1
|
O'Connor CE, Zhang F, Neufeld A, Prado O, Simmonds SP, Fortin CL, Johansson F, Mene J, Saxton SH, Kopyeva I, Gregorio NE, James Z, DeForest CA, Wayne EC, Witten DM, Stevens KR. Bioprinted platform for parallelized screening of engineered microtissues in vivo. Cell Stem Cell 2025; 32:838-853.e6. [PMID: 40168987 DOI: 10.1016/j.stem.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 12/19/2024] [Accepted: 03/04/2025] [Indexed: 04/03/2025]
Abstract
Human engineered tissues hold great promise for therapeutic tissue regeneration and repair. Yet, development of these technologies often stalls at the stage of in vivo studies due to the complexity of engineered tissue formulations, which are often composed of diverse cell populations and material elements, along with the tedious nature of in vivo experiments. We introduce a "plug and play" platform called parallelized host apposition for screening tissues in vivo (PHAST). PHAST enables parallelized in vivo testing of 43 three-dimensional microtissues in a single 3D-printed device. Using PHAST, we screen microtissue formations with varying cellular and material components and identify formulations that support vascular graft-host inosculation and engineered liver tissue function in vivo. Our studies reveal that the cellular population(s) that should be included in engineered tissues for optimal in vivo performance is material dependent. PHAST could thus accelerate development of human tissue therapies for clinical regeneration and repair.
Collapse
Affiliation(s)
- Colleen E O'Connor
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Fan Zhang
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Anna Neufeld
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Olivia Prado
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Susana P Simmonds
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Chelsea L Fortin
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA; Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
| | - Fredrik Johansson
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Jonathan Mene
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Sarah H Saxton
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Irina Kopyeva
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Nicole E Gregorio
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Zachary James
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Cole A DeForest
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA; Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth C Wayne
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Daniela M Witten
- Department of Statistics, University of Washington, Seattle, WA, USA; Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Kelly R Stevens
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA; Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98195, USA; Brotman Baty Institute, Seattle, WA 98195, USA.
| |
Collapse
|
2
|
Wang S, Wang X, Wang Y. The Progress and Promise of Lineage Reprogramming Strategies for Liver Regeneration. Cell Mol Gastroenterol Hepatol 2024; 18:101395. [PMID: 39218152 PMCID: PMC11530608 DOI: 10.1016/j.jcmgh.2024.101395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The liver exhibits remarkable regenerative capacity. However, the limited ability of primary human hepatocytes to proliferate in vitro, combined with a compromised regenerative capacity induced by pathological conditions in vivo, presents significant obstacles to effective liver regeneration following liver injuries and diseases. Developing strategies to compensate for the loss of endogenous hepatocytes is crucial for overcoming these challenges, and this remains an active area of investigation. Lineage reprogramming, the process of directly converting one cell type into another bypassing the intermediate pluripotent state, has emerged as a promising method for generating specific cell types for therapeutic purposes in regenerative medicine. Here, we discuss the recent progress and emergent technologies in lineage reprogramming into hepatic cells, and their potential applications in enhancing liver regeneration or treating liver disease models. We also address controversies and challenges that confront this field.
Collapse
Affiliation(s)
- Shuyong Wang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China.
| | - Xuan Wang
- Hepatopancreatobiliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Yunfang Wang
- Hepatopancreatobiliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Beijing, China.
| |
Collapse
|
3
|
Zhang J, Chen X, Chai Y, Zhuo C, Xu Y, Xue T, Shao D, Tao Y, Li M. 3D Printing of a Vascularized Mini-Liver Based on the Size-Dependent Functional Enhancements of Cell Spheroids for Rescue of Liver Failure. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309899. [PMID: 38380546 PMCID: PMC11077657 DOI: 10.1002/advs.202309899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Indexed: 02/22/2024]
Abstract
The emerging stem cell-derived hepatocyte-like cells (HLCs) are the alternative cell sources of hepatocytes for treatment of highly lethal acute liver failure (ALF). However, the hostile local environment and the immature cell differentiation may compromise their therapeutic efficacy. To this end, human adipose-derived mesenchymal stromal/stem cells (hASCs) are engineered into different-sized multicellular spheroids and co-cultured with 3D coaxially and hexagonally patterned human umbilical vein endothelial cells (HUVECs) in a liver lobule-like manner to enhance their hepatic differentiation efficiency. It is found that small-sized hASC spheroids, with a diameter of ≈50 µm, show superior pro-angiogenic effects and hepatic differentiation compared to the other counterparts. The size-dependent functional enhancements are mediated by the Wnt signaling pathway. Meanwhile, co-culture of hASCs with HUVECs, at a HUVECs/hASCs seeding density ratio of 2:1, distinctly promotes hepatic differentiation and vascularization both in vitro and in vivo, especially when endothelial cells are patterned into hollow hexagons. After subcutaneous implantation, the mini-liver, consisting of HLC spheroids and 3D-printed interconnected vasculatures, can effectively improve liver regeneration in two ALF animal models through amelioration of local oxidative stress and inflammation, reduction of liver necrosis, as well as increase of cell proliferation, thereby showing great promise for clinical translation.
Collapse
Affiliation(s)
- Jiabin Zhang
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| | - Xiaodie Chen
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhou510630China
| | - Yurong Chai
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhou510630China
| | - Chenya Zhuo
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| | - Tiantian Xue
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhou510630China
| | - Dan Shao
- Institute of Life SciencesSchool of MedicineSouth China University of TechnologyGuangzhou510006China
| | - Yu Tao
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| |
Collapse
|
4
|
Lagasse E, Levin M. Future medicine: from molecular pathways to the collective intelligence of the body. Trends Mol Med 2023; 29:687-710. [PMID: 37481382 PMCID: PMC10527237 DOI: 10.1016/j.molmed.2023.06.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/24/2023]
Abstract
The remarkable anatomical homeostasis exhibited by complex living organisms suggests that they are inherently reprogrammable information-processing systems that offer numerous interfaces to their physiological and anatomical problem-solving capacities. We briefly review data suggesting that the multiscale competency of living forms affords a new path for biomedicine that exploits the innate collective intelligence of tissues and organs. The concept of tissue-level allostatic goal-directedness is already bearing fruit in clinical practice. We sketch a roadmap towards 'somatic psychiatry' by using advances in bioelectricity and behavioral neuroscience to design methods that induce self-repair of structure and function. Relaxing the assumption that cellular control mechanisms are static, exploiting powerful concepts from cybernetics, behavioral science, and developmental biology may spark definitive solutions to current biomedical challenges.
Collapse
Affiliation(s)
- Eric Lagasse
- McGowan Institute for Regenerative Medicine and Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
5
|
Lee SML, Kern A, Jauch KW, Thasler R, Niess H, Thasler WE. Cold Preservation of Human Hepatocytes with High Viability. Biopreserv Biobank 2023; 21:367-377. [PMID: 36355346 DOI: 10.1089/bio.2021.0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Freshly isolated human hepatocytes are an important model for translational research, validation of experiments done in animals, and preclinical studies. Human hepatocyte isolation often cannot be carried out easily on demand in common research laboratories, and researchers often collaborate to share hepatocytes or outsource hepatocyte isolations. As a prerequisite for such a strategy, hepatocytes have to maintain their phenotypes after transport. Therefore, this study aimed to determine if overnight storage or shipment of hepatocytes affects their quality when viability, adherence, and cytochrome P450 (CYP) activities are considered. Hepatocytes were stored overnight or shipped to a collaborator in a cold storage solution on wet ice. On the next day, viability of hepatocytes was assessed before plating the cells to determine adherence. Hepatocytes were also cultured in a sandwich culture to determine CYP activities and inducibility. The results showed that although viability (79% ± 0.7% on isolation) was significantly decreased by overnight storage or shipment by 11% (p < 0.001) or 15% (p < 0.001), respectively, the viability of hepatocytes the next day at above 64% ± 2.2% remained sufficiently high for further experiments. In addition, hepatocytes stored for 18 or 24 hours were adherent the next day, and a high confluence of 81% ± 10% to 91% ± 4% was achieved after 48 hours in culture when hepatocytes were adhered on collagen-coated plates. Furthermore, CYP enzyme activities were inducible and not affected by variables such as fibrosis, age, type of operation, steatosis, and body mass index. However, our data would suggest that the type of cancer (primary/secondary), sex (male/female), hypertension, glutamic oxaloacetic transaminase activity, partial thromboplastin time, and size of perfused liver had significant effects (p < 0.05) on induction of some CYP enzymes. In conclusion, human hepatocyte isolation can be carried out at a centralized site and shared between multiple researchers, increasing flexibility and access to a representative human liver in vitro model.
Collapse
Affiliation(s)
- Serene M L Lee
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
- HTCR-Services GmbH, Munich, Germany
| | - Armin Kern
- Drug Metabolism and Pharmacokinetics, Research and Development, Bayer AG, Wuppertal, Germany
| | - Karl-Walter Jauch
- Medical Directorate, University Hospital, LMU Munich, Munich, Germany
- Human Tissue and Cell Research Foundation, Regensburg, Germany
| | | | - Hanno Niess
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang E Thasler
- Human Tissue and Cell Research Foundation, Regensburg, Germany
- Department of General, Visceral and Minimally Invasive Surgery, Red Cross Hospital Munich, Munich, Germany
| |
Collapse
|
6
|
Hu XH, Chen L, Wu H, Tang YB, Zheng QM, Wei XY, Wei Q, Huang Q, Chen J, Xu X. Cell therapy in end-stage liver disease: replace and remodel. Stem Cell Res Ther 2023; 14:141. [PMID: 37231461 DOI: 10.1186/s13287-023-03370-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Liver disease is prevalent worldwide. When it reaches the end stage, mortality rises to 50% or more. Although liver transplantation has emerged as the most efficient treatment for end-stage liver disease, its application has been limited by the scarcity of donor livers. The lack of acceptable donor organs implies that patients are at high risk while waiting for suitable livers. In this scenario, cell therapy has emerged as a promising treatment approach. Most of the time, transplanted cells can replace host hepatocytes and remodel the hepatic microenvironment. For instance, hepatocytes derived from donor livers or stem cells colonize and proliferate in the liver, can replace host hepatocytes, and restore liver function. Other cellular therapy candidates, such as macrophages and mesenchymal stem cells, can remodel the hepatic microenvironment, thereby repairing the damaged liver. In recent years, cell therapy has transitioned from animal research to early human studies. In this review, we will discuss cell therapy in end-stage liver disease treatment, especially focusing on various cell types utilized for cell transplantation, and elucidate the processes involved. Furthermore, we will also summarize the practical obstacles of cell therapy and offer potential solutions.
