way) of continuous interaction, as well as PK analysis of diclofenac metabolism. This study illustrates several generalizable design and operational principles for implementing multi-MPS “physiome-on-a-chip” approaches in drug discovery.
Collapse
Affiliation(s)
- Collin D Edington
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Wen Li Kelly Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emily Geishecker
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Timothy Kassis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Luis R Soenksen
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brij M Bhushan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | - Christian Maass
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nikolaos Tsamandouras
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jorge Valdez
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christi D Cook
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | - Jiajie Yu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emily Suter
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael Shockley
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jason Velazquez
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeremy J Velazquez
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linda Stockdale
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Julia P Papps
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Iris Lee
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicholas Vann
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mario Gamboa
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew E LaBarge
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zhe Zhong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laurie A Boyer
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rebecca L Carrier
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Catherine Communal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Steven R Tannenbaum
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | - David L Trumper
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Murat Cirit
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
35
|
Da-Silva F, Boulenc X, Vermet H, Compigne P, Gerbal-Chaloin S, Daujat-Chavanieu M, Klieber S, Poulin P. Improving Prediction of Metabolic Clearance Using Quantitative Extrapolation of Results Obtained From Human Hepatic Micropatterned Cocultures Model and by Considering the Impact of Albumin Binding. J Pharm Sci 2018. [PMID: 29524447 DOI: 10.1016/j.xphs.2018.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The objective was to compare, with the same data set, the predictive performance of 3 in vitro assays of hepatic clearance (CL), namely, micropatterned cocultures (also referring to HepatoPac®) and suspension as well as monolayer hepatocytes to define which assay is the most accurate. Furthermore, existing in vitro-to-in vivo extrapolation (IVIVE) methods were challenged to verify which method is the most predictive (i.e., direct scaling method without binding correction, conventional method based either on the unbound fraction in plasma (fup) according to the free-drug hypothesis, or based on an fup value adjusted for the albumin [ALB]-facilitated hepatic uptake phenomenon). Accordingly, the role of ALB binding was specifically challenged, and consequently, the ALB production was monitored in parallel to the metabolic stability. The ALB concentration data were used to compare the in vitro assays and to adjust the value of fup of each drug to mimic the ALB-facilitated hepatic uptake phenomenon. The results confirmed that the direct and conventional IVIVE methods generally overpredicted and underpredicted the CL in vivo in humans, respectively. However, the underprediction of the conventional IVIVE method based on fup was significantly reduced from data generated with the HepatoPac® system compared with the 2 other in vitro assays, which is possibly because that system is producing ALB at a rate much closer to the in vivo condition in liver. Hence, these observations suggest that the presence of more ALB molecules per hepatocyte in that HepatoPac® system may have facilitated the hepatic uptake of several bound drugs because their intrinsic CL was increased instead of being decreased by the ALB binding effect. Accordingly, the IVIVE method based on the fup value adjusted for the ALB-facilitated uptake phenomenon gave the lowest prediction bias from the statistical analyses. This study indicated that the HepatoPac® system combined with the adjusted value of fup was the most reliable IVIVE method and revealed the importance of quantifying the in vitro-to-in vivo variation of ALB concentration to improve the CL predictions, which would help any future physiologically based pharmacokinetics modeling exercise.
Collapse
Affiliation(s)
- Franck Da-Silva
- Sanofi R&D, Montpellier, France; Institute for Regenerative Medicine and Biotherapy, Université et CHU de Montpellier, INSERM, Montpellier, France
| | | | | | | | - Sabine Gerbal-Chaloin
- Institute for Regenerative Medicine and Biotherapy, Université et CHU de Montpellier, INSERM, Montpellier, France
| | - Martine Daujat-Chavanieu
- Institute for Regenerative Medicine and Biotherapy, Université et CHU de Montpellier, INSERM, Montpellier, France
| | | | - Patrick Poulin
- Consultant, Patrick Poulin Inc., Québec City, Canada; Associate professor, School of Public Health, IRSPUM, Université de Montréal, Canada
| |
Collapse
|
36
|
Yajima Y, Lee CN, Yamada M, Utoh R, Seki M. Development of a perfusable 3D liver cell cultivation system via bundling-up assembly of cell-laden microfibers. J Biosci Bioeng 2018; 126:111-118. [PMID: 29502942 DOI: 10.1016/j.jbiosc.2018.01.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/10/2018] [Accepted: 01/31/2018] [Indexed: 01/20/2023]
Abstract
Although the reconstruction of functional 3D liver tissue models in vitro presents numerous challenges, it is in great demand for drug development, regenerative medicine, and physiological studies. Here we propose a new approach to perform perfusion cultivation of liver cells by assembling cell-laden hydrogel microfibers. HepG2 cells were densely packed into the core of sandwich-type anisotropic microfibers, which were produced using microfluidic devices. The obtained microfibers were bundled up and packed into a perfusion chamber, and perfusion cultivation was performed. We evaluated cell viability and functions, and also monitored the oxygen consumption. Furthermore, fibers covered with vascular endothelial cells were united during the perfusion culture, to form vascular network-like conduits between fibers. The presented technique can structurally mimic the hepatic lobule in vivo and could prove to be a useful model for various biomedical research applications.
