51
|
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: 24] [Impact Index Per Article: 8.0] [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
|
52
|
Cell therapy for advanced liver diseases: Repair or rebuild. J Hepatol 2021; 74:185-199. [PMID: 32976865 DOI: 10.1016/j.jhep.2020.09.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 08/18/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Advanced liver disease presents a significant worldwide health and economic burden and accounts for 3.5% of global mortality. When liver disease progresses to organ failure the only effective treatment is liver transplantation, which necessitates lifelong immunosuppression and carries associated risks. Furthermore, the shortage of suitable donor organs means patients may die waiting for a suitable transplant organ. Cell therapies have made their way from animal studies to a small number of early clinical trials. Herein, we review the current state of cell therapies for liver disease and the mechanisms underpinning their actions (to repair liver tissue or rebuild functional parenchyma). We also discuss cellular therapies that are on the clinical horizon and challenges that must be overcome before routine clinical use is a possibility.
Collapse
|
53
|
Michielin F, Giobbe GG, Luni C, Hu Q, Maroni I, Orford MR, Manfredi A, Di Filippo L, David AL, Cacchiarelli D, De Coppi P, Eaton S, Elvassore N. The Microfluidic Environment Reveals a Hidden Role of Self-Organizing Extracellular Matrix in Hepatic Commitment and Organoid Formation of hiPSCs. Cell Rep 2020; 33:108453. [PMID: 33264615 PMCID: PMC8237389 DOI: 10.1016/j.celrep.2020.108453] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/26/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022] Open
Abstract
The specification of the hepatic identity during human liver development is strictly controlled by extrinsic signals, yet it is still not clear how cells respond to these exogenous signals by activating secretory cascades, which are extremely relevant, especially in 3D self-organizing systems. Here, we investigate how the proteins secreted by human pluripotent stem cells (hPSCs) in response to developmental exogenous signals affect the progression from endoderm to the hepatic lineage, including their competence to generate nascent hepatic organoids. By using microfluidic confined environment and stable isotope labeling with amino acids in cell culture-coupled mass spectrometry (SILAC-MS) quantitative proteomic analysis, we find high abundancy of extracellular matrix (ECM)-associated proteins. Hepatic progenitor cells either derived in microfluidics or exposed to exogenous ECM stimuli show a significantly higher potential of forming hepatic organoids that can be rapidly expanded for several passages and further differentiated into functional hepatocytes. These results prove an additional control over the efficiency of hepatic organoid formation and differentiation for downstream applications.
Collapse
Affiliation(s)
- Federica Michielin
- Great Ormond Street Institute of Child Health, University College London, WC1N1EH London, UK
| | - Giovanni G Giobbe
- Great Ormond Street Institute of Child Health, University College London, WC1N1EH London, UK
| | - Camilla Luni
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, 201210 Shanghai, China
| | - Qianjiang Hu
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, 201210 Shanghai, China
| | - Ida Maroni
- Department of Industrial Engineering, University of Padova, 35131 Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Michael R Orford
- Great Ormond Street Institute of Child Health, University College London, WC1N1EH London, UK
| | - Anna Manfredi
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, 80078 Pozzuoli, Italy
| | | | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, WC1E 6AU London, UK
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, 80078 Pozzuoli, Italy; Department of Translational Medicine, University of Naples "Federico II," 80131 Naples, Italy
| | - Paolo De Coppi
- Great Ormond Street Institute of Child Health, University College London, WC1N1EH London, UK; Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital, WC1N 3JH London, UK
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, WC1N1EH London, UK
| | - Nicola Elvassore
- Great Ormond Street Institute of Child Health, University College London, WC1N1EH London, UK; Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, 201210 Shanghai, China; Department of Industrial Engineering, University of Padova, 35131 Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), 35129 Padova, Italy.
| |
Collapse
|
54
|
Lee J, Park D, Seo Y, Chung JJ, Jung Y, Kim SH. Organ-Level Functional 3D Tissue Constructs with Complex Compartments and their Preclinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002096. [PMID: 33103834 DOI: 10.1002/adma.202002096] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/16/2020] [Indexed: 06/11/2023]
Abstract
There is an increasing interest in organ-level 3D tissue constructs, owing to their mirroring of in vivo-like features. This has resulted in a wide range of preclinical applications to obtain cell- or tissue-specific responses. Additionally, the development and improvement of sophisticated technologies, such as organoid generation, microfluidics, hydrogel engineering, and 3D printing, have enhanced 3D tissue constructs to become more elaborate. In particular, recent studies have focused on including complex compartments, i.e., vascular and innervation structured 3D tissue constructs, which mimic the nature of the human body in that all tissues/organs are interconnected and physiological phenomena are mediated through vascular and neural systems. Here, the strategies are categorized according to the number of dimensions (0D, 1D, 2D, and 3D) of the starting materials for scaling up, and novel approaches to introduce increased complexity in 3D tissue constructs are highlighted. Recent advances in preclinical applications are also investigated to gain insight into the future direction of 3D tissue construct research. Overcoming the challenges in improving organ-level functional 3D tissue constructs both in vitro and in vivo will ultimately become a life-saving tool in the biomedical field.
