1
|
Urzì O, Gasparro R, Costanzo E, De Luca A, Giavaresi G, Fontana S, Alessandro R. Three-Dimensional Cell Cultures: The Bridge between In Vitro and In Vivo Models. Int J Mol Sci 2023; 24:12046. [PMID: 37569426 PMCID: PMC10419178 DOI: 10.3390/ijms241512046] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Although historically, the traditional bidimensional in vitro cell system has been widely used in research, providing much fundamental information regarding cellular functions and signaling pathways as well as nuclear activities, the simplicity of this system does not fully reflect the heterogeneity and complexity of the in vivo systems. From this arises the need to use animals for experimental research and in vivo testing. Nevertheless, animal use in experimentation presents various aspects of complexity, such as ethical issues, which led Russell and Burch in 1959 to formulate the 3R (Replacement, Reduction, and Refinement) principle, underlying the urgent need to introduce non-animal-based methods in research. Considering this, three-dimensional (3D) models emerged in the scientific community as a bridge between in vitro and in vivo models, allowing for the achievement of cell differentiation and complexity while avoiding the use of animals in experimental research. The purpose of this review is to provide a general overview of the most common methods to establish 3D cell culture and to discuss their promising applications. Three-dimensional cell cultures have been employed as models to study both organ physiology and diseases; moreover, they represent a valuable tool for studying many aspects of cancer. Finally, the possibility of using 3D models for drug screening and regenerative medicine paves the way for the development of new therapeutic opportunities for many diseases.
Collapse
Affiliation(s)
- Ornella Urzì
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Roberta Gasparro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Elisa Costanzo
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Angela De Luca
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche, 40136 Bologna, Italy; (A.D.L.); (G.G.)
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche, 40136 Bologna, Italy; (A.D.L.); (G.G.)
| | - Simona Fontana
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Riccardo Alessandro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| |
Collapse
|
2
|
Aasadollahei N, Rezaei N, Golroo R, Agarwal T, Vosough M, Piryaei A. Bioengineering liver microtissues for modeling non-alcoholic fatty liver disease. EXCLI JOURNAL 2023; 22:367-391. [PMID: 37223084 PMCID: PMC10201011 DOI: 10.17179/excli2022-5892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/16/2023] [Indexed: 05/25/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the world's most common chronic liver disease. However, due to the lack of reliable in vitro NAFLD models, drug development studies have faced many limitations, and there is no food and drug administration-approved medicine for NAFLD treatment. A functional biomimetic in vitro human liver model requires an optimized natural microenvironment using appropriate cellular composition, to provide constructive cell-cell interactions, and niche-specific bio-molecules to supply crucial cues as cell-matrix interplay. Such a suitable liver model could employ appropriate and desired biochemical, mechanical, and physical properties similar to native tissue. Moreover, bioengineered three-dimensional tissues, specially microtissues and organoids, and more recently using infusion-based cultivation systems such as microfluidics can mimic natural tissue conditions and facilitate the exchange of nutrients and soluble factors to improve physiological function in the in vitro generated constructs. This review highlights the key players involved in NAFLD initiation and progression and discussed the available cells and matrices for in vitro NAFLD modeling. The strategies for optimizing the liver microenvironment to generate a powerful and biomimetic in vitro NAFLD model were described as well. Finally, the current challenges and future perospective for promotion in this subject were discussed.
Collapse
Affiliation(s)
- Negar Aasadollahei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Niloufar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reihaneh Golroo
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tarun Agarwal
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
A comprehensive transcriptomic comparison of hepatocyte model systems improves selection of models for experimental use. Commun Biol 2022; 5:1094. [PMID: 36241695 PMCID: PMC9568534 DOI: 10.1038/s42003-022-04046-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/28/2022] [Indexed: 11/08/2022] Open
Abstract
The myriad of available hepatocyte in vitro models provides researchers the possibility to select hepatocyte-like cells (HLCs) for specific research goals. However, direct comparison of hepatocyte models is currently challenging. We systematically searched the literature and compared different HLCs, but reported functions were limited to a small subset of hepatic functions. To enable a more comprehensive comparison, we developed an algorithm to compare transcriptomic data across studies that tested HLCs derived from hepatocytes, biliary cells, fibroblasts, and pluripotent stem cells, alongside primary human hepatocytes (PHHs). This revealed that no HLC covered the complete hepatic transcriptome, highlighting the importance of HLC selection. HLCs derived from hepatocytes had the highest transcriptional resemblance to PHHs regardless of the protocol, whereas the quality of fibroblasts and PSC derived HLCs varied depending on the protocol used. Finally, we developed and validated a web application (HLCompR) enabling comparison for specific pathways and addition of new HLCs. In conclusion, our comprehensive transcriptomic comparison of HLCs allows selection of HLCs for specific research questions and can guide improvements in culturing conditions.
Collapse
|
4
|
Ahmed U, Ahmed R, Masoud MS, Tariq M, Ashfaq UA, Augustine R, Hasan A. Stem cells based in vitro models: trends and prospects in biomaterials cytotoxicity studies. Biomed Mater 2021; 16:042003. [PMID: 33686970 DOI: 10.1088/1748-605x/abe6d8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Advanced biomaterials are increasingly used for numerous medical applications from the delivery of cancer-targeted therapeutics to the treatment of cardiovascular diseases. The issues of foreign body reactions induced by biomaterials must be controlled for preventing treatment failure. Therefore, it is important to assess the biocompatibility and cytotoxicity of biomaterials on cell culture systems before proceeding to in vivo studies in animal models and subsequent clinical trials. Direct use of biomaterials on animals create technical challenges and ethical issues and therefore, the use of non-animal models such as stem cell cultures could be useful for determination of their safety. However, failure to recapitulate the complex in vivo microenvironment have largely restricted stem cell cultures for testing the cytotoxicity of biomaterials. Nevertheless, properties of stem cells such as their self-renewal and ability to differentiate into various cell lineages make them an ideal candidate for in vitro screening studies. Furthermore, the application of stem cells in biomaterials screening studies may overcome the challenges associated with the inability to develop a complex heterogeneous tissue using primary cells. Currently, embryonic stem cells, adult stem cells, and induced pluripotent stem cells are being used as in vitro preliminary biomaterials testing models with demonstrated advantages over mature primary cell or cell line based in vitro models. This review discusses the status and future directions of in vitro stem cell-based cultures and their derivatives such as spheroids and organoids for the screening of their safety before their application to animal models and human in translational research.
Collapse
Affiliation(s)
- Uzair Ahmed
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000 Punjab, Pakistan
| | | | | | | | | | | | | |
Collapse
|
5
|
Gurevich I, Burton SA, Munn C, Ohshima M, Goedland ME, Czysz K, Rajesh D. iPSC-derived hepatocytes generated from NASH donors provide a valuable platform for disease modeling and drug discovery. Biol Open 2020; 9:bio055087. [PMID: 33268331 PMCID: PMC7758638 DOI: 10.1242/bio.055087] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/16/2020] [Indexed: 12/17/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects 30-40% of adults and 10% of children in the US. About 20% of people with NAFLD develop non-alcoholic steatohepatitis (NASH), which may lead to cirrhosis and liver cancer, and is projected to be a leading cause of liver transplantation in the near future. Human induced pluripotent stem cells (iPSC) from NASH patients are useful for generating a large number of hepatocytes for NASH modeling applications and identification of potential drug targets. We developed a novel defined in vitro differentiation process to generate cryopreservable hepatocytes using an iPSC panel of NASH donors and apparently healthy normal (AHN) controls. iPSC-derived hepatocytes displayed stage specific phenotypic markers, hepatocyte morphology, with bile canaliculi. Importantly, both fresh and cryopreserved definitive endoderm and hepatoblasts successfully differentiated to pure and functional hepatocytes with increased CYP3A4 activity in response to rifampicin and lipid accumulation upon fatty acid (FA) treatment. End-stage hepatocytes integrated into three-dimensional (3D) liver organoids and demonstrated increased levels of albumin secretion compared to aggregates consisting of hepatocytes alone. End-stage hepatocytes derived from NASH donors demonstrated spontaneous lipidosis without FA supplementation, recapitulating a feature of NASH hepatocytes in vivo Cryopreserved hepatocytes generated by this protocol across multiple donors will provide a critical cell source to facilitate the fundamental understanding of NAFLD/NASH biology and potential high throughput screening applications for preclinical evaluation of therapeutic targets.