Collapse
Affiliation(s)
- Yuya Yajima
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Chu Ning Lee
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Masumi Yamada
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Rie Utoh
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Minoru Seki
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
37
|
Kamiya K, Abe Y, Inoue K, Osaki T, Kawano R, Miki N, Takeuchi S. Well-Controlled Cell-Trapping Systems for Investigating Heterogeneous Cell-Cell Interactions. Adv Healthc Mater 2018; 7:e1701208. [PMID: 29369539 DOI: 10.1002/adhm.201701208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/13/2017] [Indexed: 11/10/2022]
Abstract
Microfluidic systems have been developed for patterning single cells to study cell-cell interactions. However, patterning multiple types of cells to understand heterogeneous cell-cell interactions remains difficult. Here, it is aimed to develop a cell-trapping device to assemble multiple types of cells in the well-controlled order and morphology. This device mainly comprises a parylene sheet for assembling cells and a microcomb for controlling the cell-trapping area. The cell-trapping area is controlled by moving the parylene sheet on an SU-8 microcomb using tweezers. Gentle downward flow is used as a driving force for the cell-trapping. The assembly of cells on a parylene sheet with round and line-shaped apertures is demonstrated. The cell-cell contacts of the trapped cells are then investigated by direct cell-cell transfer of calcein via connexin nanopores. Finally, using the device with a system for controlling the cell-trapping area, three different types of cells in the well-controlled order are assembled. The correct cell order rate obtained using the device is 27.9%, which is higher than that obtained without the sliding parylene system (0.74%). Furthermore, the occurrence of cell-cell contact between the three cell types assembled is verified. This cell-patterning device will be a useful tool for investigating heterogeneous cell-cell interactions.
Collapse
Affiliation(s)
- Koki Kamiya
- Artificial Cell Membrane Systems Group Kanagawa Institute of Industrial Science and Technology KSP EAST 303, 3‐2‐1, Sakado Takatsu‐ku, Kawasaki Kanagawa 213‐0012 Japan
| | - Yuta Abe
- Artificial Cell Membrane Systems Group Kanagawa Institute of Industrial Science and Technology KSP EAST 303, 3‐2‐1, Sakado Takatsu‐ku, Kawasaki Kanagawa 213‐0012 Japan
- School of Integrated Design Engineering Keio University 3‐14‐1, Hiyoshi, Kohoku‐ku Yokohama Kanagawa 223‐8522 Japan
| | - Kosuke Inoue
- Artificial Cell Membrane Systems Group Kanagawa Institute of Industrial Science and Technology KSP EAST 303, 3‐2‐1, Sakado Takatsu‐ku, Kawasaki Kanagawa 213‐0012 Japan
- School of Integrated Design Engineering Keio University 3‐14‐1, Hiyoshi, Kohoku‐ku Yokohama Kanagawa 223‐8522 Japan
| | - Toshihisa Osaki
- Artificial Cell Membrane Systems Group Kanagawa Institute of Industrial Science and Technology KSP EAST 303, 3‐2‐1, Sakado Takatsu‐ku, Kawasaki Kanagawa 213‐0012 Japan
- Institute of Industrial Science The University of Tokyo 4‐6‐1 Komaba Meguro‐ku Tokyo 153‐8505 Japan
| | - Ryuji Kawano
- Artificial Cell Membrane Systems Group Kanagawa Institute of Industrial Science and Technology KSP EAST 303, 3‐2‐1, Sakado Takatsu‐ku, Kawasaki Kanagawa 213‐0012 Japan
| | - Norihisa Miki
- Artificial Cell Membrane Systems Group Kanagawa Institute of Industrial Science and Technology KSP EAST 303, 3‐2‐1, Sakado Takatsu‐ku, Kawasaki Kanagawa 213‐0012 Japan
- Department of Mechanical Engineering Keio University 3‐14‐1, Hiyoshi, Kohoku‐ku Yokohama Kanagawa 223‐8522 Japan
| | - Shoji Takeuchi
- Artificial Cell Membrane Systems Group Kanagawa Institute of Industrial Science and Technology KSP EAST 303, 3‐2‐1, Sakado Takatsu‐ku, Kawasaki Kanagawa 213‐0012 Japan
- Institute of Industrial Science The University of Tokyo 4‐6‐1 Komaba Meguro‐ku Tokyo 153‐8505 Japan
| |
Collapse
|
38
|
Ortega-Prieto AM, Skelton JK, Wai SN, Large E, Lussignol M, Vizcay-Barrena G, Hughes D, Fleck RA, Thursz M, Catanese MT, Dorner M. 3D microfluidic liver cultures as a physiological preclinical tool for hepatitis B virus infection. Nat Commun 2018; 9:682. [PMID: 29445209 PMCID: PMC5813240 DOI: 10.1038/s41467-018-02969-8] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 01/09/2018] [Indexed: 12/21/2022] Open
Abstract
With more than 240 million people infected, hepatitis B virus (HBV) is a major health concern. The inability to mimic the complexity of the liver using cell lines and regular primary human hepatocyte (PHH) cultures pose significant limitations for studying host/pathogen interactions. Here, we describe a 3D microfluidic PHH system permissive to HBV infection, which can be maintained for at least 40 days. This system enables the recapitulation of all steps of the HBV life cycle, including the replication of patient-derived HBV and the maintenance of HBV cccDNA. We show that innate immune and cytokine responses following infection with HBV mimic those observed in HBV-infected patients, thus allowing the dissection of pathways important for immune evasion and validation of biomarkers. Additionally, we demonstrate that the co-culture of PHH with other non-parenchymal cells enables the identification of the cellular origin of immune effectors, thus providing a valuable preclinical platform for HBV research.