Collapse
Affiliation(s)
- Jaeseo Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - DoYeun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Yoojin Seo
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Justin J Chung
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Soo Hyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| |
Collapse
|
55
|
Lee SW, Jung DJ, Jeong GS. Gaining New Biological and Therapeutic Applications into the Liver with 3D In Vitro Liver Models. Tissue Eng Regen Med 2020; 17:731-745. [PMID: 32207030 PMCID: PMC7710770 DOI: 10.1007/s13770-020-00245-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Three-dimensional (3D) cell cultures with architectural and biomechanical properties similar to those of natural tissue have been the focus for generating liver tissue. Microarchitectural organization is believed to be crucial to hepatic function, and 3D cell culture technologies have enabled the construction of tissue-like microenvironments, thereby leading to remarkable progress in vitro models of human tissue and organs. Recently, to recapitulate the 3D architecture of tissues, spheroids and organoids have become widely accepted as new practical tools for 3D organ modeling. Moreover, the combination of bioengineering approach offers the promise to more accurately model the tissue microenvironment of human organs. Indeed, the employment of sophisticated bioengineered liver models show long-term viability and functional enhancements in biochemical parameters and disease-orient outcome. RESULTS Various 3D in vitro liver models have been proposed as a new generation of liver medicine. Likewise, new biomedical engineering approaches and platforms are available to more accurately replicate the in vivo 3D microarchitectures and functions of living organs. This review aims to highlight the recent 3D in vitro liver model systems, including micropatterning, spheroids, and organoids that are either scaffold-based or scaffold-free systems. Finally, we discuss a number of challenges that will need to be addressed moving forward in the field of liver tissue engineering for biomedical applications. CONCLUSION The ongoing development of biomedical engineering holds great promise for generating a 3D biomimetic liver model that recapitulates the physiological and pathological properties of the liver and has biomedical applications.
Collapse
Affiliation(s)
- Sang Woo Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Da Jung Jung
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Gi Seok Jeong
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
- Department of Convergence Medicine, University of Ulsan College of Medicine, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
56
|
Chang PH, Chao HM, Chern E, Hsu SH. Chitosan 3D cell culture system promotes naïve-like features of human induced pluripotent stem cells: A novel tool to sustain pluripotency and facilitate differentiation. Biomaterials 2020; 268:120575. [PMID: 33341735 DOI: 10.1016/j.biomaterials.2020.120575] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/03/2020] [Accepted: 11/21/2020] [Indexed: 12/14/2022]
Abstract
A simplified and cost-effective culture system for maintaining the pluripotency of human induced pluripotent stem cells (hiPSCs) is crucial for stem cell applications. Although recombinant protein-based feeder-free hiPSC culture systems have been developed, their manufacturing processes are expensive and complicated, which hinders hiPSC technology progress. Chitosan, a versatile biocompatible polysaccharide, has been reported as a biomaterial for three-dimensional (3D) cell culture system that promotes the physiological activities of mesenchymal stem cells and cancer cells. In the current study, we demonstrated that chitosan membranes sustained proliferation and pluripotency of hiPSCs in long-term culture (up to 365 days). Moreover, using vitronectin as the comparison group, the pluripotency of hiPSCs grown on the membranes was altered into a naïve-like state, which, for pluripotent stem cells, is an earlier developmental stage with higher stemness. On the chitosan membranes, hiPSCs self-assembled into 3D spheroids with an average diameter of ~100 μm. These hiPSC spheroids could be directly differentiated into lineage-specific cells from the three germ layers with 3D structures. Collectively, chitosan membranes not only promoted the naïve pluripotent features of hiPSCs but also provided a novel 3D differentiation platform. This convenient biomaterial-based culture system may enable the effective expansion and accessibility of hiPSCs for regenerative medicine, disease modeling, and drug screening.
Collapse
Affiliation(s)
- Po-Hsiang Chang
- niChe Lab for Stem Cell and Regenerative Medicine, Department of Biochemical Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Hsiao-Mei Chao
- niChe Lab for Stem Cell and Regenerative Medicine, Department of Biochemical Science and Technology, National Taiwan University, Taipei 10617, Taiwan; Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Edward Chern
- niChe Lab for Stem Cell and Regenerative Medicine, Department of Biochemical Science and Technology, National Taiwan University, Taipei 10617, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 10617, Taiwan.
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
57
|
Kulkeaw K, Tubsuwan A, Tongkrajang N, Whangviboonkij N. Generation of human liver organoids from pluripotent stem cell-derived hepatic endoderms. PeerJ 2020; 8:e9968. [PMID: 33133779 PMCID: PMC7580584 DOI: 10.7717/peerj.9968] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 08/26/2020] [Indexed: 12/18/2022] Open
Abstract
Background The use of a personalized liver organoid derived from human-induced pluripotent stem cells (HuiPSCs) is advancing the use of in vitro disease models for the design of specific, effective therapies for individuals. Collecting patient peripheral blood cells for HuiPSC generation is preferable because it is less invasive; however, the capability of blood cell-derived HuiPSCs for hepatic differentiation and liver organoid formation remains uncertain. Moreover, the currently available methods for liver organoid formation require a multistep process of cell differentiation or a combination of hepatic endodermal, endothelial and mesenchymal cells, which is a major hurdle for the application of personalized liver organoids in high-throughput testing of drug toxicity and safety. To demonstrate the capability of blood cell-derived HuiPSCs for liver organoid formation without support from endothelial and mesenchymal cells. Methods The peripheral blood-derived HuiPSCs first differentiated into hepatic endoderm (HE) in two-dimensional (2D) culture on Matrigel-coated plates under hypoxia for 10 days. The HE was then collected and cultured in 3D culture using 50% Matrigel under ambient oxygen. The maturation of hepatocytes was further induced by adding hepatocyte growth medium containing HGF and oncostatin M on top of the 3D culture and incubating the culture for an additional 12–17 days. The function of the liver organoids was assessed using expression analysis of hepatocyte-specific gene and proteins. Albumin (ALB) synthesis, glycogen and lipid storage, and metabolism of indocyanine were evaluated. The spatial distribution of albumin was examined using immunofluorescence and confocal microscopy. Results CD34+ hematopoietic cell-derived HuiPSCs were capable of differentiating into definitive endoderm expressing SOX17 and FOXA2, hepatic endoderm expressing FOXA2, hepatoblasts expressing AFP and hepatocytes expressing ALB. On day 25 of the 2D culture, cells expressed SOX17, FOXA2, AFP and ALB, indicating the presence of cellular heterogeneity. In contrast, the hepatic endoderm spontaneously formed a spherical, hollow structure in a 3D culture of 50% Matrigel, whereas hepatoblasts and hepatocytes could not form. Microscopic observation showed a single layer of polygonal-shaped cells arranged in a 3D structure. The hepatic endoderm-derived organoid synthesis ALB at a higher level than the 2D culture but did not express definitive endoderm-specific SOX17, indicating the greater maturity of the hepatocytes in the liver organoids. Confocal microscopic images and quantitative ELISA confirmed albumin synthesis in the cytoplasm of the liver organoid and its secretion. Overall, 3D culture of the hepatic endoderm is a relatively fast, simple, and less laborious way to generate liver organoids from HuiPSCs that is more physiologically relevant than 2D culture.
