151
|
A Novel Orthotopic Liver Cancer Model for Creating a Human-like Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13163997. [PMID: 34439154 PMCID: PMC8394300 DOI: 10.3390/cancers13163997] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma is the most common form of liver cancer. The lack of models that resemble actual tumor development in patients, limits the research to improve the diagnosis rate and develop new treatments. This study describes a novel mouse model that involves organoid formation and an implantation technique. This mouse model shares human genetic profiles and factors around the tumor, resembling the actual tumor development in patients. We demonstrate the roles of different cell types around the tumor, in promoting tumor growth, using this model. This model will be useful to understand the tumor developmental process, drug testing, diagnosis, prognosis, and treatment development. Abstract Hepatocellular carcinoma (HCC) is the most common form of liver cancer. This study aims to develop a new method to generate an HCC mouse model with a human tumor, and imitates the tumor microenvironment (TME) of clinical patients. Here, we have generated functional, three-dimensional sheet-like human HCC organoids in vitro, using luciferase-expressing Huh7 cells, human iPSC-derived endothelial cells (iPSC-EC), and human iPSC-derived mesenchymal cells (iPSC-MC). The HCC organoid, capped by ultra-purified alginate gel, was implanted into the disrupted liver using an ultrasonic homogenizer in the immune-deficient mouse, which improved the survival and engraftment rate. We successfully introduced different types of controllable TME into the model and studied the roles of TME in HCC tumor growth. The results showed the role of the iPSC-EC and iPSC-MC combination, especially the iPSC-MC, in promoting HCC growth. We also demonstrated that liver fibrosis could promote HCC tumor growth. However, it is not affected by non-alcoholic fatty liver disease. Furthermore, the implantation of HCC organoids to humanized mice demonstrated that the immune response is important in slowing down tumor growth at an early stage. In conclusion, we have created an HCC model that is useful for studying HCC development and developing new treatment options in the future.
Collapse
|
152
|
Cao D, Ge JY, Wang Y, Oda T, Zheng YW. Hepatitis B virus infection modeling using multi-cellular organoids derived from human induced pluripotent stem cells. World J Gastroenterol 2021; 27:4784-4801. [PMID: 34447226 PMCID: PMC8371505 DOI: 10.3748/wjg.v27.i29.4784] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/30/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic infection with hepatitis B virus (HBV) remains a global health concern despite the availability of vaccines. To date, the development of effective treatments has been severely hampered by the lack of reliable, reproducible, and scalable in vitro modeling systems that precisely recapitulate the virus life cycle and represent virus-host interactions. With the progressive understanding of liver organogenesis mechanisms, the development of human induced pluripotent stem cell (iPSC)-derived hepatic sources and stromal cellular compositions provides novel strategies for personalized modeling and treatment of liver disease. Further, advancements in three-dimensional culture of self-organized liver-like organoids considerably promote in vitro modeling of intact human liver tissue, in terms of both hepatic function and other physiological characteristics. Combined with our experiences in the investigation of HBV infections using liver organoids, we have summarized the advances in modeling reported thus far and discussed the limitations and ongoing challenges in the application of liver organoids, particularly those with multi-cellular components derived from human iPSCs. This review provides general guidelines for establishing clinical-grade iPSC-derived multi-cellular organoids in modeling personalized hepatitis virus infection and other liver diseases, as well as drug testing and transplantation therapy.
Collapse
Affiliation(s)
- Di Cao
- Institute of Regenerative Medicine and Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Jian-Yun Ge
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and School of Biotechnology and Heath Sciences, Wuyi University, Jiangmen 529020, Guangdong Province, China
| | - Yun Wang
- Institute of Regenerative Medicine and Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Yun-Wen Zheng
- Institute of Regenerative Medicine and Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and School of Biotechnology and Heath Sciences, Wuyi University, Jiangmen 529020, Guangdong Province, China
- School of Medicine, Yokohama City University, Yokohama 234-0006, Kanagawa, Japan
| |
Collapse
|
153
|
Lam DTUH, Dan YY, Chan YS, Ng HH. Emerging liver organoid platforms and technologies. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:27. [PMID: 34341842 PMCID: PMC8329140 DOI: 10.1186/s13619-021-00089-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/17/2021] [Indexed: 06/13/2023]
Abstract
Building human organs in a dish has been a long term goal of researchers in pursue of physiologically relevant models of human disease and for replacement of worn out and diseased organs. The liver has been an organ of interest for its central role in regulating body homeostasis as well as drug metabolism. An accurate liver replica should contain the multiple cell types found in the organ and these cells should be spatially organized to resemble tissue structures. More importantly, the in vitro model should recapitulate cellular and tissue level functions. Progress in cell culture techniques and bioengineering approaches have greatly accelerated the development of advance 3-dimensional (3D) cellular models commonly referred to as liver organoids. These 3D models described range from single to multiple cell type containing cultures with diverse applications from establishing patient-specific liver cells to modeling of chronic liver diseases and regenerative therapy. Each organoid platform is advantageous for specific applications and presents its own limitations. This review aims to provide a comprehensive summary of major liver organoid platforms and technologies developed for diverse applications.
Collapse
Affiliation(s)
- Do Thuy Uyen Ha Lam
- Laboratory of precision disease therapeutics, Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore
| | - Yock Young Dan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore
- Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Hospital, 5 Lower Kent Ridge Road, Singapore, 119074, Singapore
| | - Yun-Shen Chan
- Laboratory of precision disease therapeutics, Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
| | - Huck-Hui Ng
- Laboratory of precision disease therapeutics, Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore.
- Department of Biochemistry, National University of Singapore, Singapore, 117559, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore.
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117597, Singapore.
| |
Collapse
|
154
|
Luce E, Messina A, Duclos-Vallée JC, Dubart-Kupperschmitt A. Advanced Techniques and Awaited Clinical Applications for Human Pluripotent Stem Cell Differentiation into Hepatocytes. Hepatology 2021; 74:1101-1116. [PMID: 33420753 PMCID: PMC8457237 DOI: 10.1002/hep.31705] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/16/2020] [Accepted: 12/19/2020] [Indexed: 12/22/2022]
Abstract
Liver transplantation is currently the only curative treatment for several liver diseases such as acute liver failure, end-stage liver disorders, primary liver cancers, and certain genetic conditions. Unfortunately, despite improvements to transplantation techniques, including live donor transplantation, the number of organs available remains insufficient to meet patient needs. Hepatocyte transplantation has enabled some encouraging results as an alternative to organ transplantation, but primary hepatocytes are little available and cannot be amplified using traditional two-dimensional culture systems. Indeed, although recent studies have tended to show that three-dimensional culture enables long-term hepatocyte culture, it is still agreed that, like most adult primary cell types, hepatocytes remain refractory to in vitro expansion. Because of their exceptional properties, human pluripotent stem cells (hPSCs) can be amplified indefinitely and differentiated into any cell type, including liver cells. While many teams have worked on hepatocyte differentiation, there has been a consensus that cells obtained after hPSC differentiation have more fetal than adult hepatocyte characteristics. New technologies have been used to improve the differentiation process in recent years. This review discusses the technical improvements made to hepatocyte differentiation protocols and the clinical approaches developed to date and anticipated in the near future.
Collapse
Affiliation(s)
- Eléanor Luce
- INSERMUniversité Paris-SaclayUnité Mixte de Recherche (UMR_S) 1193VillejuifFrance.,Fédération Hospitalo-Universitaire HépatinovHôpital Paul-BrousseVillejuifFrance
| | - Antonietta Messina
- INSERMUniversité Paris-SaclayUnité Mixte de Recherche (UMR_S) 1193VillejuifFrance.,Fédération Hospitalo-Universitaire HépatinovHôpital Paul-BrousseVillejuifFrance
| | - Jean-Charles Duclos-Vallée
- INSERMUniversité Paris-SaclayUnité Mixte de Recherche (UMR_S) 1193VillejuifFrance.,Fédération Hospitalo-Universitaire HépatinovHôpital Paul-BrousseVillejuifFrance
| | - Anne Dubart-Kupperschmitt
- INSERMUniversité Paris-SaclayUnité Mixte de Recherche (UMR_S) 1193VillejuifFrance.,Fédération Hospitalo-Universitaire HépatinovHôpital Paul-BrousseVillejuifFrance
| |
Collapse
|
155
|
Tsuchida T, Murata S, Hasegawa S, Mikami S, Enosawa S, Hsu HC, Fukuda A, Okamoto S, Mori A, Matsuo M, Kawakatsu Y, Matsunari H, Nakano K, Nagashima H, Taniguchi H. Investigation of Clinical Safety of Human iPS Cell-Derived Liver Organoid Transplantation to Infantile Patients in Porcine Model. Cell Transplant 2021; 29:963689720964384. [PMID: 33103476 PMCID: PMC7784600 DOI: 10.1177/0963689720964384] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transplantation of liver organoids has been investigated as a treatment alternative to liver transplantation for chronic liver disease. Transportal approach can be considered as a method of delivering organoids to the liver. It is important to set the allowable organoid amount and verify translocation by intraportal transplantation. We first examined the transplantation tolerance and translocation of porcine fetal liver-derived allogeneic organoids using piglets. Fetal liver-derived organoids generated from the Kusabira Orange-transduced pig were transplanted to the 10-day-old piglet liver through the left branch of the portal vein. All recipients survived without any observable adverse events. In contrast, both local and main portal pressures increased transiently during transplantation. In necropsy samples, Kusabira Orange-positive donor cells were detected primarily in the target lobe of the liver and partly in other areas, including the lungs and brain. As we confirmed the transplantation allowance by porcine fetal liver-derived organoids, we performed intraportal transplantation of human-induced pluripotent stem cell (iPSC)-derived liver organoid, which we plan to use in clinical trials, and portal pressure and translocation were investigated. Human iPSC-derived liver organoids were transplanted into the same 10-day-old piglet. Portal hypertension and translocation of human iPSC-derived liver organoids to the lungs were observed in one of two transplanted animals. Translocation occurred in the piglet in which patent ductus venosus (PDV) was observed. Therefore, a 28-day-old piglet capable of surgically ligating PDV was used, and after the PDV was ligated, human iPSC-derived liver organoids with the amount of which is scheduled in clinical trials were transplanted. This procedure inhibited the translocation of human iPSC-derived liver organoids to extrahepatic sites without no portal hypertension. In conclusion, human iPSC-derived liver organoids can be safely transplanted through the portal vein. Ligation of the ductus venosus prior to transplantation was effective in inhibiting extrahepatic translocation in newborns and infants.
Collapse
Affiliation(s)
- Tomonori Tsuchida
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Soichiro Murata
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shunsuke Hasegawa
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoshi Mikami
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shin Enosawa
- Division for Advanced Medical Sciences, National Center for Child Health and Development, Tokyo, Japan
| | - Huai-Che Hsu
- Division for Advanced Medical Sciences, National Center for Child Health and Development, Tokyo, Japan
| | - Akinari Fukuda
- Department of Transplantation Surgery, Organ Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akihiro Mori
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Megumi Matsuo
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yumi Kawakatsu
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hitomi Matsunari
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Kazuaki Nakano
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Hiroshi Nagashima
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Division of Regenerative Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
156
|
Abdalkader R, Chaleckis R, Meister I, Zhang P, Wheelock CE, Kamei KI. Untargeted LC-MS Metabolomics for the Analysis of Micro-scaled Extracellular Metabolites from Hepatocytes. ANAL SCI 2021; 37:1049-1052. [PMID: 33342928 DOI: 10.2116/analsci.20n032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Metabolome analysis in micro physiological models is a challenge due to the low volume of the cell culture medium (CCM). Here, we report a LC-MS-based untargeted metabolomics protocol for the detection of hepatocyte extracellular metabolites from micro-scale samples of CCM. Using a single LC-MS method we have detected 57 metabolites of which 27 showed >2-fold shifts after 72-hour incubation. We demonstrate that micro-scale CCM samples can be used for modelling micro-physiological temporal dynamics in metabolite intensities.