Collapse
Affiliation(s)
- Xin-Hao Hu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lan Chen
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hao Wu
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yang-Bo Tang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Qiu-Min Zheng
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Xu-Yong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qiang Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qi Huang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jian Chen
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Xiao Xu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
7
|
Wang Y, Zheng Q, Sun Z, Wang C, Cen J, Zhang X, Jin Y, Wu B, Yan T, Wang Z, Gu Q, Lv X, Nan J, Wu Z, Sun W, Pan G, Zhang L, Hui L, Cai X. Reversal of liver failure using a bioartificial liver device implanted with clinical-grade human-induced hepatocytes. Cell Stem Cell 2023; 30:617-631.e8. [PMID: 37059100 DOI: 10.1016/j.stem.2023.03.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/18/2023] [Accepted: 03/15/2023] [Indexed: 04/16/2023]
Abstract
Liver resection is the first-line treatment for primary liver cancers, providing the potential for a cure. However, concerns about post-hepatectomy liver failure (PHLF), a leading cause of death following extended liver resection, have restricted the population of eligible patients. Here, we engineered a clinical-grade bioartificial liver (BAL) device employing human-induced hepatocytes (hiHeps) manufactured under GMP conditions. In a porcine PHLF model, the hiHep-BAL treatment showed a remarkable survival benefit. On top of the supportive function, hiHep-BAL treatment restored functions, specifically ammonia detoxification, of the remnant liver and facilitated liver regeneration. Notably, an investigator-initiated study in seven patients with extended liver resection demonstrated that hiHep-BAL treatment was well tolerated and associated with improved liver function and liver regeneration, meeting the primary outcome of safety and feasibility. These encouraging results warrant further testing of hiHep-BAL for PHLF, the success of which would broaden the population of patients eligible for liver resection.
Collapse
Affiliation(s)
- Yifan Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou 310016, China; Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou 310016, China
| | - Qiang Zheng
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Zhen Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chenhua Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Science, Shanghai 200031, China
| | - Jin Cen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Science, Shanghai 200031, China
| | - Xinjie Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yan Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Baihua Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Science, Shanghai 200031, China
| | - Tingting Yan
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Ziyuan Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Qiuxia Gu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xingyu Lv
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Junjie Nan
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Zhongyu Wu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Wenbin Sun
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ludi Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Science, Shanghai 200031, China.
| | - Lijian Hui
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Science, Shanghai 200031, China.
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou 310016, China; Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou 310016, China.
| |
Collapse
|
8
|
Highly efficient fabrication of functional hepatocyte spheroids by a magnetic system for the rescue of acute liver failure. Biomaterials 2023; 294:122014. [PMID: 36709644 DOI: 10.1016/j.biomaterials.2023.122014] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/21/2023]
Abstract
Engineering hepatocytes as multicellular cell spheroids can improve their viability after implantation in vivo for effective rescue of the devastating acute liver failure (ALF). However, there is still a lack of straightforward methods for efficient generation of functional hepatocyte spheroids. In this study, a magnetic system, consisting of magnetic microwell arrays and magnet blocks, is developed to realize magnetically controlled 3D cell capture and spatial confinement-mediated cell aggregation. The cell spheroids with smaller size show superior hepatic functions than the larger-sized counterparts. Notably, the intrinsic magnetism of magnetic microwell arrays can regulate superoxide anions in hepatocyte spheroids and herein promote various biological processes, including antioxidation, hepatocyte-related functions, and pro-angiogenic potential. Ectopic implantation of the functional cell spheroids in ALF-challenged mice significantly prolongs the animal survival, ameliorates inflammation, and promotes liver regeneration. Hence, application of the magnetic system for generation of functionally enhanced hepatocyte spheroids holds great potential for clinical translation in the future.
Collapse
|
9
|
Kupffer Cells as a Target for Immunotherapy. J 2022. [DOI: 10.3390/j5040036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Kupffer cells (KCs) are resident macrophages in the liver. Recent studies have revealed that KCs are closely related to inflammatory liver diseases, including nonalcoholic liver diseases (NAFLD). From this point of view, KC transplantation can be a candidate for immunotherapy against inflammatory diseases. Similar to general macrophages, KCs show several different phenotypes according to their environment. Activated KCs are involved in either proinflammatory responses or anti-inflammatory responses. Thus, to manipulate KCs for immunotherapy, it is crucial to control the direction of KC activation. Here, we summarize the outlook and the issues hindering immunotherapy using KC transplantation.
Collapse
|
10
|
Cellular Therapies in Pediatric Liver Diseases. Cells 2022; 11:cells11162483. [PMID: 36010561 PMCID: PMC9406752 DOI: 10.3390/cells11162483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/30/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Liver transplantation is the gold standard for the treatment of pediatric end-stage liver disease and liver based metabolic disorders. Although liver transplant is successful, its wider application is limited by shortage of donor organs, surgical complications, need for life long immunosuppressive medication and its associated complications. Cellular therapies such as hepatocytes and mesenchymal stromal cells (MSCs) are currently emerging as an attractive alternative to liver transplantation. The aim of this review is to present the existing world experience in hepatocyte and MSC transplantation and the potential for future effective applications of these modalities of treatment.
Collapse
|
11
|
Han B, Francipane MG, Cheikhi A, Johnson J, Chen F, Chen R, Lagasse E. Fat-associated lymphoid clusters as expandable niches for ectopic liver development. Hepatology 2022; 76:357-371. [PMID: 34890068 PMCID: PMC9546108 DOI: 10.1002/hep.32277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/01/2021] [Accepted: 12/04/2021] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Hepatocyte transplantation holds great promise as an alternative approach to whole-organ transplantation. Intraportal and intrasplenic cell infusions are primary hepatocyte transplantation delivery routes for this procedure. However, patients with severe liver diseases often have disrupted liver and spleen architectures, which introduce risks in the engraftment process. We previously demonstrated i.p. injection of hepatocytes as an alternative route of delivery that could benefit this subpopulation of patients, particularly if less invasive and low-risk procedures are required; and we have established that lymph nodes may serve as extrahepatic sites for hepatocyte engraftment. However, whether other niches in the abdominal cavity support the survival and proliferation of the transplanted hepatocytes remains unclear. APPROACH AND RESULTS Here, we showed that hepatocytes transplanted by i.p. injection engraft and generate ectopic liver tissues in fat-associated lymphoid clusters (FALCs), which are adipose tissue-embedded, tertiary lymphoid structures localized throughout the peritoneal cavity. The FALC-engrafted hepatocytes formed functional ectopic livers that rescued tyrosinemic mice from liver failure. Consistently, analyses of ectopic and native liver transcriptomes revealed a selective ectopic compensatory gene expression of hepatic function-controlling genes in ectopic livers, implying a regulated functional integration between the two livers. The lack of FALCs in the abdominal cavity of immunodeficient tyrosinemic mice hindered ectopic liver development, whereas the restoration of FALC formation through bone marrow transplantation restored ectopic liver development in these mice. Accordingly, induced abdominal inflammation increased FALC numbers, which improved hepatocyte engraftment and accelerated the recovery of tyrosinemic mice from liver failure. CONCLUSIONS Abdominal FALCs are essential extrahepatic sites for hepatocyte engraftment after i.p. transplantation and, as such, represent an easy-to-access and expandable niche for ectopic liver regeneration when adequate growth stimulus is present.
Collapse
Affiliation(s)
- Bing Han
- McGowan Institute for Regenerative Medicine and Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Maria Giovanna Francipane
- McGowan Institute for Regenerative Medicine and Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA,Ri.MED FoundationPalermoItaly
| | - Amin Cheikhi
- McGowan Institute for Regenerative Medicine and Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Joycelyn Johnson
- McGowan Institute for Regenerative Medicine and Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Fei Chen
- McGowan Institute for Regenerative Medicine and Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA,Department of Histology and EmbryologySchool of Basic SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Ruoyu Chen
- Computer SchoolBeijing Information Science and Technology UniversityBeijingChina
| | - Eric Lagasse
- McGowan Institute for Regenerative Medicine and Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
12
|
Li X, Wang Y, Yang H, Dai Y. Liver and Hepatocyte Transplantation: What Can Pigs Contribute? Front Immunol 2022; 12:802692. [PMID: 35095885 PMCID: PMC8795512 DOI: 10.3389/fimmu.2021.802692] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/30/2021] [Indexed: 12/25/2022] Open
Abstract
About one-fifth of the population suffers from liver diseases in China, meaning that liver disorders are prominent causative factors relating to the Chinese mortality rate. For patients with end-stage liver diseases such as hepatocellular carcinoma or acute liver diseases with life-threatening liver dysfunction, allogeneic liver transplantation is the only life-saving treatment. Hepatocyte transplantation is a promising alternative for patients with acute liver failure or those considered high risk for major surgery, particularly for the bridge-to-transplant period. However, the lack of donors has become a serious global problem. The clinical application of porcine xenogeneic livers and hepatocytes remains a potential solution to alleviate the donor shortage. Pig grafts of xenotransplantation play roles in providing liver support in recipients, together with the occurrence of rejection, thrombocytopenia, and blood coagulation dysfunction. In this review, we present an overview of the development, potential therapeutic impact, and remaining barriers in the clinical application of pig liver and hepatocyte xenotransplantation to humans and non-human primates. Donor pigs with optimized genetic modification combinations and highly effective immunosuppressive regimens should be further explored to improve the outcomes of xenogeneic liver and hepatocyte transplantation.