Collapse
Affiliation(s)
- Kasem Kulkeaw
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Alisa Tubsuwan
- Stem Cell Research Group, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Nongnat Tongkrajang
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Narisara Whangviboonkij
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
58
|
Stem Cells and Hydrogels for Liver Tissue Engineering: Synergistic Cure for Liver Regeneration. Stem Cell Rev Rep 2020; 16:1092-1104. [DOI: 10.1007/s12015-020-10060-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 02/06/2023]
|
59
|
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: 7] [Impact Index Per Article: 1.8] [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
|
60
|
Wang Y, Liu H, Zhang M, Wang H, Chen W, Qin J. One-step synthesis of composite hydrogel capsules to support liver organoid generation from hiPSCs. Biomater Sci 2020; 8:5476-5488. [PMID: 32914807 DOI: 10.1039/d0bm01085e] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Advances in biomaterials, especially in hydrogels, have offered great opportunities for stem cell organoid engineering with higher controllability and fidelity. Here, we propose a novel strategy for one-step synthesis of composite hydrogel capsules (CHCs) that enable engineering liver organoids from human induced pluripotent stem cells (hiPSCs) in an oil-free droplet microfluidic system. The CHCs composed of a fibrin hydrogel core and an alginate-chitosan composite shell are synthesized by an enzymatic crosslinking reaction and electrostatic complexation within stable aqueous emulsions. The proposed CHCs exhibit high uniformity with biocompatibility, stability and high-throughput properties, as well as defined compositions. Moreover, the established system enables 3D culture, differentiation and self-organized formation of liver organoids in a continuous process by encapsulating hepatocyte-like cells derived from hiPSCs. The encapsulated liver organoids consisting of hepatocyte- and cholangiocyte-like cells show favorable cell viability and growth with consistent size. Furthermore, they maintain proper liver-specific functions including urea synthesis and albumin secretion, replicating the key features of the human liver. By combining stem cell biology, defined hydrogels and the droplet microfluidic technique, the proposed system is easy-to-operate, scalable and stable to engineer stem cell organoids, which may offer a robust platform to advance organoid research and translational applications.
Collapse
Affiliation(s)
- Yaqing Wang
- School of Chemistry, Dalian University of Technology, Dalian 116024, P.R. China.
| | | | | | | | | | | |
Collapse
|
61
|
Zhang Y, De Mets R, Monzel C, Acharya V, Toh P, Chin JFL, Van Hul N, Ng IC, Yu H, Ng SS, Tamir Rashid S, Viasnoff V. Biomimetic niches reveal the minimal cues to trigger apical lumen formation in single hepatocytes. NATURE MATERIALS 2020; 19:1026-1035. [PMID: 32341512 DOI: 10.1038/s41563-020-0662-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 03/11/2020] [Indexed: 06/11/2023]
Abstract
The symmetry breaking of protein distribution and cytoskeleton organization is an essential aspect for the development of apicobasal polarity. In embryonic cells this process is largely cell autonomous, while differentiated epithelial cells collectively polarize during epithelium formation. Here, we demonstrate that the de novo polarization of mature hepatocytes does not require the synchronized development of apical poles on neighbouring cells. De novo polarization at the single-cell level by mere contact with the extracellular matrix and immobilized cadherin defining a polarizing axis. The creation of these single-cell liver hemi-canaliculi allows unprecedented imaging resolution and control and over the lumenogenesis process. We show that the density and localization of cadherins along the initial cell-cell contact act as key triggers of the reorganization from lateral to apical actin cortex. The minimal cues necessary to trigger the polarization of hepatocytes enable them to develop asymmetric lumens with ectopic epithelial cells originating from the kidney, breast or colon.
Collapse
Affiliation(s)
- Yue Zhang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Richard De Mets
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Cornelia Monzel
- Experimental Medical Physics, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Pearlyn Toh
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Jasmine Fei Li Chin
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Noémi Van Hul
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Inn Chuan Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Hanry Yu
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Soon Seng Ng
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - S Tamir Rashid
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
- Institute for Liver Studies, King's College Hospital, King's College London, London, UK
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Department of Biological Science, National University of Singapore, Singapore, Singapore.