Collapse
Affiliation(s)
- A M Ortega-Prieto
- Section of Virology, Department of Medicine, Imperial College London, London, W2 1PG, UK
| | - J K Skelton
- Section of Virology, Department of Medicine, Imperial College London, London, W2 1PG, UK
| | - S N Wai
- Section of Virology, Department of Medicine, Imperial College London, London, W2 1PG, UK
- Section of Hepatology, Department of Medicine, Imperial College London, London, W2 1NY, UK
| | - E Large
- CN Bio Innovations Ltd, Welwyn Garden City, AL7 3AX, UK
| | - M Lussignol
- Department of Infectious Diseases, King's College London, London, WC2R 2LS, UK
| | - G Vizcay-Barrena
- Centre For Ultrastructural Imaging, King's College London, London, WC2R 2LS, UK
| | - D Hughes
- CN Bio Innovations Ltd, Welwyn Garden City, AL7 3AX, UK
| | - R A Fleck
- Centre For Ultrastructural Imaging, King's College London, London, WC2R 2LS, UK
| | - M Thursz
- Section of Hepatology, Department of Medicine, Imperial College London, London, W2 1NY, UK
| | - M T Catanese
- Department of Infectious Diseases, King's College London, London, WC2R 2LS, UK
| | - M Dorner
- Section of Virology, Department of Medicine, Imperial College London, London, W2 1PG, UK.
| |
Collapse
|
39
|
Kyffin JA, Sharma P, Leedale J, Colley HE, Murdoch C, Mistry P, Webb SD. Impact of cell types and culture methods on the functionality of in vitro liver systems - A review of cell systems for hepatotoxicity assessment. Toxicol In Vitro 2018; 48:262-275. [PMID: 29408671 DOI: 10.1016/j.tiv.2018.01.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/26/2018] [Accepted: 01/27/2018] [Indexed: 12/21/2022]
Abstract
Xenobiotic safety assessment is an area that impacts a multitude of different industry sectors such as medicinal drugs, agrochemicals, industrial chemicals, cosmetics and environmental contaminants. As such there are a number of well-developed in vitro, in vivo and in silico approaches to evaluate their properties and potential impact on the environment and to humans. Additionally, there is the continual investment in multidisciplinary scientists to explore non-animal surrogate technologies to predict specific toxicological outcomes and to improve our understanding of the biological processes regarding the toxic potential of xenobiotics. Here we provide a concise, critical evaluation of a number of in vitro systems utilised to assess the hepatotoxic potential of xenobiotics.
Collapse
Affiliation(s)
- Jonathan A Kyffin
- Department of Applied Mathematics, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Parveen Sharma
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Building, Ashton Street, University of Liverpool, L69 3GE, United Kingdom.
| | - Joseph Leedale
- EPSRC Liverpool Centre for Mathematics in Healthcare, Department of Mathematical Sciences, Peach Street, University of Liverpool, L69 7ZL, United Kingdom
| | - Helen E Colley
- School of Clinical Dentistry, Claremont Crescent, University of Sheffield, Sheffield S10 2TA, United Kingdom
| | - Craig Murdoch
- School of Clinical Dentistry, Claremont Crescent, University of Sheffield, Sheffield S10 2TA, United Kingdom
| | - Pratibha Mistry
- Syngenta Ltd., Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, United Kingdom
| | - Steven D Webb
- Department of Applied Mathematics, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, United Kingdom
| |
Collapse
|
40
|
Macdonald NP, Menachery A, Reboud J, Cooper JM. Creating tissue on chip constructs in microtitre plates for drug discovery. RSC Adv 2018; 8:9603-9610. [PMID: 35540822 PMCID: PMC9078682 DOI: 10.1039/c8ra00849c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 02/16/2018] [Indexed: 11/30/2022] Open
Abstract
We report upon a novel coplanar dielectrophoresis (DEP) based cell patterning system for generating transferrable hepatic cell constructs, resembling a liver-lobule, in culture. The use of paper reinforced gel substrates provided sufficient strength to enable these constructs to be transfered into 96-well plates for long term functional studies, including in the future, drug development studies. Experimental results showed that hepatic cells formed DEP field-induced structures corresponding to an array of lobule-mimetic patterns. Hepatic viability was observed over a period of 3 days by the use of a fluorescent cell staining technique, whilst the liver specific functionality of albumin secretion showed a significant enhancement due to the layer patterning of cell lines (HepG2/C3A), compared to 2D patterned cells and un-patterned control. This “build and transfer” concept could, in future, also be adapted for the layer-by-layer construction of organs-on-chip in microtitre formats. We report upon a novel coplanar dielectrophoresis (DEP) based cell patterning system for generating transferrable hepatic cell constructs, resembling a liver-lobule, in culture.![]()