Collapse
Affiliation(s)
- Igor Gurevich
- Life Science R&D Division, FUJIFILM Cellular Dynamics, Inc., 525 Science Drive, Madison, WI 53711, USA
| | - Sarah A Burton
- Life Science R&D Division, FUJIFILM Cellular Dynamics, Inc., 525 Science Drive, Madison, WI 53711, USA
| | - Christie Munn
- Life Science R&D Division, FUJIFILM Cellular Dynamics, Inc., 525 Science Drive, Madison, WI 53711, USA
| | - Makiko Ohshima
- Life Science R&D Division, FUJIFILM Cellular Dynamics, Inc., 525 Science Drive, Madison, WI 53711, USA
| | - Madelyn E Goedland
- Life Science R&D Division, FUJIFILM Cellular Dynamics, Inc., 525 Science Drive, Madison, WI 53711, USA
| | - Katherine Czysz
- Life Science R&D Division, FUJIFILM Cellular Dynamics, Inc., 525 Science Drive, Madison, WI 53711, USA
| | - Deepika Rajesh
- Life Science R&D Division, FUJIFILM Cellular Dynamics, Inc., 525 Science Drive, Madison, WI 53711, USA
| |
Collapse
|
6
|
Okuyama S, Kawamura F, Kubiura M, Tsuji S, Osaki M, Kugoh H, Oshimura M, Kazuki Y, Tada M. Real-time fluorometric evaluation of hepatoblast proliferation in vivo and in vitro using the expression of CYP3A7 coding for human fetus-specific P450. Pharmacol Res Perspect 2020; 8:e00642. [PMID: 32886454 PMCID: PMC7507068 DOI: 10.1002/prp2.642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022] Open
Abstract
The fields of drug discovery and regenerative medicine require large numbers of adult human primary hepatocytes. For this purpose, it is desirable to use hepatocyte-like cells (HLCs) differentiated from human pluripotent stem cells (PSCs). Premature hepatoblast-like cells (HB-LCs) differentiated from PSCs provide an intermediate source and steady supply of newly mature HLCs. To develop an efficient HB-LC induction method, we constructed a red fluorescent reporter, CYP3A7R, in which DsRed is placed under the transcriptional control of CYP3A7 coding for a human fetus-type P450 enzyme. Before using this reporter in human cells, we created transgenic mice using mouse embryonic stem cells (ESCs) carrying a CYP3A7R transgene and confirmed that CYP3A7R was specifically expressed in fetal and newborn livers and reactivated in the adult liver in response to hepatic regeneration. Moreover, we optimized the induction procedure of HB-LCs from transgenic mouse ESCs using semi-quantitative fluorometric evaluation. Activation of Wnt signaling together with chromatin modulation prior to Activin A treatment greatly improved the induction efficiency of HB-LCs. BMP2 and 1.7% dimethyl sulfoxide induced selective proliferation of HB-LCs, which matured to HLCs. Therefore, CYP3A7R will provide a fluorometric evaluation system for high content screening of chemicals that induce HB-LC differentiation, hepatocyte regeneration, and hepatotoxicity when it is introduced into human PSCs.
Collapse
Affiliation(s)
- Shota Okuyama
- Stem Cells & Reprogramming LaboratoryDepartment of BiologyFaculty of ScienceToho UniversityFunabashiJapan
| | - Fumihiko Kawamura
- Stem Cells & Reprogramming LaboratoryDepartment of BiologyFaculty of ScienceToho UniversityFunabashiJapan
- Institute of Regenerative Medicine and BiofunctionGraduate School of Medical ScienceTottori UniversityYonagoJapan
| | - Musashi Kubiura
- Stem Cells & Reprogramming LaboratoryDepartment of BiologyFaculty of ScienceToho UniversityFunabashiJapan
| | - Saori Tsuji
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Mitsuhiko Osaki
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Hiroyuki Kugoh
- Institute of Regenerative Medicine and BiofunctionGraduate School of Medical ScienceTottori UniversityYonagoJapan
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Mitsuo Oshimura
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Yasuhiro Kazuki
- Institute of Regenerative Medicine and BiofunctionGraduate School of Medical ScienceTottori UniversityYonagoJapan
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Masako Tada
- Stem Cells & Reprogramming LaboratoryDepartment of BiologyFaculty of ScienceToho UniversityFunabashiJapan
| |
Collapse
|
7
|
Sengupta S, Johnson B, Seirup M, Ardalani H, Duffin B, Barrett-Wilt GA, Stewart R, Thomson JA. Co-culture with mouse embryonic fibroblasts improves maintenance of metabolic function of human small hepatocyte progenitor cells. Curr Res Toxicol 2020; 1:70-84. [PMID: 34345838 PMCID: PMC8320630 DOI: 10.1016/j.crtox.2020.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Derivation and culture of small hepatocyte progenitor cells (SHPCs) capable of proliferating in vitro has been described in rodents and recently in humans. These cells are capable of engrafting in injured livers, however, they display de-differentiated morphology and reduced xenobiotic metabolism activity in culture over passages. Here we report that SHPCs derived from adult primary human hepatocytes (PHHs) and cultured on mouse embryonic fibroblasts (MEFs) not only display differentiated morphology and exhibit gene expression profiles similar to adult PHHs, but importantly, they retain their phenotype over several passages. Further, unlike previous reports, where extensive manipulations of culture conditions are required to convert SHPCs to metabolically functional hepatocytes, SHPCs in our co-culture system maintain expression of xenobiotic metabolism-associated genes. We show that SHPCs in co-culture are able to perform xenobiotic metabolism at rates equal to their parent PHHs as evidenced by the metabolism of acetaminophen to all of its major metabolites. In summary, we present an improved co-culture system that allows generation of SHPCs from adult PHHs that maintain their differentiated phenotype over multiple passages. Our findings would be useful for expansion of limited PHHs for use in studies of drug metabolism and toxicity testing.
Collapse
Affiliation(s)
- Srikumar Sengupta
- Morgridge Institute for Research, Madison, WI, United States of America
| | - Brian Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America.,Institute for Quantitative Health Science and Engineering, Departments of Pharmacology & Toxicology and Biomedical Engineering, Michigan State University, East Lansing, MI, United States of America
| | - Morten Seirup
- Morgridge Institute for Research, Madison, WI, United States of America.,Dianomi Therapeutics, Madison, WI, United States of America
| | - Hamisha Ardalani
- Morgridge Institute for Research, Madison, WI, United States of America.,Beckman Coulter Life Sciences, San Jose, CA, United States of America
| | - Bret Duffin
- Morgridge Institute for Research, Madison, WI, United States of America
| | - Gregory A Barrett-Wilt
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI, United States of America
| | - James A Thomson
- Morgridge Institute for Research, Madison, WI, United States of America.,Department of Cell & Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America.,Department of Molecular, Cellular, & Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, United States of America
| |
Collapse
|
8
|
Khalil AS, Xie AW, Johnson HJ, Murphy WL. Sustained release and protein stabilization reduce the growth factor dosage required for human pluripotent stem cell expansion. Biomaterials 2020; 248:120007. [PMID: 32302801 PMCID: PMC8445021 DOI: 10.1016/j.biomaterials.2020.120007] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/24/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
Abstract
Translation of human pluripotent stem cell (hPSC)-derived therapies to the clinic demands scalable, cost-effective methods for cell expansion. Culture media currently used for hPSC expansion rely on high concentrations and frequent supplementation of recombinant growth factors due to their short half-life at physiological temperatures. Here, we developed a biomaterial strategy using mineral-coated microparticles (MCMs) to sustain delivery of basic fibroblast growth factor (bFGF), a thermolabile protein critical for hPSC pluripotency and proliferation. We show that the MCMs stabilize bFGF against thermally induced activity loss and provide more efficient sustained release of active growth factor compared to polymeric carriers commonly used for growth factor delivery. Using a statistically driven optimization approach called Design of Experiments, we generated a bFGF-loaded MCM formulation that supported hPSC expansion over 25 passages without the need for additional bFGF supplementation to the media, resulting in greater than 80% reduction in bFGF usage compared to standard approaches. This materials-based strategy to stabilize and sustain delivery of a thermolabile growth factor has broad potential to reduce costs associated with recombinant protein supplements in scalable biomanufacturing of emerging cell therapies.