Collapse
Affiliation(s)
- Xiaoxue Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Ying Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Haiyuan Yang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yifan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
13
|
Lin WL, Mizobuchi M, Kawahigashi M, Nakahashi O, Maekawa Y, Sakai T. Functional kupffer cells migrate to the liver from the intraperitoneal cavity. Biochem Biophys Rep 2021; 27:101103. [PMID: 34458593 PMCID: PMC8379421 DOI: 10.1016/j.bbrep.2021.101103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/28/2021] [Accepted: 08/11/2021] [Indexed: 11/23/2022] Open
Abstract
We established a method of KC transplantation by intraperitoneal (i.p.) injection using EGFP-expressing cells (EGFP-KCs) and normal KCs. The novel method is easier and less invasive than conventional methods so that it is not only technically advantageous but also ethically preferable for experiments using animals. We demonstrated that KCs migrated to the liver following i.p. Injection. Engraftment in the liver was not observed for peritoneal macrophages (pMPs). This suggests that KCs migrate to the liver via a sorting mechanism. KC injection decreased the KC number at 24 h and then recovered the KCs at 10 days to a normal level. Additionally, recovery to the normal level by KC injection was observed in mice with KC depletion induced by GdCl3. These results suggest that a regulatory mechanism exists for controlling the number of KCs.
Collapse
Affiliation(s)
- Wen-Ling Lin
- Institute for Health Sciences, Tokushima Bunri University, 180 Nishihama-bouji, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Mizuki Mizobuchi
- Institute for Health Sciences, Tokushima Bunri University, 180 Nishihama-bouji, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Mina Kawahigashi
- Institute for Health Sciences, Tokushima Bunri University, 180 Nishihama-bouji, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Otoki Nakahashi
- Institute for Health Sciences, Tokushima Bunri University, 180 Nishihama-bouji, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Yuuki Maekawa
- Institute for Health Sciences, Tokushima Bunri University, 180 Nishihama-bouji, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Takashi Sakai
- Institute for Health Sciences, Tokushima Bunri University, 180 Nishihama-bouji, Yamashiro-cho, Tokushima, 770-8514, Japan
| |
Collapse
|
14
|
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: 83] [Impact Index Per Article: 20.8] [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
|
15
|
Fontes P, Komori J, Lopez R, Marsh W, Lagasse E. Development of Ectopic Livers by Hepatocyte Transplantation Into Swine Lymph Nodes. Liver Transpl 2020; 26:1629-1643. [PMID: 32810371 PMCID: PMC7756213 DOI: 10.1002/lt.25872] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/01/2020] [Accepted: 08/09/2020] [Indexed: 12/16/2022]
Abstract
Orthotopic liver transplantation continues to be the only effective therapy for patients with end-stage liver disease. Unfortunately, many of these patients are not considered transplant candidates, lacking effective therapeutic options that would address both the irreversible progression of their hepatic failure and the control of their portal hypertension. In this prospective study, a swine model was exploited to induce subacute liver failure. Autologous hepatocytes, isolated from the left hepatic lobe, were transplanted into the mesenteric lymph nodes (LNs) by direct cell injection. At 30-60 days after transplantation, hepatocyte engraftment in LNs was successfully identified in all transplanted animals with the degree of ectopic liver mass detected being proportional to the induced native liver injury. These ectopic livers developed within the LNs showed remarkable histologic features of swine hepatic lobules, including the formation of sinusoids and bile ducts. On the basis of our previous tyrosinemic mouse model and the present pig models of induced subacute liver failure, the generation of auxiliary liver tissue using the LNs as hepatocyte engraftment sites represents a potential therapeutic approach to supplement declining hepatic function in the treatment of liver disease.
Collapse
Affiliation(s)
- Paulo Fontes
- WVU MedicineDepartment of SurgerySchool of MedicineWest Virginia UniversityMorgantownWV,LyGenesis, Inc.PittsburghPA
| | - Junji Komori
- McGowan Institute for Regenerative MedicineDepartment of PathologySchool of MedicineUniversity of PittsburghPittsburghPA,Department of SurgeryTakamatsu Red Cross HospitalKagawaJapan
| | - Roberto Lopez
- WVU MedicineDepartment of SurgerySchool of MedicineWest Virginia UniversityMorgantownWV,LyGenesis, Inc.PittsburghPA
| | - Wallis Marsh
- WVU MedicineDepartment of SurgerySchool of MedicineWest Virginia UniversityMorgantownWV
| | - Eric Lagasse
- LyGenesis, Inc.PittsburghPA,McGowan Institute for Regenerative MedicineDepartment of PathologySchool of MedicineUniversity of PittsburghPittsburghPA
| |
Collapse
|
16
|
Nicolas CT, Kaiser RA, Hickey RD, Allen KL, Du Z, VanLith CJ, Guthman RM, Amiot B, Suksanpaisan L, Han B, Francipane MG, Cheikhi A, Jiang H, Bansal A, Pandey MK, Garg I, Lowe V, Bhagwate A, O’Brien D, Kocher JPA, DeGrado TR, Nyberg SL, Lagasse E, Lillegard JB. Ex Vivo Cell Therapy by Ectopic Hepatocyte Transplantation Treats the Porcine Tyrosinemia Model of Acute Liver Failure. Mol Ther Methods Clin Dev 2020; 18:738-750. [PMID: 32913881 PMCID: PMC7452193 DOI: 10.1016/j.omtm.2020.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/07/2020] [Indexed: 11/19/2022]
Abstract
The effectiveness of cell-based therapies to treat liver failure is often limited by the diseased liver environment. Here, we provide preclinical proof of concept for hepatocyte transplantation into lymph nodes as a cure for liver failure in a large-animal model with hereditary tyrosinemia type 1 (HT1), a metabolic liver disease caused by deficiency of fumarylacetoacetate hydrolase (FAH) enzyme. Autologous porcine hepatocytes were transduced ex vivo with a lentiviral vector carrying the pig Fah gene and transplanted into mesenteric lymph nodes. Hepatocytes showed early (6 h) and durable (8 months) engraftment in lymph nodes, with reproduction of vascular and hepatic microarchitecture. Subsequently, hepatocytes migrated to and repopulated the native diseased liver. The corrected cells generated sufficient liver mass to clinically ameliorate the acute liver failure and HT1 disease as early as 97 days post-transplantation. Integration site analysis defined the corrected hepatocytes in the liver as a subpopulation of hepatocytes from lymph nodes, indicating that the lymph nodes served as a source for healthy hepatocytes to repopulate a diseased liver. Therefore, ectopic transplantation of healthy hepatocytes cures this pig model of liver failure and presents a promising approach for the development of cures for liver disease in patients.
Collapse
Affiliation(s)
- Clara T. Nicolas
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Faculty of Medicine, University of Barcelona, Barcelona, Spain
- Department of Surgery, University of Alabama Birmingham, Birmingham, AL, USA
| | - Robert A. Kaiser
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Children’s Hospitals and Clinics of Minnesota, Midwest Fetal Care Center, Minneapolis, MN, USA
| | | | - Kari L. Allen
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Zeji Du
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Rebekah M. Guthman
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Medical College of Wisconsin, Wausau, WI, USA
| | - Bruce Amiot
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Bing Han
- McGowan Institute for Regenerative Medicine and Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maria Giovanna Francipane
- McGowan Institute for Regenerative Medicine and Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- Ri.MED Foundation, Palermo, Italy
| | - Amin Cheikhi
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huailei Jiang
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Aditya Bansal
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Ishan Garg
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Val Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Aditya Bhagwate
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Daniel O’Brien
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Jean-Pierre A. Kocher
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | - Scott L. Nyberg
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Eric Lagasse
- McGowan Institute for Regenerative Medicine and Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joseph B. Lillegard
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Children’s Hospitals and Clinics of Minnesota, Midwest Fetal Care Center, Minneapolis, MN, USA
- Pediatric Surgical Associates, Minneapolis, MN, USA
| |
Collapse
|
17
|
Furuta T, Furuya K, Zheng YW, Oda T. Novel alternative transplantation therapy for orthotopic liver transplantation in liver failure: A systematic review. World J Transplant 2020; 10:64-78. [PMID: 32257850 PMCID: PMC7109592 DOI: 10.5500/wjt.v10.i3.64] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/10/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Orthotopic liver transplantation (OLT) is the only treatment for end-stage liver failure; however, graft shortage impedes its applicability. Therefore, studies investigating alternative therapies are plenty. Nevertheless, no study has comprehensively analyzed these therapies from different perspectives. AIM To summarize the current status of alternative transplantation therapies for OLT and to support future research. METHODS A systematic literature search was performed using PubMed, Cochrane Library and EMBASE for articles published between January 2010 and 2018, using the following MeSH terms: [(liver transplantation) AND cell] OR [(liver transplantation) AND differentiation] OR [(liver transplantation) AND organoid] OR [(liver transplantation) AND xenotransplantation]. Various types of studies describing therapies to replace OLT were retrieved for full-text evaluation. Among them, we selected articles including in vivo transplantation. RESULTS A total of 89 studies were selected. There are three principle forms of treatment for liver failure: Xeno-organ transplantation, scaffold-based transplantation, and cell transplantation. Xeno-organ transplantation was covered in 14 articles, scaffold-based transplantation was discussed in 22 articles, and cell transplantation was discussed in 53 articles. Various types of alternative therapies were discussed: Organ liver, 25 articles; adult hepatocytes, 31 articles; fetal hepatocytes, three articles; mesenchymal stem cells (MSCs), 25 articles; embryonic stem cells, one article; and induced pluripotent stem cells, three articles and other sources. Clinical applications were discussed in 12 studies: Cell transplantation using hepatocytes in four studies, five studies using umbilical cord-derived MSCs, three studies using bone marrow-derived MSCs, and two studies using hematopoietic stem cells. CONCLUSION The clinical applications are present only for cell transplantation. Scaffold-based transplantation is a comprehensive treatment combining organ and cell transplantations, which warrants future research to find relevant clinical applications.