Collapse
Affiliation(s)
- N. P. Macdonald
- ARC Centre of Excellence for Electromaterials Science (ACES)
- Australian Centre for Research on Separation Science (ACROSS)
- School of Chemistry
- Faculty of Science, Engineering and Technology
- University of Tasmania
| | - A. Menachery
- The Advanced Microfluidics and Microdevices Laboratory (AMMLab)
- New York University Abu Dhabi
- Abu Dhabi
- UAE
| | - J. Reboud
- Division of Biomedical Engineering
- School of Engineering
- University of Glasgow
- Glasgow
- UK
| | - J. M. Cooper
- Division of Biomedical Engineering
- School of Engineering
- University of Glasgow
- Glasgow
- UK
| |
Collapse
|
41
|
Cole BK, Feaver RE, Wamhoff BR, Dash A. Non-alcoholic fatty liver disease (NAFLD) models in drug discovery. Expert Opin Drug Discov 2017; 13:193-205. [PMID: 29190166 DOI: 10.1080/17460441.2018.1410135] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The progressive disease spectrum of non-alcoholic fatty liver disease (NAFLD), which includes non-alcoholic steatohepatitis (NASH), is a rapidly emerging public health crisis with no approved therapy. The diversity of various therapies under development highlights the lack of consensus around the most effective target, underscoring the need for better translatable preclinical models to study the complex progressive disease and effective therapies. Areas covered: This article reviews published literature of various mouse models of NASH used in preclinical studies, as well as complex organotypic in vitro and ex vivo liver models being developed. It discusses translational challenges associated with both kinds of models, and describes some of the studies that validate their application in NAFLD. Expert opinion: Animal models offer advantages of understanding drug distribution and effects in a whole body context, but are limited by important species differences. Human organotypic in vitro and ex vivo models with physiological relevance and translatability need to be used in a tiered manner with simpler screens. Leveraging newer technologies, like metabolomics, proteomics, and transcriptomics, and the future development of validated disease biomarkers will allow us to fully utilize the value of these models to understand disease and evaluate novel drugs in isolation or combination.
Collapse
Affiliation(s)
| | | | | | - Ajit Dash
- b Early Development Safety , Genentech Inc , South San Francisco , CA , USA
| |
Collapse
|
42
|
Kim D, Ryu J, Son M, Oh J, Chung K, Lee S, Lee J, Ahn J, Min J, Ahn J, Kang HM, Kim J, Jung C, Kim N, Cho H. A liver-specific gene expression panel predicts the differentiation status of in vitro hepatocyte models. Hepatology 2017; 66. [PMID: 28640507 PMCID: PMC5698781 DOI: 10.1002/hep.29324] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
UNLABELLED Alternative cell sources, such as three-dimensional organoids and induced pluripotent stem cell-derived cells, might provide a potentially effective approach for both drug development applications and clinical transplantation. For example, the development of cell sources for liver cell-based therapy has been increasingly needed, and liver transplantation is performed for the treatment for patients with severe end-stage liver disease. Differentiated liver cells and three-dimensional organoids are expected to provide new cell sources for tissue models and revolutionary clinical therapies. However, conventional experimental methods confirming the expression levels of liver-specific lineage markers cannot provide complete information regarding the differentiation status or degree of similarity between liver and differentiated cell sources. Therefore, in this study, to overcome several issues associated with the assessment of differentiated liver cells and organoids, we developed a liver-specific gene expression panel (LiGEP) algorithm that presents the degree of liver similarity as a "percentage." We demonstrated that the percentage calculated using the LiGEP algorithm was correlated with the developmental stages of in vivo liver tissues in mice, suggesting that LiGEP can correctly predict developmental stages. Moreover, three-dimensional cultured HepaRG cells and human pluripotent stem cell-derived hepatocyte-like cells showed liver similarity scores of 59.14% and 32%, respectively, although general liver-specific markers were detected. CONCLUSION Our study describes a quantitative and predictive model for differentiated samples, particularly liver-specific cells or organoids; and this model can be further expanded to various tissue-specific organoids; our LiGEP can provide useful information and insights regarding the differentiation status of in vitro liver models. (Hepatology 2017;66:1662-1674).