Collapse
Affiliation(s)
- Rodi Abdalkader
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Romanas Chaleckis
- Gunma University Initiative for Advanced Research (GIAR), Gunma University.,Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institute
| | - Isabel Meister
- Gunma University Initiative for Advanced Research (GIAR), Gunma University.,Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institute
| | - Pei Zhang
- Gunma University Initiative for Advanced Research (GIAR), Gunma University.,Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institute
| | - Craig E Wheelock
- Gunma University Initiative for Advanced Research (GIAR), Gunma University.,Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institute
| | - Ken-Ichiro Kamei
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University.,Wuya College of Innovation, Shenyang Pharmaceutical University
| |
Collapse
|
157
|
Khajavi M, Hashemi M, Kalalinia F. Recent advances in optimization of liver decellularization procedures used for liver regeneration. Life Sci 2021; 281:119801. [PMID: 34229008 DOI: 10.1016/j.lfs.2021.119801] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
Severe liver diseases have been considered the most common causes of adult deaths worldwide. Until now, liver transplantation is known as the only effective treatment for end stage liver disease. However, it is associated with several problems, most importantly, the side effects of immunosuppressive drugs that should be used after transplantation, and the shortage of tissue donors compared to the increasing number of patients requiring liver transplantation. Currently, tissue/organ decellularization as a new approach in tissue engineering is becoming a valid substitute for managing these kinds of problems. Decellularization of a whole liver is an attractive procedure to create three-dimensional (3D) scaffolds that micro-architecturally and structurally are similar to the native one and could support the repair or replacement of damaged or injured tissue. In this review, the different methods used for decellularization of liver tissue have been reviewed. In addition, the current approaches to overcome the challenges in these techniques are discussed.
Collapse
Affiliation(s)
- Mohaddeseh Khajavi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Hashemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Kalalinia
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
158
|
Engineering the Vasculature of Stem-Cell-Derived Liver Organoids. Biomolecules 2021; 11:biom11070966. [PMID: 34208902 PMCID: PMC8301828 DOI: 10.3390/biom11070966] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 12/28/2022] Open
Abstract
The vasculature of stem-cell-derived liver organoids can be engineered using methods that recapitulate embryonic liver development. Hepatic organoids with a vascular network offer great application prospects for drug screening, disease modeling, and therapeutics. However, the application of stem cell-derived organoids is hindered by insufficient vascularization and maturation. Here, we review different theories about the origin of hepatic cells and the morphogenesis of hepatic vessels to provide potential approaches for organoid generation. We also review the main protocols for generating vascularized liver organoids from stem cells and consider their potential and limitations in the generation of vascularized liver organoids.
Collapse
|
159
|
Sağraç D, Şişli HB, Şenkal S, Hayal TB, Şahin F, Doğan A. Organoids in Tissue Transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:45-64. [PMID: 34164796 DOI: 10.1007/5584_2021_647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Improvements in stem cell-based research and genetic modification tools enable stem cell-based tissue regeneration applications in clinical therapies. Although inadequate cell numbers in culture, invasive isolation procedures, and poor survival rates after transplantation remain as major challenges, cell-based therapies are useful tools for tissue regeneration.Organoids hold a great promise for tissue regeneration, organ and disease modeling, drug testing, development, and genetic profiling studies. Establishment of 3D cell culture systems eliminates the disadvantages of 2D models in terms of cell adaptation and tissue structure and function. Organoids possess the capacity to mimic the specific features of tissue architecture, cell-type composition, and the functionality of real organs while preserving the advantages of simplified and easily accessible cell culture models. Thus, organoid technology might emerge as an alternative to cell and tissue transplantation. Although transplantation of various organoids in animal models has been demonstrated, liöitations related to vascularized structure formation, cell viability and functionality remain as obstacles in organoid-based transplantation therapies. Clinical applications of organoid-based transplantations might be possible in the near future, when limitations related to cell viability and tissue integration are solved. In this review, the literature was analyzed and discussed to explore the current status of organoid-based transplantation studies.
Collapse
Affiliation(s)
- Derya Sağraç
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Hatice Burcu Şişli
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Selinay Şenkal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Taha Bartu Hayal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
160
|
Zhu X, Zhang B, He Y, Bao J. Liver Organoids: Formation Strategies and Biomedical Applications. Tissue Eng Regen Med 2021; 18:573-585. [PMID: 34132985 DOI: 10.1007/s13770-021-00357-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 02/05/2023] Open
Abstract
The liver is the most important digestive organ in the body. Several studies have explored liver biology and diseases related to the liver. However, most of these studies have only explored liver development, mechanism of liver regeneration and pathophysiology of liver diseases mainly based on two-dimensional (2D) cell lines and animal models. Traditional 2D cell lines do not represent the complex three-dimensional tissue architecture whereas animal models are limited by inter-species differences. These shortcomings limit understanding of liver biology and diseases. Liver organoid technology is effective in elucidating structural and physiological characteristics and basic tissue-level functions of liver tissue. In this review, formation strategies and a wide range of applications in biomedicine of liver organoid are summarized. Liver organoids are derived from single type cell culture, such as induced pluripotent stem cells (iPSCs), adult stem cells, primary hepatocytes, and primary cholangiocytes and multi-type cells co-culture, such as iPSC-derived hepatic endoderm cells co-cultured with mesenchymal stem cells and umbilical cord-derived endothelial cells. In vitro studies report that liver organoids are a promising model for regenerative medicine, organogenesis, liver regeneration, disease modelling, drug screening and personalized treatment. Liver organoids are a promising in vitro model for basic research and for development of clinical therapeutic interventions for hepatopathy.
Collapse
Affiliation(s)
- Xinglong Zhu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan Province, China
| | - Bingqi Zhang
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan Province, China
| | - Yuting He
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan Province, China
| | - Ji Bao
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
161
|
Ogoke O, Yousef O, Ott C, Kalinousky A, Lin W, Shamul C, Ross S, Parashurama N. Modeling Liver Organogenesis by Recreating Three-Dimensional Collective Cell Migration: A Role for TGFβ Pathway. Front Bioeng Biotechnol 2021; 9:621286. [PMID: 34211963 PMCID: PMC8239196 DOI: 10.3389/fbioe.2021.621286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 04/21/2021] [Indexed: 12/29/2022] Open
Abstract
Three-dimensional (3D) collective cell migration (CCM) is critical for improving liver cell therapies, eliciting mechanisms of liver disease, and modeling human liver development and organogenesis. Mechanisms of CCM differ in 2D vs. 3D systems, and existing models are limited to 2D or transwell-based systems, suggesting there is a need for improved 3D models of CCM. To recreate liver 3D CCM, we engineered in vitro 3D models based upon a morphogenetic transition that occurs during liver organogenesis, which occurs rapidly between E8.5 and E9.5 (mouse). During this morphogenetic transition, 3D CCM exhibits co-migration (multiple cell types), thick-strand interactions with surrounding septum transversum mesenchyme (STM), branching morphogenesis, and 3D interstitial migration. Here, we engineer several 3D in vitro culture systems, each of which mimics one of these processes in vitro. In mixed spheroids bearing both liver cells and uniquely MRC-5 (fetal lung) fibroblasts, we observed evidence of co-migration, and a significant increase in length and number of liver spheroid protrusions, which was highly sensitive to transforming growth factor beta 1 (TGFβ1) stimulation. In MRC-5-conditioned medium (M-CM) experiments, we observed dose-dependent branching morphogenesis associated with an upregulation of Twist1, which was inhibited by a broad TGFβ inhibitor. In models in which liver spheroids and MRC-5 spheroids were co-cultured, we observed complex strand morphogenesis, whereby thin, linear, 3D liver cell strands attach to the MRC-5 spheroid, anchor and thicken to form permanent and thick anchoring contacts between the two spheroids. In these spheroid co-culture models, we also observed spheroid fusion and strong evidence for interstitial migration. In conclusion, we present several novel cultivation systems that recreate distinct features of liver 3D CCM. These methodologies will greatly improve our molecular, cellular, and tissue-scale understanding of liver organogenesis, liver diseases like cancer, and liver cell therapy, and will also serve as a tool to bridge conventional 2D studies and preclinical in vivo studies.
Collapse
Affiliation(s)
- Ogechi Ogoke
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, United States
| | - Osama Yousef
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, United States
| | - Cortney Ott
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, United States
| | - Allison Kalinousky
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, United States
| | - Wayne Lin
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, United States
| | - Claire Shamul
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, United States
| | - Shatoni Ross
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, United States
| | - Natesh Parashurama
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, United States.,Department of Biomedical Engineering, University at Buffalo (State University of New York), Buffalo, NY, United States.,Clinical and Translational Research Center, University at Buffalo (State University of New York), Buffalo, NY, United States
| |
Collapse
|
162
|
Tanimizu N, Ichinohe N, Sasaki Y, Itoh T, Sudo R, Yamaguchi T, Katsuda T, Ninomiya T, Tokino T, Ochiya T, Miyajima A, Mitaka T. Generation of functional liver organoids on combining hepatocytes and cholangiocytes with hepatobiliary connections ex vivo. Nat Commun 2021; 12:3390. [PMID: 34099675 PMCID: PMC8185093 DOI: 10.1038/s41467-021-23575-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
In the liver, the bile canaliculi of hepatocytes are connected to intrahepatic bile ducts lined with cholangiocytes, which remove cytotoxic bile from the liver tissue. Although liver organoids have been reported, it is not clear whether the functional connection between hepatocytes and cholangiocytes is recapitulated in those organoids. Here, we report the generation of a hepatobiliary tubular organoid (HBTO) using mouse hepatocyte progenitors and cholangiocytes. Hepatocytes form the bile canalicular network and secrete metabolites into the canaliculi, which are then transported into the biliary tubular structure. Hepatocytes in HBTO acquire and maintain metabolic functions including albumin secretion and cytochrome P450 activities, over the long term. In this study, we establish functional liver tissue incorporating a bile drainage system ex vivo. HBTO enable us to reproduce the transport of hepatocyte metabolites in liver tissue, and to investigate the way in which the two types of epithelial cells establish functional connections. Combining mouse hepatocyte progenitors and cholangiocytes ex vivo, the authors form an organoid that can drain bile ex vivo and transport metabolites, as in the liver.
