1
|
Umezawa A, Fukuda A, Horikawa R, Uchida H, Enosawa S, Oishi Y, Nakamura N, Sasaki K, Yanagi Y, Shimizu S, Nakao T, Kodama T, Sakamoto S, Hayakawa I, Akiyama S, Saku N, Miyata S, Ite K, Javaregowda PK, Toyoda M, Nonaka H, Nakamura K, Ito Y, Fukuhara Y, Miyazaki O, Nosaka S, Nakabayashi K, Haga C, Yoshioka T, Masuda A, Ohkura T, Yamazaki-Inoue M, Machida M, Abutani-Sakamoto R, Miyajima S, Akutsu H, Matsubara Y, Igarashi T, Kasahara M. First-in-human clinical study of an embryonic stem cell product for urea cycle disorders. Stem Cell Res Ther 2025; 16:120. [PMID: 40050977 PMCID: PMC11887382 DOI: 10.1186/s13287-025-04162-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/21/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND This study assesses the safety and efficacy of hepatocyte-like cell (HLC) infusion therapy derived from human embryonic stem cells as bridging therapy for neonatal-onset urea cycle disorders (UCD). The research includes both preclinical and clinical evaluations to determine the feasibility of HLC infusion as a therapeutic option for safer pediatric liver transplantation. METHODS Preclinical studies were conducted to validate the safety, biodistribution, and ammonia metabolism capabilities of HLCs using SCID mice models of UCD and extensive animal studies. In the clinical trial, five neonates with UCD received HLC infusions, intending to maintain metabolic stability and exceed a target weight of over 6 kg, which is considered necessary for safer liver transplantation. RESULTS Preclinical studies demonstrated that HLCs successfully engrafted in the liver without adverse migration or tumor formation and effectively elongated survival. Clinically, all five neonates exceeded the target weight of 6 kg while maintaining metabolic stability and successfully bridging to transplantation. Post-transplantation follow-up revealed stable growth, metabolic control, and no neurological complications. CONCLUSIONS The combined preclinical and clinical findings support HLC infusion as a viable bridge therapy for neonates with UCD, providing metabolic support to achieve safer weight thresholds for transplantation. While promising, careful monitoring remains essential, particularly for potential complications such as thrombus formation. TRIAL REGISTRATION jRCT, jRCT1090220412. Registered on 27 February 2019, https://jrct.niph.go.jp/en-latest-detail/jRCT1090220412 (originally registered in JMACCT (JMA-IIA00412)).
Collapse
Affiliation(s)
- Akihiro Umezawa
- National Center for Child Health and Development Research Institute, Setagaya, Japan.
- Department of Advanced Pediatric Medicine (National Center for Child Health and Development), Tohoku University School of Medicine, Sendai, Japan.
| | - Akinari Fukuda
- National Center for Child Health and Development, Setagaya, Japan
| | - Reiko Horikawa
- National Center for Child Health and Development, Setagaya, Japan
| | - Hajime Uchida
- National Center for Child Health and Development, Setagaya, Japan
| | - Shin Enosawa
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Yoshie Oishi
- National Center for Child Health and Development, Setagaya, Japan
| | - Naoko Nakamura
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Kengo Sasaki
- National Center for Child Health and Development, Setagaya, Japan
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yusuke Yanagi
- National Center for Child Health and Development, Setagaya, Japan
| | - Seiichi Shimizu
- National Center for Child Health and Development, Setagaya, Japan
| | - Toshimasa Nakao
- National Center for Child Health and Development, Setagaya, Japan
| | - Tasuku Kodama
- National Center for Child Health and Development, Setagaya, Japan
| | - Seisuke Sakamoto
- National Center for Child Health and Development, Setagaya, Japan
| | - Itaru Hayakawa
- National Center for Child Health and Development, Setagaya, Japan
| | - Saeko Akiyama
- National Center for Child Health and Development Research Institute, Setagaya, Japan
- Department of Advanced Pediatric Medicine (National Center for Child Health and Development), Tohoku University School of Medicine, Sendai, Japan
| | - Noriaki Saku
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Shoko Miyata
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Kenta Ite
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Palaksha Kanive Javaregowda
- National Center for Child Health and Development Research Institute, Setagaya, Japan
- SDM Research Institute for Biomedical Sciences, A Constituent Unit of Shri Dharmasthala Manjunatheshwara University, Dharwad, India
| | - Masashi Toyoda
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Hidenori Nonaka
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Kazuaki Nakamura
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Yoshikazu Ito
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | | | - Osamu Miyazaki
- National Center for Child Health and Development, Setagaya, Japan
| | - Shunsuke Nosaka
- National Center for Child Health and Development, Setagaya, Japan
| | - Kazuhiko Nakabayashi
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Chizuko Haga
- National Center for Child Health and Development, Setagaya, Japan
| | - Takako Yoshioka
- National Center for Child Health and Development, Setagaya, Japan
| | - Akira Masuda
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Takashi Ohkura
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Mayu Yamazaki-Inoue
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Masakazu Machida
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Rie Abutani-Sakamoto
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Shoko Miyajima
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Hidenori Akutsu
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Yoichi Matsubara
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Takashi Igarashi
- National Center for Child Health and Development Research Institute, Setagaya, Japan
| | - Mureo Kasahara
- National Center for Child Health and Development, Setagaya, Japan.
| |
Collapse
|
2
|
Sun Z, Yuan X, Wu J, Wang C, Zhang K, Zhang L, Hui L. Hepatocyte transplantation: The progress and the challenges. Hepatol Commun 2023; 7:e0266. [PMID: 37695736 PMCID: PMC10497249 DOI: 10.1097/hc9.0000000000000266] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/26/2023] [Indexed: 09/13/2023] Open
Abstract
Numerous studies have shown that hepatocyte transplantation is a promising approach for liver diseases, such as liver-based metabolic diseases and acute liver failure. However, it lacks strong evidence to support the long-term therapeutic effects of hepatocyte transplantation in clinical practice. Currently, major hurdles include availability of quality-assured hepatocytes, efficient engraftment and repopulation, and effective immunosuppressive regimens. Notably, cell sources have been advanced recently by expanding primary human hepatocytes by means of dedifferentiation in vitro. Moreover, the transplantation efficiency was remarkably improved by the established preparative hepatic irradiation in combination with hepatic mitogenic stimuli regimens. Finally, immunosuppression drugs, including glucocorticoid and inhibitors for co-stimulating signals of T cell activation, were proposed to prevent innate and adaptive immune rejection of allografted hepatocytes. Despite remarkable progress, further studies are required to improve in vitro cell expansion technology, develop clinically feasible preconditioning regimens, and further optimize immunosuppression regimens or establish ex vivo gene correction-based autologous hepatocyte transplantation.
Collapse
Affiliation(s)
- Zhen Sun
- 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 Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiang Yuan
- 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 Sciences, Shanghai, China
| | - Jingqi 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 Sciences, Shanghai, 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 Sciences, Shanghai, China
| | - Kun 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 Sciences, Shanghai, 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 Sciences, Shanghai, China
| | - Lijian Hui
- 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 Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
3
|
Kok G, Ilcken EF, Houwen RH, Lindemans CA, Nieuwenhuis EE, Spierings E, Fuchs SA. The Effect of Genetic HLA Matching on Liver Transplantation Outcome: A Systematic Review and Meta-Analysis. ANNALS OF SURGERY OPEN 2023; 4:e334. [PMID: 37746594 PMCID: PMC10513352 DOI: 10.1097/as9.0000000000000334] [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: 11/21/2022] [Accepted: 08/11/2023] [Indexed: 09/26/2023] Open
Abstract
Objective We aim to investigate the effects of genetically based HLA matching on patient and graft survival, and acute and chronic rejection after liver transplantation. Background Liver transplantation is a common treatment for patients with end-stage liver disease. In contrast to most other solid organ transplantations, there is no conclusive evidence supporting human leukocyte antigen (HLA) matching for liver transplantations. With emerging alternatives such as transplantation of bankable (stem) cells, HLA matching becomes feasible, which may decrease the need for immunosuppressive therapy and improve transplantation outcomes. Methods We systematically searched the PubMed, Embase, and Cochrane databases and performed a meta-analysis investigating the effect of genetic HLA matching on liver transplantation outcomes (acute/chronic rejection, graft failure, and mortality). Results We included 14 studies with 2682 patients. HLA-C mismatching significantly increased the risk of acute rejection (full mismatching: risk ratio = 1.90, 95% confidence interval = 1.08 to 3.33, P = 0.03; partial mismatching: risk ratio = 1.33, 95% confidence interval = 1.07 to 1.66, P = 0.01). We did not discern any significant effect of HLA mismatching per locus on acute rejection for HLA-A, -B, -DR, and -DQ, nor on chronic rejection, graft failure, or mortality for HLA-DR, and -DQ. Conclusions We found evidence that genetic HLA-C matching reduces the risk of acute rejection after liver transplantation while matching for other loci does not reduce the risk of acute rejection, chronic rejection, graft failure, or mortality.
Collapse
Affiliation(s)
- Gautam Kok
- From the Department of Metabolic Diseases, Wilhelmina Children’s Hospital, Utrecht, The Netherlands
| | - Eveline F. Ilcken
- From the Department of Metabolic Diseases, Wilhelmina Children’s Hospital, Utrecht, The Netherlands
| | - Roderick H.J. Houwen
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, Utrecht, The Netherlands
| | - Caroline A. Lindemans
- Department of Immunology, Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Hematopoietic Cell Transplantation, Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Edward E.S. Nieuwenhuis
- Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, Utrecht, The Netherlands
| | - Eric Spierings
- Center of Translational Immunology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Sabine A. Fuchs
- From the Department of Metabolic Diseases, Wilhelmina Children’s Hospital, Utrecht, The Netherlands
| |
Collapse
|
4
|
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
|
5
|
Anand H, Nulty J, Dhawan A. Cell therapy in congenital inherited hepatic disorders. Best Pract Res Clin Gastroenterol 2021; 56-57:101772. [PMID: 35331403 DOI: 10.1016/j.bpg.2021.101772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 01/31/2023]
Abstract
Congenital inherited hepatic disorders (CIHDs) are a set of diverse and heterogeneous group of genetic disorders leading to a defect in an enzyme or transporter. Most of these disorders are currently treated by liver transplantation as standard of care. Improved surgical techniques and post-operative care has led to a wider availability and success of liver transplantation program worldwide. However liver transplantation has its own limitations due to invasive surgery and lifelong use of immunosuppressive agents. Our experience from auxiliary liver transplantation (where right or the left lobe of the patient liver is replaced with a healthy liver donor) demonstrated successful treatment of the underlying defect of noncirrhotic metabolic disorder suggesting that whole liver replacement may not be necessary to achieve a change in phenotype. Large number of animal studies in human models of CIHD have shown success of hepatocyte transplantation leading to its human use. This review addresses the current state of human hepatocyte transplantation in the management of CIHDs with bottlenecks to its wider application and future perspectives.