Collapse
Affiliation(s)
- Andrew S Khalil
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Angela W Xie
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Hunter J Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA; Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
9
|
Guida P, Piscitelli E, Marrese M, Martino V, Cirillo V, Guarino V, Angeli E, Cocola C, Pelucchi P, Repetto L, Firpo G, Karnavas T, Gotte M, Gritzapis A, D'Albore M, Repetto D, Pezzuoli D, Missitzis I, Porta G, Bertalot G, Bellipanni G, Zucchi I, Ambrosio L, Valbusa U, Reinbold RA. Integrating Microstructured Electrospun Scaffolds in an Open Microfluidic System for in Vitro Studies of Human Patient-Derived Primary Cells. ACS Biomater Sci Eng 2020; 6:3649-3663. [PMID: 33463182 DOI: 10.1021/acsbiomaterials.0c00352] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Recent studies have suggested that microenvironmental stimuli play a significant role in regulating cellular proliferation and migration, as well as in modulating self-renewal and differentiation processes of mammary cells with stem cell (SCs) properties. Recent advances in micro/nanotechnology and biomaterial synthesis/engineering currently enable the fabrication of innovative tissue culture platforms suitable for maintenance and differentiation of SCs in vitro. Here, we report the design and fabrication of an open microfluidic device (OMD) integrating removable poly(ε-caprolactone) (PCL) based electrospun scaffolds, and we demonstrate that the OMD allows investigation of the behavior of human cells during in vitro culture in real time. Electrospun scaffolds with modified surface topography and chemistry can influence attachment, proliferation, and differentiation of mammary SCs and epigenetic mechanisms that maintain luminal cell identity as a function of specific morphological or biochemical cues imparted by tailor-made fiber post-treatments. Meanwhile, the OMD architecture allows control of cell seeding and culture conditions to collect more accurate and informative in vitro assays. In perspective, integrated systems could be tailor-made to mimic specific physiological conditions of the local microenvironment and then analyze the response from screening specific drugs for more effective diagnostics, long-term prognostics, and disease intervention in personalized medicine.
Collapse
Affiliation(s)
- Patrizia Guida
- Department of Physics, University of Genoa, via Dodecaneso 33, 16146 Genoa, Italy
| | - Eleonora Piscitelli
- Institute of Biomedical Technologies, National Research Council of Italy, via F.lli Cervi 93, 20090 Segrate, Milan, Italy
| | - Marica Marrese
- Institute of Composite and Biomedical Materials, National Research Council of Italy, Mostra D'Oltremare, Pad. 20, viale Kennedy 54, 80125 Naples, Italy
| | - Valentina Martino
- Institute of Biomedical Technologies, National Research Council of Italy, via F.lli Cervi 93, 20090 Segrate, Milan, Italy
| | - Valentina Cirillo
- Institute of Composite and Biomedical Materials, National Research Council of Italy, Mostra D'Oltremare, Pad. 20, viale Kennedy 54, 80125 Naples, Italy
| | - Vincenzo Guarino
- Institute for Polymers, Composites & Biomaterials, National Research Council of Italy, Mostra d'Oltremare, Pad. 20, viale Kennedy 54, 80125 Naples, Italy
| | - Elena Angeli
- Department of Physics, University of Genoa, via Dodecaneso 33, 16146 Genoa, Italy
| | - Cinzia Cocola
- Institute of Biomedical Technologies, National Research Council of Italy, via F.lli Cervi 93, 20090 Segrate, Milan, Italy.,Consorzio Italbiotech, via Fantoli 15/16, 20138 Milan, Italy
| | - Paride Pelucchi
- Institute of Biomedical Technologies, National Research Council of Italy, via F.lli Cervi 93, 20090 Segrate, Milan, Italy
| | - Luca Repetto
- Department of Physics, University of Genoa, via Dodecaneso 33, 16146 Genoa, Italy
| | - Giuseppe Firpo
- Department of Physics, University of Genoa, via Dodecaneso 33, 16146 Genoa, Italy
| | - Theodoros Karnavas
- Columbia University, Department of Genetics & Development, New York, 10032 United States
| | - Martin Gotte
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany
| | - Angelos Gritzapis
- Department of Breast Cancer Surgery, Hospital "Agios Savvas", Leoforos Alexandras Avenue, 171, 11522 Athens, Greece
| | - Marietta D'Albore
- Institute of Composite and Biomedical Materials, National Research Council of Italy, Mostra D'Oltremare, Pad. 20, viale Kennedy 54, 80125 Naples, Italy
| | - Diego Repetto
- Department of Physics, University of Genoa, via Dodecaneso 33, 16146 Genoa, Italy
| | - Denise Pezzuoli
- Department of Physics, University of Genoa, via Dodecaneso 33, 16146 Genoa, Italy
| | - Ioannis Missitzis
- Department of Breast Cancer Surgery, Hospital "Agios Savvas", Leoforos Alexandras Avenue, 171, 11522 Athens, Greece
| | - Giovanni Porta
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Giovanni Bertalot
- IEO, European Institute of Oncology IRCCS, via G Ripamonti, 435, 20141 Milan, Italy
| | - Gianfranco Bellipanni
- Center for Biotechnology, Sbarro Institute for Research and Molecular Medicine and Department of Biology, Temple University, Philadelphia 19122, United States
| | - Ileana Zucchi
- Institute of Biomedical Technologies, National Research Council of Italy, via F.lli Cervi 93, 20090 Segrate, Milan, Italy
| | - Luigi Ambrosio
- Institute for Polymers, Composites & Biomaterials, National Research Council of Italy, Mostra d'Oltremare, Pad. 20, viale Kennedy 54, 80125 Naples, Italy
| | - Ugo Valbusa
- Department of Physics, University of Genoa, via Dodecaneso 33, 16146 Genoa, Italy
| | - Rolland A Reinbold
- Institute of Biomedical Technologies, National Research Council of Italy, via F.lli Cervi 93, 20090 Segrate, Milan, Italy
| |
Collapse
|
10
|
Hu C, Zhao L, Wu Z, Li L. Transplantation of mesenchymal stem cells and their derivatives effectively promotes liver regeneration to attenuate acetaminophen-induced liver injury. Stem Cell Res Ther 2020; 11:88. [PMID: 32106875 PMCID: PMC7047366 DOI: 10.1186/s13287-020-01596-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/29/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
Acetaminophen (APAP)-induced injury is a common clinical phenomenon that not only occurs in a dose-dependent manner but also occurs in some idiosyncratic individuals in a dose-independent manner. APAP overdose generally results in acute liver injury via the initiation of oxidative stress, endoplasmic reticulum (ER) stress, autophagy, liver inflammation, and microcirculatory dysfunction. Liver transplantation is the only effective strategy for treating APAP-induced liver failure, but liver transplantation is inhibited by scarce availability of donor liver grafts, acute graft rejection, lifelong immunosuppression, and unbearable costs. Currently, N-acetylcysteine (NAC) effectively restores liver functions early after APAP intake, but it does not protect against APAP-induced injury at the late stage. An increasing number of animal studies have demonstrated that mesenchymal stem cells (MSCs) significantly attenuate acute liver injury through their migratory capacity, hepatogenic differentiation, immunoregulatory capacity, and paracrine effects in acute liver failure (ALF). In this review, we comprehensively discuss the mechanisms of APAP overdose-induced liver injury and current therapies for treating APAP-induced liver injury. We then comprehensively summarize recent studies about transplantation of MSC and MSC derivatives for treating APAP-induced liver injury. We firmly believe that MSCs and their derivatives will effectively promote liver regeneration and liver injury repair in APAP overdose-treated animals and patients. To this end, MSC-based therapies may serve as an effective strategy for patients who are waiting for liver transplantation during the early and late stages of APAP-induced ALF in the near future.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lingfei Zhao
- Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zhongwen Wu
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lanjuan Li
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
11
|
Messina A, Luce E, Hussein M, Dubart-Kupperschmitt A. Pluripotent-Stem-Cell-Derived Hepatic Cells: Hepatocytes and Organoids for Liver Therapy and Regeneration. Cells 2020; 9:cells9020420. [PMID: 32059501 PMCID: PMC7072243 DOI: 10.3390/cells9020420] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/04/2020] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
The liver is a very complex organ that ensures numerous functions; it is thus susceptible to multiple types of damage and dysfunction. Since 1983, orthotopic liver transplantation (OLT) has been considered the only medical solution available to patients when most of their liver function is lost. Unfortunately, the number of patients waiting for OLT is worryingly increasing, and extracorporeal liver support devices are not yet able to counteract the problem. In this review, the current and expected methodologies in liver regeneration are briefly analyzed. In particular, human pluripotent stem cells (hPSCs) as a source of hepatic cells for liver therapy and regeneration are discussed. Principles of hPSC differentiation into hepatocytes are explored, along with the current limitations that have led to the development of 3D culture systems and organoid production. Expected applications of these organoids are discussed with particular attention paid to bio artificial liver (BAL) devices and liver bio-fabrication.