Collapse
Affiliation(s)
- Tomoaki Furuta
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba-shi 305-8575, Ibaraki, Japan
| | - Kinji Furuya
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba-shi 305-8575, Ibaraki, Japan
| | - Yun-Wen Zheng
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba-shi 305-8575, Ibaraki, Japan
- Institute of Regenerative Medicine and Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
- Department of Regenerative Medicine, School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
- Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba-shi 305-8575, Ibaraki, Japan
| |
Collapse
|
18
|
Stevens KR, Scull MA, Ramanan V, Fortin CL, Chaturvedi RR, Knouse KA, Xiao JW, Fung C, Mirabella T, Chen AX, McCue MG, Yang MT, Fleming HE, Chung K, de Jong YP, Chen CS, Rice CM, Bhatia SN. In situ expansion of engineered human liver tissue in a mouse model of chronic liver disease. Sci Transl Med 2018; 9:9/399/eaah5505. [PMID: 28724577 DOI: 10.1126/scitranslmed.aah5505] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 07/12/2016] [Accepted: 03/06/2017] [Indexed: 12/29/2022]
Abstract
Control of both tissue architecture and scale is a fundamental translational roadblock in tissue engineering. An experimental framework that enables investigation into how architecture and scaling may be coupled is needed. We fabricated a structurally organized engineered tissue unit that expanded in response to regenerative cues after implantation into mice with liver injury. Specifically, we found that tissues containing patterned human primary hepatocytes, endothelial cells, and stromal cells in a degradable hydrogel expanded more than 50-fold over the course of 11 weeks in mice with injured livers. There was a concomitant increase in graft function as indicated by the production of multiple human liver proteins. Histologically, we observed the emergence of characteristic liver stereotypical microstructures mediated by coordinated growth of hepatocytes in close juxtaposition with a perfused vasculature. We demonstrated the utility of this system for probing the impact of multicellular geometric architecture on tissue expansion in response to liver injury. This approach is a hybrid strategy that harnesses both biology and engineering to more efficiently deploy a limited cell mass after implantation.
Collapse
Affiliation(s)
- Kelly R Stevens
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Departments of Bioengineering and Pathology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Margaret A Scull
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA
| | - Vyas Ramanan
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Chelsea L Fortin
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Departments of Bioengineering and Pathology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Ritika R Chaturvedi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristin A Knouse
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Harvard Medical School, Boston, MA 02115, USA
| | - Jing W Xiao
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA
| | - Canny Fung
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA
| | | | - Amanda X Chen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Margaret G McCue
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | | | - Heather E Fleming
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Kwanghun Chung
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Ype P de Jong
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA.,Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Christopher S Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Bioengineering, Boston University, Boston, MA 02215, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA. .,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
19
|
Hickey RD, Mao SA, Glorioso J, Elgilani F, Amiot B, Chen H, Rinaldo P, Marler R, Jiang H, DeGrado TR, Suksanpaisan L, O'Connor MK, Freeman BL, Ibrahim SH, Peng KW, Harding CO, Ho CS, Grompe M, Ikeda Y, Lillegard JB, Russell SJ, Nyberg SL. Curative ex vivo liver-directed gene therapy in a pig model of hereditary tyrosinemia type 1. Sci Transl Med 2017; 8:349ra99. [PMID: 27464750 DOI: 10.1126/scitranslmed.aaf3838] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 07/05/2016] [Indexed: 12/23/2022]
Abstract
We tested the hypothesis that ex vivo hepatocyte gene therapy can correct the metabolic disorder in fumarylacetoacetate hydrolase-deficient (Fah(-/-)) pigs, a large animal model of hereditary tyrosinemia type 1 (HT1). Recipient Fah(-/-) pigs underwent partial liver resection and hepatocyte isolation by collagenase digestion. Hepatocytes were transduced with one or both of the lentiviral vectors expressing the therapeutic Fah and the reporter sodium-iodide symporter (Nis) genes under control of the thyroxine-binding globulin promoter. Pigs received autologous transplants of hepatocytes by portal vein infusion. After transplantation, the protective drug 2-(2-nitro-4-trifluoromethylbenzyol)-1,3 cyclohexanedione (NTBC) was withheld from recipient pigs to provide a selective advantage for expansion of corrected FAH(+) cells. Proliferation of transplanted cells, assessed by both immunohistochemistry and noninvasive positron emission tomography imaging of NIS-labeled cells, demonstrated near-complete liver repopulation by gene-corrected cells. Tyrosine and succinylacetone levels improved to within normal range, demonstrating complete correction of tyrosine metabolism. In addition, repopulation of the Fah(-/-) liver with transplanted cells inhibited the onset of severe fibrosis, a characteristic of nontransplanted Fah(-/-) pigs. This study demonstrates correction of disease in a pig model of metabolic liver disease by ex vivo gene therapy. To date, ex vivo gene therapy has only been successful in small animal models. We conclude that further exploration of ex vivo hepatocyte genetic correction is warranted for clinical use.
Collapse
Affiliation(s)
- Raymond D Hickey
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA. Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | - Shennen A Mao
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Jaime Glorioso
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Faysal Elgilani
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Bruce Amiot
- Brami Biomedical Inc., Coon Rapids, MN 55433, USA
| | - Harvey Chen
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Piero Rinaldo
- Division of Laboratory Genetics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ronald Marler
- Department of Comparative Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Huailei Jiang
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Lukkana Suksanpaisan
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA. Imanis Life Sciences, Rochester, MN 55902, USA
| | | | - Brittany L Freeman
- Division of Pediatric Gastroenterology, Mayo Clinic, Rochester, MN 55905, USA
| | - Samar H Ibrahim
- Division of Pediatric Gastroenterology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kah Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Cary O Harding
- Department of Molecular and Medical Genetics and Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Chak-Sum Ho
- Histocompatibility Laboratory, Gift of Life Michigan, Ann Arbor, MI 48108, USA
| | - Markus Grompe
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Yasuhiro Ikeda
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Joseph B Lillegard
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA. Midwest Fetal Care Center, Children's Hospitals and Clinics of Minnesota, Minneapolis, MN 55404, USA
| | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Scott L Nyberg
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
20
|
Emerging advancements in liver regeneration and organogenesis as tools for liver replacement. Curr Opin Organ Transplant 2017; 21:581-587. [PMID: 27755169 DOI: 10.1097/mot.0000000000000365] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Although the liver possesses a unique, innate ability to regenerate through mass compensation, transplantation remains the only therapy when damage outpaces regeneration, or liver metabolic capacity is irreversibly impacted. Recent insight from developmental biology has greatly influenced the advancement of alternative options to transplantation in these settings. RECENT FINDINGS Factors known to direct liver cell specification, expansion, and differentiation have been used to generate hepatocyte-like cells from stem and somatic cells for developing cell therapies. Additionally, interactions between hepatic epithelial and nonepithelial cells key to establishing hepatic architecture have been used in tissue engineering approaches to advance self-organizing hepatic organoids and bioartificial liver devices. Simultaneously, recent clinically applicable advances in human hepatocyte transplantation and promotion of innate hepatic regeneration have been limited. SUMMARY Although mature hepatocytes have the potential to bridge to, or replace whole organ transplantation, limits in the ability to obtain healthy cells, stabilize in-vitro expansion, cryopreserve, and alleviate rejection, still exist. Alternative sources for generating hepatocytes hold promise for cell therapy and tissue engineering. These may allow generation of autologous or universal donor cells that eliminate the need for immunosuppression; however, limits exist regarding hepatocyte maturity and efficacy at liver repopulation, as well as applicability to human chronic liver disease.
Collapse
|
21
|
Iwase H, Liu H, Schmelzer E, Ezzelarab M, Wijkstrom M, Hara H, Lee W, Singh J, Long C, Lagasse E, Gerlach JC, Cooper DKC, Gridelli B. Transplantation of hepatocytes from genetically engineered pigs into baboons. Xenotransplantation 2017; 24. [PMID: 28130881 DOI: 10.1111/xen.12289] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/23/2016] [Accepted: 12/27/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Some patients with acute or acute-on-chronic hepatic failure die before a suitable human liver allograft becomes available. Encouraging results have been achieved in such patients by the transplantation of human hepatocyte progenitor cells from fetal liver tissue. The aim of the study was to explore survival of hepatocytes from genetically engineered pigs after direct injection into the spleen and other selected sites in immunosuppressed baboons to monitor the immune response and the metabolic function and survival of the transplanted hepatocytes. METHODS Baboons (n=3) were recipients of GTKO/hCD46 pig hepatocytes. All three baboons received anti-thymocyte globulin (ATG) induction and tapering methylprednisolone. Baboon 1 received maintenance immunosuppressive therapy with tacrolimus and rapamycin. Baboons 2 and 3 received an anti-CD40mAb/rapamycin-based regimen that prevents sensitization to pig solid organ grafts. The baboons were euthanized 4 or 5 weeks after hepatocyte transplantation. The baboon immune response was monitored by the measurement of anti-non-Gal IgM and IgG antibodies (by flow cytometry) and CFSE-mixed lymphocyte reaction. Monitoring for hepatocyte survival and function was by (i) real-time PCR detection of porcine DNA, (ii) real-time PCR for porcine gene expression, and (iii) pig serum albumin levels (by ELISA). The sites of hepatocyte injection were examined microscopically. RESULTS Detection of porcine DNA and porcine gene expression was minimal at all sites of hepatocyte injection. Serum levels of porcine albumen were very low-500-1000-fold lower than in baboons with orthotopic pig liver grafts, and approximately 5000-fold lower than in healthy pigs. No hepatocytes or infiltrating immune cells were seen at any of the injection sites. Two baboons (Baboons 1 and 3) demonstrated a significant increase in anti-pig IgM and an even greater increase in IgG, indicating sensitization to pig antigens. DISCUSSION AND CONCLUSIONS As a result of this disappointing experience, the following points need to be considered. (i) Were the isolated pig hepatocytes functionally viable? (ii) Are pig hepatocytes more immunogenic than pig hearts, kidneys, artery patch grafts, or islets? (iii) Does injection of pig cells (antigens) into the spleen and/or lymph nodes stimulate a greater immune response than when pig tissues are grafted at other sites? (iv) Did the presence of the recipient's intact liver prevent survival and proliferation of pig hepatocytes? (v) Is pig CD47-primate SIRP-α compatibility essential? In conclusion, the transplantation of genetically engineered pig hepatocytes into multiple sites in immunosuppressed baboons was associated with very early graft failure. Considerable further study is required before clinical trials should be undertaken.