Collapse
Affiliation(s)
- Hanish Anand
- King's College Hospital NHS Trust: King's College Hospital NHS Foundation Trust, United Kingdom; DhawanLab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, UK
| | - Jessica Nulty
- King's College Hospital NHS Trust: King's College Hospital NHS Foundation Trust, United Kingdom; DhawanLab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, UK
| | - Anil Dhawan
- King's College Hospital NHS Trust: King's College Hospital NHS Foundation Trust, United Kingdom; DhawanLab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, UK.
| |
Collapse
|
6
|
Cell-Based Regeneration and Treatment of Liver Diseases. Int J Mol Sci 2021; 22:ijms221910276. [PMID: 34638617 PMCID: PMC8508969 DOI: 10.3390/ijms221910276] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022] Open
Abstract
The liver, in combination with a functional biliary system, is responsible for maintaining a great number of vital body functions. However, acute and chronic liver diseases may lead to irreversible liver damage and, ultimately, liver failure. At the moment, the best curative option for patients suffering from end-stage liver disease is liver transplantation. However, the number of donor livers required by far surpasses the supply, leading to a significant organ shortage. Cellular therapies play an increasing role in the restoration of organ function and can be integrated into organ transplantation protocols. Different types and sources of stem cells are considered for this purpose, but highly specific immune cells are also the focus of attention when developing individualized therapies. In-depth knowledge of the underlying mechanisms governing cell differentiation and engraftment is crucial for clinical implementation. Additionally, novel technologies such as ex vivo machine perfusion and recent developments in tissue engineering may hold promising potential for the implementation of cell-based therapies to restore proper organ function.
Collapse
|
7
|
Zhang W, Lanzoni G, Hani H, Overi D, Cardinale V, Simpson S, Pitman W, Allen A, Yi X, Wang X, Gerber D, Prestwich G, Lozoya O, Gaudio E, Alvaro D, Tokaz D, Dominguez-Bendala J, Adin C, Piedrahita J, Mathews K, Sethupathy P, Carpino G, He Z, Wauthier E, Reid LM. Patch grafting, strategies for transplantation of organoids into solid organs such as liver. Biomaterials 2021; 277:121067. [PMID: 34517276 DOI: 10.1016/j.biomaterials.2021.121067] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/06/2021] [Accepted: 08/08/2021] [Indexed: 12/28/2022]
Abstract
Epithelial cell therapies have been at an impasse because of inefficient methods of transplantation to solid organs. Patch grafting strategies were established enabling transplantation of ≥107th organoids/patch of porcine GFP+ biliary tree stem/progenitors into livers of wild type hosts. Grafts consisted of organoids embedded in soft (~100 Pa) hyaluronan hydrogels, both prepared in serum-free Kubota's Medium; placed against target sites; covered with a silk backing impregnated with more rigid hyaluronan hydrogels (~700 Pa); and use of the backing to tether grafts with sutures or glue to target sites. Hyaluronan coatings (~200-300 Pa) onto the serosal surface of the graft served to minimize adhesions with neighboring organs. The organ's clearance of hyaluronans enabled restoration of tissue-specific paracrine and systemic signaling, resulting in return of normal hepatic histology, with donor parenchymal cells uniformly integrated amidst host cells and that had differentiated to mature hepatocytes and cholangiocytes. Grafts containing donor mature hepatocytes, partnered with endothelia, and in the same graft biomaterials as for stem/progenitor organoids, did not engraft. Engraftment occurred if porcine liver-derived mesenchymal stem cells (MSCs) were co-transplanted with donor mature cells. RNA-seq analyses revealed that engraftment correlated with expression of matrix-metalloproteinases (MMPs), especially secreted isoforms that were found expressed strongly by organoids, less so by MSCs, and minimally, if at all, by adult cells. Engraftment with patch grafting strategies occurred without evidence of emboli or ectopic cell distribution. It was successful with stem/progenitor organoids or with cells with a source(s) of secreted MMP isoforms and offers significant potential for enabling cell therapies for solid organs.
Collapse
Affiliation(s)
- Wencheng Zhang
- Departments of Cell Biology and Physiology, Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, 27599, USA; Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University School of Medicine, 1800 Yuntai Rd, Pudong New Area, Shanghai, 200123, China
| | - Giacomo Lanzoni
- Diabetes Research Institute, U. Miami Leonard M. Miller School of Medicine, 1450 N.W. 10th Avenue, Miami, FL, 33136, USA
| | - Homayoun Hani
- Departments of Cell Biology and Physiology, Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Diletta Overi
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University, Piazzale Aldo Moro, 5, 00185, Roma RM, Italy
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Piazzale Aldo Moro, 5, 00185, Roma RM, Italy
| | - Sean Simpson
- Department of Molecular Biomedical Sciences, NCSU Colleage of Veterinary Medicine, Raleigh, NC, 27606, USA; The Comparative Medicine Institute, NCSU College of Veterinary Medicine, Raleigh, NC, 27606, USA; Department of Comparative Veterinary Anatomy, NCSU College of Veterinary Medicine, Raleigh, NC, 27606, USA
| | - Wendy Pitman
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, T7 006D Veterinary Research Tower, Box 17, Ithaca, NY, 14853, USA
| | - Amanda Allen
- Departments of Cell Biology and Physiology, Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Xianwen Yi
- Departments of Surgery, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Xicheng Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University School of Medicine, 1800 Yuntai Rd, Pudong New Area, Shanghai, 200123, China
| | - David Gerber
- Departments of Surgery, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Glenn Prestwich
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT, 84112, USA
| | - Oswaldo Lozoya
- Departments of Cell Biology and Physiology, Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, 27599, USA; Department of Biomedical Engineering, UNC School of Medicine, Chapel Hill, NC, 27599, USA.
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University, Piazzale Aldo Moro, 5, 00185, Roma RM, Italy
| | - Domenico Alvaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Piazzale Aldo Moro, 5, 00185, Roma RM, Italy
| | - Debra Tokaz
- Department of Population Health and Pathobiology, NCSU College of Veterinary Medicine, Raleigh, NC, 27606, USA
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, U. Miami Leonard M. Miller School of Medicine, 1450 N.W. 10th Avenue, Miami, FL, 33136, USA
| | - Christopher Adin
- Department of Clinical Sciences, NCSU College of Veterinary Medicine, Raleigh, NC, 27606, USA
| | - Jorge Piedrahita
- Department of Molecular Biomedical Sciences, NCSU Colleage of Veterinary Medicine, Raleigh, NC, 27606, USA; The Comparative Medicine Institute, NCSU College of Veterinary Medicine, Raleigh, NC, 27606, USA; Department of Comparative Veterinary Anatomy, NCSU College of Veterinary Medicine, Raleigh, NC, 27606, USA
| | - Kyle Mathews
- Department of Clinical Sciences, NCSU College of Veterinary Medicine, Raleigh, NC, 27606, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, T7 006D Veterinary Research Tower, Box 17, Ithaca, NY, 14853, USA
| | - Guido Carpino
- Department of Movement, Human and Health Sciences, Division of Health Sciences, University of Rome "Foro Italico", Roma, Italy
| | - Zhiying He
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University School of Medicine, 1800 Yuntai Rd, Pudong New Area, Shanghai, 200123, China
| | - Eliane Wauthier
- Departments of Cell Biology and Physiology, Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Lola M Reid
- Departments of Cell Biology and Physiology, Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
8
|
Takano C, Grubbs BH, Ishige M, Ogawa E, Morioka I, Hayakawa S, Miki T. Clinical perspective on the use of human amniotic epithelial cells to treat congenital metabolic diseases with a focus on maple syrup urine disease. Stem Cells Transl Med 2021; 10:829-835. [PMID: 33547875 PMCID: PMC8133340 DOI: 10.1002/sctm.20-0225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/16/2022] Open
Abstract
Congenital metabolic diseases are a group of hereditary disorders caused by the deficiency of a single specific enzyme activity. Without appropriate therapy, affected patients suffer severe neurologic disability and eventual death. The current mainstays of management attempt to slow disease progression, but are not curative. Several of these diseases have demonstrated significant benefits from liver transplantation; however, this approach is limited by the morbidity associated with this invasive procedure and a shortage of donor organs. Therefore, there is a need to establish a new strategy for improving the quality of a life for these patients. One potential solution is regenerative therapy using hepatocytes generated from stem cells. Herein, we discuss pertinent issues necessary for clinical application of the human amniotic epithelial cell, a type of placental stem cell. Focusing on maple syrup urine disease as an example, where liver replacement is an effective therapy, we explore this approach from a clinician's perspective.
Collapse
Affiliation(s)
- Chika Takano
- Division of Microbiology, Department of Pathology and MicrobiologyNihon University School of MedicineTokyoJapan
- Department of Pediatrics and Child HealthNihon University School of MedicineTokyoJapan
| | - Brendan H. Grubbs
- Department of Obstetrics and GynecologyKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Mika Ishige
- Department of Pediatrics and Child HealthNihon University School of MedicineTokyoJapan
| | - Erika Ogawa
- Department of Pediatrics and Child HealthNihon University School of MedicineTokyoJapan
| | - Ichiro Morioka
- Department of Pediatrics and Child HealthNihon University School of MedicineTokyoJapan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and MicrobiologyNihon University School of MedicineTokyoJapan
| | - Toshio Miki
- Department of PhysiologyNihon University School of MedicineTokyoJapan
| |
Collapse
|
9
|
Clinical progress of auxiliary liver transplantation. Eur J Gastroenterol Hepatol 2021; 33:4-8. [PMID: 32398492 DOI: 10.1097/meg.0000000000001751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
After half a century of development, auxiliary liver transplantation (ALT) technology gradually matured and major indications of ALT have been gradually expanded. This review summarized the history of ALT and introduced indications for ALT which including metabolic liver disease, fulminant hepatic failure, highly sensitized kidney transplantation, prevention of hepatic resection of small hepatic syndrome, etc.; at the same time, the hot issues related to ALT were discussed, including the regulation of hepatic portal blood flow of transplanted liver and residual liver, how to treat the graft liver and remaining liver on second stage. Additionally, the expansion of indications for ALT which included the implementation of ALT for patients with liver cancer and ALT for patients with liver cirrhosis was discussed. It was believed that ALT can greatly alleviate the contradiction of insufficient source of graft liver.
Collapse
|
10
|
Salminen AT, Allahyari Z, Gholizadeh S, McCloskey MC, Ajalik R, Cottle RN, Gaborski TR, McGrath JL. In vitro Studies of Transendothelial Migration for Biological and Drug Discovery. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:600616. [PMID: 35047883 PMCID: PMC8757899 DOI: 10.3389/fmedt.2020.600616] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammatory diseases and cancer metastases lack concrete pharmaceuticals for their effective treatment despite great strides in advancing our understanding of disease progression. One feature of these disease pathogeneses that remains to be fully explored, both biologically and pharmaceutically, is the passage of cancer and immune cells from the blood to the underlying tissue in the process of extravasation. Regardless of migratory cell type, all steps in extravasation involve molecular interactions that serve as a rich landscape of targets for pharmaceutical inhibition or promotion. Transendothelial migration (TEM), or the migration of the cell through the vascular endothelium, is a particularly promising area of interest as it constitutes the final and most involved step in the extravasation cascade. While in vivo models of cancer metastasis and inflammatory diseases have contributed to our current understanding of TEM, the knowledge surrounding this phenomenon would be significantly lacking without the use of in vitro platforms. In addition to the ease of use, low cost, and high controllability, in vitro platforms permit the use of human cell lines to represent certain features of disease pathology better, as seen in the clinic. These benefits over traditional pre-clinical models for efficacy and toxicity testing are especially important in the modern pursuit of novel drug candidates. Here, we review the cellular and molecular events involved in leukocyte and cancer cell extravasation, with a keen focus on TEM, as discovered by seminal and progressive in vitro platforms. In vitro studies of TEM, specifically, showcase the great experimental progress at the lab bench and highlight the historical success of in vitro platforms for biological discovery. This success shows the potential for applying these platforms for pharmaceutical compound screening. In addition to immune and cancer cell TEM, we discuss the promise of hepatocyte transplantation, a process in which systemically delivered hepatocytes must transmigrate across the liver sinusoidal endothelium to successfully engraft and restore liver function. Lastly, we concisely summarize the evolving field of porous membranes for the study of TEM.