Collapse
Affiliation(s)
- Dae‐Soo Kim
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea,Department of Functional GenomicsKorea University of Science and TechnologyDaejeonRepublic of Korea
| | - Jea‐Woon Ryu
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Mi‐Young Son
- Department of Functional GenomicsKorea University of Science and TechnologyDaejeonRepublic of Korea,Stem Cell Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Jung‐Hwa Oh
- Korea Institute of ToxicologyDaejeonRepublic of Korea
| | - Kyung‐Sook Chung
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea,Department of Functional GenomicsKorea University of Science and TechnologyDaejeonRepublic of Korea
| | - Sugi Lee
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea,Department of Functional GenomicsKorea University of Science and TechnologyDaejeonRepublic of Korea
| | - Jeong‐Ju Lee
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Jun‐Ho Ahn
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Ju‐Sik Min
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Jiwon Ahn
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Hyun Mi Kang
- Stem Cell Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Janghwan Kim
- Department of Functional GenomicsKorea University of Science and TechnologyDaejeonRepublic of Korea,Stem Cell Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Cho‐Rok Jung
- Department of Functional GenomicsKorea University of Science and TechnologyDaejeonRepublic of Korea,Stem Cell Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea
| | - Nam‐Soon Kim
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea,Department of Functional GenomicsKorea University of Science and TechnologyDaejeonRepublic of Korea
| | - Hyun‐Soo Cho
- Genome Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonRepublic of Korea,Department of Functional GenomicsKorea University of Science and TechnologyDaejeonRepublic of Korea
| |
Collapse
|
43
|
Liguori GR, Jeronimus BF, de Aquinas Liguori TT, Moreira LFP, Harmsen MC. * Ethical Issues in the Use of Animal Models for Tissue Engineering: Reflections on Legal Aspects, Moral Theory, Three Rs Strategies, and Harm-Benefit Analysis. Tissue Eng Part C Methods 2017; 23:850-862. [PMID: 28756735 DOI: 10.1089/ten.tec.2017.0189] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Animal experimentation requires a solid and rational moral foundation. Objective and emphatic decision-making and protocol evaluation by researchers and ethics committees remain a difficult and sensitive matter. This article presents three perspectives that facilitate a consideration of the minimally acceptable standard for animal experiments, in particular, in tissue engineering (TE) and regenerative medicine. First, we review the boundaries provided by law and public opinion in America and Europe. Second, we review contemporary moral theory to introduce the Neo-Rawlsian contractarian theory to objectively evaluate the ethics of animal experiments. Third, we introduce the importance of available reduction, replacement, and refinement strategies, which should be accounted for in moral decision-making and protocol evaluation of animal experiments. The three perspectives are integrated into an algorithmic and graphic harm-benefit analysis tool based on the most relevant aspects of animal models in TE. We conclude with a consideration of future avenues to improve animal experiments.
Collapse
Affiliation(s)
- Gabriel R Liguori
- 1 Lab for Cardiovascular Regenerative Medicine Research Group (CAVAREM), Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen , Groningen, the Netherlands .,2 Laboratory of Cardiovascular Surgery and Circulation Pathophysiology (LIM-11), Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bertus F Jeronimus
- 3 Department of Developmental Psychology, University of Groningen , Groningen, the Netherlands .,4 Department of Psychiatry, Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University of Groningen, University Medical Center Groningen , Groningen, the Netherlands
| | - Tácia T de Aquinas Liguori
- 1 Lab for Cardiovascular Regenerative Medicine Research Group (CAVAREM), Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen , Groningen, the Netherlands .,2 Laboratory of Cardiovascular Surgery and Circulation Pathophysiology (LIM-11), Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Luiz Felipe P Moreira
- 2 Laboratory of Cardiovascular Surgery and Circulation Pathophysiology (LIM-11), Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Martin C Harmsen
- 1 Lab for Cardiovascular Regenerative Medicine Research Group (CAVAREM), Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen , Groningen, the Netherlands
| |
Collapse
|
44
|
Jaroch K, Jaroch A, Bojko B. Cell cultures in drug discovery and development: The need of reliable in vitro-in vivo extrapolation for pharmacodynamics and pharmacokinetics assessment. J Pharm Biomed Anal 2017; 147:297-312. [PMID: 28811111 DOI: 10.1016/j.jpba.2017.07.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 07/16/2017] [Accepted: 07/19/2017] [Indexed: 12/21/2022]
Abstract
For ethical and cost-related reasons, use of animals for the assessment of mode of action, metabolism and/or toxicity of new drug candidates has been increasingly scrutinized in research and industrial applications. Implementation of the 3 "Rs"1; rule (Reduction, Replacement, Refinement) through development of in silico or in vitro assays has become an essential element of risk assessment. Physiologically based pharmacokinetic (PBPK2) modeling is the most potent in silico tool used for extrapolation of pharmacokinetic parameters to animal or human models from results obtained in vitro. Although, many types of in vitro assays are conducted during drug development, use of cell cultures is the most reliable one. Two-dimensional (2D) cell cultures have been a part of drug development for many years. Nowadays, their role is decreasing in favor of three-dimensional (3D) cell cultures and co-cultures. 3D cultures exhibit protein expression patterns and intercellular junctions that are closer to in vivo states in comparison to classical monolayer cultures. Co-cultures allow for examinations of the mutual influence of different cell lines. However, the complexity and high costs of co-cultures and 3D equipment exclude such methods from high-throughput screening (HTS).3In vitro absorption, distribution, metabolism, and excretion assessment, as well as drug-drug interaction (DDI), are usually performed with the use of various cell culture based assays. Progress in in silico and in vitro methods can lead to better in vitro-in vivo extrapolation (IVIVE4) outcomes and have a potential to contribute towards a significant reduction in the number of laboratory animals needed for drug research. As such, concentrated efforts need to be spent towards the development of an HTS in vitro platform with satisfactory IVIVE features.