Collapse
Affiliation(s)
- Hayato Iwase
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hong Liu
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA.,Department of General Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Eva Schmelzer
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohamed Ezzelarab
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Martin Wijkstrom
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hidetaka Hara
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Whayoung Lee
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jagjit Singh
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cassandra Long
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric Lagasse
- Department of Pathology, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jörg C Gerlach
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - David K C Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruno Gridelli
- Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| |
Collapse
|
22
|
Abstract
The ultimate treatment for end-stage renal disease (ESRD) is orthotopic transplantation. However, the demand for kidney transplantation far exceeds the number of available donor organs. While more than 100,000 Americans need a kidney, only 17,000 people receive a kidney transplant each year (National Kidney Foundation's estimations). In recent years, several regenerative medicine/tissue engineering approaches have been exploited to alleviate the kidney shortage crisis. Although these approaches have yielded promising results in experimental animal models, the kidney is a complex organ and translation into the clinical realm has been challenging to date. In this review, we will discuss cell therapy-based approaches for kidney regeneration and whole-kidney tissue engineering strategies, including our innovative approach to regenerate a functional kidney using the lymph node as an in vivo bioreactor.
Collapse
|
23
|
Pahk KJ, Mohammad GH, Malago M, Saffari N, Dhar DK. A Novel Approach to Ultrasound-Mediated Tissue Decellularization and Intra-Hepatic Cell Delivery in Rats. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:1958-1967. [PMID: 27184248 DOI: 10.1016/j.ultrasmedbio.2016.03.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 03/10/2016] [Accepted: 03/20/2016] [Indexed: 06/05/2023]
Abstract
Liver transplantation is the mainstay of treatment for end stage liver diseases, including metabolic and congenital liver diseases. The number of suitable donor organs is, however, limited, and a whole-liver transplant requires complex surgery. Cell therapy, such as intra-portal hepatocytes transplantation, has been considered as a bridging therapy to liver transplantation but has shown a mixed clinical outcome with limited success, including low level of engraftment of transplanted hepatocytes. Here, we report a novel cell delivery technique in a rat model by creating a cavity inside the liver parenchyma by non-invasive high intensity focused ultrasound histotripsy. Our in vivo experimental results together with histologic observations show that direct injection of cells inside the cavity can facilitate successful uptake, proliferation and integration of the transplanted hepatocytes in the recipient liver. We were able to restore the plasma albumin level to 50% of the normal level in Nagase analbuminemic rats (serum albumin level of the Nagase rats was initially nil) by cell therapy after high intensity focused ultrasound-mediated histotripsy. We believe that this novel technique would enable the delivery of a large number of cells into the liver to restore liver function, particularly as a treatment for metabolic liver diseases. This novel method of intra-hepatic hepatocyte transplantation might be an invaluable tool for cell therapy in the future.
Collapse
Affiliation(s)
- Ki Joo Pahk
- Department of Mechanical Engineering, University College London, London, UK
| | - Goran Hamid Mohammad
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, UK
| | - Massimo Malago
- Hepato-pancreatic-biliary and Liver Transplantation Surgery, Royal Free Hospital, University College London, London, UK
| | - Nader Saffari
- Department of Mechanical Engineering, University College London, London, UK.
| | - Dipok Kumar Dhar
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, UK; King Faisal Specialist Hospital and Research Center, Comparative Medicine Department and Organ Transplantation Center, Riyadh, Saudi Arabia
| |
Collapse
|
24
|
Francipane MG, Lagasse E. Towards Organs on Demand: Breakthroughs and Challenges in Models of Organogenesis. CURRENT PATHOBIOLOGY REPORTS 2016; 4:77-85. [PMID: 28979828 DOI: 10.1007/s40139-016-0111-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In recent years, functional three-dimensional (3D) tissue generation in vitro has been significantly advanced by tissue-engineering methods, achieving better reproduction of complex native organs compared to conventional culture systems. This review will discuss traditional 3D cell culture techniques as well as newly developed technology platforms. These recent techniques provide new possibilities in the creation of human body parts and provide more accurate predictions of tissue response to drug and chemical challenges. Given the rapid advancement in the human induced pluripotent stem cell (iPSC) field, these platforms also hold great promise in the development of patient-specific, transplantable tissues and organs on demand.
Collapse
Affiliation(s)
- Maria Giovanna Francipane
- McGowan Institute for Regenerative Medicine, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- Ri.MED Foundation, 90133 Palermo, Italy
| | - Eric Lagasse
- McGowan Institute for Regenerative Medicine, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|
25
|
de l’Hortet AC, Takeishi K, Guzman-Lepe J, Handa K, Matsubara K, Fukumitsu K, Dorko K, Presnell SC, Yagi H, Soto-Gutierrez A. Liver-Regenerative Transplantation: Regrow and Reset. Am J Transplant 2016; 16:1688-96. [PMID: 26699680 PMCID: PMC4874858 DOI: 10.1111/ajt.13678] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/04/2015] [Accepted: 11/26/2015] [Indexed: 01/25/2023]
Abstract
Liver transplantation, either a partial liver from a living or deceased donor or a whole liver from a deceased donor, is the only curative therapy for severe end-stage liver disease. Only one-third of those on the liver transplant waiting list will be transplanted, and the demand for livers is projected to increase 23% in the next 20 years. Consequently, organ availability is an absolute constraint on the number of liver transplants that can be performed. Regenerative therapies aim to enhance liver tissue repair and regeneration by any means available (cell repopulation, tissue engineering, biomaterials, proteins, small molecules, and genes). Recent experimental work suggests that liver repopulation and engineered liver tissue are best suited to the task if an unlimited availability of functional induced pluripotent stem (iPS)-derived liver cells can be achieved. The derivation of iPS cells by reprogramming cell fate has opened up new lines of investigation, for instance, the generation of iPS-derived xenogeneic organs or the possibility of simply inducing the liver to reprogram its own hepatocyte function after injury. We reviewed current knowledge about liver repopulation, generation of engineered livers and reprogramming of liver function. We also discussed the numerous barriers that have to be overcome for clinical implementation.
Collapse
Affiliation(s)
| | - K. Takeishi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - J. Guzman-Lepe
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - K. Handa
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - K. Matsubara
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - K. Fukumitsu
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - K. Dorko
- Organovo Holdings Inc., San Diego, CA
| | | | - H. Yagi
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - A. Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA,Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA,Corresponding author: Alejandro Soto-Gutierrez,
| |
Collapse
|
26
|
Yuan J, Li W, Huang J, Guo X, Li X, Lu X, Huang X, Zhang H. Transplantation of human adipose stem cell-derived hepatocyte-like cells with restricted localization to liver using acellular amniotic membrane. Stem Cell Res Ther 2015; 6:217. [PMID: 26541667 PMCID: PMC4635993 DOI: 10.1186/s13287-015-0208-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 07/31/2015] [Accepted: 08/19/2015] [Indexed: 12/14/2022] Open
Abstract
Introduction Adult stem cell-derived hepatocytes transplantation holds considerable promise for future clinical individualized therapy of liver failure or dysfunction. However, the low engraftment of the available hepatocytes in the liver disease microenvironment has been a major obstacle. Methods Acellular human amniotic membrane was developed as a three-dimensional scaffold and combined with hepatocyte-like cells derived from human adipose stem cells to engineer a hepatic tissue graft that would allow hepatocyte engraftment in the liver effectively. Results The hepatic tissue grafts maintained hepatocyte-specific gene expression and functionality in vitro. When transplanted into the surgical incision in livers for engraftment, the engineered hepatic grafts significantly decreased the degree of liver injury caused by a carbon tetrachloride treatment and generated cords that were similar to the ductal plates in the liver between the acellular human amniotic membrane and the liver of receipts at day 3 post-transplantation. The hepatic tissue grafts maintained the expression of human hepatocyte-specific markers albumin, hepatocyte nuclear factor 4α, and cytochrome P450 2B6 in the liver of receipts, and acquired human-specific drug metabolism ability at eight weeks post-transplantation. Conclusions The acellular human amniotic membrane has the ability to maintain the functional phenotype of the hepatocyte-like cells derived from human adipose stem cells. Functional acellular human amniotic membrane-hepatocytes grafts integrated with the liver decreases the acute liver injury of mice. These engineered tissue constructs may support stem cell-based individualized therapy for liver disease and for bioartificial liver establishment. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0208-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Yuan
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10, Xitoutiao, You An Men, Beijing, 100069, China.
| | - Weihong Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10, Xitoutiao, You An Men, Beijing, 100069, China.
| | - Jieqiong Huang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10, Xitoutiao, You An Men, Beijing, 100069, China.
| | - Xinyue Guo
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10, Xitoutiao, You An Men, Beijing, 100069, China.