Collapse
Affiliation(s)
- Alec T. Salminen
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Zahra Allahyari
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Shayan Gholizadeh
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Molly C. McCloskey
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Raquel Ajalik
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Renee N. Cottle
- Bioengineering, Clemson University, Clemson, SC, United States
| | - Thomas R. Gaborski
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - James L. McGrath
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| |
Collapse
|
11
|
Papatheodoridi M, Mazza G, Pinzani M. Regenerative hepatology: In the quest for a modern prometheus? Dig Liver Dis 2020; 52:1106-1114. [PMID: 32868215 DOI: 10.1016/j.dld.2020.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022]
Abstract
As liver-related morbidity and mortality is rising worldwide and orthotopic liver transplantation (OLT) remains the only standard-of-care for end-stage liver disease or acute liver failure, shortage of donor organs is becoming more prominent. Importantly, advances in regenerative Hepatology and liver bioengineering are bringing new hope to the possibility of restoring impaired hepatic functionality in the presence of acute or chronic liver failure. Hepatocyte transplantation and artificial liver-support systems were the first strategies used in regenerative hepatology but have presented various types of efficiency limitations restricting their widespread use. In parallel, liver bioengineering has been a rapidly developing field bringing continuously novel advancements in biomaterials, three dimensional (3D) scaffolds, cell sources and relative methodologies for creating bioengineered liver tissue. The current major task in liver bioengineering is to build small implantable liver mass for treating inherited metabolic disorders, bioengineered bile ducts for congenital biliary defects and large bioengineered liver organs for transplantation, as substitutes to donor-organs, in cases of acute or acute-on-chronic liver failure. This review aims to summarize the state-of-the-art and upcoming technologies of regenerative Hepatology that are emerging as promising alternatives to the current standard-of care in liver disease.
Collapse
Affiliation(s)
- Margarita Papatheodoridi
- Sheila Sherlock Liver Unit, Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Giuseppe Mazza
- Sheila Sherlock Liver Unit, Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Massimo Pinzani
- Sheila Sherlock Liver Unit, Institute for Liver and Digestive Health, University College London, London, United Kingdom.
| |
Collapse
|
12
|
Nguyen MP, Jain V, Iansante V, Mitry RR, Filippi C, Dhawan A. Clinical application of hepatocyte transplantation: current status, applicability, limitations, and future outlook. Expert Rev Gastroenterol Hepatol 2020; 14:185-196. [PMID: 32098516 DOI: 10.1080/17474124.2020.1733975] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: Hepatocyte transplantation (HT) is a promising alternative to liver transplantation for the treatment of liver-based metabolic diseases and acute liver failure (ALF). However, shortage of good-quality liver tissues, early cell loss post-infusion, reduced cell engraftment and function restricts clinical application.Areas covered: A comprehensive literature search was performed to cover pre-clinical and clinical HT studies. The review discusses the latest developments to address HT limitations: cell sources from marginal/suboptimal donors to neonatal livers, differentiating pluripotent stem cells into hepatocyte-like cells, in vitro expansion, prevention of immune response to transplanted cells by encapsulation or using innate immunity-inhibiting agents, and enhancing engraftment through partial hepatectomy or irradiation.Expert opinion: To date, published data are highly encouraging specially the alginate-encapsulated hepatocyte treatment of children with ALF. Hepatocyte functions can be further improved through co-culturing with mesenchymal stromal cells. Moreover, ex-vivo genetic correction will enable the use of autologous cells in future personalized medicine.
Collapse
Affiliation(s)
- Minh Phuong Nguyen
- Dhawan Lab. at the Mowat Labs, Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Vandana Jain
- Dhawan Lab. at the Mowat Labs, Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Valeria Iansante
- Dhawan Lab. at the Mowat Labs, Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Ragai R Mitry
- Dhawan Lab. at the Mowat Labs, Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Celine Filippi
- Dhawan Lab. at the Mowat Labs, Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Anil Dhawan
- Dhawan Lab. at the Mowat Labs, Institute of Liver Studies, King's College Hospital, London, United Kingdom
| |
Collapse
|
13
|
Kruitwagen HS, Oosterhoff LA, van Wolferen ME, Chen C, Nantasanti Assawarachan S, Schneeberger K, Kummeling A, van Straten G, Akkerdaas IC, Vinke CR, van Steenbeek FG, van Bruggen LW, Wolfswinkel J, Grinwis GC, Fuchs SA, Gehart H, Geijsen N, Vries RG, Clevers H, Rothuizen J, Schotanus BA, Penning LC, Spee B. Long-Term Survival of Transplanted Autologous Canine Liver Organoids in a COMMD1-Deficient Dog Model of Metabolic Liver Disease. Cells 2020; 9:cells9020410. [PMID: 32053895 PMCID: PMC7072637 DOI: 10.3390/cells9020410] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/30/2022] Open
Abstract
The shortage of liver organ donors is increasing and the need for viable alternatives is urgent. Liver cell (hepatocyte) transplantation may be a less invasive treatment compared with liver transplantation. Unfortunately, hepatocytes cannot be expanded in vitro, and allogenic cell transplantation requires long-term immunosuppression. Organoid-derived adult liver stem cells can be cultured indefinitely to create sufficient cell numbers for transplantation, and they are amenable to gene correction. This study provides preclinical proof of concept of the potential of cell transplantation in a large animal model of inherited copper toxicosis, such as Wilson’s disease, a Mendelian disorder that causes toxic copper accumulation in the liver. Hepatic progenitors from five COMMD1-deficient dogs were isolated and cultured using the 3D organoid culture system. After genetic restoration of COMMD1 expression, the organoid-derived hepatocyte-like cells were safely delivered as repeated autologous transplantations via the portal vein. Although engraftment and repopulation percentages were low, the cells survived in the liver for up to two years post-transplantation. The low engraftment was in line with a lack of functional recovery regarding copper excretion. This preclinical study confirms the survival of genetically corrected autologous organoid-derived hepatocyte-like cells in vivo and warrants further optimization of organoid engraftment and functional recovery in a large animal model of human liver disease.
Collapse
Affiliation(s)
- Hedwig S. Kruitwagen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
- Correspondence: (H.S.K.); (B.S.)
| | - Loes A. Oosterhoff
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Monique E. van Wolferen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Chen Chen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Sathidpak Nantasanti Assawarachan
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Kerstin Schneeberger
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Anne Kummeling
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Giora van Straten
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Ies C. Akkerdaas
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Christel R. Vinke
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Frank G. van Steenbeek
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Leonie W.L. van Bruggen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Jeannette Wolfswinkel
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Guy C.M. Grinwis
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands;
| | - Sabine A. Fuchs
- Division of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands;
| | - Helmuth Gehart
- Hubrecht Institute for Developmental Biology and Stem Cell Research and University Medical Center, Utrecht University, 3584 CT Utrecht, The Netherlands; (H.G.); (H.C.)
| | - Niels Geijsen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
- Hubrecht Institute for Developmental Biology and Stem Cell Research and University Medical Center, Utrecht University, 3584 CT Utrecht, The Netherlands; (H.G.); (H.C.)
| | - Robert G. Vries
- Hubrecht Organoid Technology (HUB), 3584 CT Utrecht, The Netherlands;
| | - Hans Clevers
- Hubrecht Institute for Developmental Biology and Stem Cell Research and University Medical Center, Utrecht University, 3584 CT Utrecht, The Netherlands; (H.G.); (H.C.)
| | - Jan Rothuizen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Baukje A. Schotanus
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Louis C. Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (L.A.O.); (M.E.v.W.); (C.C.); (S.N.A.); (K.S.); (A.K.); (G.v.S.); (I.C.A.); (C.R.V.); (F.G.v.S.); (L.W.L.v.B.); (J.W.); (N.G.); (J.R.); (B.A.S.); (L.C.P.)
- Correspondence: (H.S.K.); (B.S.)
| |
Collapse
|
14
|
Häberle J, Burlina A, Chakrapani A, Dixon M, Karall D, Lindner M, Mandel H, Martinelli D, Pintos-Morell G, Santer R, Skouma A, Servais A, Tal G, Rubio V, Huemer M, Dionisi-Vici C. Suggested guidelines for the diagnosis and management of urea cycle disorders: First revision. J Inherit Metab Dis 2019; 42:1192-1230. [PMID: 30982989 DOI: 10.1002/jimd.12100] [Citation(s) in RCA: 294] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 02/06/2023]
Abstract
In 2012, we published guidelines summarizing and evaluating late 2011 evidence for diagnosis and therapy of urea cycle disorders (UCDs). With 1:35 000 estimated incidence, UCDs cause hyperammonemia of neonatal (~50%) or late onset that can lead to intellectual disability or death, even while effective therapies do exist. In the 7 years that have elapsed since the first guideline was published, abundant novel information has accumulated, experience on newborn screening for some UCDs has widened, a novel hyperammonemia-causing genetic disorder has been reported, glycerol phenylbutyrate has been introduced as a treatment, and novel promising therapeutic avenues (including gene therapy) have been opened. Several factors including the impact of the first edition of these guidelines (frequently read and quoted) may have increased awareness among health professionals and patient families. However, under-recognition and delayed diagnosis of UCDs still appear widespread. It was therefore necessary to revise the original guidelines to ensure an up-to-date frame of reference for professionals and patients as well as for awareness campaigns. This was accomplished by keeping the original spirit of providing a trans-European consensus based on robust evidence (scored with GRADE methodology), involving professionals on UCDs from nine countries in preparing this consensus. We believe this revised guideline, which has been reviewed by several societies that are involved in the management of UCDs, will have a positive impact on the outcomes of patients by establishing common standards, and spreading and harmonizing good practices. It may also promote the identification of knowledge voids to be filled by future research.
Collapse
Affiliation(s)
- Johannes Häberle
- University Children's Hospital Zurich and Children's Research Centre, Zurich, Switzerland
| | - Alberto Burlina
- Division of Inborn Metabolic Disease, Department of Pediatrics, University Hospital Padua, Padova, Italy
| | - Anupam Chakrapani
- Department of Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Marjorie Dixon
- Dietetics, Great Ormond Street Hospital for Children, NHS Trust, London, UK
| | - Daniela Karall
- Clinic for Pediatrics, Division of Inherited Metabolic Disorders, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Lindner
- University Children's Hospital, Frankfurt am Main, Germany
| | - Hanna Mandel
- Institute of Human Genetics and metabolic disorders, Western Galilee Medical Center, Nahariya, Israel
| | - Diego Martinelli
- Division of Metabolism, Bambino Gesù Children's Hospital, Rome, Italy
| | - Guillem Pintos-Morell
- Centre for Rare Diseases, University Hospital Vall d'Hebron, Barcelona, Spain
- CIBERER_GCV08, Research Institute IGTP, Barcelona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | - René Santer
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anastasia Skouma
- Institute of Child Health, Agia Sofia Children's Hospital, Athens, Greece
| | - Aude Servais
- Service de Néphrologie et maladies métaboliques adulte Hôpital Necker 149, Paris, France
| | - Galit Tal
- The Ruth Rappaport Children's Hospital, Rambam Medical Center, Haifa, Israel
| | - Vicente Rubio
- Instituto de Biomedicina de Valencia (IBV-CSIC), Centro de Investigación Biomédica en Red para Enfermedades Raras (CIBERER), Valencia, Spain
| | - Martina Huemer
- University Children's Hospital Zurich and Children's Research Centre, Zurich, Switzerland
- Department of Paediatrics, Landeskrankenhaus Bregenz, Bregenz, Austria
| | | |
Collapse
|
15
|
Kruitwagen HS, Fieten H, Penning LC. Towards Bioengineered Liver Stem Cell Transplantation Studies in a Preclinical Dog Model for Inherited Copper Toxicosis. Bioengineering (Basel) 2019; 6:E88. [PMID: 31557851 PMCID: PMC6955979 DOI: 10.3390/bioengineering6040088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/13/2019] [Accepted: 09/17/2019] [Indexed: 01/20/2023] Open
Abstract
Wilson Disease is a rare autosomal recessive liver disorder in humans. Although its clinical presentation and age of onset are highly variable, hallmarks include signs of liver disease, neurological features and so-called Kayser-Fleischer rings in the eyes of the patient. Hepatic copper accumulation leads to liver disease and eventually to liver cirrhosis. Treatment options include life-long copper chelation therapy and/or decrease in copper intake. Eventually liver transplantations are indicated. Although clinical outcome of liver transplantations is favorable, the lack of suitable donor livers hampers large numbers of transplantations. As an alternative, cell therapies with hepatocytes or liver stem cells are currently under investigation. Stem cell biology in relation to pets is in its infancy. Due to the specific population structure of dogs, canine copper toxicosis is frequently encountered in various dog breeds. Since the histology and clinical presentation resemble Wilson Disease, we combined genetics, gene-editing, and matrices-based stem cell cultures to develop a translational preclinical transplantation model for inherited copper toxicosis in dogs. Here we describe the roadmap followed, starting from the discovery of a causative copper toxicosis mutation in a specific dog breed and culminating in transplantation of genetically-engineered autologous liver stem cells.