- Centre National de la Recherche Scientifique Unité Mixte Internationale, Singapore, Singapore.
| |
Collapse
|
62
|
Zeng F, Zhang Y, Han X, Weng J, Gao Y. Liver Buds and Liver Organoids: New Tools for Liver Development, Disease and Medical Application. Stem Cell Rev Rep 2020; 15:774-784. [PMID: 31863336 DOI: 10.1007/s12015-019-09909-z] [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] [Indexed: 12/17/2022]
Abstract
The current understanding and effective treatment of liver disease is far from satisfactory. Liver organoids and liver buds (LBs) transforming cell culture from two dimensions(2D) to three dimensions(3D) has provided infinite possibilities for stem cells to use in clinic. Recent technological advances in the 3D culture have shown the potentiality of liver organoids and LBs as the promising tool to model in vitro liver diseases. The induced LBs and liver organoids provide a platform for cell-based therapy, liver disease models, liver organogenesis and drugs screening. And its genetic heterogeneity supplies a way for the realization of precision medicine.
Collapse
Affiliation(s)
- Fanhong Zeng
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China
| | - Yue Zhang
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China
| | - Xu Han
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China
| | - Jun Weng
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China.
| | - Yi Gao
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China.
| |
Collapse
|
63
|
Functionalisation of a heat-derived and bio-inert albumin hydrogel with extracellular matrix by air plasma treatment. Sci Rep 2020; 10:12429. [PMID: 32709918 PMCID: PMC7382478 DOI: 10.1038/s41598-020-69301-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 06/22/2020] [Indexed: 12/23/2022] Open
Abstract
Albumin-based hydrogels are increasingly attractive in tissue engineering because they provide a xeno-free, biocompatible and potentially patient-specific platform for tissue engineering and drug delivery. The majority of research on albumin hydrogels has focused on bovine serum albumin (BSA), leaving human serum albumin (HSA) comparatively understudied. Different gelation methods are usually employed for HSA and BSA, and variations in the amino acid sequences of HSA and BSA exist; these account for differences in the hydrogel properties. Heat-induced gelation of aqueous HSA is the easiest method of synthesizing HSA hydrogels however hydrogel opacity and poor cell attachment limit their usefulness in downstream applications. Here, a solution to this problem is presented. Stable and translucent HSA hydrogels were created by controlled thermal gelation and the addition of sodium chloride. The resulting bio-inert hydrogel was then subjected to air plasma treatment which functionalised its surface, enabling the attachment of basement membrane matrix (Geltrex). In vitro survival and proliferation studies of foetal human osteoblasts subsequently demonstrated good biocompatibility of functionalised albumin hydrogels compared to untreated samples. Thus, air plasma treatment enables functionalisation of inert heat-derived HSA hydrogels with extracellular matrix proteins and these may be used as a xeno-free platform for biomedical research or cell therapy.
Collapse
|
64
|
Ding C, Chen X, Kang Q, Yan X. Biomedical Application of Functional Materials in Organ-on-a-Chip. Front Bioeng Biotechnol 2020; 8:823. [PMID: 32793573 PMCID: PMC7387427 DOI: 10.3389/fbioe.2020.00823] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/29/2020] [Indexed: 01/06/2023] Open
Abstract
The organ-on-a-chip (OOC) technology has been utilized in a lot of biomedical fields such as fundamental physiological and pharmacological researches. Various materials have been introduced in OOC and can be broadly classified into inorganic, organic, and hybrid materials. Although PDMS continues to be the preferred material for laboratory research, materials for OOC are constantly evolving and progressing, and have promoted the development of OOC. This mini review provides a summary of the various type of materials for OOC systems, focusing on the progress of materials and related fabrication technologies within the last 5 years. The advantages and drawbacks of these materials in particular applications are discussed. In addition, future perspectives and challenges are also discussed.
Collapse
Affiliation(s)
- Chizhu Ding
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Xiang Chen
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Qinshu Kang
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Xianghua Yan
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| |
Collapse
|
65
|
Liu H, Wang Y, Wang H, Zhao M, Tao T, Zhang X, Qin J. A Droplet Microfluidic System to Fabricate Hybrid Capsules Enabling Stem Cell Organoid Engineering. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903739. [PMID: 32537414 PMCID: PMC7284190 DOI: 10.1002/advs.201903739] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 05/14/2023]
Abstract
Organoids derived from self-organizing stem cells represent a major technological breakthrough with the potential to revolutionize biomedical research. However, building high-fidelity organoids in a reproducible and high-throughput manner remains challenging. Here, a droplet microfluidic system is developed for controllable fabrication of hybrid hydrogel capsules, which allows for massive 3D culture and formation of functional and uniform islet organoids derived from human-induced pluripotent stem cells (hiPSCs). In this all-in-water microfluidic system, an array of droplets is utilized as templates for one-step fabrication of binary capsules relying on interfacial complexation of oppositely charged Na-alginate (NaA) and chitosan (CS). The produced hybrid capsules exhibit high uniformity, and are biocompatible, stable, and permeable. The established system enables capsule production, 3D culture, and self-organizing formation of human islet organoids in a continuous process by encapsulating pancreatic endocrine cells from hiPSCs. The generated islet organoids contain islet-specific α- and β-like cells with high expression of pancreatic hormone specific genes and proteins. Moreover, they exhibit sensitive glucose-stimulated insulin secretion function, demonstrating the capability of these binary capsules to engineer human organoids from hiPSCs. The proposed system is scalable, easy-to-operate, and stable, which can offer a robust platform for advancing human organoids research and translational applications.