Collapse
Affiliation(s)
- Naoki Tanimizu
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Norihisa Ichinohe
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasushi Sasaki
- Biology Division, Department of Liberal Arts and Sciences, Center for Medical Education, Sapporo Medical University, Sapporo, Japan.,Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tohru Itoh
- Laboratory of Stem Cell Therapy, The Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Ryo Sudo
- Department of System Design Engineering, Keio University, Yokohama, Japan
| | - Tomoko Yamaguchi
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Takeshi Katsuda
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Takafumi Ninomiya
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Tokino
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Atsushi Miyajima
- Laboratory of Stem Cell Therapy, The Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Toshihiro Mitaka
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
163
|
Danoy M, Tauran Y, Poulain S, Jellali R, Bruce J, Leduc M, Le Gall M, Koui Y, Arakawa H, Gilard F, Gakiere B, Kato Y, Plessy C, Kido T, Miyajima A, Sakai Y, Leclerc E. Investigation of the hepatic development in the coculture of hiPSCs-derived LSECs and HLCs in a fluidic microenvironment. APL Bioeng 2021; 5:026104. [PMID: 34027283 PMCID: PMC8116060 DOI: 10.1063/5.0041227] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/02/2021] [Indexed: 12/29/2022] Open
Abstract
Interactions between the different liver cell types are critical to the maintenance or induction of their function in vitro. In this work, human-induced Pluripotent Stem Cells (hiPSCs)-derived Liver Sinusoidal Endothelial Cells (LSECs) and Hepatocytes-Like Cells (HLCs) were cultured and matured in a microfluidic environment. Both cell populations were differentiated in Petri dishes, detached, and inoculated in microfluidic biochips. In cocultures of both cell types, the tissue has exhibited a higher production of albumin (3.19 vs 5.31 μg/mL/106 cells in monocultures and cocultures) as well as a higher inducibility CYP450 over monocultures of HLCs. Tubular-like structures composed of LSECs and positive for the endothelial marker PECAM1, as well as a tissue more largely expressing Stabilin-2 were detected in cocultures only. In contrast, monocultures exhibited no network and less specific endothelial markers. The transcriptomic analysis did not reveal a marked difference between the profiles of both culture conditions. Nevertheless, the analysis allowed us to highlight different upstream regulators in cocultures (SP1, EBF1, and GATA3) and monocultures (PML, MECP2, and NRF1). In cocultures, the multi-omics dataset after 14 days of maturation in biochips has shown the activation of signaling related to hepatic maturation, angiogenesis, and tissue repair. In this condition, inflammatory signaling was also found to be reduced when compared to monocultures as illustrated by the activation of NFKB and by the detection of several cytokines involved in tissue injury in the latter. Finally, the extracted biological processes were discussed regarding the future development of a new generation of human in vitro hepatic models.
Collapse
Affiliation(s)
- Mathieu Danoy
- Authors to whom correspondence should be addressed: and
| | | | - Stephane Poulain
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Rachid Jellali
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu, CS 60319, 60203 Compiègne Cedex, Compiegne, France
| | - Johanna Bruce
- Plateforme protéomique 3P5, Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Marjorie Leduc
- Plateforme protéomique 3P5, Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Morgane Le Gall
- Plateforme protéomique 3P5, Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Yuta Koui
- Laboratory of Stem Cell Therapy, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Hiroshi Arakawa
- Laboratory of Molecular Pharmacokinetics, Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa City, Ishikawa 920-1192, Japan
| | - Francoise Gilard
- Institute of Plant Sciences Paris-Saclay (IPS2), UMR 9213/UMR1403, CNRS, INRA, Université Paris-Sud, Université d'Evry, Université Paris-Diderot, Sorbonne Paris-Cité, Saclay Plant Sciences, Bâtiment 630, 91405 Orsay, France
| | - Bertrand Gakiere
- Institute of Plant Sciences Paris-Saclay (IPS2), UMR 9213/UMR1403, CNRS, INRA, Université Paris-Sud, Université d'Evry, Université Paris-Diderot, Sorbonne Paris-Cité, Saclay Plant Sciences, Bâtiment 630, 91405 Orsay, France
| | - Yukio Kato
- Laboratory of Molecular Pharmacokinetics, Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa City, Ishikawa 920-1192, Japan
| | - Charles Plessy
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Taketomo Kido
- Laboratory of Stem Cell Therapy, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Atsushi Miyajima
- Laboratory of Stem Cell Therapy, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, graduate school of Engineering, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Eric Leclerc
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
164
|
Yao T, Zhang Y, Lv M, Zang G, Ng SS, Chen X. Advances in 3D cell culture for liver preclinical studies. Acta Biochim Biophys Sin (Shanghai) 2021; 53:643-651. [PMID: 33973620 DOI: 10.1093/abbs/gmab046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Indexed: 11/13/2022] Open
Abstract
The 3D cell culture model is an indispensable tool in the study of liver biology in the field of health and disease and the development of clinically relevant products for liver therapies. The 3D culture model captures critical factors of the microenvironmental niche required by hepatocytes for exhibiting optimal phenotypes, thus enabling the pursuit of a range of preclinical studies that are not entirely feasible in conventional 2D cell models. In this review, we highlight the major attributes associated with and the components needed for the development of a functional 3D liver culture model for a range of applications.
Collapse
Affiliation(s)
- Ting Yao
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Yi Zhang
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Mengjiao Lv
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Guoqing Zang
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Soon Seng Ng
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W2 1PG, UK
| | - Xiaohua Chen
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| |
Collapse
|
165
|
Individual Control and Quantification of 3D Spheroids in a High-Density Microfluidic Droplet Array. Cell Rep 2021; 31:107670. [PMID: 32460010 PMCID: PMC7262598 DOI: 10.1016/j.celrep.2020.107670] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 11/20/2019] [Accepted: 04/29/2020] [Indexed: 12/29/2022] Open
Abstract
As three-dimensional cell culture formats gain in popularity, there emerges a need for tools that produce vast amounts of data on individual cells within the spheroids or organoids. Here, we present a microfluidic platform that provides access to such data by parallelizing the manipulation of individual spheroids within anchored droplets. Different conditions can be applied in a single device by triggering the merging of new droplets with the spheroid-containing drops. This allows cell-cell interactions to be initiated for building microtissues, studying stem cells’ self-organization, or observing antagonistic interactions. It also allows the spheroids’ physical or chemical environment to be modulated, as we show by applying a drug over a large range of concentrations in a single parallelized experiment. This convergence of microfluidics and image acquisition leads to a data-driven approach that allows the heterogeneity of 3D culture behavior to be addressed across the scales, bridging single-cell measurements with population measurements. Microfluidic droplet pairs sequentially trapped in capillary anchors before merging 1 spheroid/droplet, with microenvironment modulations driven by droplet merging A wide range of drug concentrations tested on hepatic-like spheroids in a single chip Data-driven approach unravels 3D tissue-level dynamic drug response
Collapse
|
166
|
Marsee A, Roos FJM, Verstegen MMA, Gehart H, de Koning E, Lemaigre F, Forbes SJ, Peng WC, Huch M, Takebe T, Vallier L, Clevers H, van der Laan LJW, Spee B. Building consensus on definition and nomenclature of hepatic, pancreatic, and biliary organoids. Cell Stem Cell 2021; 28:816-832. [PMID: 33961769 PMCID: PMC11699540 DOI: 10.1016/j.stem.2021.04.005] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hepatic, pancreatic, and biliary (HPB) organoids are powerful tools for studying development, disease, and regeneration. As organoid research expands, the need for clear definitions and nomenclature describing these systems also grows. To facilitate scientific communication and consistent interpretation, we revisit the concept of an organoid and introduce an intuitive classification system and nomenclature for describing these 3D structures through the consensus of experts in the field. To promote the standardization and validation of HPB organoids, we propose guidelines for establishing, characterizing, and benchmarking future systems. Finally, we address some of the major challenges to the clinical application of organoids.
Collapse
Affiliation(s)
- Ary Marsee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Floris J M Roos
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Helmuth Gehart
- Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Eelco de Koning
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, the Netherlands; Leiden University Medical Center, Department of Medicine, Leiden, the Netherlands
| | - Frédéric Lemaigre
- Université Catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Stuart J Forbes
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Weng Chuan Peng
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, and Center for Stem Cell, and Organoid Medicine (CuSTOM), Cincinnati Children Hospital Medical Center, Cincinnati, OH, USA; Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ludovic Vallier
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, Cambridgeshire, UK; Department of Surgery, University of Cambridge and National Institute for Health Research Cambridge Biomedical Research Center, Cambridge, UK
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, the Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
167
|
Harrison SP, Baumgarten SF, Verma R, Lunov O, Dejneka A, Sullivan GJ. Liver Organoids: Recent Developments, Limitations and Potential. Front Med (Lausanne) 2021; 8:574047. [PMID: 34026769 PMCID: PMC8131532 DOI: 10.3389/fmed.2021.574047] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Liver cell types derived from induced pluripotent stem cells (iPSCs) share the potential to investigate development, toxicity, as well as genetic and infectious disease in ways currently limited by the availability of primary tissue. With the added advantage of patient specificity, which can play a role in all of these areas. Many iPSC differentiation protocols focus on 3 dimensional (3D) or organotypic differentiation, as these offer the advantage of more closely mimicking in vivo systems including; the formation of tissue like architecture and interactions/crosstalk between different cell types. Ultimately such models have the potential to be used clinically and either with or more aptly, in place of animal models. Along with the development of organotypic and micro-tissue models, there will be a need to co-develop imaging technologies to enable their visualization. A variety of liver models termed "organoids" have been reported in the literature ranging from simple spheres or cysts of a single cell type, usually hepatocytes, to those containing multiple cell types combined during the differentiation process such as hepatic stellate cells, endothelial cells, and mesenchymal cells, often leading to an improved hepatic phenotype. These allow specific functions or readouts to be examined such as drug metabolism, protein secretion or an improved phenotype, but because of their relative simplicity they lack the flexibility and general applicability of ex vivo tissue culture. In the liver field these are more often constructed rather than developed together organotypically as seen in other organoid models such as brain, kidney, lung and intestine. Having access to organotypic liver like surrogates containing multiple cell types with in vivo like interactions/architecture, would provide vastly improved models for disease, toxicity and drug development, combining disciplines such as microfluidic chip technology with organoids and ultimately paving the way to new therapies.
Collapse
Affiliation(s)
- Sean Philip Harrison
- Hybrid Technology Hub–Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Saphira Felicitas Baumgarten
- Hybrid Technology Hub–Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Rajneesh Verma
- Hybrid Technology Hub–Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, Prague, Czechia
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, Prague, Czechia
| | - Gareth John Sullivan
- Hybrid Technology Hub–Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
- Norwegian Center for Stem Cell Research, Oslo University Hospital, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
168
|
Khoshdel-Rad N, Zahmatkesh E, Moeinvaziri F, Haghparast N, Baharvand H, Aghdami N, Moghadasali R. Promoting Maturation of Human Pluripotent Stem Cell-Derived Renal Microtissue by Incorporation of Endothelial and Mesenchymal Cells. Stem Cells Dev 2021; 30:428-440. [PMID: 33787359 DOI: 10.1089/scd.2020.0189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Directed differentiation of human pluripotent stem cells (hPSCs) uses a growing number of small molecules and growth factors required for in vitro generation of renal lineage cells. Although current protocols are relatively inefficient or expensive. The first objective of the present work was to establish a new differentiation protocol for generating renal precursors. We sought to determine if inducer of definitive endoderm 1 (IDE1), a cost-effective small molecule, can be used to replace activin A. Gene expression data showed significantly increased expressions of nephrogenic markers in cells differentiated with 20 nM IDE1 compared with cells differentiated with activin A. Thus, renal lineage cells could be generated by this alternative approach. Afterward, we determined whether coculture of endothelial and mesenchymal cells could increase the maturation of three-dimensional (3D) renal structures. For this purpose, we employed a 3D coculture system in which hPSC-derived kidney precursors were cocultured with endothelial cells (ECs) and mesenchymal stem cells (MSCs), hereafter named RMEM (renal microtissue derived from coculture of renal precursors with endothelial and mesenchymal stem cells). hPSC-derived kidney precursors were cultured either alone [renal microtissue (RM)] or in coculture with human umbilical vein endothelial cells and human bone marrow-derived mesenchymal stem cells at an approximate ratio of 10:7:2, respectively. Immunofluorescent staining showed expressions of kidney-specific markers synaptopodin, LTL, and E-cadherin, as well as CD31+ ECs that were distributed throughout the RMEMs. Quantitative real-time polymerase chain reaction analysis confirmed a significant increase in gene expressions of the renal-specific markers in RMEMs compared with RMs. These findings demonstrated that renal precursors cocultured with endothelial and MSCs showed greater maturity compared with RMs. Moreover, ex ovo transplantation induced further maturation in the RMEM constructs. Our novel approach enabled the generation of RMEM that could potentially be used in high-throughput drug screening and nephrotoxicology studies.