Collapse
Affiliation(s)
- Hedwig S Kruitwagen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584CM Utrecht, The Netherlands.
| | - Hille Fieten
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584CM Utrecht, The Netherlands.
| | - Louis C Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584CM Utrecht, The Netherlands.
| |
Collapse
|
16
|
Iansante V, Chandrashekran A, Dhawan A. Cell-based liver therapies: past, present and future. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0229. [PMID: 29786563 DOI: 10.1098/rstb.2017.0229] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2017] [Indexed: 12/16/2022] Open
Abstract
Liver transplantation represents the standard treatment for people with an end-stage liver disease and some liver-based metabolic disorders; however, shortage of liver donor tissues limits its availability. Furthermore, whole liver replacement eliminates the possibility of using native liver as a possible target for future gene therapy in case of liver-based metabolic defects. Cell therapy has emerged as a potential alternative, as cells can provide the hepatic functions and engraft in the liver parenchyma. Various options have been proposed, including human or other species hepatocytes, hepatocyte-like cells derived from stem cells or more futuristic alternatives, such as combination therapies with different cell types, organoids and cell-biomaterial combinations. In this review, we aim to give an overview of the cell therapies developed so far, highlighting preclinical and/or clinical achievements as well as the limitations that need to be overcome to make them fully effective and safe for clinical applications.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Valeria Iansante
- Dhawan Lab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London at King's College Hospital, London SE5 9PJ, UK
| | - Anil Chandrashekran
- Dhawan Lab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London at King's College Hospital, London SE5 9PJ, UK
| | - Anil Dhawan
- Dhawan Lab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London at King's College Hospital, London SE5 9PJ, UK
| |
Collapse
|
17
|
Fujiyoshi J, Yamaza H, Sonoda S, Yuniartha R, Ihara K, Nonaka K, Taguchi T, Ohga S, Yamaza T. Therapeutic potential of hepatocyte-like-cells converted from stem cells from human exfoliated deciduous teeth in fulminant Wilson's disease. Sci Rep 2019; 9:1535. [PMID: 30733544 PMCID: PMC6367569 DOI: 10.1038/s41598-018-38275-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/20/2018] [Indexed: 02/08/2023] Open
Abstract
Wilson’s disease (WD) is an inherited metabolic disease arising from ATPase copper transporting beta gene (ATP7B) mutation. Orthotoropic liver transplantation is the only radical treatment of fulminant WD, although appropriate donors are lacking at the onset of emergency. Given the hepatogenic capacity and tissue-integration/reconstruction ability in the liver of stem cells from human exfoliated deciduous teeth (SHED), SHED have been proposed as a source for curing liver diseases. We hypothesized the therapeutic potential of SHED and SHED-converted hepatocyte-like- cells (SHED-Heps) for fulminant WD. SHED and SHED-Heps were transplanted into WD model Atp7b-mutated Long-Evans Cinnamon (LEC) rats received copper overloading to induce a lethal fulminant liver failure. Due to the superior copper tolerance via ATP7B, SHED-Hep transplantation gave more prolonged life-span of fulminant LEC rats than SHED transplantation. The integrated ATP7B-expressing SHED-Heps showed more therapeutic effects on to restoring the hepatic dysfunction and tissue damages in the recipient liver than the integrated naïve SHED without ATP7B expression. Moreover, SHED-Heps could reduce copper-induced oxidative stress via ATP7B- independent stanniocalcin 1 secretion in the fulminant LEC rats, suggesting a possible role for paracrine effect of the integrated SHED-Heps. Taken together, SHED-Heps offer a potential of functional restoring, bridging, and preventive approaches for treating fulminant WD.
Collapse
Affiliation(s)
- Junko Fujiyoshi
- Department of Pediatrics, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Haruyoshi Yamaza
- Department of Pediatric Dentistry, Kyushu University Graduate School of Dental Science, Fukuoka, 812-8582, Japan
| | - Soichiro Sonoda
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, Fukuoka, 812-8582, Japan
| | - Ratih Yuniartha
- Department of Pediatric Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Kenji Ihara
- Department of Pediatrics, Faculty of Medicine, Oita University, Yuhu, 879-5593, Japan
| | - Kazuaki Nonaka
- Department of Pediatric Dentistry, Kyushu University Graduate School of Dental Science, Fukuoka, 812-8582, Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Takayoshi Yamaza
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, Fukuoka, 812-8582, Japan.
| |
Collapse
|
18
|
Domino Hepatocyte Transplantation: A Therapeutic Alternative for the Treatment of Acute Liver Failure. Can J Gastroenterol Hepatol 2018; 2018:2593745. [PMID: 30065914 PMCID: PMC6051327 DOI: 10.1155/2018/2593745] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/25/2018] [Accepted: 05/31/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is a severe syndrome with an elevated mortality rate, ranging from 40 to 80 %. Currently, liver transplantation is the only definitive treatment for these patients and new therapies aiming to treat ALF include artificial organs implant and stem cells therapy, for example. However, a major limitation of liver donors exists. Living donor liver transplantation (LDLT), split liver transplantation (SLT), and domino liver transplantation (DLT) are some of the available alternatives to treat ALF patients, but these do not reduce the number of patients on waiting lists. Herein, we discuss domino hepatocyte transplantation (DHT) using livers that would not meet transplantation criteria. METHODS We conducted a literature search on PubMed/Medline using acute liver failure, liver transplantation, hepatocyte transplantation, and domino liver transplantation as key words. RESULTS New sources of biochemically functional hepatocytes and therapeutic treatments, in parallel to organ transplantation, may improve liver injury recovery and decrease mortality rates. Moreover, the literature reports hepatocyte transplantation as a therapeutic alternative for organ shortage. However, a major challenge remains for a wide clinical application of hepatocytes therapy, i.e., the availability of sufficient amounts of cells for transplantation. Ideally, hepatocytes isolated from livers rejected for transplantation may be a promising alternative for this problem. CONCLUSION Our review suggests that DHT may be an excellent strategy to increase cell supplies for hepatocyte transplantation.
Collapse
|
19
|
Lee CA, Sinha S, Fitzpatrick E, Dhawan A. Hepatocyte transplantation and advancements in alternative cell sources for liver-based regenerative medicine. J Mol Med (Berl) 2018; 96:469-481. [PMID: 29691598 PMCID: PMC5988761 DOI: 10.1007/s00109-018-1638-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/07/2018] [Accepted: 04/11/2018] [Indexed: 12/16/2022]
Abstract
Human hepatocyte transplantation has been actively perused as an alternative to liver replacement for acute liver failure and liver-based metabolic defects. Current challenges in this field include a limited cell source, reduced cell viability following cryopreservation and poor engraftment of cells into the recipient liver with consequent limited life span. As a result, alternative stem cell sources such as pluripotent stem cells, fibroblasts, hepatic progenitor cells, amniotic epithelial cells and mesenchymal stem/stromal cells (MSCs) can be used to generate induced hepatocyte like cells (HLC) with each technique exhibiting advantages and disadvantages. HLCs may have comparable function to primary human hepatocytes and could offer patient-specific treatment. However, long-term functionality of transplanted HLCs and the potential oncogenic risks of using stem cells have yet to be established. The immunomodulatory effects of MSCs are promising, and multiple clinical trials are investigating their effect in cirrhosis and acute liver failure. Here, we review the current status of hepatocyte transplantation, alternative cell sources to primary human hepatocytes and their potential in liver regeneration. We also describe recent clinical trials using hepatocytes derived from stem cells and their role in improving the phenotype of several liver diseases.
Collapse
Affiliation(s)
- Charlotte A Lee
- Dhawan Lab, Institute of Liver Studies, King's College London at King's College Hospital NHS Foundation trust, London, UK
| | - Siddharth Sinha
- Dhawan Lab, Institute of Liver Studies, King's College London at King's College Hospital NHS Foundation trust, London, UK
| | - Emer Fitzpatrick
- Paediatric Liver GI and Nutrition Centre, King's College London at King's College Hospital NHS Foundation Trust, London, UK
| | - Anil Dhawan
- Paediatric Liver GI and Nutrition Centre, King's College London at King's College Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
20
|
Kohli R, Cortes M, Heaton ND, Dhawan A. Liver transplantation in children: state of the art and future perspectives. Arch Dis Child 2018; 103:192-198. [PMID: 28918383 DOI: 10.1136/archdischild-2015-310023] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022]
Abstract
In this review, we provide a state of the art of liver transplantation in children, as the procedure is now carried out for more than 30 years and most of our paediatric colleagues are managing these patients jointly with liver transplant centres. Our goal for this article is to enhance the understanding of the liver transplant process that a child and his family goes through while explaining the surgical advances and the associated complications that could happen in the immediate or long-term follow-up. We have deliberately introduced the theme that 'liver transplant is a disease' and 'not a cure', to emphasise the need for adherence with immunosuppression, a healthy lifestyle and lifelong medical follow-up.
Collapse
Affiliation(s)
- Rohit Kohli
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Miriam Cortes
- Department of Adult and Pediatric Liver Transplant Surgery, Institute of Liver Studies, King's College Hospital, London, UK
| | - N D Heaton
- Department of Adult and Pediatric Liver Transplant Surgery, Institute of Liver Studies, King's College Hospital, London, UK
| | - Anil Dhawan
- Pediatrics Liver GI and Nutrition Centre and MowatLabs, King's College Hospital, London, UK
| |
Collapse
|
21
|
Iansante V, Mitry RR, Filippi C, Fitzpatrick E, Dhawan A. Human hepatocyte transplantation for liver disease: current status and future perspectives. Pediatr Res 2018; 83:232-240. [PMID: 29149103 DOI: 10.1038/pr.2017.284] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/02/2017] [Indexed: 12/16/2022]
Abstract
Liver transplantation is the accepted treatment for patients with acute liver failure and liver-based metabolic disorders. However, donor organ shortage and lifelong need for immunosuppression are the main limitations to liver transplantation. In addition, loss of the native liver as a target organ for future gene therapy for metabolic disorders limits the futuristic treatment options, resulting in the need for alternative therapeutic strategies. A potential alternative to liver transplantation is allogeneic hepatocyte transplantation. Over the last two decades, hepatocyte transplantation has made the transition from bench to bedside. Standardized techniques have been established for isolation, culture, and cryopreservation of human hepatocytes. Clinical hepatocyte transplantation safety and short-term efficacy have been proven; however, some major hurdles-mainly concerning shortage of donor organs, low cell engraftment, and lack of a long-lasting effect-need to be overcome to widen its clinical applications. Current research is aimed at addressing these problems, with the ultimate goal of increasing hepatocyte transplantation efficacy in clinical applications.