Collapse
Affiliation(s)
- Haitao Liu
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinDivision of BiotechnologyCAS Key Laboratory of SSACDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinUniversity of Chinese Academy of SciencesBeijing100049China
| | - Yaqing Wang
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinDivision of BiotechnologyCAS Key Laboratory of SSACDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinUniversity of Chinese Academy of SciencesBeijing100049China
| | - Hui Wang
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinDivision of BiotechnologyCAS Key Laboratory of SSACDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinUniversity of Chinese Academy of SciencesBeijing100049China
| | - Mengqian Zhao
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinDivision of BiotechnologyCAS Key Laboratory of SSACDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinUniversity of Chinese Academy of SciencesBeijing100049China
| | - Tingting Tao
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinDivision of BiotechnologyCAS Key Laboratory of SSACDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xu Zhang
- Dr. X. ZhangDepartment of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Jianhua Qin
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinDivision of BiotechnologyCAS Key Laboratory of SSACDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- H. Liu, Y. Wang, H. Wang, M. Zhao, T. Tao, Prof. J. QinUniversity of Chinese Academy of SciencesBeijing100049China
- Prof. J. QinInstitute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
- Prof. J. QinCAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| |
Collapse
|
66
|
Yap KK, Gerrand YW, Dingle AM, Yeoh GC, Morrison WA, Mitchell GM. Liver sinusoidal endothelial cells promote the differentiation and survival of mouse vascularised hepatobiliary organoids. Biomaterials 2020; 251:120091. [PMID: 32408048 DOI: 10.1016/j.biomaterials.2020.120091] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 04/15/2020] [Accepted: 05/02/2020] [Indexed: 02/08/2023]
Abstract
The structural and physiological complexity of currently available liver organoids is limited, thereby reducing their relevance for drug studies, disease modelling, and regenerative therapy. In this study we combined mouse liver progenitor cells (LPCs) with mouse liver sinusoidal endothelial cells (LSECs) to generate hepatobiliary organoids with liver-specific vasculature. Organoids consisting of 5x103 cells were created from either LPCs, or a 1:1 combination of LPC/LSECs. LPC organoids demonstrated mild hepatobiliary differentiation in vitro with minimal morphological change; in contrast LPC/LSEC organoids developed clusters of polygonal hepatocyte-like cells and biliary ducts over a 7 day period. Hepatic (albumin, CPS1, CYP3A11) and biliary (GGT1) genes were significantly upregulated in LPC/LSEC organoids compared to LPC organoids over 7 days, as was albumin secretion. LPC/LSEC organoids also had significantly higher in vitro viability compared to LPC organoids. LPC and LPC/LSEC organoids were transplanted into vascularised chambers created in Fah-/-/Rag2-/-/Il2rg-/- mice (50 LPC organoids, containing 2.5x105 LPCs, and 100 LPC/LSEC organoids, containing 2.5x105 LPCs). At 2 weeks, minimal LPCs survived in chambers with LPC organoids, but robust hepatobiliary ductular tissue was present in LPC/LSEC organoids. Morphometric analysis demonstrated a 115-fold increase in HNF4α+ cells in LPC/LSEC organoid chambers (17.26 ± 4.34 cells/mm2 vs 0.15 ± 0.15 cells/mm2, p = 0.018), and 42-fold increase in Sox9+ cells in LPC/LSEC organoid chambers (28.29 ± 6.05 cells/mm2 vs 0.67 ± 0.67 cells/mm2, p = 0.011). This study presents a novel method to develop vascularised hepatobiliary organoids, with both in vitro and in vivo results confirming that incorporating LSECs with LPCs into organoids significantly increases the differentiation of hepatobiliary tissue within organoids and their survival post-transplantation.
Collapse
Affiliation(s)
- Kiryu K Yap
- O'Brien Institute, Department of St Vincent's Institute, Victoria, Australia; University of Melbourne Department of Surgery, St Vincent's Hospital Melbourne, Victoria, Australia.
| | - Yi-Wen Gerrand
- O'Brien Institute, Department of St Vincent's Institute, Victoria, Australia
| | - Aaron M Dingle
- O'Brien Institute, Department of St Vincent's Institute, Victoria, Australia
| | - George C Yeoh
- Harry Perkins Institute of Medical Research & Centre for Medical Research, University of Western Australia, Western Australia, Australia
| | - Wayne A Morrison
- O'Brien Institute, Department of St Vincent's Institute, Victoria, Australia; University of Melbourne Department of Surgery, St Vincent's Hospital Melbourne, Victoria, Australia; Australian Catholic University, Victoria, Australia
| | - Geraldine M Mitchell
- O'Brien Institute, Department of St Vincent's Institute, Victoria, Australia; University of Melbourne Department of Surgery, St Vincent's Hospital Melbourne, Victoria, Australia; Australian Catholic University, Victoria, Australia
| |
Collapse
|
67
|
Carpenter R, Macres D, Kwak JG, Daniel K, Lee J. Fabrication of Bioactive Inverted Colloidal Crystal Scaffolds Using Expanded Polystyrene Beads. Tissue Eng Part C Methods 2020; 26:143-155. [PMID: 32031058 PMCID: PMC7099427 DOI: 10.1089/ten.tec.2019.0333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/03/2020] [Indexed: 12/19/2022] Open
Abstract
Inverted colloidal crystal (ICC) hydrogel scaffolds have emerged as a new class of three-dimensional cell culture matrix that represents a unique opportunity to reproduce lymphoid tissue microenvironments. ICC geometry promotes the formation of stromal cell networks and their interaction with hematopoietic cells, a core cellular process in lymphoid tissues. When subdermally implanted, ICC hydrogel scaffolds direct unique foreign body responses to form a vascularized stromal tissue with prolonged attraction of hematopoietic cells, which together resemble lymphoid tissue microenvironments. While conceptually simple, fabrication of ICC hydrogel scaffold requires multiple steps and laborious handling of delicate materials. Here, we introduce a facile route for ICC hydrogel scaffold fabrication using expanded polystyrene (EPS) beads. EPS beads shrink and fuse in a tunable manner under pressurized thermal conditions, which serves as colloidal crystal templates for ICC scaffold fabrication. Inclusion of collagen in the precursor solution greatly simplified preparation of bioactive hydrogel scaffolds. The resultant EPS-templated bioactive ICC hydrogel scaffolds demonstrate characteristic features required for lymphoid tissue modeling in both in vitro and in vivo settings. We envision that the presented method will facilitate widespread implementation of ICC hydrogel scaffolds for lymphoid tissue engineering and other emerging applications. Impact statement Inverted colloidal crystal (ICC) hydrogel scaffolds have emerged as a new class of three-dimensional cell culture matrix that represents a unique opportunity for lymphoid tissue modeling and other emerging novel bioengineering applications. While conceptually simple, fabrication of the ICC hydrogel scaffold requires multiple steps and laborious handling of delicate materials with highly toxic chemicals. The presented method for ICC hydrogel scaffold fabrication using expanded polystyrene (EPS) beads is simple, cost-effective, and involves less toxic chemicals than conventional methods, while retaining comparable biological significance. We envision that EPS bead-based hydrogel scaffold fabrication will greatly facilitate the widespread implementation of ICC hydrogel scaffolds and their practical applications.