Collapse
Affiliation(s)
- Niloofar Khoshdel-Rad
- Department of Stem Cells and Developmental Biology, 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
| | - Ensieh Zahmatkesh
- Department of Stem Cells and Developmental Biology, 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
| | - Farideh Moeinvaziri
- Department of Stem Cells and Developmental Biology, 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
| | - Newsha Haghparast
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, 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
| |
Collapse
|
169
|
Fritsche E, Haarmann-Stemmann T, Kapr J, Galanjuk S, Hartmann J, Mertens PR, Kämpfer AAM, Schins RPF, Tigges J, Koch K. Stem Cells for Next Level Toxicity Testing in the 21st Century. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006252. [PMID: 33354870 DOI: 10.1002/smll.202006252] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/13/2020] [Indexed: 06/12/2023]
Abstract
The call for a paradigm change in toxicology from the United States National Research Council in 2007 initiates awareness for the invention and use of human-relevant alternative methods for toxicological hazard assessment. Simple 2D in vitro systems may serve as first screening tools, however, recent developments infer the need for more complex, multicellular organotypic models, which are superior in mimicking the complexity of human organs. In this review article most critical organs for toxicity assessment, i.e., skin, brain, thyroid system, lung, heart, liver, kidney, and intestine are discussed with regards to their functions in health and disease. Embracing the manifold modes-of-action how xenobiotic compounds can interfere with physiological organ functions and cause toxicity, the need for translation of such multifaceted organ features into the dish seems obvious. Currently used in vitro methods for toxicological applications and ongoing developments not yet arrived in toxicity testing are discussed, especially highlighting the potential of models based on embryonic stem cells and induced pluripotent stem cells of human origin. Finally, the application of innovative technologies like organs-on-a-chip and genome editing point toward a toxicological paradigm change moves into action.
Collapse
Affiliation(s)
- Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
- Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | | | - Julia Kapr
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Saskia Galanjuk
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Hartmann
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Peter R Mertens
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, Magdeburg, 39106, Germany
| | - Angela A M Kämpfer
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Roel P F Schins
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Katharina Koch
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| |
Collapse
|
170
|
Frum T, Spence JR. hPSC-derived organoids: models of human development and disease. J Mol Med (Berl) 2021; 99:463-473. [PMID: 32857169 PMCID: PMC7914270 DOI: 10.1007/s00109-020-01969-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/30/2020] [Accepted: 08/18/2020] [Indexed: 12/18/2022]
Abstract
Organoids derived from human pluripotent stem cells (hPSCs) have emerged as important models for investigating human-specific aspects of development and disease. Here we discuss hPSC-derived organoids through the lens of development-highlighting how stages of human development align with the development of hPSC-derived organoids in the tissue culture dish. Using hPSC-derived lung and intestinal organoids as examples, we discuss the value and application of such systems for understanding human biology, as well as strategies for enhancing organoid complexity and maturity.
Collapse
Affiliation(s)
- Tristan Frum
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jason R Spence
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA.
| |
Collapse
|
171
|
Zubareva EV, Nadezhdin SV, Nadezhdina NA, Belyaeva VS, Burda YE, Avtina TV, Gudyrev OS, Kolesnik IM, Kulikova SY, Mishenin MO. 3D organotypic cell structures for drug development and Microorganism-Host interaction research. RESEARCH RESULTS IN PHARMACOLOGY 2021. [DOI: 10.3897/rrpharmacology.7.62118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: The article describes a new method of tissue engineering, which is based on the use of three-dimensional multicellular constructs consisting of stem cells that mimic the native tissue in vivo – organoids.
3D cell cultures: The currently existing model systems of three-dimensional cultures are described.
Characteristics of organoids and strategies for their culturing: The main approaches to the fabrication of 3D cell constructs using pluripotent (embryonic and induced) stem cells or adult stem cells are described.
Brain organoids (Cerebral organoids): Organoids of the brain, which are used to study the development of the human brain, are characterized, with the description of biology of generating region-specific cerebral organoids.
Lung organoids: Approaches to the generation of lung organoids are described, by means of pluripotent stem cells and lung tissue cell lines.
Liver organoids: The features of differentiation of stem cells into hepatocyte-like cells and the creation of 3D hepatic organoids are characterized.
Intestinal organoids: The formation of small intestine organoids from stem cells is described.
Osteochondral organoids: Fabrication of osteochondral organoids is characterised.
Use of organoids as test systems for drugs screening: The information on drug screening using organoids is provided.
Using organoids to model infectious diseases and study adaptive responses of microorganisms when interacting with the host: The use of organoids for modeling infectious diseases and studying the adaptive responses of microorganisms when interacting with the host organism is described.
Conclusion: The creation of three-dimensional cell structures that reproduce the structural and functional characteristics of tissue in vivo, makes it possible to study the biology of the body’s development, the features of intercellular interactions, screening drugs and co-cultivating with viruses, bacteria and parasites.
Collapse
|
172
|
Yuzuriha A, Nakamura S, Sugimoto N, Kihara S, Nakagawa M, Yamamoto T, Sekiguchi K, Eto K. Extracellular laminin regulates hematopoietic potential of pluripotent stem cells through integrin β1-ILK-β-catenin-JUN axis. Stem Cell Res 2021; 53:102287. [PMID: 33813173 DOI: 10.1016/j.scr.2021.102287] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/21/2022] Open
Abstract
Recombinant matrices have enabled feeder cell-free maintenance cultures of human pluripotent stem cells (hPSCs), with laminin 511-E8 fragment (LM511-E8) being widely used. However, we herein report that hPSCs maintained on LM511-E8 resist differentiating to multipotent hematopoietic progenitor cells (HPCs), unlike hPSCs maintained on LM421-E8 or LM121-E8. The latter two LM-E8s bound weakly to hPSCs compared with LM511-E8 and activated the canonical Wnt/β-catenin signaling pathway. Moreover, the extracellular LM-E8-dependent preferential hematopoiesis was associated with a higher expression of integrin β1 (ITGB1) and downstream integrin-linked protein kinase (ILK), β-catenin and phosphorylated JUN. Accordingly, the lower coating concentration of LM511-E8 or addition of a Wnt/β-catenin signaling activator, CHIR99021, facilitated higher HPC yield. In contrast, the inhibition of ILK, Wnt or JNK by inhibitors or mRNA knockdown suppressed the HPC yield. These findings suggest that extracellular laminin scaffolds modulate the hematopoietic differentiation potential of hPSCs by activating the ITGB1-ILK-β-catenin-JUN axis at the undifferentiated stage. Finally, the combination of low-concentrated LM511-E8 and a revised hPSC-sac method, which adds bFGF, SB431542 and heparin to the conventional method, enabled a higher yield of HPCs and higher rate for definitive hematopoiesis, suggesting a useful protocol for obtaining differentiated hematopoietic cells from hPSCs in general.
Collapse
Affiliation(s)
- Akinori Yuzuriha
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan
| | - Sou Nakamura
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan
| | - Shunsuke Kihara
- Department of Fundamental Cell Technology, CiRA, Kyoto University, Kyoto, Japan
| | - Masato Nakagawa
- Department of Life Science Frontiers, CiRA, Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, CiRA, Kyoto University, Kyoto, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan; AMED-CREST, AMED 1-7-1 Otemachi, Chiyodaku, Tokyo 100-0004, Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Japan
| | - Koji Eto
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan; Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
173
|
Rao S, Hossain T, Mahmoudi T. 3D human liver organoids: An in vitro platform to investigate HBV infection, replication and liver tumorigenesis. Cancer Lett 2021; 506:35-44. [PMID: 33675983 DOI: 10.1016/j.canlet.2021.02.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023]
Abstract
Hepatitis B Virus (HBV) infection is a leading cause of chronic liver cirrhosis and hepatocellular carcinoma (HCC) with an estimated 400 million people infected worldwide. The precise molecular mechanisms underlying HBV replication and tumorigenesis have remained largely uncharacterized due to the lack of a primary cell model to study HBV, a virus that exhibits stringent host species and cell-type specificity. Organoid technology has recently emerged as a powerful tool to investigate human diseases in a primary 3D cell-culture system that maintains the organisation and functionality of the tissue of origin. In this review, we describe the utilisation of human liver organoid platforms to study HBV. We first present the different categories of liver organoids and their demonstrated ability to support the complete HBV replication cycle. We then discuss the potential applications of liver organoids in investigating HBV infection and replication, related tumorigenesis and novel HBV-directed therapies. Liver organoids can be genetically modified, patient-derived, expanded and biobanked, thereby serving as a clinically-relevant, human, primary cell-derived platform to investigate HBV. Finally, we provide insights into the future applications of this powerful technology in the context of HBV-infection and HCC.
Collapse
Affiliation(s)
- Shringar Rao
- Department of Biochemistry, Erasmus University Medical Centre, PO Box 2040, 3000, CA, 9 Rotterdam, the Netherlands
| | - Tanvir Hossain
- Department of Biochemistry, Erasmus University Medical Centre, PO Box 2040, 3000, CA, 9 Rotterdam, the Netherlands
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Centre, PO Box 2040, 3000, CA, 9 Rotterdam, the Netherlands; Department of Pathology, Erasmus University Medical Centre, PO Box 2040, 3000, CA, Rotterdam, the Netherlands; Department of Urology, Erasmus University Medical Centre, PO Box 2040, 3000, CA, Rotterdam, the Netherlands.
| |
Collapse
|
174
|
Matsumoto R, Takahashi Y. Human pituitary development and application of iPSCs for pituitary disease. Cell Mol Life Sci 2021; 78:2069-2079. [PMID: 33206204 PMCID: PMC11071979 DOI: 10.1007/s00018-020-03692-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/11/2022]
Abstract
The pituitary plays a pivotal role in maintaining systemic homeostasis by secreting several hormones. During fetal development, the pituitary develops from the oral ectoderm in contact with the adjacent hypothalamus. This process is regulated by the fine-tuned expression of transcription and growth factors. Impairments of this process result in congenital pituitary hypoplasia leading to dysfunction of the pituitary. Although animal models such as knockout mice have helped to clarify these underlying mechanisms, the developmental processes of the human pituitary gland and the mechanisms of human pituitary disorders have not been fully understood. This is because, at least in part, of the lack of a human pituitary developmental model. Recently, methods for in vitro induction of the pituitary gland from human pluripotent stem cells were developed. These models can be utilized not only for regenerative medicine but also for human pituitary studies on developmental biology and for modeling of pituitary disorders, such as hypopituitarism and pituitary tumors. In this review, we provide an overview of recent progress in the applications of pluripotent stem cells for pituitary research and discuss further perspectives for pituitary studies.
Collapse
Affiliation(s)
- Ryusaku Matsumoto
- Department of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yutaka Takahashi
- Department of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan.