| | - Xueyang Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10, Xitoutiao, You An Men, Beijing, 100069, China.
| | - Xin Lu
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10, Xitoutiao, You An Men, Beijing, 100069, China.
| | - Xiaowu Huang
- Fu Xing Hospital, Capital Medical University, No. 20, Fu xing men wai, Beijing, 100038, China.
| | - Haiyan Zhang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10, Xitoutiao, You An Men, Beijing, 100069, China.
| |
Collapse
|
27
|
Francipane MG, Lagasse E. Pluripotent Stem Cells to Rebuild a Kidney: The Lymph Node as a Possible Developmental Niche. Cell Transplant 2015; 25:1007-23. [PMID: 26160801 DOI: 10.3727/096368915x688632] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Kidney disease poses a global challenge. Stem cell therapy may offer an alternative therapeutic approach to kidney transplantation, which is often hampered by the limited supply of donor organs. While specific surface antigen markers have yet to be identified for the analysis and purification of kidney stem/progenitor cells for research or clinical use, the reprogramming of somatic cells to pluripotent cells and their differentiation into the various kidney lineages might represent a valuable strategy to create a renewable cell source for regenerative purposes. In this review, we first provide an overview of kidney development and explore current knowledge about the role of extra- and intrarenal cells in kidney repair and organogenesis. We then discuss recent advances in the 1) differentiation of rodent and human embryonic stem cells (ESCs) into renal lineages; 2) generation of induced pluripotent stem cells (iPSCs) from renal or nonrenal (kidney patient-derived) adult cells; 3) differentiation of iPSCs into renal lineages; and 4) direct transcriptional reprogramming of adult renal cells into kidney progenitor cells. Finally, we describe the lymph node as a potential three-dimensional (3D) in vivo environment for kidney organogenesis from pluripotent stem cells.
Collapse
Affiliation(s)
- Maria Giovanna Francipane
- McGowan Institute for Regenerative Medicine, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | |
Collapse
|
28
|
Raschzok N, Sallmon H, Pratschke J, Sauer IM. MicroRNAs in liver tissue engineering - New promises for failing organs. Adv Drug Deliv Rev 2015; 88:67-77. [PMID: 26116880 DOI: 10.1016/j.addr.2015.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 06/10/2015] [Accepted: 06/16/2015] [Indexed: 12/15/2022]
Abstract
miRNA-based technologies provide attractive tools for several liver tissue engineering approaches. Herein, we review the current state of miRNA applications in liver tissue engineering. Several miRNAs have been implicated in hepatic disease and proper hepatocyte function. However, the clinical translation of these findings into tissue engineering has just begun. miRNAs have been successfully used to induce proliferation of mature hepatocytes and improve the differentiation of hepatic precursor cells. Nonetheless, miRNA-based approaches beyond cell generation have not yet entered preclinical or clinical investigations. Moreover, miRNA-based concepts for the biliary tree have yet to be developed. Further research on miRNA based modifications, however, holds the promise of enabling significant improvements to liver tissue engineering approaches due to their ability to regulate and fine-tune all biological processes relevant to hepatic tissue engineering, such as proliferation, differentiation, growth, and cell function.
Collapse
Affiliation(s)
- Nathanael Raschzok
- General, Visceral, and Transplantation Surgery, Charité - Universitätsmedizin Berlin, Germany
| | - Hannes Sallmon
- Neonatology, Charité - Universitätsmedizin Berlin, Germany
| | - Johann Pratschke
- General, Visceral, and Transplantation Surgery, Charité - Universitätsmedizin Berlin, Germany
| | - Igor M Sauer
- General, Visceral, and Transplantation Surgery, Charité - Universitätsmedizin Berlin, Germany.
| |
Collapse
|
29
|
Stevens KR, Miller JS, Blakely BL, Chen CS, Bhatia SN. Degradable hydrogels derived from PEG-diacrylamide for hepatic tissue engineering. J Biomed Mater Res A 2015; 103:3331-8. [PMID: 25851120 PMCID: PMC4890565 DOI: 10.1002/jbm.a.35478] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/02/2015] [Accepted: 04/02/2015] [Indexed: 01/19/2023]
Abstract
Engineered tissue constructs have the potential to augment or replace whole organ transplantation for the treatment of liver failure. Poly(ethylene glycol) (PEG)‐based systems are particularly promising for the construction of engineered liver tissue due to their biocompatibility and amenability to modular addition of bioactive factors. To date, primary hepatocytes have been successfully encapsulated in non‐degradable hydrogels based on PEG‐diacrylate (PEGDA). In this study, we describe a hydrogel system based on PEG‐diacrylamide (PEGDAAm) containing matrix‐metalloproteinase sensitive (MMP‐sensitive) peptide in the hydrogel backbone that is suitable for hepatocyte culture both in vitro and after implantation. By replacing hydrolytically unstable esters in PEGDA with amides in PEGDAAm, resultant hydrogels resisted non‐specific hydrolysis, while still allowing for MMP‐mediated hydrogel degradation. Optimization of polymerization conditions, hepatocellular density, and multicellular tissue composition modulated both the magnitude and longevity of hepatic function in vitro. Importantly, hepatic PEGDAAm‐based tissues survived and functioned for over 3 weeks after implantation ectopically in the intraperitoneal (IP) space of nude mice. Together, these studies suggest that MMP‐sensitive PEGDAAm‐based hydrogels may be a useful material system for applications in tissue engineering and regenerative medicine. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 103A: 3331–3338, 2015.
Collapse
Affiliation(s)
- Kelly R Stevens
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| | - Jordan S Miller
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Brandon L Blakely
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Christopher S Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Sangeeta N Bhatia
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139.,Howard Hughes Medical Institute, Cambridge, Massachusetts, 02139.,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| |
Collapse
|
30
|
New Tools in Experimental Cellular Therapy for the Treatment of Liver Diseases. CURRENT TRANSPLANTATION REPORTS 2015; 2:202-210. [PMID: 26317066 DOI: 10.1007/s40472-015-0059-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The current standard of care for end stage liver disease is orthotopic liver transplantation (OLT). Through improvement in surgical techniques, immunosuppression, and general medical care, liver transplantation has become an effective treatment over the course of the last half-century. Unfortunately, due to the limited availability of donor organs, there is a finite limit to the number of patients who will benefit from this therapy. This review will discuss current research in experimental cellular therapies for acute, chronic, and metabolic liver failure that may be appropriate when liver transplantation is not an immediate option.
Collapse
|
31
|
Hickey RD, Mao SA, Amiot B, Suksanpaisan L, Miller A, Nace R, Glorioso J, Peng KW, Ikeda Y, Russell SJ, Nyberg SL. Noninvasive 3-dimensional imaging of liver regeneration in a mouse model of hereditary tyrosinemia type 1 using the sodium iodide symporter gene. Liver Transpl 2015; 21:442-53. [PMID: 25482651 PMCID: PMC5957080 DOI: 10.1002/lt.24057] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/30/2014] [Indexed: 12/24/2022]
Abstract
Cell transplantation is a potential treatment for the many liver disorders that are currently only curable by organ transplantation. However, one of the major limitations of hepatocyte (HC) transplantation is an inability to monitor cells longitudinally after injection. We hypothesized that the thyroidal sodium iodide symporter (NIS) gene could be used to visualize transplanted HCs in a rodent model of inherited liver disease: hereditary tyrosinemia type 1. Wild-type C57Bl/6J mouse HCs were transduced ex vivo with a lentiviral vector containing the mouse Slc5a5 (NIS) gene controlled by the thyroxine-binding globulin promoter. NIS-transduced cells could robustly concentrate radiolabeled iodine in vitro, with lentiviral transduction efficiencies greater than 80% achieved in the presence of dexamethasone. Next, NIS-transduced HCs were transplanted into congenic fumarylacetoacetate hydrolase knockout mice, and this resulted in the prevention of liver failure. NIS-transduced HCs were readily imaged in vivo by single-photon emission computed tomography, and this demonstrated for the first time noninvasive 3-dimensional imaging of regenerating tissue in individual animals over time. We also tested the efficacy of primary HC spheroids engrafted in the liver. With the NIS reporter, robust spheroid engraftment and survival could be detected longitudinally after direct parenchymal injection, and this thereby demonstrated a novel strategy for HC transplantation. This work is the first to demonstrate the efficacy of NIS imaging in the field of HC transplantation. We anticipate that NIS labeling will allow noninvasive and longitudinal identification of HCs and stem cells in future studies related to liver regeneration in small and large preclinical animal models.
Collapse
Affiliation(s)
- Raymond D. Hickey
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Bruce Amiot
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Amber Miller
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Rebecca Nace
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Kah Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yasuhiro Ikeda
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
32
|
Francipane MG, Lagasse E. The lymph node as a new site for kidney organogenesis. Stem Cells Transl Med 2015; 4:295-307. [PMID: 25646529 PMCID: PMC4339853 DOI: 10.5966/sctm.2014-0208] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/23/2014] [Indexed: 12/11/2022] Open
Abstract
The shortage of organs for kidney transplantation has created the need to develop new strategies to restore renal structure and function. Given our recent finding that the lymph node (LN) can serve as an in vivo factory to generate or sustain complex structures like liver, pancreas, and thymus, we investigated whether it could also support kidney organogenesis from mouse renal embryonic tissue (metanephroi). Here we provide the first evidence that metanephroi acquired a mature phenotype upon injection into LN, and host cells likely contributed to this process. Urine-like fluid-containing cysts were observed in several grafts 12 weeks post-transplantation, indicating metanephroi transplants' ability to excrete products filtered from the blood. Importantly, the kidney graft adapted to a loss of host renal mass, speeding its development. Thus, the LN might provide a unique tool for studying the mechanisms of renal maturation, cell proliferation, and fluid secretion during cyst development. Moreover, we provide evidence that inside the LN, short-term cultured embryonic kidney cells stimulated with the Wnt agonist R-Spondin 2 gave rise to a monomorphic neuron-like cell population expressing the neuronal 200-kDa neurofilament heavy marker. This finding indicates that the LN might be used to validate the differentiation potential of candidate stem cells in regenerative nephrology.