Collapse
Affiliation(s)
- Ryan Carpenter
- Department of Chemical Engineering, Institute for Applied Life Sciences, UMass-Amherst, Amherst, Massachusetts
| | - Dalton Macres
- Department of Biomedical Engineering, UMass-Amherst, Amherst, Massachusetts
| | - Jun-Goo Kwak
- Molecular and Cellular Biology Graduate Program, UMass-Amherst, Amherst, Massachusetts
| | - Katherine Daniel
- Department of Biomedical Engineering, UMass-Amherst, Amherst, Massachusetts
| | - Jungwoo Lee
- Department of Chemical Engineering, Institute for Applied Life Sciences, UMass-Amherst, Amherst, Massachusetts
- Molecular and Cellular Biology Graduate Program, UMass-Amherst, Amherst, Massachusetts
| |
Collapse
|
68
|
Kwak JG, Lee J. Thermoresponsive Inverted Colloidal Crystal Hydrogel Scaffolds for Lymphoid Tissue Engineering. Adv Healthc Mater 2020; 9:e1901556. [PMID: 32017462 PMCID: PMC7103457 DOI: 10.1002/adhm.201901556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/05/2020] [Indexed: 12/15/2022]
Abstract
Inverted colloidal crystal (ICC) hydrogel scaffolds represent unique opportunities in modeling lymphoid tissues and expanding hematopoietic-lymphoid cells. Fully interconnected spherical pore arrays direct the formation of stromal networks and facilitate interactions between stroma and hematopoietic-lymphoid cells. However, due to the intricate architecture of these materials, release of expanded cells is restricted and requires mechanical disruption or chemical dissolution of the hydrogel scaffold. One potent biomaterials strategy to release pore-entrapped hematopoietic-lymphoid cells without breaking the scaffolds apart is to transiently increase the dimensions of these materials using stimuli-responsive polymers. Having this mindset, thermoresponsive ICC scaffolds that undergo rapid (<1 min) and substantial (>300%) diameter change over a physiological temperature range (4-37 °C) by using poly(N-isopropylacrylamide) (PNIPAM) with nanogel crosslinkers is developed. For a proof-of-concept study, the stromal niche by creating osteospheroids, aggregates of osteoblasts, and bone chips is first replicated, and subsequently Nalm-6 model hematopoietic-lymphoid cells are introduced. A sixfold increase in cell count is harvested when ICC hydrogel scaffolds are expanded without termination of the established 3D stromal cell culture. It is envisioned that thermoresponsive ICC hydrogel scaffolds will enable for scalable and sustainable ex vivo expansion of hematopoietic-lymphoid cells.
Collapse
Affiliation(s)
- Jun-Goo Kwak
- Molecular and Cellular Biology Graduate Program, Institute for Applied Life Sciences, University of Massachusetts Amherst, Life Sciences Laboratories N560, 240 Thatcher Road, Amherst, MA, 01003, USA
| | - Jungwoo Lee
- Department of Chemical Engineering, Molecular and Cellular Biology Graduate Program, Institute for Applied Life Sciences, University of Massachusetts Amherst, Life Sciences Laboratories N567, 240 Thatcher Road, Amherst, MA, 01003, USA
| |
Collapse
|
69
|
da Silva Morais A, Vieira S, Zhao X, Mao Z, Gao C, Oliveira JM, Reis RL. Advanced Biomaterials and Processing Methods for Liver Regeneration: State-of-the-Art and Future Trends. Adv Healthc Mater 2020; 9:e1901435. [PMID: 31977159 DOI: 10.1002/adhm.201901435] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Liver diseases contribute markedly to the global burden of mortality and disease. The limited organ disposal for orthotopic liver transplantation results in a continuing need for alternative strategies. Over the past years, important progress has been made in the field of tissue engineering (TE). Many of the early trials to improve the development of an engineered tissue construct are based on seeding cells onto biomaterial scaffolds. Nowadays, several TE approaches have been developed and are applied to one vital organ: the liver. Essential elements must be considered in liver TE-cells and culturing systems, bioactive agents or growth factors (GF), and biomaterials and processing methods. The potential of hepatocytes, mesenchymal stem cells, and others as cell sources is demonstrated. They need engineered biomaterial-based scaffolds with perfect biocompatibility and bioactivity to support cell proliferation and hepatic differentiation as well as allowing extracellular matrix deposition and vascularization. Moreover, they require a microenvironment provided using conventional or advanced processing technologies in order to supply oxygen, nutrients, and GF. Herein the biomaterials and the conventional and advanced processing technologies, including cell-sheets process, 3D bioprinting, and microfluidic systems, as well as the future trends in these major fields are discussed.