Collapse
Affiliation(s)
- Karol Jaroch
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2 Street, 85-089 Bydgoszcz, Poland
| | - Alina Jaroch
- Department and Institute of Nutrition and Dietetics, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Dębowa 3 Street, 85-626 Bydgoszcz, Poland; Department and Clinic of Geriatrics, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Curie Sklodowskiej 9 Street, 85-094 Bydgoszcz, Poland
| | - Barbara Bojko
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2 Street, 85-089 Bydgoszcz, Poland.
| |
Collapse
|
45
|
Mazza G, Al-Akkad W, Telese A, Longato L, Urbani L, Robinson B, Hall A, Kong K, Frenguelli L, Marrone G, Willacy O, Shaeri M, Burns A, Malago M, Gilbertson J, Rendell N, Moore K, Hughes D, Notingher I, Jell G, Del Rio Hernandez A, De Coppi P, Rombouts K, Pinzani M. Rapid production of human liver scaffolds for functional tissue engineering by high shear stress oscillation-decellularization. Sci Rep 2017; 7:5534. [PMID: 28717194 PMCID: PMC5514140 DOI: 10.1038/s41598-017-05134-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/09/2017] [Indexed: 01/07/2023] Open
Abstract
The development of human liver scaffolds retaining their 3-dimensional structure and extra-cellular matrix (ECM) composition is essential for the advancement of liver tissue engineering. We report the design and validation of a new methodology for the rapid and accurate production of human acellular liver tissue cubes (ALTCs) using normal liver tissue unsuitable for transplantation. The application of high shear stress is a key methodological determinant accelerating the process of tissue decellularization while maintaining ECM protein composition, 3D-architecture and physico-chemical properties of the native tissue. ALTCs were engineered with human parenchymal and non-parenchymal liver cell lines (HepG2 and LX2 cells, respectively), human umbilical vein endothelial cells (HUVEC), as well as primary human hepatocytes and hepatic stellate cells. Both parenchymal and non-parenchymal liver cells grown in ALTCs exhibited markedly different gene expression when compared to standard 2D cell cultures. Remarkably, HUVEC cells naturally migrated in the ECM scaffold and spontaneously repopulated the lining of decellularized vessels. The metabolic function and protein synthesis of engineered liver scaffolds with human primary hepatocytes reseeded under dynamic conditions were maintained. These results provide a solid basis for the establishment of effective protocols aimed at recreating human liver tissue in vitro.
Collapse
Affiliation(s)
- Giuseppe Mazza
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK.
| | - Walid Al-Akkad
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - Andrea Telese
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - Lisa Longato
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - Luca Urbani
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute for Child Health. University College London, London, UK
| | - Benjamin Robinson
- Department of Bioengineering, Cellular and Molecular Biomechanics. Imperial College, London, UK
| | - Andrew Hall
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - Kenny Kong
- School of Physics and Astronomy, University of Nottingham, Nottingham, UK
| | - Luca Frenguelli
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - Giusi Marrone
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - Oliver Willacy
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - Mohsen Shaeri
- CN Bio Innovations Limited. BioPark Hertfordshire, Broadwater Road, Welwyn Garden City, Hertfordshire, UK
| | - Alan Burns
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute for Child Health. University College London, London, UK
- Department of Clinical Genetics, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Massimo Malago
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - Janet Gilbertson
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Royal Free Hospital. University College London, London, UK
| | - Nigel Rendell
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Royal Free Hospital. University College London, London, UK
| | - Kevin Moore
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - David Hughes
- CN Bio Innovations Limited. BioPark Hertfordshire, Broadwater Road, Welwyn Garden City, Hertfordshire, UK
| | - Ioan Notingher
- School of Physics and Astronomy, University of Nottingham, Nottingham, UK
| | - Gavin Jell
- Center for Nanotechnology and Regenerative Medicine, Division of Surgery and Interventional Science. University College London, London, UK
| | | | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute for Child Health. University College London, London, UK
| | - Krista Rombouts
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Royal Free Hospital. University College London, London, UK
| |
Collapse
|
46
|
Sarkar U, Ravindra KC, Large E, Young CL, Rivera-Burgos D, Yu J, Cirit M, Hughes DJ, Wishnok JS, Lauffenburger DA, Griffith LG, Tannenbaum SR. Integrated Assessment of Diclofenac Biotransformation, Pharmacokinetics, and Omics-Based Toxicity in a Three-Dimensional Human Liver-Immunocompetent Coculture System. Drug Metab Dispos 2017; 45:855-866. [PMID: 28450578 PMCID: PMC5469400 DOI: 10.1124/dmd.116.074005] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 04/21/2017] [Indexed: 12/13/2022] Open
Abstract