- Department of Diabetes and Endocrinology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8522, Japan.
| |
Collapse
|
175
|
Development of a genetically modified hepatoma cell line with heat-inducible high liver function. Cytotechnology 2021; 73:353-362. [PMID: 34149171 DOI: 10.1007/s10616-021-00457-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 02/05/2021] [Indexed: 01/15/2023] Open
Abstract
Hepatoma cells are a promising cell source for the construction of bioartificial liver (BAL) systems owing to their high proliferative capability. However, their low liver function compared with primary hepatocytes is a major problem. In a previous study, we established a genetically modified hepatoma cell line, Hepa/8F5, in which eight liver-enriched transcription factor (LETF) genes were transduced into mouse hepatoma Hepa1-6 cells using a drug-inducible transactivator system. These cells proliferate actively under normal culture conditions, meaning that large quantities can be prepared easily. When the overexpression of the LETFs is induced by the addition of an inducer drug, cell growth stops and cell morphology changes with concomitant high expression of liver functions. However, the liver functions largely depend on the presence of the inducer drug, which must be continuously added to maintain these enhanced functions. In the present study, we attempted to modify the method of induction of LETF overexpression in Hepa/8F5 cells to remove the requirement for continual drug addition. To this end, we constructed a system in which the artificial transactivator was transcribed and amplified under the control of a heat-shock protein promoter, and introduced the system into the genome of Hepa/8F5 cells. In our modified cell line, heat-triggered LETF expression was confirmed to induce high liver function. After drug-screening of transfected cells, we established a hepatoma cell line (Hepa/HS), which exhibited high, heat-inducible liver functions. The Hepa/HS cells may represent a new cell source for hepatic studies such as the construction of BAL systems. Supplementary Information The online version of this article (10.1007/s10616-021-00457-4) contains supplementary material, which is available to authorized users.
Collapse
|
176
|
Establishment of Human Leukocyte Antigen-Mismatched Immune Responses after Transplantation of Human Liver Bud in Humanized Mouse Models. Cells 2021; 10:cells10020476. [PMID: 33672150 PMCID: PMC7927063 DOI: 10.3390/cells10020476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/05/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Humanized mouse models have contributed significantly to human immunology research. In transplant immunity, human immune cell responses to donor grafts have not been reproduced in a humanized animal model. To elicit human T-cell immune responses, we generated immune-compromised nonobese diabetic/Shi-scid, IL-2RγKO Jic (NOG) with a homozygous expression of human leukocyte antigen (HLA) class I heavy chain (NOG-HLA-A2Tg) mice. After the transplantation of HLA-A2 human hematopoietic stem cells into NOG-HLA-A2Tg, we succeeded in achieving alloimmune responses after the HLA-mismatched human-induced pluripotent stem cell (hiPSC)-derived liver-like tissue transplantation. This immune response was inhibited by administering tacrolimus. In this model, we reproduced allograft rejection after the human iPSC-derived liver-like tissue transplantation. Human tissue transplantation on the humanized mouse liver surface is a good model that can predict T-cell-mediated cellular rejection that may occur when organ transplantation is performed.
Collapse
|
177
|
Serras AS, Rodrigues JS, Cipriano M, Rodrigues AV, Oliveira NG, Miranda JP. A Critical Perspective on 3D Liver Models for Drug Metabolism and Toxicology Studies. Front Cell Dev Biol 2021; 9:626805. [PMID: 33732695 PMCID: PMC7957963 DOI: 10.3389/fcell.2021.626805] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
The poor predictability of human liver toxicity is still causing high attrition rates of drug candidates in the pharmaceutical industry at the non-clinical, clinical, and post-marketing authorization stages. This is in part caused by animal models that fail to predict various human adverse drug reactions (ADRs), resulting in undetected hepatotoxicity at the non-clinical phase of drug development. In an effort to increase the prediction of human hepatotoxicity, different approaches to enhance the physiological relevance of hepatic in vitro systems are being pursued. Three-dimensional (3D) or microfluidic technologies allow to better recapitulate hepatocyte organization and cell-matrix contacts, to include additional cell types, to incorporate fluid flow and to create gradients of oxygen and nutrients, which have led to improved differentiated cell phenotype and functionality. This comprehensive review addresses the drug-induced hepatotoxicity mechanisms and the currently available 3D liver in vitro models, their characteristics, as well as their advantages and limitations for human hepatotoxicity assessment. In addition, since toxic responses are greatly dependent on the culture model, a comparative analysis of the toxicity studies performed using two-dimensional (2D) and 3D in vitro strategies with recognized hepatotoxic compounds, such as paracetamol, diclofenac, and troglitazone is performed, further highlighting the need for harmonization of the respective characterization methods. Finally, taking a step forward, we propose a roadmap for the assessment of drugs hepatotoxicity based on fully characterized fit-for-purpose in vitro models, taking advantage of the best of each model, which will ultimately contribute to more informed decision-making in the drug development and risk assessment fields.
Collapse
Affiliation(s)
- Ana S. Serras
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Madalena Cipriano
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Armanda V. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Nuno G. Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
178
|
Garreta E, Kamm RD, Chuva de Sousa Lopes SM, Lancaster MA, Weiss R, Trepat X, Hyun I, Montserrat N. Rethinking organoid technology through bioengineering. NATURE MATERIALS 2021; 20:145-155. [PMID: 33199860 DOI: 10.1038/s41563-020-00804-4] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 08/18/2020] [Indexed: 06/11/2023]
Abstract
In recent years considerable progress has been made in the development of faithful procedures for the differentiation of human pluripotent stem cells (hPSCs). An important step in this direction has also been the derivation of organoids. This technology generally relies on traditional three-dimensional culture techniques that exploit cell-autonomous self-organization responses of hPSCs with minimal control over the external inputs supplied to the system. The convergence of stem cell biology and bioengineering offers the possibility to provide these stimuli in a controlled fashion, resulting in the development of naturally inspired approaches to overcome major limitations of this nascent technology. Based on the current developments, we emphasize the achievements and ongoing challenges of bringing together hPSC organoid differentiation, bioengineering and ethics. This Review underlines the need for providing engineering solutions to gain control of self-organization and functionality of hPSC-derived organoids. We expect that this knowledge will guide the community to generate higher-grade hPSC-derived organoids for further applications in developmental biology, drug screening, disease modelling and personalized medicine.
Collapse
Affiliation(s)
- Elena Garreta
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Roger D Kamm
- Department of Biological Engineering and Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | | | | | - Ron Weiss
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Xavier Trepat
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain
| | - Insoo Hyun
- Department of Bioethics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for Bioethics, Harvard Medical School, Boston, MA, USA
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
179
|
Kong J, Wen S, Cao W, Yue P, Xu X, Zhang Y, Luo L, Chen T, Li L, Wang F, Tao J, Zhou G, Luo S, Liu A, Bao F. Lung organoids, useful tools for investigating epithelial repair after lung injury. Stem Cell Res Ther 2021; 12:95. [PMID: 33516265 PMCID: PMC7846910 DOI: 10.1186/s13287-021-02172-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/17/2021] [Indexed: 02/07/2023] Open
Abstract
Organoids are derived from stem cells or organ-specific progenitors. They display structures and functions consistent with organs in vivo. Multiple types of organoids, including lung organoids, can be generated. Organoids are applied widely in development, disease modelling, regenerative medicine, and other multiple aspects. Various human pulmonary diseases caused by several factors can be induced and lead to different degrees of lung epithelial injury. Epithelial repair involves the participation of multiple cells and signalling pathways. Lung organoids provide an excellent platform to model injury to and repair of lungs. Here, we review the recent methods of cultivating lung organoids, applications of lung organoids in epithelial repair after injury, and understanding the mechanisms of epithelial repair investigated using lung organoids. By using lung organoids, we can discover the regulatory mechanisms related to the repair of lung epithelia. This strategy could provide new insights for more effective management of lung diseases and the development of new drugs.
Collapse
Affiliation(s)
- Jing Kong
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China.,The School of Medicine, Kunming University, Kunming, 650214, China
| | - Shiyuan Wen
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Wenjing Cao
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Peng Yue
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xin Xu
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Yu Zhang
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Lisha Luo
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Taigui Chen
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Lianbao Li
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Feng Wang
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Jian Tao
- The School of Medicine, Kunming University, Kunming, 650214, China
| | - Guozhong Zhou
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Suyi Luo
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Aihua Liu
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Yunnan Province Key Laboratory of Children's Major Diseases Research, The Children's Hospital of Kunming, Kunming Medical University, Kunming, 650030, China.
| | - Fukai Bao
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China.
| |
Collapse
|
180
|
Jin M, Yi X, Liao W, Chen Q, Yang W, Li Y, Li S, Gao Y, Peng Q, Zhou S. Advancements in stem cell-derived hepatocyte-like cell models for hepatotoxicity testing. Stem Cell Res Ther 2021; 12:84. [PMID: 33494782 PMCID: PMC7836452 DOI: 10.1186/s13287-021-02152-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Drug-induced liver injury (DILI) is one of the leading causes of clinical trial failures and high drug attrition rates. Currently, the commonly used hepatocyte models include primary human hepatocytes (PHHs), animal models, and hepatic cell lines. However, these models have disadvantages that include species-specific differences or inconvenient cell extraction methods. Therefore, a novel, inexpensive, efficient, and accurate model that can be applied to drug screening is urgently needed. Owing to their self-renewable ability, source abundance, and multipotent competence, stem cells are stable sources of drug hepatotoxicity screening models. Because 3D culture can mimic the in vivo microenvironment more accurately than can 2D culture, the former is commonly used for hepatocyte culture and drug screening. In this review, we introduce the different sources of stem cells used to generate hepatocyte-like cells and the models for hepatotoxicity testing that use stem cell-derived hepatocyte-like cells.
Collapse
Affiliation(s)
- Meixian Jin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Xiao Yi
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Liao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qi Chen
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Wanren Yang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yang Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shao Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yi Gao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qing Peng
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Shuqin Zhou
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
181
|
Wang L, Ye Z, Jang YY. Convergence of human pluripotent stem cell, organoid, and genome editing technologies. Exp Biol Med (Maywood) 2021; 246:861-875. [PMID: 33467883 DOI: 10.1177/1535370220985808] [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: 12/26/2022] Open
Abstract
The last decade has seen many exciting technological breakthroughs that greatly expanded the toolboxes for biological and biomedical research, yet few have had more impact than induced pluripotent stem cells and modern-day genome editing. These technologies are providing unprecedented opportunities to improve physiological relevance of experimental models, further our understanding of developmental processes, and develop novel therapies. One of the research areas that benefit greatly from these technological advances is the three-dimensional human organoid culture systems that resemble human tissues morphologically and physiologically. Here we summarize the development of human pluripotent stem cells and their differentiation through organoid formation. We further discuss how genetic modifications, genome editing in particular, were applied to answer basic biological and biomedical questions using organoid cultures of both somatic and pluripotent stem cell origins. Finally, we discuss the potential challenges of applying human pluripotent stem cell and organoid technologies for safety and efficiency evaluation of emerging genome editing tools.