Collapse
Affiliation(s)
- Alain da Silva Morais
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Sílvia Vieira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Xinlian Zhao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Joaquim M. Oliveira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| | - Rui L. Reis
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
70
|
Liu H, Wang Y, Cui K, Guo Y, Zhang X, Qin J. Advances in Hydrogels in Organoids and Organs-on-a-Chip. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902042. [PMID: 31282047 DOI: 10.1002/adma.201902042] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/25/2019] [Indexed: 05/10/2023]
Abstract
Significant advances in materials, microscale technology, and stem cell biology have enabled the construction of 3D tissues and organs, which will ultimately lead to more effective diagnostics and therapy. Organoids and organs-on-a-chip (OOC), evolved from developmental biology and bioengineering principles, have emerged as major technological breakthrough and distinct model systems to revolutionize biomedical research and drug discovery by recapitulating the key structural and functional complexity of human organs in vitro. There is growing interest in the development of functional biomaterials, especially hydrogels, for utilization in these promising systems to build more physiologically relevant 3D tissues with defined properties. The remarkable properties of defined hydrogels as proper extracellular matrix that can instruct cellular behaviors are presented. The recent trend where functional hydrogels are integrated into organoids and OOC systems for the construction of 3D tissue models is highlighted. Future opportunities and perspectives in the development of advanced hydrogels toward accelerating organoids and OOC research in biomedical applications are also discussed.
Collapse
Affiliation(s)
- Haitao Liu
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqing Wang
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kangli Cui
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqiong Guo
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xu Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Jianhua Qin
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
71
|
Corbett JL, Duncan SA. iPSC-Derived Hepatocytes as a Platform for Disease Modeling and Drug Discovery. Front Med (Lausanne) 2019; 6:265. [PMID: 31803747 PMCID: PMC6873655 DOI: 10.3389/fmed.2019.00265] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022] Open
Abstract
The liver is one of the largest organs in the body and is responsible for a diverse repertoire of metabolic processes. Such processes include the secretion of serum proteins, carbohydrate and lipid metabolism, bile acid and urea synthesis, detoxification of drugs and metabolic waste products, and vitamin and carbohydrate storage. Currently, liver disease is one of the most prevalent causes of mortality in the USA with congenital liver defects contributing to a significant proportion of these deaths. Historically the study of liver disease has been hampered by a shortage of organ donors, the subsequent scarcity of healthy tissue, and the failure of animal models to fully recapitulate human liver function. In vitro culture of hepatocytes has also proven difficult because primary hepatocytes rapidly de-differentiate in culture. Recent advances in stem cell technology have facilitated the generation of induced pluripotent stem cells (iPSCs) from various somatic cell types from patients. Such cells can be differentiated to a liver cell fate, essentially providing a limitless supply of cells with hepatocyte characteristics that can mimic the pathophysiology of liver disease. Furthermore, development of the CRISPR-Cas9 system, as well as advancement of miniaturized differentiation platforms has facilitated the development of high throughput models for the investigation of hepatocyte differentiation and drug discovery. In this review, we will explore the latest advances in iPSC-based disease modeling and drug screening platforms and examine how this technology is being used to identify new pharmacological interventions, and to advance our understanding of liver development and mechanisms of disease. We will cover how iPSC technology is being used to develop predictive models for rare diseases and how information gained from large in vitro screening experiments can be used to directly inform clinical investigation.
Collapse
Affiliation(s)
| | - Stephen A. Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
72
|
Agarwal T, Subramanian B, Maiti TK. Liver Tissue Engineering: Challenges and Opportunities. ACS Biomater Sci Eng 2019; 5:4167-4182. [PMID: 33417776 DOI: 10.1021/acsbiomaterials.9b00745] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver tissue engineering aims at the possibility of reproducing a fully functional organ for the treatment of acute and chronic liver disorders. Approaches in this field endeavor to replace organ transplantation (gold standard treatment for liver diseases in a clinical setting) with in vitro developed liver tissue constructs. However, the complexity of the liver microarchitecture and functionality along with the limited supply of cellular components of the liver pose numerous challenges. This review provides a comprehensive outlook onto how the physicochemical, mechanobiological, and spatiotemporal aspects of the substrates could be tuned to address current challenges in the field. We also highlight the strategic advancements made in the field so far for the development of artificial liver tissue. We further showcase the currently available prototypes in research and clinical trials, which shows the hope for the future of liver tissue engineering.