Collapse
Affiliation(s)
- V Iansante
- DhawanLab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, UK
| | - R R Mitry
- DhawanLab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, UK
| | - C Filippi
- DhawanLab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, UK
| | - E Fitzpatrick
- DhawanLab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, UK
| | - A Dhawan
- DhawanLab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, UK
| |
Collapse
|
22
|
Meyburg J, Opladen T, Spiekerkötter U, Schlune A, Schenk JP, Schmidt J, Weitz J, Okun J, Bürger F, Omran TB, Abdoh G, Al Rifai H, Monavari A, Konstantopoulou V, Kölker S, Yudkoff M, Hoffmann GF. Human heterologous liver cells transiently improve hyperammonemia and ureagenesis in individuals with severe urea cycle disorders. J Inherit Metab Dis 2018; 41:81-90. [PMID: 29027067 DOI: 10.1007/s10545-017-0097-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/19/2017] [Accepted: 09/22/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Urea cycle disorders (UCDs) still have a poor prognosis despite several therapeutic advancements. As liver transplantation can provide a cure, liver cell therapy (LCT) might be a new therapeutic option in these patients. METHODS Twelve patients with severe UCDs were included in this prospective clinical trial. Patients received up to six infusions of cryopreserved human heterologous liver cells via a surgically placed catheter in the portal vein. Portal vein pressure, portal vein flow, and vital signs were monitored continuously. Calcineurin inhibitors and steroids were used for immunosuppression. In four patients, ureagenesis was determined with stable isotopes. Number and severity of hyperammonemic events and side effects of immunosuppression were analyzed during an observation period of up to 2 years. RESULTS No study-related mortality was observed. The application catheter dislocated in two children. No significant side effects of catheter application or cell infusion were noted in the other ten patients. The overall incidence of infections did not differ significantly from a historical control group, and no specific side effects of immunosuppression were found. Seven patients were treated per protocol and could be analyzed for efficacy. Severe metabolic crises could be prevented in all of these patients, moderate crises in four of seven. Ureagenesis increased after cell infusion in all patients investigated. CONCLUSIONS We found a favorable safety profile with respect to catheter placement, intraportal liver cell infusion, and immunosuppression. More than half of the children treated per protocol experienced metabolic stabilization and could be safely bridged to liver transplantation.
Collapse
Affiliation(s)
- Jochen Meyburg
- Centre for Child and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, D-69120, Heidelberg, Germany.
| | - Thomas Opladen
- Centre for Child and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, D-69120, Heidelberg, Germany
| | - Ute Spiekerkötter
- Department of General Pediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
- Division of General Pediatrics, University Children's Hospital, Freiburg, Germany
| | - Andrea Schlune
- Department of General Pediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Jens-Peter Schenk
- Division of Pediatric Radiology, Department of Diagnostic and Interventional Radiology, University Hospital, Heidelberg, Germany
| | - Jan Schmidt
- Department of Visceral and Transplant Surgery, University Hospital, Heidelberg, Germany
| | - Jürgen Weitz
- Department of Visceral and Transplant Surgery, University Hospital, Heidelberg, Germany
| | - Jürgen Okun
- Centre for Child and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, D-69120, Heidelberg, Germany
| | - Friederike Bürger
- Centre for Child and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, D-69120, Heidelberg, Germany
| | - Tawfeg Ben Omran
- Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
| | - Ghassan Abdoh
- Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
| | - Hilal Al Rifai
- Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
| | - Ahmad Monavari
- National Centre of Inherited Metabolic Disorders, Dublin, Ireland
| | - Vassiliki Konstantopoulou
- Division of Pulmonology, Allergology, Endocrinology, and Metabolic Medicine, University Children's Hospital, Vienna, Austria
| | - Stefan Kölker
- Centre for Child and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, D-69120, Heidelberg, Germany
| | - Marc Yudkoff
- Division of Metabolic Diseases, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Georg F Hoffmann
- Centre for Child and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, D-69120, Heidelberg, Germany
| |
Collapse
|
23
|
Fagg WS, Liu N, Yang MJ, Cheng K, Chung E, Kim JS, Wu G, Fair J. Magnetic Targeting of Stem Cell Derivatives Enhances Hepatic Engraftment into Structurally Normal Liver. Cell Transplant 2017; 26:1868-1877. [PMID: 29390880 PMCID: PMC5802632 DOI: 10.1177/0963689717737320] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 08/23/2017] [Accepted: 09/01/2017] [Indexed: 12/15/2022] Open
Abstract
Attaining consistent robust engraftment in the structurally normal liver is an obstacle for cellular transplantation. Most experimental approaches to increase transplanted cells' engraftment involve recipient-centered deleterious methods such as partial hepatectomy or irradiation which may be unsuitable in the clinic. Here, we present a cell-based strategy that increases engraftment into the structurally normal liver using a combination of magnetic targeting and proliferative endoderm progenitor (EPs) cells. Magnetic labeling has little effect on cell viability and differentiation, but in the presence of magnetic targeting, it increases the initial dwell time of transplanted EPs into the undamaged liver parenchyma. Consequently, greater cell retention in the liver is observed concomitantly with fewer transplanted cells in the lungs. These highly proliferative cells then significantly increase their biomass over time in the liver parenchyma, approaching nearly 4% of total liver cells 30 d after transplant. Therefore, the cell-based mechanisms of increased initial dwell time through magnetic targeting combined with high rate of proliferation in situ yield significant engraftment in the undamaged liver.
Collapse
Affiliation(s)
- W. Samuel Fagg
- Transplant Division, Department of Surgery, University of Texas Medical Branch Galveston, TX, USA
- Shriners Hospital for Children, University of Texas Medical Branch, Galveston, TX, USA
| | - Naiyou Liu
- Transplant Division, Department of Surgery, University of Texas Medical Branch Galveston, TX, USA
- Shriners Hospital for Children, University of Texas Medical Branch, Galveston, TX, USA
| | - Ming-Jim Yang
- Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Ke Cheng
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eric Chung
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jae-Sung Kim
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gordon Wu
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jeffrey Fair
- Transplant Division, Department of Surgery, University of Texas Medical Branch Galveston, TX, USA
- Shriners Hospital for Children, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
24
|
Squires JE, Soltys KA, McKiernan P, Squires RH, Strom SC, Fox IJ, Soto-Gutierrez A. Clinical Hepatocyte Transplantation: What Is Next? CURRENT TRANSPLANTATION REPORTS 2017; 4:280-289. [PMID: 29732274 DOI: 10.1007/s40472-017-0165-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose of review Significant recent scientific developments have occurred in the field of liver repopulation and regeneration. While techniques to facilitate liver repopulation with donor hepatocytes and different cell sources have been studied extensively in the laboratory, in recent years clinical hepatocyte transplantation (HT) and liver repopulation trials have demonstrated new disease indications and also immunological challenges that will require the incorporation of a fresh look and new experimental approaches. Recent findings Growth advantage and regenerative stimulus are necessary to allow donor hepatocytes to proliferate. Current research efforts focus on mechanisms of donor hepatocyte expansion in response to liver injury/preconditioning. Moreover, latest clinical evidence shows that important obstacles to HT include optimizing engraftment and limited duration of effectiveness, with hepatocytes being lost to immunological rejection. We will discuss alternatives for cellular rejection monitoring, as well as new modalities to follow cellular graft function and near-to-clinical cell sources. Summary HT partially corrects genetic disorders for a limited period of time and has been associated with reversal of ALF. The main identified obstacles that remain to make HT a curative approach include improving engraftment rates, and methods for monitoring cellular graft function and rejection. This review aims to discuss current state-of-the-art in clinical HT and provide insights into innovative approaches taken to overcome these obstacles.
Collapse
Affiliation(s)
- James E Squires
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Kyle A Soltys
- Thomas E. Starzl Transplant Institute, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Patrick McKiernan
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Robert H Squires
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Stephen C Strom
- Karolinska Institutet, Department of Laboratory Medicine, Division of Pathology, Stockholm, Sweden
| | - Ira J Fox
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
25
|
Tatsumi K, Okano T. Hepatocyte Transplantation: Cell Sheet Technology for Liver Cell Transplantation. CURRENT TRANSPLANTATION REPORTS 2017; 4:184-192. [PMID: 28932649 PMCID: PMC5577064 DOI: 10.1007/s40472-017-0156-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Purpose of Review We will review the recent developments of cell sheet technology as a feasible tissue engineering approach. Specifically, we will focus on the technological advancement for engineering functional liver tissue using cell sheet technology, and the associated therapeutic effect of cell sheets for liver diseases, highlighting hemophilia. Recent Findings Cell-based therapies using hepatocytes have recently been explored as a new therapeutic modality for patients with many forms of liver disease. We have developed a cell sheet technology, which allows cells to be harvested in a monolithic layer format. We have succeeded in fabricating functional liver tissues in mice by stacking the cell sheets composed of primary hepatocytes. As a curative measure for hemophilia, we have also succeeded in treating hemophilia mice by transplanting of cells sheets composed of genetically modified autologous cells. Summary Tissue engineering using cell sheet technology provides the opportunity to create new therapeutic options for patients with various types of liver diseases.
Collapse
Affiliation(s)
- Kohei Tatsumi
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511 Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan.,Cell Sheet Tissue Engineering Center and Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112 USA
| |
Collapse
|
26
|
El Baz H, Demerdash Z, Kamel M, Atta S, Salah F, Hassan S, Hammam O, Khalil H, Meshaal S, Raafat I. Transplant of Hepatocytes, Undifferentiated Mesenchymal Stem Cells, and In Vitro Hepatocyte-Differentiated Mesenchymal Stem Cells in a Chronic Liver Failure Experimental Model: A Comparative Study. EXP CLIN TRANSPLANT 2017; 16:81-89. [PMID: 28585911 DOI: 10.6002/ect.2016.0226] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Liver transplant is the cornerstone line of treatment for chronic liver diseases; however, the long list of complications and obstacles stand against this operation. Searching for new modalities for treatment of chronic liver illness is a must. In the present research, we aimed to compare the effects of transplant of undifferentiated human mesenchymal stem cells, in vitro differentiated mesenchymal stem cells, and adult hepatocytes in an experimental model of chronic liver failure. MATERIALS AND METHODS Undifferentiated human cord blood mesenchymal stem cells were isolated, pro-pagated, and characterized by morphology, gene expression analysis, and flow cytometry of surface markers and in vitro differentiated into hepatocyte-like cells. Rat hepatocytes were isolated by double perfusion technique. An animal model of chronic liver failure was developed, and undifferentiated human cord blood mesenchymal stem cells, in vitro hepato-genically differentiated mesenchymal stem cells, or freshly isolated rat hepatocytes were transplanted into a CCL4 cirrhotic experimental model. Animals were killed 3 months after transplant, and liver functions and histopathology were assessed. RESULTS Compared with the cirrhotic control group, the 3 cell-treated groups showed improved alanine aminotransferase, aspartate aminotransferase, albumin, and bilirubin levels, with best results shown in the hepatocyte-treated group. Histopathologic examination of the treated groups showed improved fibrosis, with best results obtained in the undifferentiated mesenchymal stem cell-treated group. CONCLUSIONS Both adult hepatocytes and cord blood mesenchymal stem cells proved to be promising candidates for cell-based therapy in liver regeneration on an experimental level. Improved liver function was evident in the hepatocyte-treated group, and fibrosis control was more evident in the undifferentiated mesenchymal stem cell-treated group.