In vitro hepatocyte culture systems have inherent limitations in capturing known human drug toxicities that arise from complex immune responses. Therefore, we established and characterized a liver immunocompetent coculture model and evaluated diclofenac (DCF) metabolic profiles, in vitro-in vivo clearance correlations, toxicological responses, and acute phase responses using liquid chromatography-tandem mass spectrometry. DCF biotransformation was assessed after 48 hours of culture, and the major phase I and II metabolites were similar to the in vivo DCF metabolism profile in humans. Further characterization of secreted bile acids in the medium revealed that a glycine-conjugated bile acid was a sensitive marker of dose-dependent toxicity in this three-dimensional liver microphysiological system. Protein markers were significantly elevated in the culture medium at high micromolar doses of DCF, which were also observed previously for acute drug-induced toxicity in humans. In this immunocompetent model, lipopolysaccharide treatment evoked an inflammatory response that resulted in a marked increase in the overall number of acute phase proteins. Kupffer cell-mediated cytokine release recapitulated an in vivo proinflammatory response exemplified by a cohort of 11 cytokines that were differentially regulated after lipopolysaccharide induction, including interleukin (IL)-1β, IL-1Ra, IL-6, IL-8, IP-10, tumor necrosis factor-α, RANTES (regulated on activation normal T cell expressed and secreted), granulocyte colony-stimulating factor, macrophage colony-stimulating factor, macrophage inflammatory protein-1β, and IL-5. In summary, our findings indicate that three-dimensional liver microphysiological systems may serve as preclinical investigational platforms from the perspective of the discovery of a set of clinically relevant biomarkers including potential reactive metabolites, endogenous bile acids, excreted proteins, and cytokines to predict early drug-induced liver toxicity in humans.
Collapse
Affiliation(s)
- Ujjal Sarkar
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Kodihalli C Ravindra
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Emma Large
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Carissa L Young
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Dinelia Rivera-Burgos
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Jiajie Yu
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Murat Cirit
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - David J Hughes
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - John S Wishnok
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Douglas A Lauffenburger
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Linda G Griffith
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Steven R Tannenbaum
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| |
Collapse
|
47
|
Miles JR, Laughlin TD, Sargus-Patino CN, Pannier AK. In vitro porcine blastocyst development in three-dimensional alginate hydrogels. Mol Reprod Dev 2017; 84:775-787. [PMID: 28407335 DOI: 10.1002/mrd.22814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/07/2017] [Indexed: 11/08/2022]
Abstract
Appropriate embryonic and fetal development significantly impact pregnancy success and, therefore, the efficiency of swine production. The pre-implantation period of porcine pregnancy is characterized by several developmental hallmarks, which are initiated by the dramatic morphological change that occurs as pig blastocysts elongate from spherical to filamentous blastocysts. Deficiencies in blastocyst elongation contribute to approximately 20% of embryonic loss, and have a direct influence on within-litter birth weight variation. Although factors identified within the uterine environment may play a role in blastocyst elongation, little is known about the exact mechanisms by which porcine (or other species') blastocysts initiate and progress through the elongation process. This is partly due to the difficulty of replicating elongation in vitro, which would allow for its study in a controlled environment and in real-time. We developed a three dimensional (3-D) culture system using alginate hydrogel matrices that can encapsulate pig blastocysts, maintain viability and blastocyst architecture, and facilitate reproducible morphological changes with corresponding expression of steroidogenic enzyme transcripts and estrogen production, consistent with the initiation of elongation in vivo. This review highlights key aspects of the pre-implantation period of porcine pregnancy and the difficulty of studying blastocyst elongation in vivo or by using in vitro systems. This review also provides insights on the utility of 3-D hydrogels to study blastocyst elongation continuously and in real-time as a complementary and confirmatory approach to in vivo analysis.
Collapse
Affiliation(s)
- Jeremy R Miles
- USDA, U.S. Meat Animal Research Center (USMARC), Clay Center, Nebraska
| | - Taylor D Laughlin
- Department of Biological Systems Engineering, University of Nebraska-Lincoln (UNL), Lincoln, Nebraska
| | - Catherine N Sargus-Patino
- Department of Biological Systems Engineering, University of Nebraska-Lincoln (UNL), Lincoln, Nebraska
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln (UNL), Lincoln, Nebraska
| |
Collapse
|
48
|
Ng SS, Xiong A, Nguyen K, Masek M, No DY, Elazar M, Shteyer E, Winters MA, Voedisch A, Shaw K, Rashid ST, Frank CW, Cho NJ, Glenn JS. Long-term culture of human liver tissue with advanced hepatic functions. JCI Insight 2017; 2:90853. [PMID: 28570275 DOI: 10.1172/jci.insight.90853] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 04/27/2017] [Indexed: 01/16/2023] Open
Abstract
A major challenge for studying authentic liver cell function and cell replacement therapies is that primary human hepatocytes rapidly lose their advanced function in conventional, 2-dimensional culture platforms. Here, we describe the fabrication of 3-dimensional hexagonally arrayed lobular human liver tissues inspired by the liver's natural architecture. The engineered liver tissues exhibit key features of advanced differentiation, such as human-specific cytochrome P450-mediated drug metabolism and the ability to support efficient infection with patient-derived inoculums of hepatitis C virus. The tissues permit the assessment of antiviral agents and maintain their advanced functions for over 5 months in culture. This extended functionality enabled the prediction of a fatal human-specific hepatotoxicity caused by fialuridine (FIAU), which had escaped detection by preclinical models and short-term clinical studies. The results obtained with the engineered human liver tissue in this study provide proof-of-concept determination of human-specific drug metabolism, demonstrate the ability to support infection with human hepatitis virus derived from an infected patient and subsequent antiviral drug testing against said infection, and facilitate detection of human-specific drug hepatotoxicity associated with late-onset liver failure. Looking forward, the scalability and biocompatibility of the scaffold are also ideal for future cell replacement therapeutic strategies.