Collapse
Affiliation(s)
- Antonietta Messina
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
| | - Eléanor Luce
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
| | - Marwa Hussein
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
| | - Anne Dubart-Kupperschmitt
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
- Correspondence: ; Tel.: +33-145595138
| |
Collapse
|
12
|
Ardalani H, Sengupta S, Harms V, Vickerman V, Thomson JA, Murphy WL. 3-D culture and endothelial cells improve maturity of human pluripotent stem cell-derived hepatocytes. Acta Biomater 2019; 95:371-381. [PMID: 31362140 DOI: 10.1016/j.actbio.2019.07.047] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 07/19/2019] [Accepted: 07/25/2019] [Indexed: 12/18/2022]
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived hepatocytes (iHEP) offer an attractive alternative to primary human hepatocytes (PHH) for drug toxicity studies, as PHHs are limited in supply, vary in their metabolic activity between donors, and rapidly lose their functionality in vitro. However, one of the major drawbacks with iHEP cells in drug safety studies is their decreased phenotypic maturity, with lower liver specific enzyme activity compared with that of PHH. Here we evaluated the effects of 3D culture and non-parenchymal cells on the maturation of iHEPs. We describe a serum-free, chemically defined 3D in vitro model using iHEP cells, which is compatible with automation and conventional assay plates. The iHEP cells cultured in this model form polarized aggregates with functional bile canaliculi and strongly increased expression of albumin, urea and genes encoding phase I and II drug metabolism enzymes and bile transporters. Cytochrome P450-mediated metabolism is significantly higher in 3D iHEP aggregates compared to 2D iHEP culture. Furthermore, addition of human liver sinusoidal endothelial cells (sECs) and iPS-derived endothelial cells (iECs) improved mature hepatocyte function and CYP450 enzyme activity. Also, ECs formed endothelial networks within the hepatic 3D cultures, mimicking aspects of an in vivo architecture. Collectively, these results suggest that the iHEP/EC aggregates described here may have the potential to be used for many applications, including as an in vitro model to study liver diseases associated with sinusoidal endothelial cells. STATEMENT OF SIGNIFICANCE: iPS-derived hepatocytes provide an inexhaustible source of cells for drug screening, toxicology studies and cell-based therapies, but lack mature phenotype of adult primary human hepatocytes (PHH). Herein, we show that 3D culture of iPS-derived hepatocytes and their co-culture with human sinusoidal endothelial cells (sECs) to improve their maturity.
Collapse
Affiliation(s)
- Hamisha Ardalani
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA; Morgridge Institute for Research, Madison, WI, USA
| | | | - Victoria Harms
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, WI, USA
| | | | - James A Thomson
- Morgridge Institute for Research, Madison, WI, USA; Cell and Regenerative Biology, University of Wisconsin-Madison, WI, USA; Department of Molecular, Cellular, and Developmental Biology, University of California-Santa Barbara, CA, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, WI, USA.
| |
Collapse
|
13
|
Kim TW, Che JH, Yun JW. Use of stem cells as alternative methods to animal experimentation in predictive toxicology. Regul Toxicol Pharmacol 2019; 105:15-29. [DOI: 10.1016/j.yrtph.2019.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022]
|
14
|
Natale A, Vanmol K, Arslan A, Van Vlierberghe S, Dubruel P, Van Erps J, Thienpont H, Buzgo M, Boeckmans J, De Kock J, Vanhaecke T, Rogiers V, Rodrigues RM. Technological advancements for the development of stem cell-based models for hepatotoxicity testing. Arch Toxicol 2019; 93:1789-1805. [PMID: 31037322 DOI: 10.1007/s00204-019-02465-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/18/2019] [Indexed: 02/07/2023]
Abstract
Stem cells are characterized by their self-renewal capacity and their ability to differentiate into multiple cell types of the human body. Using directed differentiation strategies, stem cells can now be converted into hepatocyte-like cells (HLCs) and therefore, represent a unique cell source for toxicological applications in vitro. However, the acquired hepatic functionality of stem cell-derived HLCs is still significantly inferior to primary human hepatocytes. One of the main reasons for this is that most in vitro models use traditional two-dimensional (2D) setups where the flat substrata cannot properly mimic the physiology of the human liver. Therefore, 2D-setups are progressively being replaced by more advanced culture systems, which attempt to replicate the natural liver microenvironment, in which stem cells can better differentiate towards HLCs. This review highlights the most recent cell culture systems, including scaffold-free and scaffold-based three-dimensional (3D) technologies and microfluidics that can be employed for culture and hepatic differentiation of stem cells intended for hepatotoxicity testing. These methodologies have shown to improve in vitro liver cell functionality according to the in vivo liver physiology and allow to establish stem cell-based hepatic in vitro platforms for the accurate evaluation of xenobiotics.
Collapse
Affiliation(s)
- Alessandra Natale
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Koen Vanmol
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | - Aysu Arslan
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Jürgen Van Erps
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | - Hugo Thienpont
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | | | - Joost Boeckmans
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Joery De Kock
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Vera Rogiers
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Robim M Rodrigues
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
15
|
Novel in vitro dynamic metabolic system for predicting the human pharmacokinetics of tolbutamide. Acta Pharmacol Sin 2018; 39:1522-1532. [PMID: 29644999 DOI: 10.1038/aps.2017.201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/09/2017] [Indexed: 12/12/2022] Open
Abstract
Liver metabolism is commonly considered the major determinant in drug discovery and development. Many in vitro drug metabolic studies have been developed and applied to understand biotransformation. However, these methods have disadvantages, resulting in inconsistencies between in vivo and in vitro experiments. A major factor is that they are static systems that do not consider the transport process in the liver. Here we developed an in vitro dynamic metabolic system (Bio-PK metabolic system) to mimic the human pharmacokinetics of tolbutamide. Human liver microsomes (HLMs) encapsulated in a F127'-Acr-Bis hydrogel (FAB hydrogel) were placed in the incubation system. A microdialysis sampling technique was used to monitor the metabolic behavior of tolbutamide in hydrogels. The measured results in the system were used to fit the in vitro intrinsic clearance of tolbutamide with a mathematical model. Then, a PBPK model that integrated the corresponding in vitro intrinsic clearance was developed to verify the system. Compared to the traditional incubation method, reasonable PK profiles and the in vivo clearance of tolbutamide could be predicted by integrating the intrinsic clearance of tolbutamide obtained from the Bio-PK metabolic system into the PBPK model. The predicted maximum concentration (Cmax), area under the concentration-time curve (AUC), time to reach the maximum plasma concentration (Tmax) and in vivo clearance were consistent with the clinically observed data. This novel in vitro dynamic metabolic system can compensate for some limitations of traditional incubation methods; it may provide a new method for screening compounds and predicting pharmacokinetics in the early stages, supporting the development of compounds.