Collapse
Affiliation(s)
- Hanan El Baz
- From the Immunology Department, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
mTOR Inhibition Suppresses Posttransplant Alloantibody Production Through Direct Inhibition of Alloprimed B Cells and Sparing of CD8+ Antibody-Suppressing T cells. Transplantation 2017; 100:1898-906. [PMID: 27362313 DOI: 10.1097/tp.0000000000001291] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND De novo alloantibodies (donor-specific antibody) contribute to antibody-mediated rejection and poor long-term graft survival. Because the development of donor-specific antibody is associated with early graft loss of cell transplants and reduced long-term survival of solid organ transplants, we hypothesized that conventional immunosuppressives, calcineurin inhibitors (CNi), and mammalian target of rapamycin inhibitors (mTORi), may not be as effective for suppression of humoral alloimmunity as for cell-mediated immunity. METHODS Wild-type or CD8-depleted mice were transplanted with allogeneic hepatocytes. Recipients were treated with mTORi and/or CNi and serially monitored for alloantibody and graft survival. The direct effect of mTORi and CNi on alloprimed B cell function was investigated in Rag1 mice adoptively transferred with alloprimed IgG1 B cells. The efficacy of mTORi and/or CNi to suppress CD8-mediated cytotoxicity of IgG1 B cells was evaluated in in vitro and in vivo cytotoxicity assays. RESULTS Mammalian target of rapamycin inhibitors, but not CNi, reduced alloantibody production in transplant recipients, directly suppressed alloantibody production by alloprimed IgG1 B cells and delayed graft rejection in both low and high alloantibody producers. Combination treatment with mTORi and CNi resulted in loss of the inhibitory effect observed for mTORi monotherapy in part due to CNi suppression of CD8 T cells which downregulate alloantibody production (CD8 TAb-supp cells). CONCLUSIONS Our data support that mTORi is a potent inhibitor of humoral immunity through suppression of alloprimed B cells and preservation of CD8 TAb-supp cells. In contrast, alloantibody is readily detected in CNi-treated recipients because CNi does not suppress alloprimed B cells and interferes with downregulatory CD8 TAb-supp cells.
Collapse
|
28
|
Wadström J, Ericzon BG, Halloran PF, Bechstein WO, Opelz G, Serón D, Grinyó J, Loupy A, Kuypers D, Mariat C, Clancy M, Jardine AG, Guirado L, Fellström B, O'Grady J, Pirenne J, O'Leary JG, Aluvihare V, Trunečka P, Baccarani U, Neuberger J, Soto-Gutierrez A, Geissler EK, Metzger M, Gray M. Advancing Transplantation: New Questions, New Possibilities in Kidney and Liver Transplantation. Transplantation 2017; 101 Suppl 2S:S1-S41. [PMID: 28125449 DOI: 10.1097/tp.0000000000001563] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jonas Wadström
- 1 Karolinska University Hospital, Stockholm, Sweden. 2 Karolinska Institutet, Stockholm, Sweden. 3 Alberta Transplant Applied Genomics Centre, Edmonton, Canada. 4 Frankfurt University Hospital and Clinics, Frankfurt, Germany. 5 University of Heidelberg, Heidelberg, Germany. 6 Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain. 7 Red de Investigación Renal (REDinREN), Instituto Carlos III, Madrid, Spain. 8 Hospital Universitari de Bellvitge, University of Barcelona, Spain. 9 Service de Néphrologie-Transplantation, Hôpital Necker, Paris, France. 10 University Hospitals Leuven, Leuven, Belgium. 11 University Hospital of Saint-Etienne, Jean Monnet University, France. 12 Western Infirmary, Glasgow, United Kingdom. 13 Fundació Puigvert, Barcelona, Spain. 14 University of Uppsala, Uppsala, Sweden. 15 King's College Hospital, London, United Kingdom. 16 Baylor University Medical Center Dallas, Dallas, TX. 17 Transplantcenter, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic. 18 Department of Medical and Biological Sciences, University Hospital of Udine, Udine, Italy. 19 Liver Unit, Queen Elizabeth Hospital, Birmingham, United Kingdom. 20 Directorate of Organ Donation and Transplantation, NHS Blood and Transplant, Bristol, United Kingdom. 21 Department of Pathology, University of Pittsburgh, Pittsburgh, PA. 22 Experimental Surgery, University Hospital Regensburg, University of Regensburg, Regensburg, Germany. 23 Ahead of Time GmbH, Starnberg, Germany. 24 Better Value Healthcare, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Orthotopic liver transplantation remains the only proven cure for end-stage liver failure. Despite significant advances in the field, the clinical demand for donor organs far outweighs the supply. Hepatocyte transplantation has been proposed as an alternative approach to whole liver transplant in select diseases. Several international centers have reported experimental trials of human hepatocyte transplantation in acute liver failure and liver-based metabolic disorders. This chapter provides an introduction to hepatocyte transplantation from both a technical and clinical perspective. We will also focus on the special needs of pediatric patients, since historically the majority of clinical hepatocyte transplants have involved infants and children.
Collapse
|
30
|
Lee PC, Truong B, Vega-Crespo A, Gilmore WB, Hermann K, Angarita SA, Tang JK, Chang KM, Wininger AE, Lam AK, Schoenberg BE, Cederbaum SD, Pyle AD, Byrne JA, Lipshutz GS. Restoring Ureagenesis in Hepatocytes by CRISPR/Cas9-mediated Genomic Addition to Arginase-deficient Induced Pluripotent Stem Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e394. [PMID: 27898091 PMCID: PMC5155330 DOI: 10.1038/mtna.2016.98] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022]
Abstract
Urea cycle disorders are incurable enzymopathies that affect nitrogen metabolism and typically lead to hyperammonemia. Arginase deficiency results from a mutation in Arg1, the enzyme regulating the final step of ureagenesis and typically results in developmental disabilities, seizures, spastic diplegia, and sometimes death. Current medical treatments for urea cycle disorders are only marginally effective, and for proximal disorders, liver transplantation is effective but limited by graft availability. Advances in human induced pluripotent stem cell research has allowed for the genetic modification of stem cells for potential cellular replacement therapies. In this study, we demonstrate a universally-applicable CRISPR/Cas9-based strategy utilizing exon 1 of the hypoxanthine-guanine phosphoribosyltransferase locus to genetically modify and restore arginase activity, and thus ureagenesis, in genetically distinct patient-specific human induced pluripotent stem cells and hepatocyte-like derivatives. Successful strategies restoring gene function in patient-specific human induced pluripotent stem cells may advance applications of genetically modified cell therapy to treat urea cycle and other inborn errors of metabolism.
Collapse
Affiliation(s)
- Patrick C Lee
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Brian Truong
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Agustin Vega-Crespo
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - W Blake Gilmore
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kip Hermann
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Stephanie Ak Angarita
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jonathan K Tang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Katherine M Chang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Austin E Wininger
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Alex K Lam
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Benjamen E Schoenberg
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Stephen D Cederbaum
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, California, USA.,Semel Institute for Neuroscience, UCLA, Los Angeles, California, USA
| | - April D Pyle
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - James A Byrne
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Gerald S Lipshutz
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, California, USA.,Semel Institute for Neuroscience, UCLA, Los Angeles, California, USA.,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
31
|
Alternative Cell Sources to Adult Hepatocytes for Hepatic Cell Therapy. Methods Mol Biol 2016; 1506:17-42. [PMID: 27830543 DOI: 10.1007/978-1-4939-6506-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Adult hepatocyte transplantation is limited by scarce availability of suitable donor liver tissue for hepatocyte isolation. New cell-based therapies are being developed to supplement whole-organ liver transplantation, to reduce the waiting-list mortality rate, and to obtain more sustained and significant metabolic correction. Fetal livers and unsuitable neonatal livers for organ transplantation have been proposed as potential useful sources of hepatic cells for cell therapy. However, the major challenge is to use alternative cell sources for transplantation that can be derived from reproducible methods. Different types of stem cells with hepatic differentiation potential are eligible for generating large numbers of functional hepatocytes for liver cell therapy to treat degenerative disorders, inborn hepatic metabolic diseases, and organ failure. Clinical trials are designed to fully establish the safety profile of such therapies and to define target patient groups and standardized protocols.
Collapse
|
32
|
Lee CA, Dhawan A, Smith RA, Mitry RR, Fitzpatrick E. Instant Blood-Mediated Inflammatory Reaction in Hepatocyte Transplantation: Current Status and Future Perspectives. Cell Transplant 2016; 25:1227-36. [PMID: 26996786 DOI: 10.3727/096368916x691286] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hepatocyte transplantation (HT) is emerging as a promising alternative to orthotopic liver transplantation (OLT) in patients with certain liver-based metabolic disease and acute liver failure. Hepatocytes are generally infused into the portal venous system, from which they migrate into the liver cell plates of the native organ. One of the major hurdles to the sustained success of this therapy is early cell loss, with up to 70% of hepatocytes lost immediately following infusion. This is largely thought to be due to the instant blood-mediated inflammatory reaction (IBMIR), resulting in the activation of complement and coagulation pathways. Transplanted hepatocytes produce and release tissue factor (TF), which activates the coagulation pathway, leading to the formation of thrombin and fibrin clots. Thrombin can further activate a number of complement proteins, leading to the activation of the membrane attack complex (MAC) and subsequent hepatocyte cell death. Inflammatory cells including granulocytes, monocytes, Kupffer cells, and natural killer (NK) cells have been shown to cluster around transplanted hepatocytes, leading to their rapid clearance shortly after transplantation. Current research aims to improve cell engraftment and prevent early cell loss. This has been proven successful in vitro using pharmacological interventions such as melagatran, low-molecular-weight dextran sulphate, and N-acetylcysteine (NAC). Effective inhibition of IBMIR would significantly improve hepatocyte engraftment, proliferation, and function, providing successful treatment for patients with liver-based metabolic diseases.
Collapse
Affiliation(s)
- Charlotte A Lee
- Institute of Liver Studies, King's College London, School of Life Sciences and Medicine, King's College Hospital, London, UK
| | | | | | | | | |
Collapse
|
33
|
Cantz T, Sharma AD, Ott M. Concise review: cell therapies for hereditary metabolic liver diseases-concepts, clinical results, and future developments. Stem Cells 2016; 33:1055-62. [PMID: 25524146 DOI: 10.1002/stem.1920] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/23/2014] [Accepted: 11/07/2014] [Indexed: 12/11/2022]
Abstract
The concept of cell-based therapies for inherited metabolic liver diseases has been introduced for now more than 40 years in animal experiments, but controlled clinical data in humans are still not available. In the era of dynamic developments in stem cell science, the "right" cell for transplantation is considered as an important key for successful treatment. Do we aim to transplant mature hepatocytes or do we consider the liver as a stem/progenitor-driven organ and replenish the diseased liver with genetically normal stem/progenitor cells? Although conflicting results from cell tracing and transplantation experiments have recently emerged about the existence and role of stem/progenitor cells in the liver, their overall contribution to parenchymal cell homeostasis and tissue repair is limited. Accordingly, engraftment and repopulation efficacies of extrahepatic and liver-derived stem/progenitor cell types are considered to be lower compared to mature hepatocytes. On the basis of these results, we will discuss the current clinical cell transplantation programs for inherited metabolic liver diseases and future developments in liver cell therapy.