Collapse
Affiliation(s)
- Soon Seng Ng
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.,School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Anming Xiong
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Khanh Nguyen
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Marilyn Masek
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.,Department of Pathology, Stanford University School of Medicine
| | - Da Yoon No
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.,Department of Bioengineering, Stanford University, Stanford California, USA
| | - Menashe Elazar
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Eyal Shteyer
- Department of Pediatric Gastroenterology and Nutrition, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Mark A Winters
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | | | - Kate Shaw
- Department of Obstetrics and Gynecology
| | - Sheikh Tamir Rashid
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Curtis W Frank
- Department of Chemical Engineering, Stanford University, Stanford California, USA
| | - Nam Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jeffrey S Glenn
- Division of Gastroenterology and Hepatology, Department of Medicine.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
49
|
Starokozhko V, Groothuis GMM. Challenges on the road to a multicellular bioartificial liver. J Tissue Eng Regen Med 2017; 12:e227-e236. [PMID: 27943623 DOI: 10.1002/term.2385] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/28/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022]
Abstract
Over recent years, the progress in the development of a bioartificial liver (BAL) as an extracorporeal device or as a tissue engineered transplantable organ has been immense. However, many important BAL characteristics that are necessary to meet clinical demands have not been sufficiently addressed. This review describes the existing challenges in the development of a BAL for clinical applications, highlighting multicellularity and sinusoidal microarchitecture as crucial BAL characteristics to fulfil various liver functions. Currently available sources of nonparenchymal liver cells, such as endothelial cells, cholangiocytes and macrophages, used in BAL development are defined. Also, we discuss the recent studies on the reconstruction of the complex liver sinusoid microarchitecture using various liver cell types. Moreover, we highlight some other aspects of a BAL, such as liver zonation and formation of a vascular as well as biliary network for an adequate delivery, biotransformation and removal of substrates and waste products. Finally, the benefits of a multicellular BAL for the pharmaceutical industry are briefly described. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Viktoriia Starokozhko
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands
| | - Geny M M Groothuis
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands
| |
Collapse
|
50
|
Hughes DJ, Kostrzewski T, Sceats EL. Opportunities and challenges in the wider adoption of liver and interconnected microphysiological systems. Exp Biol Med (Maywood) 2017; 242:1593-1604. [PMID: 28504617 DOI: 10.1177/1535370217708976] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Liver disease represents a growing global health burden. The development of in vitro liver models which allow the study of disease and the prediction of metabolism and drug-induced liver injury in humans remains a challenge. The maintenance of functional primary hepatocytes cultures, the parenchymal cell of the liver, has historically been difficult with dedifferentiation and the consequent loss of hepatic function limiting utility. The desire for longer term functional liver cultures sparked the development of numerous systems, including collagen sandwiches, spheroids, micropatterned co-cultures and liver microphysiological systems. This review will focus on liver microphysiological systems, often referred to as liver-on-a-chip, and broaden to include platforms with interconnected microphysiological systems or multi-organ-chips. The interconnection of microphysiological systems presents the opportunity to explore system level effects, investigate organ cross talk, and address questions which were previously the preserve of animal experimentation. As a field, microphysiological systems have reached a level of maturity suitable for commercialization and consequent evaluation by a wider community of users, in academia and the pharmaceutical industry. Here scientific, operational, and organizational considerations relevant to the wider adoption of microphysiological systems will be discussed. Applications in which microphysiological systems might offer unique scientific insights or enable studies currently feasible only with animal models are described, and challenges which might be addressed to enable wider adoption of the technologies are highlighted. A path forward which envisions the development of microphysiological systems in partnerships between academia, vendors and industry, is proposed. Impact statement Microphysiological systems are in vitro models of human tissues and organs. These systems have advanced rapidly in recent years and are now being commercialized. To achieve wide adoption in the biological and pharmaceutical research communities, microphysiological systems must provide unique insights which translate to humans. This will be achieved by identifying key applications and making microphysiological systems intuitive to use.
Collapse
Affiliation(s)
- David J Hughes
- CN Bio Innovations Limited, Welwyn Garden City AL73AX, UK
| | | | - Emma L Sceats
- CN Bio Innovations Limited, Welwyn Garden City AL73AX, UK
| |
Collapse
|