Collapse
|
16
|
Davis BM, Loghin ER, Conway KR, Zhang X. Automated Closed-System Expansion of Pluripotent Stem Cell Aggregates in a Rocking-Motion Bioreactor. SLAS Technol 2018; 23:364-373. [PMID: 29481762 DOI: 10.1177/2472630318760745] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pluripotent stem cell suspension aggregates have proven to be an efficient and phenotypically stable means for expansion and directed differentiation. Bioreactor systems with automation of perfusion, fluidization, and gas exchange are essential for scaling up pluripotent stem cell cultures. Since stem cell pluripotency and differentiation are affected by both chemical and physical signals, we investigated a low-shear method for the expansion of cells in a rocking-motion bioreactor. The rocking motion drives continual mixing and aeration, and the single-use disposable bioreactors avoid issues around contamination during seeding, medium exchange, passage, and cell harvest. Serial passaging from a 150 mL to a 1 L scale was demonstrated, achieving cell densities of up to 4 million cells/mL. In an average of 13 experiments, pluripotent stem cell aggregates expanded 5.7-fold (with maximal 9.5-fold expansion) and maintained 97% viability over 4 days in a rocking bioreactor culture. In seven experiments with improved culture conditions, the average expansion was 6.8-fold. Maintenance of pluripotency was confirmed by differentiation to all three germ layers and surface marker expression, and the expanded aggregates maintained a stable normal karyotype. The automation associated with the rocking platform bioreactor required no user intervention during the 4-day culture, providing hands-off expansion of pluripotent stem cells.
Collapse
Affiliation(s)
- Brian M Davis
- 1 GE Global Research Center, Biosciences Organization, Niskayuna, NY, USA
| | - Evelina R Loghin
- 1 GE Global Research Center, Biosciences Organization, Niskayuna, NY, USA
| | - Kenneth R Conway
- 1 GE Global Research Center, Biosciences Organization, Niskayuna, NY, USA
| | - Xiaohua Zhang
- 2 GE Global Research Center, Mechanical Systems Organization, Niskayuna, NY, USA
| |
Collapse
|
17
|
Delp J, Gutbier S, Cerff M, Zasada C, Niedenführ S, Zhao L, Smirnova L, Hartung T, Borlinghaus H, Schreiber F, Bergemann J, Gätgens J, Beyss M, Azzouzi S, Waldmann T, Kempa S, Nöh K, Leist M. Stage-specific metabolic features of differentiating neurons: Implications for toxicant sensitivity. Toxicol Appl Pharmacol 2017; 354:64-80. [PMID: 29278688 DOI: 10.1016/j.taap.2017.12.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 01/08/2023]
Abstract
Developmental neurotoxicity (DNT) may be induced when chemicals disturb a key neurodevelopmental process, and many tests focus on this type of toxicity. Alternatively, DNT may occur when chemicals are cytotoxic only during a specific neurodevelopmental stage. The toxicant sensitivity is affected by the expression of toxicant targets and by resilience factors. Although cellular metabolism plays an important role, little is known how it changes during human neurogenesis, and how potential alterations affect toxicant sensitivity of mature vs. immature neurons. We used immature (d0) and mature (d6) LUHMES cells (dopaminergic human neurons) to provide initial answers to these questions. Transcriptome profiling and characterization of energy metabolism suggested a switch from predominantly glycolytic energy generation to a more pronounced contribution of the tricarboxylic acid cycle (TCA) during neuronal maturation. Therefore, we used pulsed stable isotope-resolved metabolomics (pSIRM) to determine intracellular metabolite pool sizes (concentrations), and isotopically non-stationary 13C-metabolic flux analysis (INST 13C-MFA) to calculate metabolic fluxes. We found that d0 cells mainly use glutamine to fuel the TCA. Furthermore, they rely on extracellular pyruvate to allow continuous growth. This metabolic situation does not allow for mitochondrial or glycolytic spare capacity, i.e. the ability to adapt energy generation to altered needs. Accordingly, neuronal precursor cells displayed a higher sensitivity to several mitochondrial toxicants than mature neurons differentiated from them. In summary, this study shows that precursor cells lose their glutamine dependency during differentiation while they gain flexibility of energy generation and thereby increase their resistance to low concentrations of mitochondrial toxicants.
Collapse
Affiliation(s)
- Johannes Delp
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Simon Gutbier
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Martin Cerff
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich 52425, Germany
| | - Christin Zasada
- Max-Delbrück-Center of Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Sebastian Niedenführ
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich 52425, Germany
| | - Liang Zhao
- Johns Hopkins University, Bloomberg School of Public Health, Center for Alternatives to Animal Testing (CAAT), Baltimore, MD, USA
| | - Lena Smirnova
- Johns Hopkins University, Bloomberg School of Public Health, Center for Alternatives to Animal Testing (CAAT), Baltimore, MD, USA
| | - Thomas Hartung
- Johns Hopkins University, Bloomberg School of Public Health, Center for Alternatives to Animal Testing (CAAT), Baltimore, MD, USA
| | - Hanna Borlinghaus
- Department of Computer and Information Science, University of Konstanz, Konstanz, Germany
| | - Falk Schreiber
- Department of Computer and Information Science, University of Konstanz, Konstanz, Germany; Faculty of Information Technology, Monash University, Melbourne, Australia
| | - Jörg Bergemann
- Department of Life Sciences, Albstadt-Sigmaringen University of Applied Sciences, Sigmaringen, Germany
| | - Jochem Gätgens
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich 52425, Germany
| | - Martin Beyss
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich 52425, Germany
| | - Salah Azzouzi
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich 52425, Germany
| | - Tanja Waldmann
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Stefan Kempa
- Max-Delbrück-Center of Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Katharina Nöh
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich 52425, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; CAAT-Europe, University of Konstanz, Konstanz 78457, Germany.
| |
Collapse
|
18
|
CYP3A5 Mediates Effects of Cocaine on Human Neocorticogenesis: Studies using an In Vitro 3D Self-Organized hPSC Model with a Single Cortex-Like Unit. Neuropsychopharmacology 2017; 42:774-784. [PMID: 27534267 PMCID: PMC5240177 DOI: 10.1038/npp.2016.156] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 05/26/2016] [Accepted: 08/10/2016] [Indexed: 12/31/2022]
Abstract
Because of unavoidable confounding variables in the direct study of human subjects, it has been difficult to unravel the effects of prenatal cocaine exposure on the human fetal brain, as well as the cellular and biochemical mechanisms involved. Here, we propose a novel approach using a human pluripotent stem cell (hPSC)-based 3D neocortical organoid model. This model retains essential features of human neocortical development by encompassing a single self-organized neocortical structure, without including an animal-derived gelatinous matrix. We reported previously that prenatal cocaine exposure to rats during the most active period of neural progenitor proliferation induces cytoarchitectural changes in the embryonic neocortex. We also identified a role of CYP450 and consequent oxidative ER stress signaling in these effects. However, because of differences between humans and rodents in neocorticogenesis and brain CYP metabolism, translation of the research findings from the rodent model to human brain development is uncertain. Using hPSC 3D neocortical organoids, we demonstrate that the effects of cocaine are mediated through CYP3A5-induced generation of reactive oxygen species, inhibition of neocortical progenitor cell proliferation, induction of premature neuronal differentiation, and interruption of neural tissue development. Furthermore, knockdown of CYP3A5 reversed these cocaine-induced pathological phenotypes, suggesting CYP3A5 as a therapeutic target to mitigate the deleterious neurodevelopmental effects of prenatal cocaine exposure in humans. Moreover, 3D organoid methodology provides an innovative platform for identifying adverse effects of abused psychostimulants and pharmaceutical agents, and can be adapted for use in neurodevelopmental disorders with genetic etiologies.