Collapse
|
73
|
Adult and iPS-derived non-parenchymal cells regulate liver organoid development through differential modulation of Wnt and TGF-β. Stem Cell Res Ther 2019; 10:258. [PMID: 31416480 PMCID: PMC6694663 DOI: 10.1186/s13287-019-1367-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/15/2019] [Accepted: 07/31/2019] [Indexed: 01/03/2023] Open
Abstract
Background Liver organoid technology holds great promises to be used in large-scale population-based drug screening and in future regenerative medicine strategies. Recently, some studies reported robust protocols for generating isogenic liver organoids using liver parenchymal and non-parenchymal cells derived from induced pluripotent stem cells (iPS) or using isogenic adult primary non-parenchymal cells. However, the use of whole iPS-derived cells could represent great challenges for a translational perspective. Methods Here, we evaluated the influence of isogenic versus heterogenic non-parenchymal cells, using iPS-derived or adult primary cell lines, in the liver organoid development. We tested four groups comprised of all different combinations of non-parenchymal cells for the liver functionality in vitro. Gene expression and protein secretion of important hepatic function markers were evaluated. Additionally, liver development-associated signaling pathways were tested. Finally, organoid label-free proteomic analysis and non-parenchymal cell secretome were performed in all groups at day 12. Results We show that liver organoids generated using primary mesenchymal stromal cells and iPS-derived endothelial cells expressed and produced significantly more albumin and showed increased expression of CYP1A1, CYP1A2, and TDO2 while presented reduced TGF-β and Wnt signaling activity. Proteomics analysis revealed that major shifts in protein expression induced by this specific combination of non-parenchymal cells are related to integrin profile and TGF-β/Wnt signaling activity. Conclusion Aiming the translation of this technology bench-to-bedside, this work highlights the role of important developmental pathways that are modulated by non-parenchymal cells enhancing the liver organoid maturation. Electronic supplementary material The online version of this article (10.1186/s13287-019-1367-x) contains supplementary material, which is available to authorized users.
Collapse
|
74
|
Bioengineering an Artificial Human Blood⁻Brain Barrier in Rodents. Bioengineering (Basel) 2019; 6:bioengineering6020038. [PMID: 31052208 PMCID: PMC6630638 DOI: 10.3390/bioengineering6020038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/15/2022] Open
Abstract
Our group has recently created a novel in-vivo human brain organoid vascularized with human iPSC-derived endothelial cells. In this review article, we discuss the challenges of creating a perfused human brain organoid model in an immunosuppressed rodent host and discuss potential applications for neurosurgical disease modeling.
Collapse
|
75
|
Blackford SJ, Ng SS, Segal JM, King AJ, Austin AL, Kent D, Moore J, Sheldon M, Ilic D, Dhawan A, Mitry RR, Rashid ST. Validation of Current Good Manufacturing Practice Compliant Human Pluripotent Stem Cell-Derived Hepatocytes for Cell-Based Therapy. Stem Cells Transl Med 2019; 8:124-137. [PMID: 30456803 PMCID: PMC6344902 DOI: 10.1002/sctm.18-0084] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/22/2018] [Accepted: 09/25/2018] [Indexed: 01/04/2023] Open
Abstract
Recent advancements in the production of hepatocytes from human pluripotent stem cells (hPSC-Heps) afford tremendous possibilities for treatment of patients with liver disease. Validated current good manufacturing practice (cGMP) lines are an essential prerequisite for such applications but have only recently been established. Whether such cGMP lines are capable of hepatic differentiation is not known. To address this knowledge gap, we examined the proficiency of three recently derived cGMP lines (two hiPSC and one hESC) to differentiate into hepatocytes and their suitability for therapy. hPSC-Heps generated using a chemically defined four-step hepatic differentiation protocol uniformly demonstrated highly reproducible phenotypes and functionality. Seeding into a 3D poly(ethylene glycol)-diacrylate fabricated inverted colloid crystal scaffold converted these immature progenitors into more advanced hepatic tissue structures. Hepatic constructs could also be successfully encapsulated into the immune-privileged material alginate and remained viable as well as functional upon transplantation into immune competent mice. This is the first report we are aware of demonstrating cGMP-compliant hPSCs can generate cells with advanced hepatic function potentially suitable for future therapeutic applications. Stem Cells Translational Medicine 2019;8:124&14.
Collapse
Affiliation(s)
- Samuel J.I. Blackford
- Centre for Stem Cells and Regenerative MedicineKing's College LondonLondonUnited Kingdom
| | - Soon Seng Ng
- Centre for Stem Cells and Regenerative MedicineKing's College LondonLondonUnited Kingdom
| | - Joe M. Segal
- Centre for Stem Cells and Regenerative MedicineKing's College LondonLondonUnited Kingdom
| | - Aileen J.F. King
- Diabetes Research GroupFaculty of Life Sciences & Medicine, King's College LondonLondonUnited Kingdom
| | - Amazon L. Austin
- Diabetes Research GroupFaculty of Life Sciences & Medicine, King's College LondonLondonUnited Kingdom
| | - Deniz Kent
- Centre for Stem Cells and Regenerative MedicineKing's College LondonLondonUnited Kingdom
| | - Jennifer Moore
- RUCDR Infinite BiologicsRutgers UniversityNew BrunswickNew JerseyUSA
| | - Michael Sheldon
- RUCDR Infinite BiologicsRutgers UniversityNew BrunswickNew JerseyUSA
| | - Dusko Ilic
- Stem Cell Laboratory, Department of Women and Children's HealthFaculty of Life Sciences and Medicine, King's College LondonLondonUnited Kingdom
| | - Anil Dhawan
- Institute for Liver StudiesKing's College Hospital, King's College LondonLondonUnited Kingdom
| | - Ragai R. Mitry
- Institute for Liver StudiesKing's College Hospital, King's College LondonLondonUnited Kingdom
| | - S. Tamir Rashid
- Centre for Stem Cells and Regenerative MedicineKing's College LondonLondonUnited Kingdom
- Institute for Liver StudiesKing's College Hospital, King's College LondonLondonUnited Kingdom
| |
Collapse
|