Collapse
Affiliation(s)
- Lin Wang
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Zhaohui Ye
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Yoon-Young Jang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, John Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
182
|
Yasui R, Sekine K, Yamaguchi K, Furukawa Y, Taniguchi H. Robust parameter design of human induced pluripotent stem cell differentiation protocols defines lineage-specific induction of anterior-posterior gut tube endodermal cells. Stem Cells 2021; 39:429-442. [PMID: 33400835 DOI: 10.1002/stem.3326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/07/2020] [Indexed: 12/29/2022]
Abstract
Tissues and cells derived from pluripotent stem cells (PSC) are likely to become widely used in disease modeling, drug screening, and regenerative medicine. For these applications, the in vitro PSC differentiation process must be elaborately investigated and controlled to reliably obtain the desired end products. However, because traditional experimental methods, such as one factor at a time or brute-force approaches, are impractical for detailed screening of complex PSC cultivation conditions, more strategic and effective screening based on statistical design of experiments (DOE) ought to be indispensable. Among various DOE approaches, we regard robust parameter design (RPD) as particularly suited for differentiation protocol optimization due to its suitability for multifactorial screening. We confirmed the adaptability of RPD for investigating human induced PSC lineage specification toward anterior-posterior gut tube endodermal cells and clarified both the contribution of each cell signaling pathway and the effect of cell signaling condition alteration on marker RNA expression levels, while increasing the efficiency of the screening in 243-fold (18 vs 4374) compared with that of a brute-force approach. Specific induction of anterior foregut, hepatic, pancreatic, or mid-hindgut cells was achieved using seven iPSC strains with the optimal culture protocols established on the basis of RPD analysis. RPD has the potential to enable efficient construction and optimization of PSC differentiation protocols, and its use is recommended from fundamental research to mass production of PSC-derived products.
Collapse
Affiliation(s)
- Ryota Yasui
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.,Fundamental Research Laboratory, Eiken Chemical Co., Ltd., Nogi, Tochigi, Japan
| | - Keisuke Sekine
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.,Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Cancer Cell Systems, National Cancer Center Research Institute, Tokyo, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.,Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
183
|
Park IS. Synergistic effect of three-dimensional coculture and photobiomodulation therapy on vascularized liver spheroid formation by stem cells. J Cell Physiol 2021; 236:5865-5874. [PMID: 33432611 DOI: 10.1002/jcp.30270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/28/2022]
Abstract
Despite studies reporting functional differentiation of liver cells, a three-dimensional, vascularized liver organ has yet to be developed from mesenchymal stem cells. We investigated whether treatment with photobiomodulation (PBM) before three-dimensional liver spheroid transplantation improved the recovery of liver function via stimulation of angiogenesis and hepatocyte differentiation. Liver spheroids composed of hepatic, endothelial, and mesenchymal cells were subjected to PBM therapy. To evaluate the in vivo therapeutic effect of the liver spheroids treated with PBM, phosphate-buffered saline, liver spheroid, and PBM-treated liver spheroid were transplanted into a damaged host liver using conventional chimeric mouse models. To further characterize the maturation of transplanted PBM-liver spheroid compared with the newly generated non-PBM-liver spheroid or human liver tissues, the expression profiles of mature liver signature genes were analyzed. Liver spheroids expressed hepatocyte growth factors, including vascular endothelial growth factor and angiogenic factors. The cells in liver spheroid compensated for the low viability and improved the function of hepatocytes. Here, we demonstrate the formation of vascularized and functional human liver spheroid from human adipose-derived stem cells by transplantation of liver tissue created in vitro. Albumin secretion by PBM-treated liver spheroid was higher on Day 28 compared with liver spheroid-seeded transplant group. PBM-liver spheroids serve as individual vascularization units, promoting the simultaneous development of new microvascular networks at different locations inside the implanted tissue constructs. The vasculature in the liver spheroid transplants became functional by connecting to the host vessels within 48 h. These PBM-liver spheroids may be useful in designing artificial three-dimensional hepatic tissue constructs and in cell therapy with limited numbers of human hepatocytes.
Collapse
Affiliation(s)
- In-Su Park
- Cell Therapy Center, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
184
|
Danoy M, Tauran Y, Poulain S, Jellali R, Bruce J, Leduc M, Le Gall M, Gilard F, Kido T, Arakawa H, Araya K, Mori D, Kato Y, Kusuhara H, Plessy C, Miyajima A, Sakai Y, Leclerc E. Multi-omics analysis of hiPSCs-derived HLCs matured on-chip revealed patterns typical of liver regeneration. Biotechnol Bioeng 2021; 118:3716-3732. [PMID: 33404112 DOI: 10.1002/bit.27667] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/13/2020] [Accepted: 12/20/2020] [Indexed: 12/17/2022]
Abstract
Maturation of human-induced pluripotent stem cells (hiPSCs)-derived hepatocytes-like cells (HLCs) toward a complete hepatocyte phenotype remains a challenge as primitiveness patterns are still commonly observed. In this study, we propose a modified differentiation protocol for those cells which includes a prematuration in Petri dishes and a maturation in microfluidic biochip. For the first time, a large range of biomolecular families has been extracted from the same sample to combine transcriptomic, proteomic, and metabolomic analysis. After integration, these datasets revealed specific molecular patterns and highlighted the hepatic regeneration profile in biochips. Overall, biochips exhibited processes of cell proliferation and inflammation (via TGFB1) coupled with anti-fibrotic signaling (via angiotensin 1-7, ATR-2, and MASR). Moreover, cultures in this condition displayed physiological lipid-carbohydrate homeostasis (notably via PPAR, cholesterol metabolism, and bile synthesis) coupled with cell respiration through advanced oxidative phosphorylation (through the overexpression of proteins from the third and fourth complex). The results presented provide an original overview of the complex mechanisms involved in liver regeneration using an advanced in vitro organ-on-chip technology.
Collapse
Affiliation(s)
- Mathieu Danoy
- CNRS UMI 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, Tokyo, Japan.,Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Yannick Tauran
- CNRS UMI 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, Tokyo, Japan.,Laboratoire des Multimatériaux et Interfaces, UMR CNRS 5615, Univ Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Stéphane Poulain
- RIKEN Center for Integrative Medical Science, Yokohama, Kanagawa, Japan.,Biomedical Microsystems Lab, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Rachid Jellali
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319-60203 Compiègne Cedex, Compiègne, France
| | - Johanna Bruce
- Plateforme 3P5 Proteomi'ic, Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 22 rue Méchain, Paris, France
| | - Marjorie Leduc
- Plateforme 3P5 Proteomi'ic, Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 22 rue Méchain, Paris, France
| | - Morgane Le Gall
- Plateforme 3P5 Proteomi'ic, Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 22 rue Méchain, Paris, France
| | - Francoise Gilard
- Plateforme Métabolisme Métabolome, Institute of Plant Sciences Paris-Saclay (IPS2), CNRS, INRA, Univ. Paris-Sud, Univ. Evry, Univ. Paris-Diderot, Univ. Paris Saclay, Gif-sur-Yvette Cedex, France
| | - Taketomo Kido
- Laboratory of Stem Cell Therapy, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Arakawa
- Laboratory of Molecular Pharmacokinetics, Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa City, Ishikawa, Japan
| | - Karin Araya
- Laboratory of Molecular Pharmacokinetics, Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa City, Ishikawa, Japan
| | - Daiki Mori
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukio Kato
- Laboratory of Molecular Pharmacokinetics, Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa City, Ishikawa, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Charles Plessy
- RIKEN Center for Integrative Medical Science, Yokohama, Kanagawa, Japan
| | - Atsushi Miyajima
- Laboratory of Stem Cell Therapy, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Eric Leclerc
- CNRS UMI 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, Tokyo, Japan.,Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319-60203 Compiègne Cedex, Compiègne, France
| |
Collapse
|
185
|
Funata M, Nio Y, Erion DM, Thompson WL, Takebe T. The promise of human organoids in the digestive system. Cell Death Differ 2021; 28:84-94. [PMID: 33204011 PMCID: PMC7852589 DOI: 10.1038/s41418-020-00661-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
The advent of organoid technology has enabled scientists and clinicians to utilize cells from primary tissues or pluripotent stem cells (PSCs) to grow self-organizing tissue systems, thus attaining cellular diversity, spatial organization, and functionality as found within digestive tracts. The development of human gastrointestinal (GI) and hepato-biliary-pancreatic organoids as an in-a-dish model present novel opportunities to study humanistic mechanisms of organogenesis, regeneration and pathogenesis. Herein, we review the recent portfolios of primary tissue-derived and PSC-derived organoids in the digestive systems. We also discuss the promise and challenges in disease modeling and drug development applications for digestive disorders.
Collapse
Affiliation(s)
- Masaaki Funata
- T-CiRA Discovery, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
- Takeda-CiRA Joint Program, Fujisawa City, Kanagawa, Japan
| | - Yasunori Nio
- T-CiRA Discovery, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
- Takeda-CiRA Joint Program, Fujisawa City, Kanagawa, Japan
| | - Derek M Erion
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 35 Landsdowne Street, Cambridge, MA, 02139, USA
| | - Wendy L Thompson
- Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Takanori Takebe
- Takeda-CiRA Joint Program, Fujisawa City, Kanagawa, Japan.
- Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
- Communication Design Center, Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan.
| |
Collapse
|
186
|
Thompson WL, Takebe T. Human liver model systems in a dish. Dev Growth Differ 2021; 63:47-58. [PMID: 33423319 PMCID: PMC7940568 DOI: 10.1111/dgd.12708] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
The human adult liver has a multi-cellular structure consisting of large lobes subdivided into lobules containing portal triads and hepatic cords lined by specialized blood vessels. Vital hepatic functions include filtering blood, metabolizing drugs, and production of bile and blood plasma proteins like albumin, among many other functions, which are generally dependent on the location or zone in which the hepatocyte resides in the liver. Due to the liver's intricate structure, there are many challenges to design differentiation protocols to generate more mature functional hepatocytes from human stem cells and maintain the long-term viability and functionality of primary hepatocytes. To this end, recent advancements in three-dimensional (3D) stem cell culture have accelerated the generation of a human miniature liver system, also known as liver organoids, with polarized epithelial cells, supportive cell types and extra-cellular matrix deposition by translating knowledge gained in studies of animal organogenesis and regeneration. To facilitate the efforts to study human development and disease using in vitro hepatic models, a thorough understanding of state-of-art protocols and underlying rationales is essential. Here, we review rapidly evolving 3D liver models, mainly focusing on organoid models differentiated from human cells.
Collapse
Affiliation(s)
- Wendy L. Thompson
- Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM). Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM). Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229-3039, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
- Communication Design Center, Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Japan
| |
Collapse
|
187
|
Bove G, Mehnert AK, Dao Thi VL. iPSCs for modeling hepatotropic pathogen infections. IPSCS FOR STUDYING INFECTIOUS DISEASES 2021:149-213. [DOI: 10.1016/b978-0-12-823808-0.00013-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
188
|
Oliveira AG, Fiorotto R. Novel approaches to liver disease diagnosis and modeling. Transl Gastroenterol Hepatol 2021; 6:19. [PMID: 33824923 PMCID: PMC7829068 DOI: 10.21037/tgh-20-109] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Lack of a prompt and accurate diagnosis remains on top of the list of challenges faced by patients with rare liver diseases. Although rare liver diseases affect a significant percentage of the population as a group, when taken singularly they represent unique diseases and the approaches used for diagnosis of common liver diseases are insufficient. However, the development of new methods for the acquisition of molecular and clinical data (i.e., genomic, proteomics, metabolomics) and computational tools for their analysis and integration, together with advances in modeling diseases using stem cell-based technology [i.e., induced pluripotent stem cells (iPSCs) and tissue organoids] represent a promising and powerful tool to improve the clinical management of these patients. This is the goal of precision medicine, a novel approach of modern medicine that aims at delivering a specific treatment based on disease-specific biological insights and individual profile. This review will discuss the application and advances of these technologies and how they represent a new opportunity in hepatology.