Collapse
Affiliation(s)
- Maria Giovanna Francipane
- Department of Pathology, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Ri.MED Foundation, Palermo, Italy
| | - Eric Lagasse
- Department of Pathology, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Ri.MED Foundation, Palermo, Italy
| |
Collapse
|
33
|
Francipane MG, Lagasse E. Maturation of embryonic tissues in a lymph node: a new approach for bioengineering complex organs. Organogenesis 2015; 10:323-31. [PMID: 25531035 PMCID: PMC4750546 DOI: 10.1080/15476278.2014.995509] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Given our recent finding that the lymph node (LN) can serve as an in vivo factory to generate complex structures like liver, pancreas, and thymus, we investigated whether LN could also support early development and maturation from several mid-embryonic (E14.5/15.5) mouse tissues including brain, thymus, lung, stomach, and intestine. Here we observed brain maturation in LN by showing the emergence of astrocytes with well-developed branching processes. Thymus maturation in LN was monitored by changes in host immune cells. Finally, newly terminally differentiated mucus-producing cells were identified in ectopic tissues generated by transplantation of lung, stomach and intestine in LN. Thus, we speculate the LN offers a unique approach to study the intrinsic and extrinsic differentiation potential of cells and tissues during early development, and provides a new site for bioengineering complex body parts.
Collapse
Key Words
- 21wEcT, 21-week ectopic thymus
- 2D, 2-dimensional
- 3D, 3-dimensional
- 3wEcI, 3-week ectopic intestine
- 3wEcL, 3-week ectopic lung
- 3wEcS, 3-week ectopic stomach
- 6wEcT, 6-week ectopic thymus
- AdT, adult thymus
- Aire, autoimmune regulator
- CgA, chromogranin A
- E14.5/15.5, embryonic day 14.5 to 15.5
- ECM, extracellular matrix
- ER-TR7, reticular fibroblasts and reticular fibers
- EmI, embryonic intestine
- EmL, embryonic lung
- EmS, embryonic stomach
- EmT, embryonic thymus
- EpCAM1, epithelial cell adhesion molecule 1
- FACS, fluorescence-activated cell sorting
- FAH, fumarylacetoacetate hydrolase
- GFAPδ, glial fibrillary acid protein delta
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- K5, keratin 5
- K8, keratin 8
- LN, lymph node
- MAP-2, Microtubule-associated protein 2
- bioreactor
- cTEC, cortical thymic epithelial cell
- chimerism
- development
- lymph node
- mTEC, medullary thymic epithelial cell
- mTOR, mammalian target of rapamycin
- terminal differentiation
Collapse
Affiliation(s)
- Maria Giovanna Francipane
- a McGowan Institute for Regenerative Medicine; Department of Pathology ; University of Pittsburgh School of Medicine ; Pittsburgh , PA USA
| | | |
Collapse
|
34
|
Logan GJ, de Alencastro G, Alexander IE, Yeoh GC. Exploiting the unique regenerative capacity of the liver to underpin cell and gene therapy strategies for genetic and acquired liver disease. Int J Biochem Cell Biol 2014; 56:141-52. [PMID: 25449261 DOI: 10.1016/j.biocel.2014.10.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/15/2014] [Accepted: 10/21/2014] [Indexed: 02/06/2023]
Abstract
The number of genetic or acquired diseases of the liver treatable by organ transplantation is ever-increasing as transplantation techniques improve placing additional demands on an already limited organ supply. While cell and gene therapies are distinctly different modalities, they offer a synergistic alternative to organ transplant due to distinct architectural and physiological properties of the liver. The hepatic blood supply and fenestrated endothelial system affords relatively facile accessibility for cell and/or gene delivery. More importantly, however, the remarkable capacity of hepatocytes to proliferate and repopulate the liver creates opportunities for new treatments based on emerging technologies. This review will summarise current understanding of liver regeneration, describe clinical and experimental cell and gene therapeutic modalities and discuss critical challenges to translate these new technologies to wider clinical utility. This article is part of a Directed Issue entitled: "Regenerative Medicine: the challenge of translation".
Collapse
Affiliation(s)
- Grant J Logan
- Gene Therapy Research Unit of The Children's Medical Research Institute and The Children's Hospital at Westmead, Australia
| | - Gustavo de Alencastro
- Gene Therapy Research Unit of The Children's Medical Research Institute and The Children's Hospital at Westmead, Australia
| | - Ian E Alexander
- Gene Therapy Research Unit of The Children's Medical Research Institute and The Children's Hospital at Westmead, Australia; University of Sydney Discipline of Paediatrics and Child Health, Westmead, NSW 2145, Australia
| | - George C Yeoh
- The Centre for Medical Research, Harry Perkins Institute of Medical Research, Crawley, WA 6009, Australia.
| |
Collapse
|
35
|
|
36
|
Fox IJ, Daley GQ, Goldman SA, Huard J, Kamp TJ, Trucco M. Stem cell therapy. Use of differentiated pluripotent stem cells as replacement therapy for treating disease. Science 2014; 345:1247391. [PMID: 25146295 PMCID: PMC4329726 DOI: 10.1126/science.1247391] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pluripotent stem cells (PSCs) directed to various cell fates holds promise as source material for treating numerous disorders. The availability of precisely differentiated PSC-derived cells will dramatically affect blood component and hematopoietic stem cell therapies and should facilitate treatment of diabetes, some forms of liver disease and neurologic disorders, retinal diseases, and possibly heart disease. Although an unlimited supply of specific cell types is needed, other barriers must be overcome. This review of the state of cell therapies highlights important challenges. Successful cell transplantation will require optimizing the best cell type and site for engraftment, overcoming limitations to cell migration and tissue integration, and occasionally needing to control immunologic reactivity, as well as a number of other challenges. Collaboration among scientists, clinicians, and industry is critical for generating new stem cell-based therapies.
Collapse
Affiliation(s)
- Ira J Fox
- Department of Surgery, Children's Hospital of Pittsburgh and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - George Q Daley
- Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA. Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Broad Institute, Cambridge, MA, USA. Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, The University of Rochester Medical Center, Rochester, NY, USA. Center for Basic and Translational Neuroscience, University of Copenhagen, Denmark
| | - Johnny Huard
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Timothy J Kamp
- Stem Cell and Regenerative Medicine Center, Cellular and Molecular Arrhythmia Research Program, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Massimo Trucco
- Division of Immunogenetics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
37
|
Abstract
Despite the tremendous hurdles presented by the complexity of the liver's structure and function, advances in liver physiology, stem cell biology and reprogramming, and the engineering of tissues and devices are accelerating the development of cell-based therapies for treating liver disease and liver failure. This State of the Art Review discusses both the near- and long-term prospects for such cell-based therapies and the unique challenges for clinical translation.
Collapse
Affiliation(s)
- Sangeeta N Bhatia
- Institute for Medical Engineering & Science at MIT, Department of Electrical Engineering and Computer Science, David H. Koch Institute at MIT, and the Howard Hughes Medical Institute, Cambridge, MA 02139, USA. Division of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Gregory H Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kenneth S Zaret
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ira J Fox
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, and McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15224, USA
| |
Collapse
|
38
|
Abstract
Effective utilization of three-dimensional printing for tissue and organ engineering remains nontrivial. Here, Jordan Miller identifies key challenges and discusses conceptual targets on the horizon. How structure relates to function—across spatial scales, from the single molecule to the whole organism—is a central theme in biology. Bioengineers, however, wrestle with the converse question: will function follow form? That is, we struggle to approximate the architecture of living tissues experimentally, hoping that the structure we create will lead to the function we desire. A new means to explore the relationship between form and function in living tissue has arrived with three-dimensional printing, but the technology is not without limitations.
Collapse
Affiliation(s)
- Jordan S. Miller
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW This review outlines the concept of cell-based therapy to restore tissue function, and addresses four key points to consider in cell transplantation: source, surveillance, safety, and site. Whereas each point is essential, additional attention should be given to transplantation sites if cell therapy is going to be successful in the clinic. Various ectopic locations are discussed, and the strengths and weaknesses of each are compared as suitable candidates for cell therapy. RECENT FINDINGS Studies in rodents often demonstrate cell transplantation and engraftment in ectopic sites, with little evidence to suggest why it may also work in humans. For example, transplantation to the subcapsular space of the kidney is often performed in rodents, but has not been a good predictor of clinical success. Recent work has shown that the lymph node may be a good site for transplantation of multiple tissue types, and several reasons are highlighted as to why it should be considered for future studies. SUMMARY The use of cell-based therapy in the clinic has been hampered by the lack of appropriate sites for transplantation. The lymph node is a promising alternative for cell transplantation, and offers hope for clinical application.
Collapse
|
40
|
Abstract
The treatment of end-stage liver disease and acute liver failure remains a clinically relevant issue. Although orthotopic liver transplantation is a well-established procedure, whole-organ transplantation is invasive and increasingly limited by the unavailability of suitable donor organs. Artificial and bioartificial liver support systems have been developed to provide an alternative to whole organ transplantation, but despite three decades of scientific efforts, the results are still not convincing with respect to clinical outcome. In this Review, conceptual limitations of clinically available liver support therapy systems are discussed. Furthermore, alternative concepts, such as hepatocyte transplantation, and cutting-edge developments in the field of liver support strategies, including the repopulation of decellularized organs and the biofabrication of entirely new organs by printing techniques or induced organogenesis are analysed with respect to clinical relevance. Whereas hepatocyte transplantation shows promising clinical results, at least for the temporary treatment of inborn metabolic diseases, so far data regarding implantation of engineered hepatic tissue have only emerged from preclinical experiments. However, the evolving techniques presented here raise hope for bioengineered liver support therapies in the future.