Collapse
|
19
|
Goldring C, Antoine DJ, Bonner F, Crozier J, Denning C, Fontana RJ, Hanley NA, Hay DC, Ingelman-Sundberg M, Juhila S, Kitteringham N, Silva-Lima B, Norris A, Pridgeon C, Ross JA, Sison Young R, Tagle D, Tornesi B, van de Water B, Weaver RJ, Zhang F, Park BK. Stem cell-derived models to improve mechanistic understanding and prediction of human drug-induced liver injury. Hepatology 2017; 65:710-721. [PMID: 27775817 PMCID: PMC5266558 DOI: 10.1002/hep.28886] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/01/2016] [Indexed: 01/12/2023]
Abstract
Current preclinical drug testing does not predict some forms of adverse drug reactions in humans. Efforts at improving predictability of drug-induced tissue injury in humans include using stem cell technology to generate human cells for screening for adverse effects of drugs in humans. The advent of induced pluripotent stem cells means that it may ultimately be possible to develop personalized toxicology to determine interindividual susceptibility to adverse drug reactions. However, the complexity of idiosyncratic drug-induced liver injury means that no current single-cell model, whether of primary liver tissue origin, from liver cell lines, or derived from stem cells, adequately emulates what is believed to occur during human drug-induced liver injury. Nevertheless, a single-cell model of a human hepatocyte which emulates key features of a hepatocyte is likely to be valuable in assessing potential chemical risk; furthermore, understanding how to generate a relevant hepatocyte will also be critical to efforts to build complex multicellular models of the liver. Currently, hepatocyte-like cells differentiated from stem cells still fall short of recapitulating the full mature hepatocellular phenotype. Therefore, we convened a number of experts from the areas of preclinical and clinical hepatotoxicity and safety assessment, from industry, academia, and regulatory bodies, to specifically explore the application of stem cells in hepatotoxicity safety assessment and to make recommendations for the way forward. In this short review, we particularly discuss the importance of benchmarking stem cell-derived hepatocyte-like cells to their terminally differentiated human counterparts using defined phenotyping, to make sure the cells are relevant and comparable between labs, and outline why this process is essential before the cells are introduced into chemical safety assessment. (Hepatology 2017;65:710-721).
Collapse
Affiliation(s)
- Christopher Goldring
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Daniel J. Antoine
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | | | - Jonathan Crozier
- European Partnership for Alternative Approaches to Animal Testing (EPAA), Brussels, Belgium
| | - Chris Denning
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Robert J. Fontana
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Neil A. Hanley
- Centre for Endocrinology & Diabetes, University of Manchester; Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre Manchester, UK
| | - David C. Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, UK
| | | | - Satu Juhila
- R&D, In Vitro Biology, Orion Pharma, Espoo, Finland
| | - Neil Kitteringham
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | | | - Alan Norris
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Chris Pridgeon
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - James A. Ross
- MRC Centre for Regenerative Medicine, University of Edinburgh, UK
| | - Rowena Sison Young
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Danilo Tagle
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Belen Tornesi
- Abbvie Global Pharmaceutical Research and Development, North Chicago, IL, USA
| | - Bob van de Water
- Faculty of Science, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, University of Leiden, Netherlands
| | - Richard J. Weaver
- Institut de Recherches Internationales Servier (I.R.I.S), Suresnes, 92284, Cedex France
| | - Fang Zhang
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - B. Kevin Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| |
Collapse
|
20
|
Yao X, Yin N, Faiola F. Stem cell toxicology: a powerful tool to assess pollution effects on human health. Natl Sci Rev 2016. [DOI: 10.1093/nsr/nww089] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AbstractEnvironmental pollution is a global problem; the lack of comprehensive toxicological assessments may lead to increased health risks. To fully understand the health effects of pollution, it is paramount to implement fast, efficient and specific toxicity screening that relies on human models rather than on time-consuming, expensive and often inaccurate tests involving live animals. Human stem cell toxicology represents a valid alternative to traditional toxicity assays because it takes advantage of the ability of stem cells to differentiate into multiple cell types and tissues of the human body. Thus, this branch of toxicology provides a possibility to assess cellular, embryonic, developmental, reproductive and functional toxicity in vitro within a single system highly relevant to human physiology. In this review, we describe the development, performance and future perspectives of stem cell toxicology, with an emphasis on how it can meet the increasing challenges posed by environmental pollution in the modern world.
Collapse
Affiliation(s)
- Xinglei Yao
- Stake Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nuoya Yin
- Stake Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Francesco Faiola
- Stake Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
21
|
Takayama K, Mizuguchi H. Generation of human pluripotent stem cell-derived hepatocyte-like cells for drug toxicity screening. Drug Metab Pharmacokinet 2016; 32:12-20. [PMID: 28012798 DOI: 10.1016/j.dmpk.2016.10.408] [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] [Received: 09/07/2016] [Revised: 10/03/2016] [Accepted: 10/17/2016] [Indexed: 01/22/2023]
Abstract
Because drug-induced liver injury is one of the main reasons for drug development failures, it is important to perform drug toxicity screening in the early phase of pharmaceutical development. Currently, primary human hepatocytes are most widely used for the prediction of drug-induced liver injury. However, the sources of primary human hepatocytes are limited, making it difficult to supply the abundant quantities required for large-scale drug toxicity screening. Therefore, there is an urgent need for a novel unlimited, efficient, inexpensive, and predictive model which can be applied for large-scale drug toxicity screening. Human embryonic stem (ES) cells and induced pluripotent stem (iPS) cells are able to replicate indefinitely and differentiate into most of the body's cell types, including hepatocytes. It is expected that hepatocyte-like cells generated from human ES/iPS cells (human ES/iPS-HLCs) will be a useful tool for drug toxicity screening. To apply human ES/iPS-HLCs to various applications including drug toxicity screening, homogenous and functional HLCs must be differentiated from human ES/iPS cells. In this review, we will introduce the current status of hepatocyte differentiation technology from human ES/iPS cells and a novel method to predict drug-induced liver injury using human ES/iPS-HLCs.
Collapse
Affiliation(s)
- Kazuo Takayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; The Keihanshin Consortium for Fostering the Next Generation of Global Leaders in Research (K-CONNEX), Kyoto University, Kyoto 606-8302, Japan; PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan; Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan.
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
22
|
Chitrangi S, Nair P, Khanna A. 3D engineered In vitro hepatospheroids for studying drug toxicity and metabolism. Toxicol In Vitro 2016; 38:8-18. [PMID: 27794450 DOI: 10.1016/j.tiv.2016.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/14/2016] [Accepted: 10/24/2016] [Indexed: 01/29/2023]
Abstract
Drug toxicity is one of the reasons for late stage drug attrition, because of hepatotoxicity. Various in vitro liver models like primary human hepatocytes, immortalized human hepatic cell lines, liver slices and microsomes have been used; but limited by viability, hepatic gene expression and function. The 3D-engineered construct of hepatocyte-like-cells (HLCs) differentiated from stem cells, may provide a limitless source of hepatocytes with improved reproducibility. Towards this end, we used hepatospheroids (diameter=50-80μm) differentiated from human-umbilical-cord-mesenchymal stem cells (hUC-MSCs) on 3D scaffold GEVAC (Gelatin-vinyl-acetate-copolymer) as in vitro model for studying drug metabolism/toxicity. Our data demonstrated that hUC-MSCs-derived-hepatospheroids cultured on GEVAC expressed significantly higher drug-metabolizing enzymes (CYPs) both at mRNA and activity level compared to 2D culture, using HR-LC/MS. We further showed that hepatospheroids convert phenacetin (by CYP1A2) and testosterone (by CYP3A4) to their human-specific metabolites acetaminophen and 6β-hydroxytestosterone with a predictive clearance rate of 0.011ml/h/106 cells and 0.021ml/h/106 cells respectively, according to first-order kinetics. Hepatotoxicity was confirmed by exposing hepatospheroids to ethanol and acetaminophen; ROS generation, cell viability, cytoskeleton structure, elevation of liver function enzymes, i.e. AST and ALT, was analyzed. To the best of our knowledge, this is the first report to use hUC-MSCs-derived-hepatospheroids on GEVAC as in vitro model for drug metabolism/toxicity study; which can replace the conventional 2D-models used in drug development.
Collapse
Affiliation(s)
- Swati Chitrangi
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM'S NMIMS (Deemed-to-be ) University, V. L Mehta road, Vile Parle (West), Mumbai 400056, Maharashtra, India
| | - Prabha Nair
- Division of Tissue Engineering and Regeneration Technologies, Biomedical Technology Wing, Shree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695012, Kerala, India
| | - Aparna Khanna
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM'S NMIMS (Deemed-to-be ) University, V. L Mehta road, Vile Parle (West), Mumbai 400056, Maharashtra, India.
| |
Collapse
|
23
|
Chaudhari P, Prasad N, Tian L, Jang YY. Determination of Functional Activity of Human iPSC-Derived Hepatocytes by Measurement of CYP Metabolism. Methods Mol Biol 2016; 1357:383-94. [PMID: 25410290 DOI: 10.1007/7651_2014_145] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The advent of induced pluripotent stem cell (iPSC) technology has enabled the modeling of an array of specific human disease phenotypes, aiding in the increasingly important and indispensable understanding of disease progression and pathogenesis. Pluripotent stem cell-derived hepatocytes present a new avenue for drug screening and personalized drug testing toward precision medicine. CYP450 microsomal enzymes play a critical role in drug metabolism. Hence, CYP activity measurement of iPSC-derived hepatocytes is a vital prerequisite, to ensure metabolic functionality before proceeding to drug testing. Herein, we describe the protocol for measurement of different CYP450 enzyme activities in human iPSC-derived hepatocytes.