Collapse
Affiliation(s)
- André G. Oliveira
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Romina Fiorotto
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, USA
| |
Collapse
|
189
|
Abstract
Organoids are in vitro miniaturized and simplified model systems of organs that have gained enormous interest for modelling tissue development and disease, and for personalized medicine, drug screening and cell therapy. Despite considerable success in culturing physiologically relevant organoids, challenges remain to achieve real-life applications. In particular, the high variability of self-organizing growth and restricted experimental and analytical access hamper the translatability of organoid systems. In this Review, we argue that many limitations of traditional organoid culture can be addressed by engineering approaches at all levels of organoid systems. We investigate cell surface and genetic engineering approaches, and discuss stem cell niche engineering based on the design of matrices that allow spatiotemporal control of organoid growth and shape-guided morphogenesis. We examine how microfluidic approaches and lessons learnt from organs-on-a-chip enable the integration of mechano-physiological parameters and increase accessibility of organoids to improve functional readouts. Applying engineering principles to organoids increases reproducibility and provides experimental control, which will, ultimately, be required to enable clinical translation.
Collapse
Affiliation(s)
- Moritz Hofer
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Matthias P. Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science (SB), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
190
|
Current and Future Perspectives of the Use of Organoids in Radiobiology. Cells 2020; 9:cells9122649. [PMID: 33317153 PMCID: PMC7764598 DOI: 10.3390/cells9122649] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
The majority of cancer patients will be treated with radiotherapy, either alone or together with chemotherapy and/or surgery. Optimising the balance between tumour control and the probability of normal tissue side effects is the primary goal of radiation treatment. Therefore, it is imperative to understand the effects that irradiation will have on both normal and cancer tissue. The more classical lab models of immortal cell lines and in vivo animal models have been fundamental to radiobiological studies to date. However, each of these comes with their own limitations and new complementary models are required to fill the gaps left by these traditional models. In this review, we discuss how organoids, three-dimensional tissue-resembling structures derived from tissue-resident, embryonic or induced pluripotent stem cells, overcome the limitations of these models and thus have a growing importance in the field of radiation biology research. The roles of organoids in understanding radiation-induced tissue responses and in moving towards precision medicine are examined. Finally, the limitations of organoids in radiobiology and the steps being made to overcome these limitations are considered.
Collapse
|
191
|
Farzaneh Z, Abbasalizadeh S, Asghari-Vostikolaee MH, Alikhani M, Cabral JMS, Baharvand H. Dissolved oxygen concentration regulates human hepatic organoid formation from pluripotent stem cells in a fully controlled bioreactor. Biotechnol Bioeng 2020; 117:3739-3756. [PMID: 32725885 DOI: 10.1002/bit.27521] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/11/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022]
Abstract
Developing technologies for scalable production of human organoids has gained increased attention for "organoid medicine" and drug discovery. We developed a scalable and integrated differentiation process for generation of hepatic organoid from human pluripotent stem cells (hPSCs) in a fully controlled stirred tank bioreactor with 150 ml working volume by application of physiological oxygen concentrations in different liver tissue zones. We found that the 20-40% dissolved oxygen concentration [DO] (corresponded to 30-60 mmHg pO2 within the liver tissue) significantly influences the process outcome via regulating the differentiation fate of hPSC aggregates by enhancing mesoderm induction. Regulation of the [DO] at 30% DO resulted in efficient generation of human fetal-like hepatic organoids that had a uniform size distribution and were comprised of red blood cells and functional hepatocytes, which exhibited improved liver-specific marker gene expressions, key liver metabolic functions, and, more important, higher inducible cytochrome P450 activity compared to the other trials. These hepatic organoids were successfully engrafted in an acute liver injury mouse model and produced albumin after implantation. These results demonstrated the significant impact of the dissolved oxygen concentration on hPSC hepatic differentiation fate and differentiation efficacy that should be considered ascritical translational aspect of established scalable liver organoid generation protocols for potential clinical and drug discovery applications.
Collapse
Affiliation(s)
- Zahra Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Abbasalizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Institute Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Mohammad-Hassan Asghari-Vostikolaee
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mehdi Alikhani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Joaquim M S Cabral
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Institute Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, 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
| |
Collapse
|
192
|
Cox CR, Lynch S, Goldring C, Sharma P. Current Perspective: 3D Spheroid Models Utilizing Human-Based Cells for Investigating Metabolism-Dependent Drug-Induced Liver Injury. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:611913. [PMID: 35047893 PMCID: PMC8757888 DOI: 10.3389/fmedt.2020.611913] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver injury (DILI) remains a leading cause for the withdrawal of approved drugs. This has significant financial implications for pharmaceutical companies, places increasing strain on global health services, and causes harm to patients. For these reasons, it is essential that in-vitro liver models are capable of detecting DILI-positive compounds and their underlying mechanisms, prior to their approval and administration to patients or volunteers in clinical trials. Metabolism-dependent DILI is an important mechanism of drug-induced toxicity, which often involves the CYP450 family of enzymes, and is associated with the production of a chemically reactive metabolite and/or inefficient removal and accumulation of potentially toxic compounds. Unfortunately, many of the traditional in-vitro liver models fall short of their in-vivo counterparts, failing to recapitulate the mature hepatocyte phenotype, becoming metabolically incompetent, and lacking the longevity to investigate and detect metabolism-dependent DILI and those associated with chronic and repeat dosing regimens. Nevertheless, evidence is gathering to indicate that growing cells in 3D formats can increase the complexity of these models, promoting a more mature-hepatocyte phenotype and increasing their longevity, in vitro. This review will discuss the use of 3D in vitro models, namely spheroids, organoids, and perfusion-based systems to establish suitable liver models to investigate metabolism-dependent DILI.
Collapse
Affiliation(s)
- Christopher R. Cox
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- *Correspondence: Christopher R. Cox
| | - Stephen Lynch
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Goldring
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Parveen Sharma
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| |
Collapse
|
193
|
Hendriks D, Artegiani B, Hu H, Chuva de Sousa Lopes S, Clevers H. Establishment of human fetal hepatocyte organoids and CRISPR-Cas9-based gene knockin and knockout in organoid cultures from human liver. Nat Protoc 2020; 16:182-217. [PMID: 33247284 DOI: 10.1038/s41596-020-00411-2] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022]
Abstract
The liver is composed of two epithelial cell types: hepatocytes and liver ductal cells. Culture conditions for expansion of human liver ductal cells in vitro as organoids were previously described in a protocol; however, primary human hepatocytes remained hard to expand, until recently. In this protocol, we provide full details of how we overcame this limitation, establishing culture conditions that facilitate long-term expansion of human fetal hepatocytes as organoids. In addition, we describe how to generate (multi) gene knockouts using CRISPR-Cas9 in both human fetal hepatocyte and adult liver ductal organoid systems. Using a CRISPR-Cas9 and homology-independent organoid transgenesis (CRISPR-HOT) approach, efficient gene knockin can be achieved in these systems. These gene knockin and knockout approaches, and their multiplexing, should be useful for a variety of applications, such as disease modeling, investigating gene functions and studying processes, such as cellular differentiation and cell division. The protocol to establish human fetal hepatocyte organoid cultures takes ~1-2 months. The protocols to genome engineer human liver ductal organoids and human fetal hepatocyte organoids take 2-3 months.
Collapse
Affiliation(s)
- Delilah Hendriks
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, the Netherlands. .,Oncode Institute, Utrecht, the Netherlands.
| | - Benedetta Artegiani
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, the Netherlands. .,Oncode Institute, Utrecht, the Netherlands. .,The Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Huili Hu
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, the Netherlands
| | | | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, the Netherlands. .,Oncode Institute, Utrecht, the Netherlands. .,The Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
194
|
Lucendo-Villarin B, Meseguer-Ripolles J, Drew J, Fischer L, Ma E, Flint O, Simpson KJ, Machesky LM, Mountford JC, Hay DC. Development of a cost-effective automated platform to produce human liver spheroids for basic and applied research. Biofabrication 2020; 13:015009. [PMID: 33007774 DOI: 10.1088/1758-5090/abbdb2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/02/2020] [Indexed: 12/14/2022]
Abstract
Liver disease represents an increasing cause of global morbidity and mortality. Currently, liver transplant is the only treatment curative for end-stage liver disease. Donor organs cannot meet the demand and therefore scalable treatments and new disease models are required to improve clinical intervention. Pluripotent stem cells represent a renewable source of human tissue. Recent advances in three-dimensional cell culture have provided the field with more complex systems that better mimic liver physiology and function. Despite these improvements, current cell-based models are variable in performance and expensive to manufacture at scale. This is due, in part, to the use of poorly defined or cross-species materials within the process, severely affecting technology translation. To address this issue, we have developed an automated and economical platform to produce liver tissue at scale for modelling disease and small molecule screening. Stem cell derived liver spheres were formed by combining hepatic progenitors with endothelial cells and stellate cells, in the ratios found within the liver. The resulting tissue permitted the study of human liver biology 'in the dish' and could be scaled for screening. In summary, we have developed an automated differentiation system that permits reliable self-assembly of human liver tissue for biomedical application. Going forward we believe that this technology will not only serve as anin vitroresource, and may have an important role to play in supporting failing liver function in humans.
Collapse
Affiliation(s)
- B Lucendo-Villarin
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Both authors contributed equally to this manuscript
| | - J Meseguer-Ripolles
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Both authors contributed equally to this manuscript
| | - J Drew
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
| | - L Fischer
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - E Ma
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Campus, G61 1BD, United Kingdom
| | - O Flint
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - K J Simpson
- Scottish Liver Transplant Unit, Royal Infirmary, Edinburgh EH16 4SA, United Kingdom
| | - L M Machesky
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Campus, G61 1BD, United Kingdom
| | - J C Mountford
- SNBTS, 52 Research Avenue North, Heriot-Watt Research Park, Edinburgh EH14 4BE, United Kingdom
| | - D C Hay
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Author to whom any correspondence should be addressed
| |
Collapse
|
195
|
Priester C, MacDonald A, Dhar M, Bow A. Examining the Characteristics and Applications of Mesenchymal, Induced Pluripotent, and Embryonic Stem Cells for Tissue Engineering Approaches across the Germ Layers. Pharmaceuticals (Basel) 2020; 13:E344. [PMID: 33114710 PMCID: PMC7692540 DOI: 10.3390/ph13110344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
The field of regenerative medicine utilizes a wide array of technologies and techniques for repairing and restoring function to damaged tissues. Among these, stem cells offer one of the most potent and promising biological tools to facilitate such goals. Implementation of mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) offer varying advantages based on availability and efficacy in the target tissue. The focus of this review is to discuss characteristics of these three subset stem cell populations and examine their utility in tissue engineering. In particular, the development of therapeutics that utilize cell-based approaches, divided by germinal layer to further assess research targeting specific tissues of the mesoderm, ectoderm, and endoderm. The combinatorial application of MSCs, iPSCs, and ESCs with natural and synthetic scaffold technologies can enhance the reparative capacity and survival of implanted cells. Continued efforts to generate more standardized approaches for these cells may provide improved study-to-study variations on implementation, thereby increasing the clinical translatability of cell-based therapeutics. Coupling clinically translatable research with commercially oriented methods offers the potential to drastically advance medical treatments for multiple diseases and injuries, improving the quality of life for many individuals.