Collapse
Affiliation(s)
- Tobias Cantz
- Translational Hepatology and Stem Cell Biology, Cluster of Excellence REBIRTH, Hannover, Germany; Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | | |
Collapse
|
34
|
Dhawan A. Clinical human hepatocyte transplantation: Current status and challenges. Liver Transpl 2015; 21 Suppl 1:S39-44. [PMID: 26249755 DOI: 10.1002/lt.24226] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/27/2015] [Accepted: 07/29/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Anil Dhawan
- Department of Pediatric Hepatology, Cell Therapy Unit, National Institute for Health Research/Wellcome Trust King's Clinical Research Facility, King's College Hospital, London, UK
| |
Collapse
|
35
|
Zhou H, Liu H, Ezzelarab M, Schmelzer E, Wang Y, Gerlach J, Gridelli B, Cooper DKC. Experimental hepatocyte xenotransplantation--a comprehensive review of the literature. Xenotransplantation 2015; 22:239-48. [PMID: 25950141 PMCID: PMC4519403 DOI: 10.1111/xen.12170] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 04/18/2015] [Indexed: 12/11/2022]
Abstract
Hepatocyte transplantation (Tx) is a potential therapy for certain diseases of the liver, including hepatic failure. However, there is a limited supply of human livers as a source of cells and, after isolation, human hepatocytes can be difficult to expand in culture, limiting the number available for Tx. Hepatocytes from other species, for example, the pig, have therefore emerged as a potential alternative source. We searched the literature through the end of 2014 to assess the current status of experimental research into hepatocyte xenoTx. The literature search identified 51 reports of in vivo cross-species Tx of hepatocytes in a variety of experimental models. Most studies investigated the Tx of human (n = 23) or pig (n = 19) hepatocytes. No studies explored hepatocytes from genetically engineered pigs. The spleen was the most common site of Tx (n = 23), followed by the liver (through the portal vein [n = 6]) and peritoneal cavity (n = 19). In 47 studies (92%), there was evidence of hepatocyte engraftment and function across a species barrier. The data provided by this literature search strengthen the hypothesis that xenoTx of hepatocytes is feasible and potentially successful as a clinical therapy for certain liver diseases, including hepatic failure. By excluding vascular structures, hepatocytes isolated from genetically engineered pig livers may address some of the immunological problems of xenoTx.
Collapse
Affiliation(s)
- Huidong Zhou
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Kidney Transplantation, Second Affiliated Hospital of the University of South China, Heng(1)yang, Hunan, China
| | - Hong Liu
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of General Surgery, First Hospital of Shanxi Medical University, ShanXi, China
| | - Mohamed Ezzelarab
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eva Schmelzer
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yi Wang
- Center for Kidney Transplantation, Second Affiliated Hospital of the University of South China, Heng(1)yang, Hunan, China
| | - Jörg Gerlach
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruno Gridelli
- Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - David K. C. Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
36
|
Tolosa L, López S, Pareja E, Donato MT, Myara A, Nguyen TH, Castell JV, Gómez-Lechón MJ. Human neonatal hepatocyte transplantation induces long-term rescue of unconjugated hyperbilirubinemia in the Gunn rat. Liver Transpl 2015; 21:801-11. [PMID: 25821167 DOI: 10.1002/lt.24121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 03/06/2015] [Accepted: 03/14/2015] [Indexed: 12/12/2022]
Abstract
Crigler-Najjar type 1 disease is a rare inherited metabolic disease characterized by high levels of unconjugated bilirubin due to the complete absence of hepatic uridine diphosphoglucuronate-glucuronosyltransferase activity. Hepatocyte transplantation (HT) has been proposed as an alternative treatment for Crigler-Najjar syndrome, but it is still limited by the quality and the low engraftment and repopulation ability of the cells used. Because of their attachment capability and expression of adhesion molecules as well as the higher proportion of hepatic progenitor cells, neonatal hepatocytes may have an advantage over adult cells. Adult or neonatal hepatocytes were transplanted into Gunn rats, a model for Crigler-Najjar disease. Engraftment and repopulation were studied and compared by immunofluorescence (IF). Additionally, the serum bilirubin levels, the presence of bilirubin conjugates in rat serum, and the expression of uridine diphosphate glucuronosyltransferase 1 family polypeptide A1 (UGT1A1) in rat liver samples were also analyzed. Here we show that neonatal HT results in long-term correction in Gunn rats. In comparison with adult cells, neonatal cells showed better engraftment and repopulation capability 3 days and 6 months after transplantation, respectively. Bilirubinemia decreased in the transplanted animals during the whole experimental follow-up (6 months). Bilirubin conjugates were also present in the serum of the transplanted animals. Western blots and IF confirmed the presence and expression of UGT1A1 in the liver. This work is the first to demonstrate the advantage of using neonatal hepatocytes for the treatment of Crigler-Najjar in vivo.
Collapse
Affiliation(s)
- Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Silvia López
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Eugenia Pareja
- Unidad de Cirugía Hepatobiliopancreática y Transplante Hepático, Hospital La Fe, Valencia, Spain
| | - María Teresa Donato
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Fondo de Investigaciones Sanitarias, Barcelona, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Anne Myara
- Service de Biologie, Groupe Hospitalier Saint Joseph, Paris, France
| | - Tuan Huy Nguyen
- INSERM Unités Mixtes de Recherche en Santé 1064, Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | - José Vicente Castell
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Fondo de Investigaciones Sanitarias, Barcelona, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - María José Gómez-Lechón
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Fondo de Investigaciones Sanitarias, Barcelona, Spain
| |
Collapse
|
37
|
Brassier A, Gobin S, Arnoux JB, Valayannopoulos V, Habarou F, Kossorotoff M, Servais A, Barbier V, Dubois S, Touati G, Barouki R, Lesage F, Dupic L, Bonnefont JP, Ottolenghi C, De Lonlay P. Long-term outcomes in Ornithine Transcarbamylase deficiency: a series of 90 patients. Orphanet J Rare Dis 2015; 10:58. [PMID: 25958381 PMCID: PMC4443534 DOI: 10.1186/s13023-015-0266-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 04/13/2015] [Indexed: 12/11/2022] Open
Abstract
Background The principal aim of this study was to investigate the long-term outcomes of a large cohort of patients with ornithine transcarbamylase deficiency (OTCD) who were followed up at a single medical center. Methods We analyzed clinical, biochemical and genetic parameters of 90 patients (84 families, 48 males and 42 females) with OTCD between 1971 and 2011. Results Twenty-seven patients (22 boys, 5 girls) had a neonatal presentation; 52 patients had an “intermediate” late-onset form of the disease (21 boys, 31 girls) that was revealed between 1 month and 16 years; and 11 patients (5 boys, 6 girls) presented in adulthood (16 to 55 years). Patients with a neonatal presentation had increased mortality (90% versus 13% in late-onset forms) and peak plasma ammonium (mean value: 960 μmol/L versus 500 μmol/L) and glutamine (mean value: 4110 μmol/L versus 1000 μmol/L) levels at diagnosis. All of the neonatal forms displayed a greater number of acute decompensations (mean value: 6.2/patient versus 2.5 and 1.4 in infants and adults, respectively). In the adult group, some patients even recently died at the time of presentation during their first episode of coma. Molecular analyses identified a deleterious mutation in 59/68 patients investigated. Single base substitutions were detected more frequently than deletions (69% and 12%, respectively), with a recurrent mutation identified in the late-onset groups (pArg40 His; 13% in infants, 57% in adults); inherited mutations represented half of the cases. The neurological score did not differ significantly between the patients who were alive in the neonatal or late-onset groups and did not correlate with the peak ammonia and plasma glutamine concentrations at diagnosis. However, in late-onset forms of the disease, ammonia levels adjusted according to the glutamine/citrulline ratio at diagnosis were borderline predictors of low IQ (p = 0.12 by logistic regression; area under the receiver operating characteristic curve of 76%, p <0.05). Conclusions OTCD remains a severe disease, even in adult-onset patients for whom the prevention of metabolic decompensations is crucial. The combination of biochemical markers warrants further investigations to provide additional prognostic information regarding the neurological outcomes of patients with OTCD.
Collapse
Affiliation(s)
- Anais Brassier
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France. .,Université Paris Descartes, Institut Imagine, Hôpital Necker-Enfants Malades, APHP, Paris, France.
| | - Stephanie Gobin
- Service de Génétique, Paris, France. .,Université Paris Descartes, Institut Imagine, Hôpital Necker-Enfants Malades, APHP, Paris, France.
| | - Jean Baptiste Arnoux
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France.
| | - Vassili Valayannopoulos
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France.
| | - Florence Habarou
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France. .,Service de Biochimie Métabolique, Paris, France. .,Université Paris Descartes, Institut Imagine, Hôpital Necker-Enfants Malades, APHP, Paris, France.
| | - Manoelle Kossorotoff
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France.
| | - Aude Servais
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France.
| | - Valerie Barbier
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France.
| | - Sandrine Dubois
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France.
| | - Guy Touati
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France.
| | - Robert Barouki
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France. .,Service de Biochimie Métabolique, Paris, France. .,Université Paris Descartes, Institut Imagine, Hôpital Necker-Enfants Malades, APHP, Paris, France.
| | - Fabrice Lesage
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France. .,Service de Réanimation pédiatrique, Paris, France.
| | - Laurent Dupic
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France. .,Service de Réanimation pédiatrique, Paris, France.
| | - Jean Paul Bonnefont
- Service de Génétique, Paris, France. .,Université Paris Descartes, Institut Imagine, Hôpital Necker-Enfants Malades, APHP, Paris, France.
| | - Chris Ottolenghi
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France. .,Service de Biochimie Métabolique, Paris, France. .,Université Paris Descartes, Institut Imagine, Hôpital Necker-Enfants Malades, APHP, Paris, France.
| | - Pascale De Lonlay
- Reference Center of Inherited Metabolic Diseases and units of metabolism and neurology, 149 rue de Sèvres, 75015, Paris, France. .,Université Paris Descartes, Institut Imagine, Hôpital Necker-Enfants Malades, APHP, Paris, France.
| |
Collapse
|
38
|
Gramignoli R, Vosough M, Kannisto K, Srinivasan RC, Strom SC. Clinical hepatocyte transplantation: practical limits and possible solutions. Eur Surg Res 2015; 54:162-177. [PMID: 25633583 DOI: 10.1159/000369552] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/04/2014] [Indexed: 01/05/2025]
Abstract
Since the first human hepatocyte transplants (HTx) in 1992, clinical studies have clearly established proof of principle for this therapy as a treatment for patients with acquired or inherited liver disease. Although major accomplishments have been made, there are still some specific limitations to this technology, which, if overcome, could greatly enhance the efficacy and implementation of this therapy. Here, we describe what in our view are the most significant obstacles to the clinical application of HTx and review the solutions currently proposed. The obstacles of significance include the limited number and quality of liver tissues as a cell source, the lack of clinical grade reagents, quality control evaluation of hepatocytes prior to transplantation, hypothermic storage of cells prior to transplantation, preconditioning treatments to enhance engraftment and proliferation of donor cells, tracking or monitoring cells after transplantation, and the optimal immunosuppression protocols for transplant recipients.