Collapse
Affiliation(s)
- Pooja Chaudhari
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Neha Prasad
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Lipeng Tian
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yoon-Young Jang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
24
|
Lauschke VM, Hendriks DFG, Bell CC, Andersson TB, Ingelman-Sundberg M. Novel 3D Culture Systems for Studies of Human Liver Function and Assessments of the Hepatotoxicity of Drugs and Drug Candidates. Chem Res Toxicol 2016; 29:1936-1955. [DOI: 10.1021/acs.chemrestox.6b00150] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Volker M. Lauschke
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Delilah F. G. Hendriks
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Catherine C. Bell
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Tommy B. Andersson
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Cardiovascular
and Metabolic Diseases, Innovative Medicines and Early Development
Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - Magnus Ingelman-Sundberg
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| |
Collapse
|
25
|
Cipriano M, Freyer N, Knöspel F, Oliveira NG, Barcia R, Cruz PE, Cruz H, Castro M, Santos JM, Zeilinger K, Miranda JP. Self-assembled 3D spheroids and hollow-fibre bioreactors improve MSC-derived hepatocyte-like cell maturation in vitro. Arch Toxicol 2016; 91:1815-1832. [PMID: 27590069 DOI: 10.1007/s00204-016-1838-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/24/2016] [Indexed: 01/11/2023]
Abstract
3D cultures of human stem cell-derived hepatocyte-like cells (HLCs) have emerged as promising models for short- and long-term maintenance of hepatocyte phenotype in vitro cultures by better resembling the in vivo environment of the liver and consequently increase the translational value of the resulting data. In this study, the first stage of hepatic differentiation of human neonatal mesenchymal stem cells (hnMSCs) was performed in 2D monolayer cultures for 17 days. The second stage was performed by either maintaining cells in 2D cultures for an extra 10 days, as control, or alternatively cultured in 3D as self-assembled spheroids or in multicompartment membrane bioreactor system. All systems enabled hnMSC differentiation into HLCs as shown by positive immune staining of hepatic markers CK-18, HNF-4α, albumin, the hepatic transporters OATP-C and MRP-2 as well as drug-metabolizing enzymes like CYP1A2 and CYP3A4. Similarly, all models also displayed relevant glucose, phase I and phase II metabolism, the ability to produce albumin and to convert ammonia into urea. However, EROD activity and urea production were increased in both 3D systems. Moreover, the spheroids revealed higher bupropion conversion, whereas bioreactor showed increased albumin production and capacity to biotransform diclofenac. Additionally, diclofenac resulted in an IC50 value of 1.51 ± 0.05 and 0.98 ± 0.03 in 2D and spheroid cultures, respectively. These data suggest that the 3D models tested improved HLC maturation showing a relevant biotransformation capacity and thus provide more appropriate reliable models for mechanistic studies and more predictive systems for in vitro toxicology applications.
Collapse
Affiliation(s)
- Madalena Cipriano
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Nora Freyer
- Bioreactor Group, Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Fanny Knöspel
- Bioreactor Group, Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Nuno G Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Rita Barcia
- ECBio S.A., Rua Henrique Paiva Couceiro, Nº 27, 2700-451, Amadora, Portugal
| | - Pedro E Cruz
- ECBio S.A., Rua Henrique Paiva Couceiro, Nº 27, 2700-451, Amadora, Portugal
| | - Helder Cruz
- ECBio S.A., Rua Henrique Paiva Couceiro, Nº 27, 2700-451, Amadora, Portugal
| | - Matilde Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Jorge M Santos
- ECBio S.A., Rua Henrique Paiva Couceiro, Nº 27, 2700-451, Amadora, Portugal
| | - Katrin Zeilinger
- Bioreactor Group, Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Joana P Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
26
|
Gómez-Lechón MJ, Tolosa L. Human hepatocytes derived from pluripotent stem cells: a promising cell model for drug hepatotoxicity screening. Arch Toxicol 2016; 90:2049-2061. [PMID: 27325232 DOI: 10.1007/s00204-016-1756-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 06/09/2016] [Indexed: 01/09/2023]
Abstract
Drug-induced liver injury (DILI) is a frequent cause of failure in both clinical and post-approval stages of drug development, and poses a key challenge to the pharmaceutical industry. Current animal models offer poor prediction of human DILI. Although several human cell-based models have been proposed for the detection of human DILI, human primary hepatocytes remain the gold standard for preclinical toxicological screening. However, their use is hindered by their limited availability, variability and phenotypic instability. In contrast, pluripotent stem cells, which include embryonic and induced pluripotent stem cells (iPSCs), proliferate extensively in vitro and can be differentiated into hepatocytes by the addition of soluble factors. This provides a stable source of hepatocytes for multiple applications, including early preclinical hepatotoxicity screening. In addition, iPSCs also have the potential to establish genotype-specific cells from different individuals, which would increase the predictivity of toxicity assays allowing more successful clinical trials. Therefore, the generation of human hepatocyte-like cells derived from pluripotent stem cells seems to be promising for overcoming limitations of hepatocyte preparations, and it is expected to have a substantial repercussion in preclinical hepatotoxicity risk assessment in early drug development stages.
Collapse
Affiliation(s)
- María José Gómez-Lechón
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe de Valencia, Torre A, 6ª Planta, Avenida Fernando Abril Martorell 106, 46026, Valencia, Spain.,CIBERehd, FIS, 08036, Barcelona, Spain
| | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe de Valencia, Torre A, 6ª Planta, Avenida Fernando Abril Martorell 106, 46026, Valencia, Spain.
| |
Collapse
|
27
|
Hansel MC, Davila JC, Vosough M, Gramignoli R, Skvorak KJ, Dorko K, Marongiu F, Blake W, Strom SC. The Use of Induced Pluripotent Stem Cells for the Study and Treatment of Liver Diseases. ACTA ACUST UNITED AC 2016; 67:14.13.1-14.13.27. [PMID: 26828329 DOI: 10.1002/0471140856.tx1413s67] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Liver disease is a major global health concern. Liver cirrhosis is one of the leading causes of death in the world and currently the only therapeutic option for end-stage liver disease (e.g., acute liver failure, cirrhosis, chronic hepatitis, cholestatic diseases, metabolic diseases, and malignant neoplasms) is orthotropic liver transplantation. Transplantation of hepatocytes has been proposed and used as an alternative to whole organ transplant to stabilize and prolong the lives of patients in some clinical cases. Although these experimental therapies have demonstrated promising and beneficial results, their routine use remains a challenge due to the shortage of donor livers available for cell isolation, variable quality of those tissues, the potential need for lifelong immunosuppression in the transplant recipient, and high costs. Therefore, new therapeutic strategies and more reliable clinical treatments are urgently needed. Recent and continuous technological advances in the development of stem cells suggest they may be beneficial in this respect. In this review, we summarize the history of stem cell and induced pluripotent stem cell (iPSC) technology in the context of hepatic differentiation and discuss the potential applications the technology may offer for human liver disease modeling and treatment. This includes developing safer drugs and cell-based therapies to improve the outcomes of patients with currently incurable health illnesses. We also review promising advances in other disease areas to highlight how the stem cell technology could be applied to liver diseases in the future. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Marc C Hansel
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| | - Julio C Davila
- Department of Biochemistry, University of Puerto Rico School of Medicine, Medical Sciences Campus, San Juan, Puerto Rico
| | - Massoud Vosough
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kristen J Skvorak
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kenneth Dorko
- Department of Pharmacology, Toxicology and Therapeutics, Kansas University Medical Center, Kansas City, Kansas
| | - Fabio Marongiu
- Department of Biomedical Sciences, Section of Experimental Pathology, Unit of Experimental Medicine, University of Cagliari, Cagliari, Italy
| | - William Blake
- Genetically Modified Models Center of Emphasis, Pfizer, Groton, Connecticut
| | - Stephen C Strom
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania.,Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
28
|
Galán A, Simón C. Monitoring Stemness in Long-Term hESC Cultures by Real-Time PCR. Methods Mol Biol 2016; 1307:89-104. [PMID: 25403464 DOI: 10.1007/7651_2014_131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Human embryonic stem cells (hESC) involve long-term cultures that must remain undifferentiated. The real-time PCR (RT-PCR) technique allows the relative quantification of target genes, including undifferentiation and differentiation markers when referred to a housekeeping control with the addition of a calibrator that serves as an internal control to compare different lots of reactions during the time. The main aspects will include a minimal number of cells to be analyzed, genes to be tested, and how to choose the appropriate calibrator sample and the reference gene. In this chapter, we present how to apply the RT-PCR technique, protocols for its performance, experimental setup, and software analysis, as of the gene expression of hESC lines in consecutive passages for long-term culture surveillance.