Collapse
Affiliation(s)
- Caitlin Priester
- Department of Animal Science, University of Tennessee, Knoxville, TN 37998, USA;
| | - Amber MacDonald
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Madhu Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Austin Bow
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| |
Collapse
|
196
|
He YT, Zhu XL, Li SF, Zhang BQ, Li Y, Wu Q, Zhang YL, Zhou YY, Li L, Qi YN, Bao J, Bu H. Creating rat hepatocyte organoid as an in vitro model for drug testing. World J Stem Cells 2020; 12:1184-1195. [PMID: 33178400 PMCID: PMC7596445 DOI: 10.4252/wjsc.v12.i10.1184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/15/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Liver organoids have recently been applied as models for liver disease and drug screening, especially when combined with liver-on-a-chip technologies. Compared to hepatocyte-like cells, primary hepatocytes have high functionality but cannot maintain their function when cultured in vitro. Mesenchymal stem cells (MSCs) enhance hepatocyte function and maintain hepatocyte metabolism when co-cultured with hepatocytes. MSCs can help induced pluripotent stem cells to generate an organoid structure via the MSC-based traction force triggered by extracellular matrix (ECM) proteins. In this study, primary hepatocytes were co-cultured with MSCs on a liver-derived ECM to generate liver organoids within a short duration.
AIM To create hepatocyte organoids by co-culturing primary hepatocytes with MSCs on a porcine liver extracellular matrix (PLECM) gel.
METHODS Perfusion and enzymatic hydrolysis were used to form the PLECM gel. Rat hepatocytes and human MSCs were mixed and plated on pre-solidified PLECM gel in a 48-well plate for 48 h to generate organoids. Generated organoids were evaluated through hematoxylin and eosin, periodic acid-Schiff, immuno-histological, and immunofluorescence staining, and quantitative PCR for alb, CYP450 gene markers, and urea cycle genes. Culture medium was collected to detect albumin (ALB) and urea production on days 2, 4, 6, 8, 14, and 20.
RESULTS The whole porcine liver was perfused and enzymatically hydrolyzed to form a PLECM gel. The structural components and basement membrane composition of the ECM, such as collagen type I, collagen type IV, fibronectin, and laminin, were demonstrated to be retained. Through interaction of human MSCs with the liver-derived ECM, primary hepatocytes and human MSCs assembled together into a 3D construction and generated primary hepatocyte organoids for 48 h. The mRNAs of the gene alb, the CYP450 gene markers cyp1a1, cyp1a2, and cyp3a2 as well as urea cycle genes arg-1, asl, ass-1, cps-1, nags were highly expressed in hepatocyte organoids. Long-term survival of the primary hepatocyte organoids, as well as stable functionality, was demonstrated via ALB and urea production in vitro.
CONCLUSION Our new method of creating primary hepatocyte organoids by co-culturing hepatocytes with MSCs on liver-derived ECM hydrogels could be used to develop models for liver disease and for drug screening.
Collapse
Affiliation(s)
- Yu-Ting He
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xing-Long Zhu
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Sheng-Fu Li
- Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bing-Qi Zhang
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yi Li
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qiong Wu
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yun-Lin Zhang
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yan-Yan Zhou
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li Li
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ya-Na Qi
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ji Bao
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hong Bu
- Department of Pathology, West China Hospital, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
197
|
Abstract
The organoid model represents a major breakthrough in cell biology that has revolutionised biomedical research. Organoids are 3D physiological in vitro structures that recapitulate morphological and functional features of in vivo tissues and offer significant advantages over traditional cell culture methods. Liver organoids are of particular interest because of the pleiotropy of functions exerted by the human liver, their utility to model different liver diseases, and their potential application as cell-based therapies in regenerative medicine. Moreover, because they can be derived from patient tissues, organoid models offer new perspectives in personalised medicine and drug discovery. In this review, we discuss the current liver organoid models for the study of liver disease.
Collapse
Key Words
- 3D cultures
- A1AT, alpha-1 antitrypsin
- ALD, alcohol-related liver disease
- CCA, cholangiocarcinoma
- CFTR, cystic fibrosis transmembrane conductance regulator
- CHC, combined hepato-cholangiocarcinoma
- CLD, chronic liver disease
- CTLN1, citrullinemia type 1
- Chol-orgs, cholangiocyte organoids
- Disease modelling
- EGF, epidermal growth factor
- ER, endoplasmic reticulum
- ESCs, embryonic stem cells
- FFAs, free fatty acids
- HCC, hepatocellular carcinoma
- HUVEC, human umbilical vein endothelial cells
- Hep-orgs, hepatocyte organoids
- IL-, interleukin-
- Liver disease
- MSC, mesenchymal stem cell
- NAFLD, non-alcoholic fatty liver disease
- Organoids
- PDO, patient-derived organoid
- PDX, patient-derived xenograft
- PHH, primary human hepatocyte
- PSC, primary sclerosing cholangitis
- Personalised medicine
- Preclinical models
- iPSC, induced pluripotent stem cell
Collapse
Affiliation(s)
- Sandro Nuciforo
- Department of Biomedicine, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| | - Markus H Heim
- Department of Biomedicine, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland.,Clarunis, University Center for Gastrointestinal and Liver Diseases, CH-4002 Basel, Switzerland
| |
Collapse
|
198
|
Sekine K, Ogawa S, Tsuzuki S, Kobayashi T, Ikeda K, Nakanishi N, Takeuchi K, Kanai E, Otake Y, Okamoto S, Kobayashi T, Takebe T, Taniguchi H. Generation of human induced pluripotent stem cell-derived liver buds with chemically defined and animal origin-free media. Sci Rep 2020; 10:17937. [PMID: 33087763 PMCID: PMC7578079 DOI: 10.1038/s41598-020-73908-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 09/23/2020] [Indexed: 01/09/2023] Open
Abstract
Advances in organoid technology have broadened the number of target diseases and conditions in which human induced pluripotent stem cell (iPSC)-based regenerative medicine can be applied; however, mass production of organoids and the development of chemically defined, animal origin-free (CD-AOF) media and supplements are unresolved issues that hamper the clinical applicability of these approaches. CD-AOF media and supplements ensure the quality and reproducibility of culture systems by lowering lot-to-lot variations and the risk of contamination with viruses or toxins. We previously generated liver organoids from iPSCs, namely iPSC-liver buds (iPSC-LBs), by mimicking the organogenic interactions among hepatocytes, endothelial cells (ECs), and mesenchymal cells (MCs) and recently reported the mass production of iPSC-LBs derived entirely from iPSCs (all iPSC-LBs), which should facilitate their large-scale production for the treatment of liver failure. However, in previous studies we used media originating from animals for differentiation except for the maintenance of undifferentiated iPSCs. Therefore, we developed a CD-AOF medium to generate all iPSC-LBs. We first developed a CD-AOF medium for hepatocytes, ECs, and stage-matched MCs, i.e., septum transversum mesenchyme (STM), in 2D cultures. We next generated all iPSC-LBs by incubating individual cell types in ultra-low attachment micro-dimple plates. The hepatic functions of all iPSC-LBs generated using the CD-AOF medium were equivalent to those of all iPSC-LBs generated using the conventional medium both in vitro and in vivo. Furthermore, we found that this CD-AOF medium could be used in several cell culture settings. Taken together, these results demonstrate the successful development of a CD-AOF medium suitable for all iPSC-LBs. The protocol developed in this study will facilitate the clinical applicability of all iPSC-LBs in the treatment of liver diseases.
Collapse
Affiliation(s)
- Keisuke Sekine
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan. .,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
| | - Shimpei Ogawa
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kawasaki, 210-8681, Japan
| | - Syusaku Tsuzuki
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Tatsuya Kobayashi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kazuki Ikeda
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Noriko Nakanishi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kenta Takeuchi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Eriko Kanai
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Yugo Otake
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Tsuyoshi Kobayashi
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kawasaki, 210-8681, Japan
| | - Takanori Takebe
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.,Advanced Medical Research Center, Yokohama City University, Yokohama, 236-0004, Japan.,Division of Gastroenterology, Hepatology and Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229-3039, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229-3039, USA
| | - Hideki Taniguchi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan. .,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan. .,Advanced Medical Research Center, Yokohama City University, Yokohama, 236-0004, Japan.
| |
Collapse
|
199
|
Ogoke O, Maloy M, Parashurama N. The science and engineering of stem cell-derived organoids-examples from hepatic, biliary, and pancreatic tissues. Biol Rev Camb Philos Soc 2020; 96:179-204. [PMID: 33002311 DOI: 10.1111/brv.12650] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 08/08/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
Abstract
The field of organoid engineering promises to revolutionize medicine with wide-ranging applications of scientific, engineering, and clinical interest, including precision and personalized medicine, gene editing, drug development, disease modelling, cellular therapy, and human development. Organoids are a three-dimensional (3D) miniature representation of a target organ, are initiated with stem/progenitor cells, and are extremely promising tools with which to model organ function. The biological basis for organoids is that they foster stem cell self-renewal, differentiation, and self-organization, recapitulating 3D tissue structure or function better than two-dimensional (2D) systems. In this review, we first discuss the importance of epithelial organs and the general properties of epithelial cells to provide a context and rationale for organoids of the liver, pancreas, and gall bladder. Next, we develop a general framework to understand self-organization, tissue hierarchy, and organoid cultivation. For each of these areas, we provide a historical context, and review a wide range of both biological and mathematical perspectives that enhance understanding of organoids. Next, we review existing techniques and progress in hepatobiliary and pancreatic organoid engineering. To do this, we review organoids from primary tissues, cell lines, and stem cells, and introduce engineering studies when applicable. We discuss non-invasive assessment of organoids, which can reveal the underlying biological mechanisms and enable improved assays for growth, metabolism, and function. Applications of organoids in cell therapy are also discussed. Taken together, we establish a broad scientific foundation for organoids and provide an in-depth review of hepatic, biliary and pancreatic organoids.
Collapse
Affiliation(s)
- Ogechi Ogoke
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, U.S.A
| | - Mitchell Maloy
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, U.S.A
| | - Natesh Parashurama
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, U.S.A.,Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), Buffalo, NY, U.S.A.,Department of Biomedical Engineering, University at Buffalo (State University of New York), Buffalo, NY, U.S.A
| |
Collapse
|
200
|
Sahu S, Sharan SK. Translating Embryogenesis to Generate Organoids: Novel Approaches to Personalized Medicine. iScience 2020; 23:101485. [PMID: 32864586 PMCID: PMC7441954 DOI: 10.1016/j.isci.2020.101485] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The astounding capacity of pluripotent stem cells (PSCs) to differentiate and self-organize has revolutionized the development of 3D cell culture models. The major advantage is its ability to mimic in vivo microenvironments and cellular interactions when compared with the classical 2D cell culture models. Recent innovations in generating embryo-like structures (including blastoids and gastruloids) from PSCs have advanced the experimental accessibility to understand embryogenesis with immense potential to model human development. Taking cues on how embryonic development leads to organogenesis, PSCs can also be directly differentiated to form mini-organs or organoids of a particular lineage. Organoids have opened new avenues to augment our understanding of stem cell and regenerative biology, tissue homeostasis, and disease mechanisms. In this review, we provide insights from developmental biology with a comprehensive resource of signaling pathways that in a coordinated manner form embryo-like structures and organoids. Moreover, the advent of assembloids and multilineage organoids from PSCs opens a new dimension to study paracrine function and multi-tissue interactions in vitro. Although this led to an avalanche of enthusiasm to utilize organoids for organ transplantation studies, we examine the current limitations and provide perspectives to improve reproducibility, scalability, functional complexity, and cell-type characterization. Taken together, these 3D in vitro organ-specific and patient-specific models hold great promise for drug discovery, clinical management, and personalized medicine.
Collapse
Affiliation(s)
- Sounak Sahu
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-04, 1050 Boyles Street, Frederick, MD 21702, USA
| | - Shyam K. Sharan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-33, 1050 Boyles Street, Frederick, MD 21702, USA
| |
Collapse
|