Collapse
Affiliation(s)
- Roberto Gramignoli
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
39
|
Affiliation(s)
- Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Yonsei University, Wonju College of Medicine, Wonju, Korea
| | - Soon Koo Baik
- Cell Therapy and Tissue Engineering Center, Yonsei University, Wonju College of Medicine, Wonju, Korea
- Department of Internal Medicine, Yonsei University, Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
40
|
Hansel MC, Gramignoli R, Skvorak KJ, Dorko K, Marongiu F, Blake W, Davila J, Strom SC. The history and use of human hepatocytes for the treatment of liver diseases: the first 100 patients. CURRENT PROTOCOLS IN TOXICOLOGY 2014; 62:14.12.1-23. [PMID: 25378242 PMCID: PMC4343212 DOI: 10.1002/0471140856.tx1412s62] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Orthotopic liver transplantation remains the only curative treatment for many end-stage liver diseases, yet the number of patients receiving liver transplants remains limited by the number of organs available for transplant. There is a need for alternative therapies for liver diseases. The transplantation of isolated hepatocytes (liver cells) has been used as an experimental therapy for liver disease in a limited number of cases. Recently, the 100th case of hepatocyte transplantation was reported. This review discusses the history of the hepatocyte transplant field, the major discoveries that supported and enabled the first hepatocyte transplants, and reviews the cases and outcomes of the first 100 clinical transplants. Some of the problems that limit the application or efficacy of hepatocyte transplantation are discussed, as are possible solutions to these problems. In conclusion, hepatocyte transplants have proven effective particularly in cases of metabolic liver disease where reversal or amelioration of the characteristic symptoms of the disease is easily quantified. However, no patients have been completely corrected of a metabolic liver disease for a significant amount of time by hepatocyte transplantation alone. It is likely that future developments in new sources of cells for transplantation will be required before this cellular therapy can be fully implemented and available for large numbers of patients.
Collapse
Affiliation(s)
- Marc C Hansel
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| | | | | | | | | | | | | | | |
Collapse
|
41
|
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
|
42
|
Matsuura K, Utoh R, Nagase K, Okano T. Cell sheet approach for tissue engineering and regenerative medicine. J Control Release 2014; 190:228-39. [DOI: 10.1016/j.jconrel.2014.05.024] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/10/2014] [Accepted: 05/13/2014] [Indexed: 01/06/2023]
|
43
|
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
|
44
|
Kruitwagen HS, Spee B, Schotanus BA. Hepatic progenitor cells in canine and feline medicine: potential for regenerative strategies. BMC Vet Res 2014; 10:137. [PMID: 24946932 PMCID: PMC4089933 DOI: 10.1186/1746-6148-10-137] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 12/31/2013] [Indexed: 12/17/2022] Open
Abstract
New curative therapies for severe liver disease are urgently needed in both the human and veterinary clinic. It is important to find new treatment modalities which aim to compensate for the loss of parenchymal tissue and to repopulate the liver with healthy hepatocytes. A prime focus in regenerative medicine of the liver is the use of adult liver stem cells, or hepatic progenitor cells (HPCs), for functional recovery of liver disease. This review describes recent developments in HPC research in dog and cat and compares these findings to experimental rodent studies and human pathology. Specifically, the role of HPCs in liver regeneration, key components of the HPC niche, and HPC activation in specific types of canine and feline liver disease will be reviewed. Finally, the potential applications of HPCs in regenerative medicine of the liver are discussed and a potential role is suggested for dogs as first target species for HPC-based trials.
Collapse
Affiliation(s)
- Hedwig S Kruitwagen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584 CM, Utrecht, The Netherlands.
| | | | | |
Collapse
|
45
|
Filippi C, Dhawan A. Current status of human hepatocyte transplantation and its potential for Wilson's disease. Ann N Y Acad Sci 2014; 1315:50-5. [DOI: 10.1111/nyas.12386] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Celine Filippi
- NIHR Biomedical Research Centre at Guy's and St Thomas’ NHS Foundation Trust and King's College London; London United Kingdom
- Hepatocyte Biology and Transplantation Group; Institute of Liver Studies; King's College London; London United Kingdom
| | - Anil Dhawan
- Hepatocyte Biology and Transplantation Group; Institute of Liver Studies; King's College London; London United Kingdom
- Paediatric Liver; GI and Nutrition Centre; King's College Hospital; London United Kingdom
| |
Collapse
|
46
|
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
|
47
|
Vogel KR, Kennedy AA, Whitehouse LA, Gibson KM. Therapeutic hepatocyte transplant for inherited metabolic disorders: functional considerations, recent outcomes and future prospects. J Inherit Metab Dis 2014; 37:165-76. [PMID: 24085555 PMCID: PMC3975709 DOI: 10.1007/s10545-013-9656-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/12/2013] [Accepted: 09/13/2013] [Indexed: 12/18/2022]
Abstract
The applications, outcomes and future strategies of hepatocyte transplantation (HTx) as a corrective intervention for inherited metabolic disease (IMD) are described. An overview of HTx in IMDs, as well as preclinical evaluations in rodent and other mammalian models, is summarized. Current treatments for IMDs are highlighted, along with short- and long-term outcomes and the potential for HTx to supplement or supplant these treatments. Finally, the advantages and disadvantages of HTx are presented, highlighted by long-term challenges with interorgan engraftment and expansion of transplanted cells, in addition to the future prospects of stem cell transplants. At present, the utility of HTx is represented by the potential to bridge patients with life-threatening liver disease to organ transplantation, especially as an adjuvant intervention where severe organ shortages continue to pose challenges.
Collapse
Affiliation(s)
- Kara R Vogel
- Section of Clinical Pharmacology, College of Pharmacy, Washington State University, SAC 525M, P.O. Box 1495, Spokane, WA, 99210-1495, USA
| | | | | | | |
Collapse
|
48
|
Ribes-Koninckx C, Ibars EP, Calzado Agrasot MÁ, Bonora-Centelles A, Miquel BP, Vila Carbó JJ, Aliaga ED, Pallardó JM, Gómez-Lechón MJ, Castell JV. Clinical outcome of hepatocyte transplantation in four pediatric patients with inherited metabolic diseases. Cell Transplant 2013; 21:2267-82. [PMID: 23231960 DOI: 10.3727/096368912x637505] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Hepatocyte transplantation (HT) has become an effective therapy for patients with metabolic inborn errors. We report the clinical outcome of four children with metabolic inborn errors that underwent HT, describing the cell infusion protocol and the metabolic outcome of transplanted patients. Cryopreserved hepatocytes were used as this allows scheduling of treatments. Functional competence (viability, cell attachment, major cytochrome P450 and UDP-glucuronosyltransferase 1A1 activities, and urea synthesis) and microbiological safety of cell batches were assessed prior to clinical use. Four pediatric patients with liver metabolic diseases [ornithine transcarbamylase (OTC) deficiency, Crigler-Najjar (CNI) syndrome, glycogen storage disease Ia (GSD-Ia), and tyrosinemia type I (TYR-I)] underwent HT. Indication for HT was based on severity of disease, deterioration of quality of life, and benefits for the patients, with the ultimate goal to improve their clinical status whenever liver transplantation (LT) was not indicated or to bridge LT. Cells were infused into the portal vein while monitoring portal flow. The protocol included antibiotic prophylaxis and immunosuppressant therapy. After HT, analytical data on the disease were obtained. The OTC-deficient patient showed a sustained decrease in plasma ammonia levels and increased urea production after HT. Further cell infusions could not be administered given a fatal nosocomial fungus sepsis 2 weeks after the last HT. The CNI and GSD-Ia patients improved their clinical status after HT. They displayed reduced serum bilirubin levels (by ca. 50%) and absence of hypoglycaemic episodes, respectively. In both cases, the HT contributed to stabilize their clinical status as LT was not indicated. In the infant with TYR-I, HT stabilized temporarily the biochemical parameters, resulting in the amelioration of his clinical status while diagnosis of the disease was unequivocally confirmed by full gene sequencing. In this patient, HT served as a bridge therapy to LT.
Collapse
Affiliation(s)
- Carmen Ribes-Koninckx
- Paediatric Gastroenterology and Hepatology Unit, University La Fe Hospital, Valencia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Shigeta T, Hsu HC, Enosawa S, Matsuno N, Kasahara M, Matsunari H, Umeyama K, Watanabe M, Nagashima H. Transgenic pig expressing the red fluorescent protein kusabira-orange as a novel tool for preclinical studies on hepatocyte transplantation. Transplant Proc 2013; 45:1808-10. [PMID: 23769049 DOI: 10.1016/j.transproceed.2013.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 01/15/2013] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Research on hepatocyte transplantation as an alternative or supplementary treatment for liver transplantation is progressing. However, to advance to clinical trials, confidence in the technique must be established and its safety must be validated by conducting experiments using animals of comparable sizes to humans, such as pigs. We used transgenic pigs expressing red fluorescence protein for investigating the distribution and survival of transplanted cells. MATERIALS AND METHODS Donor hepatocytes were isolated from transgenic Kusabira-Orange (KO)-expressing pigs (age, 41 days; weight, 10 kg) created by in vitro fertilization using sperm from a transgenic-cloned KO pig by Matsunari et al. and ova from a domestic pig. The hepatocyte transplant recipients were the nontransgenic, KO-negative littermates. In these recipient pigs, double lumen cannulae were inserted into the supramesenteric veins to access the hepatic portal region. KO-positive donor hepatocytes from the transgenic male pig were isolated using collagenase perfusion. Hepatocytes (1 × 10(9) cells) were transplanted through the cannula. For estimating allogeneic immunogenicity, full-thickness skin (3 × 3 cm) from the same donor was grafted orthotopically on the neck region of the recipients. Immunosuppressive treatment was not implemented. The recipient pigs were humanely killed at 7 and 39 days after transplantation, and the organs were harvested, including the lungs, heart, liver, pancreas, and kidneys. RESULTS Strong red fluorescence was detected in both the parenchymal and nonparenchymal hepatocytes of the transgenic male donor pig by fluorescent microscopy. Transplanted cells were detected in the liver and lung of the recipient pigs at 7 days after perfusion. Hepatocytes remained in the liver and lung of recipients on day 39, with lower numbers than that on day 7. CONCLUSION Transgenic pigs expressing the fluorescent protein KO serve as a useful model of cell transplantation in preclinical studies.
Collapse
Affiliation(s)
- T Shigeta
- Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hepatocyte labeling with ⁹⁹mTc-GSA: a potential non-invasive technique for tracking cell transplantation. Int J Artif Organs 2013; 35:450-7. [PMID: 22476879 DOI: 10.5301/ijao.5000096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2012] [Indexed: 11/20/2022]
Abstract
BACKGROUND Hepatocyte transplantation is a promising alternative to orthotopic liver transplantation, however, the fate of transplanted hepatocytes is not well defined. ⁹⁹mTc-galactosyl-serum albumin (⁹⁹mTc-GSA) is a clinical scintigraphic agent which is specifically taken up by the hepatocyte asialoglycoprotein receptor (ASGPR). AIMS To investigate labeling of fresh and cryopreserved human hepatocytes and fresh rat hepatocytes in vitro using ⁹⁹mTc-GSA. METHODS Human and rat hepatocytes were isolated from liver tissue by collagenase perfusion. The ASGPR were characterized using immunohistochemistry and RT-PCR. Hepatocytes were incubated with ⁹⁹mTc-GSA in suspension at 4°C and 37°C. Cell viability and function was determined using cell mitochondrial dehydrogenase (MTS) and sulphorhodamine B (SRB) assays. RESULTS Fresh and cryopreserved human hepatocytes expressed the ASGPR. Incubation of hepatocytes in suspension with ⁹⁹mTc-GSA reduced the viability of hepatocytes, but this was similar to unlabeled control cells. Greater loss of viability was seen on incubation at 37°C compared to 4°C, but there was a significantly greater uptake of ⁹⁹mTc-GSA at the physiological temperature (6.6 ± SE 0.6-fold increase, p<0.05) consistent with ASGPR-mediated endocytosis. MTS and SRB assays were not significantly affected by labeling with ⁹⁹mTc-GSA in all three cell types. A mean of 18.5% of the radioactivity was released over 120 min when ⁹⁹mTc-GSA -labeled hepatocytes were shaken in vitro at 37°C. CONCLUSIONS Human and rat hepatocytes can be labeled with ⁹⁹mTc-GSA, which may have potential application for in vivo imaging after hepatocyte transplantation.
Collapse
|