Collapse
Affiliation(s)
- Amparo Galán
- Gene Expression and RNA Metabolism, Prince Felipe Research Center (CIPF), Valencia, Spain
| | | |
Collapse
|
29
|
Araki T, Iwazaki N, Ishiguro N, Sakamoto A, Nagata K, Ohbuchi M, Moriguchi H, Motoi M, Shinkyo R, Homma T, Sakamoto S, Iwase Y, Ise R, Nakanishi Y, Uto M, Inoue T. Requirements for human iPS cell-derived hepatocytes as an alternative to primary human hepatocytes for assessing absorption, distribution, metabolism, excretion and toxicity of pharmaceuticals. ACTA ACUST UNITED AC 2016. [DOI: 10.2131/fts.3.89] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Tetsuro Araki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Non-Clinical Evaluation Expert Committee, Drug Evaluation Committee, Japan Pharmaceutical Manufacturers Association
- Laboratory for Safety Assessment and ADME, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation
| | - Norihiko Iwazaki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- DMPK Research Laboratories, Mitsubishi Tanabe Pharma Corporation
| | - Naoki Ishiguro
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Pharmacokinetics and Non-Clinical Safety, Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co., Ltd
| | - Atsushi Sakamoto
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Pharmacokinetics and Non-Clinical Safety, Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co., Ltd
| | - Keisuke Nagata
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Drug Safety Research Laboratories, Astellas Pharma Inc
| | - Masato Ohbuchi
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Analysis & Pharmacokinetics Research Laboratories, Astellas Pharma Inc
| | - Hiroyuki Moriguchi
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Analysis & Pharmacokinetics Research Laboratories, Astellas Pharma Inc
| | - Makiko Motoi
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co., Ltd
| | - Raku Shinkyo
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co., Ltd
| | - Toshiki Homma
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Kissei Pharmaceutical Co., Ltd
| | - Sakae Sakamoto
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Kissei Pharmaceutical Co., Ltd
| | - Yumiko Iwase
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Non-Clinical Evaluation Expert Committee, Drug Evaluation Committee, Japan Pharmaceutical Manufacturers Association
- Mitsubishi Tanabe Pharma Corporation
| | - Ryota Ise
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Shin Nippon Biomedical Laboratories, Ltd
| | - Yasuharu Nakanishi
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd
| | - Masahiro Uto
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd
| | - Tomoaki Inoue
- Consortium for Safety Assessment using Human iPS Cells (CSAHi)
- Non-Clinical Evaluation Expert Committee, Drug Evaluation Committee, Japan Pharmaceutical Manufacturers Association
- Safety Assessment Department, Research Division, Chugai Pharmaceutical Co., Ltd
| |
Collapse
|
30
|
Abstract
Attrition due to nonclinical safety represents a major issue for the productivity of pharmaceutical research and development (R&D) organizations, especially during the compound optimization stages of drug discovery and the early stages of clinical development. Focusing on decreasing nonclinical safety-related attrition is not a new concept, and various approaches have been experimented with over the last two decades. Front-loading testing funnels in Discovery with in vitro toxicity assays designed to rapidly identify unfavorable molecules was the approach adopted by most pharmaceutical R&D organizations a few years ago. However, this approach has also a non-negligible opportunity cost. Hence, significant refinements to the "fail early, fail often" paradigm have been proposed recently to reflect the complexity of accurately categorizing compounds with early data points without taking into account other important contextual aspects, in particular efficacious systemic and tissue exposures. This review provides an overview of toxicology approaches and models that can be used in pharmaceutical Discovery at the series/lead identification and lead optimization stages to guide and inform chemistry efforts, as well as a personal view on how to best use them to meet nonclinical safety-related attrition objectives consistent with a sustainable pharmaceutical R&D model. The scope of this review is limited to small molecules, as large molecules are associated with challenges that are quite different. Finally, a perspective on how several emerging technologies may impact toxicity evaluation is also provided.
Collapse
Affiliation(s)
- Eric A G Blomme
- Global Preclinical Safety, AbbVie Inc. , 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Yvonne Will
- Drug Safety Research and Development, Pfizer , Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
31
|
Elanzew A, Sommer A, Pusch-Klein A, Brüstle O, Haupt S. A reproducible and versatile system for the dynamic expansion of human pluripotent stem cells in suspension. Biotechnol J 2015; 10:1589-99. [PMID: 26110829 DOI: 10.1002/biot.201400757] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 03/13/2015] [Accepted: 06/23/2015] [Indexed: 11/11/2022]
Abstract
Reprogramming of patient cells to human induced pluripotent stem cells (hiPSC) has facilitated in vitro disease modeling studies aiming at deciphering the molecular and cellular mechanisms that contribute to disease pathogenesis and progression. To fully exploit the potential of hiPSC for biomedical applications, technologies that enable the standardized generation and expansion of hiPSC from large numbers of donors are required. Paralleled automated processes for the expansion of hiPSC could provide an opportunity to maximize the generation of hiPSC collections from patient cohorts while minimizing hands-on time and costs. In order to develop a simple method for the parallel expansion of human pluripotent stem cells (hPSC) we established a protocol for their cultivation as undifferentiated aggregates in a bench-top bioreactor system (BioLevitator™). We show that long-term expansion (10 passages) of hPSCs either in mTeSR or E8 medium preserved a normal karyotype, three-germ-layer differentiation potential and high expression of pluripotency-associated markers. The system enables the expansion from low inoculation densities (0.3 × 10(5) cells/mL) and provides a simplified, cost-efficient and time-saving method for the provision of hiPSC at midi-scale. Implementation of this protocol in cell production schemes has the potential to advance cell manufacturing in many areas of hiPSC-based medical research.
Collapse
Affiliation(s)
- Andreas Elanzew
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, Bonn, Germany.,LIFE&BRAIN GmbH, Bonn, Germany
| | | | | | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, Bonn, Germany. .,LIFE&BRAIN GmbH, Bonn, Germany.
| | - Simone Haupt
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, Bonn, Germany. .,LIFE&BRAIN GmbH, Bonn, Germany.
| |
Collapse
|
32
|
Curran CS, Carrillo ER, Ponik SM, Keely PJ. Collagen density regulates xenobiotic and hypoxic response of mammary epithelial cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:114-124. [PMID: 25481308 PMCID: PMC4323890 DOI: 10.1016/j.etap.2014.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 10/27/2014] [Indexed: 06/04/2023]
Abstract
Breast density, where collagen I is the dominant component, is a significant breast cancer risk factor. Cell surface integrins interact with collagen, activate focal adhesion kinase (FAK), and downstream cell signals associated with xenobiotics (AhR, ARNT) and hypoxia (HIF-1α, ARNT). We examined if mammary cells cultured in high density (HD) or low density (LD) collagen gels affected xenobiotic or hypoxic responses. ARNT production was significantly reduced by HD culture and in response to a FAK inhibitor. Consistent with a decrease in ARNT, AhR and HIF-1α reporter activation and VEGF production was lower in HD compared to LD. However, P450 production was enhanced in HD and induced by AhR and HIF-1α agonists, possibly in response to increased NF-κB activaton. Thus, collagen density differentially regulates downstream cell signals of AhR and HIF-1α by modulating the activity of FAK, the release of NF-κB transcriptional factors, and the levels of ARNT.
Collapse
Affiliation(s)
- Colleen S Curran
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| | - Esteban R Carrillo
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| | - Patricia J Keely
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| |
Collapse
|