Collapse
|
41
|
|
42
|
Willenbring H, Soto-Gutierrez A. Transplantable liver organoids made from only three ingredients. Cell Stem Cell 2014; 13:139-40. [PMID: 23910079 DOI: 10.1016/j.stem.2013.07.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Liver cell therapies using induced pluripotent stem cells (iPSCs) are in development. A recent paper in Nature by Takebe et al. expands the range of liver diseases that could be treated with iPSC-derived hepatocytes by combining them with endothelial and stromal cells to generate organoids that survive and function extrahepatically.
Collapse
Affiliation(s)
- Holger Willenbring
- Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | |
Collapse
|
43
|
Ohashi K, Okano T. Functional Tissue Engineering of the Liver and Islets. Anat Rec (Hoboken) 2013; 297:73-82. [DOI: 10.1002/ar.22810] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 01/01/2023]
Affiliation(s)
- Kazuo Ohashi
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; Shinjyuku-ku Tokyo Japan
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjyuku-ku Tokyo Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; Shinjyuku-ku Tokyo Japan
| |
Collapse
|
44
|
Michalopoulos GK, Grompe M, Theise ND. Assessing the potential of induced liver regeneration. Nat Med 2013; 19:1096-7. [PMID: 24013747 DOI: 10.1038/nm.3325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
45
|
Turner NJ, Keane TJ, Badylak SF. Lessons from developmental biology for regenerative medicine. ACTA ACUST UNITED AC 2013; 99:149-59. [DOI: 10.1002/bdrc.21040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 07/27/2013] [Accepted: 07/27/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Neill J. Turner
- McGowan Institute for Regenerative Medicine; University of Pittsburgh, Pittsburgh, Pennsylvania and Department of Surgery, University of Pittsburgh; Pittsburgh Pennsylvania
| | - Timothy J. Keane
- McGowan Institute for Regenerative Medicine; University of Pittsburgh, Pittsburgh, Pennsylvania and Department of Bioengineering, University of Pittsburgh; Pittsburgh Pennsylvania
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, and Department of Bioengineering, University of Pittsburgh; Pittsburgh Pennsylvania
| |
Collapse
|
46
|
Abstract
Because of their high proliferative capacity, resistance to cryopreservation, and ability to differentiate into hepatocyte-like cells, stem and progenitor cells have recently emerged as attractive cell sources for liver cell therapy, a technique used as an alternative to orthotopic liver transplantation in the treatment of various hepatic ailments ranging from metabolic disorders to end-stage liver disease. Although stem and progenitor cells have been isolated from various tissues, obtaining them from the liver could be an advantage for the treatment of hepatic disorders. However, the techniques available to isolate these stem/progenitor cells are numerous and give rise to cell populations with different morphological and functional characteristics. In addition, there is currently no established consensus on the tests that need to be performed to ensure the quality and safety of these cells when used clinically. The purpose of this review is to describe the different types of liver stem/progenitor cells currently reported in the literature, discuss their suitability and limitations in terms of clinical applications, and examine how the culture and transplantation techniques can potentially be improved to achieve a better clinical outcome.
Collapse
Affiliation(s)
- Catherine A. Lombard
- Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Institut de Recherche Expérimentale et Clinique, Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Julie Prigent
- Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Institut de Recherche Expérimentale et Clinique, Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Etienne M. Sokal
- Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Institut de Recherche Expérimentale et Clinique, Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| |
Collapse
|
47
|
Turner RA, Wauthier E, Lozoya O, McClelland R, Bowsher JE, Barbier C, Prestwich G, Hsu E, Gerber DA, Reid LM. Successful transplantation of human hepatic stem cells with restricted localization to liver using hyaluronan grafts. Hepatology 2013; 57:775-84. [PMID: 22996260 PMCID: PMC3583296 DOI: 10.1002/hep.26065] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 08/14/2012] [Indexed: 12/24/2022]
Abstract
Cell therapies are potential alternatives to organ transplantation for liver failure or dysfunction but are compromised by inefficient engraftment, cell dispersal to ectopic sites, and emboli formation. Grafting strategies have been devised for transplantation of human hepatic stem cells (hHpSCs) embedded into a mix of soluble signals and extracellular matrix biomaterials (hyaluronans, type III collagen, laminin) found in stem cell niches. The hHpSCs maintain a stable stem cell phenotype under the graft conditions. The grafts were transplanted into the livers of immunocompromised murine hosts with and without carbon tetrachloride treatment to assess the effects of quiescent versus injured liver conditions. Grafted cells remained localized to the livers, resulting in a larger bolus of engrafted cells in the host livers under quiescent conditions and with potential for more rapid expansion under injured liver conditions. By contrast, transplantation by direct injection or via a vascular route resulted in inefficient engraftment and cell dispersal to ectopic sites. Transplantation by grafting is proposed as a preferred strategy for cell therapies for solid organs such as the liver.
Collapse
Affiliation(s)
- Rachael A. Turner
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC,Department of Biomedical Engineering, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Eliane Wauthier
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Oswaldo Lozoya
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC,Department of Biomedical Engineering, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Randall McClelland
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - James E. Bowsher
- Department of Biomedical Engineering Duke University School of Medicine, Durham, NC
| | - Claire Barbier
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Glenn Prestwich
- Department of Medicinal Chemistry and Center for Therapeutic Biomaterials University of Utah, Salt Lake City, UT
| | - Edward Hsu
- Department of Biomedical Engineering Duke University School of Medicine, Durham, NC
| | - David A. Gerber
- Department of Surgery, University of North Carolina School of Medicine, Chapel Hill, NC,Lineberger Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Lola M. Reid
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC,Department of Biomedical Engineering, University of North Carolina School of Medicine, Chapel Hill, NC,Program in Molecular Biology and Biotechnology, University of North Carolina School of Medicine, Chapel Hill, NC,Lineberger Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC,Corresponding Author: LM Reid, UNC School of Medicine, Campus Box 7038, Glaxo Building Rms 32-35, Chapel Hill, NC 27599. Phone: 919-966-0347; FAX: 919-6112.
| |
Collapse
|
48
|
Aupet S, Simoné G, Heyd B, Bachellier P, Vidal I, Richert L, Martin H. Isolation of viable human hepatic progenitors from adult livers is possible even after 48 hours of cold ischemia. Tissue Eng Part C Methods 2013. [PMID: 23198983 DOI: 10.1089/ten.tec.2012.0237] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Liver transplantation, utilized routinely for end-stage liver disease, has been constrained by the paucity of organ donors, and is being complemented by alternative strategies such as liver cell transplantation. One of the most promising forms of liver cell transplantation is hepatic stem cell therapies, as the number of human hepatic stem cells (hHpSCs) and other early hepatic progenitor cells (HPCs) are sufficient to provide treatment for multiple patients from a single liver source. In the present study, human adult livers were exposed to cold ischemia and then processed after <24 or 48 h. Cells positive for epithelial cell adhesion molecule (EpCAM), a marker on early lineage stage HPCs, were immunoselected and counted. Approximately 100,000 EpCAM(+) cells/gram of tissue was obtained from surgical resection of livers subjected to cold ischemia up to 24 h and comparable numbers, albeit somewhat lower, were obtained from those exposed to 48 h of cold ischemia. The yields are similar to those reported from livers with minimal exposure to ischemia. When cultured on plastic dishes and in Kubota's Medium, a serum-free medium designed for early lineage stage HPCs, colonies of rapidly expanding cells formed. They were confirmed to be probable hHpSCs by their ability to survive and expand on plastic and in Kubota's Medium for months, by co-expression of EpCAM and neural cell adhesion molecule, minimal if any albumin expression, with EpCAM found throughout the cells, and no expression of alpha-fetoprotein. The yields of viable EpCAM(+) cells were surprisingly large, and the numbers from a single donor liver are sufficient to treat approximately 50-100 patients given the numbers of EpCAM(+) cells currently used in hepatic stem cell therapies. Thus, cold ischemic livers for up to 48 h are a new source of cells that might be used for liver cell therapies.
Collapse
Affiliation(s)
- Sophie Aupet
- EA4267 FDE, SFR133, Faculté de Médecine et Pharmacie, Besançon, France
| | | | | | | | | | | | | |
Collapse
|
49
|
Hammerman MR. Xenotransplantation of embryonic pig pancreas for treatment of diabetes mellitus in non-human primates. ACTA ACUST UNITED AC 2013; 6. [PMID: 24312695 DOI: 10.4236/jbise.2013.65a002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transplantation therapy for diabetes in humans is limited by the low availability of human donor whole pancreas or islets. Outcomes are complicated by immunosuppressive drug toxicity. Xenotransplantation is a strategy to overcome supply problems. Implantation of tissue obtained early during embryogenesis is a way to reduce transplant immunogenicity. Pig insulin is biologically active in humans. In that regard the pig is an appropriate xenogeneic organ donor. Insulin-producing cells originating from embryonic pig pancreas obtained very early following pancreatic primordium formation [embryonic day 28 (E28)] engraft long-term in rhesus macaques. Endocrine cells originating from embryonic pig pancreas transplanted in host mesentery migrate to mesenteric lymph nodes, engraft, differentiate and improve glucose tolerance in rhesus macaques without the need for immune suppression. Transplantation of embryonic pig pancreas is a novel approach towards beta cell replacement therapy that could be applicable to humans.
Collapse
Affiliation(s)
- Marc R Hammerman
- George M. O'Brien Center for Kidney Disease Research, Renal Division, Departments of Medicine and Cell Biology and Physiology, Washington University School of Medicine, St. Louis MO 63110
| |
Collapse
|
50
|
Zakrzewski JL, Tuckett AZ, van den Brink MRM. Transforming lymph nodes into tissue factories. Nat Biotechnol 2012; 30:958-9. [DOI: 10.1038/nbt.2388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|