1
|
Guo L, Zhang L, Xu H, Yu P, Wang Z, Lu D, Chen M, Wu B. Diurnal hepatic CYP3A11 contributes to chronotoxicity of the pyrrolizidine alkaloid retrorsine in mice. Xenobiotica 2021; 51:1019-1028. [PMID: 34311664 DOI: 10.1080/00498254.2021.1950867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
1. Retrorsine (RTS) is a pyrrolizidine alkaloid (distributed in many medicinal plants) that has significant hepatotoxicity. Here, we aimed to determine the daily variations in RTS hepatotoxicity (chronotoxicity) in mice, and to investigate the role of metabolism in generating RTS chronotoxicity.2. Acute toxicity and pharmacokinetic studies were performed with mice after RTS administration at different times of the day. Hepatotoxicity was assessed by measuring plasma ALT (alanine aminotransferase) and AST (aspartate aminotransferase) levels. mRNA and proteins were determined by qPCR and Western blotting, respectively. Time-dependent in vitro metabolism of RTS was assessed by using mouse liver microsomes.3. We found that RTS toxicity was more severe in the dark phase (zeitgeber time 14 or ZT14 and ZT18) than in the light phase (ZT2 and ZT6). This chronotoxicity was associated with a dosing time difference in the systemic exposures of RTS and a pyrrolic ester metabolite (a cause of hepatotoxicity, measured by the levels of pyrrole-GSH conjugate and pyrrole-protein adducts due to a high chemical reactivity). Moreover, the CYP3A11 (a major enzyme for RTS bioactivation) inhibitor ketoconazole decreased the production of pyrrole-GSH conjugate and abrogated diurnal rhythm in RTS metabolism. In addition, E4bp4 (a circadian regulator of Cyp3a11) ablation abolished the rhythm of CYP3A11 expression and abrogated the dosing time-dependency of RTS toxicity.4. In conclusion, RTS chronotoxicity in mice was attributed to time-varying hepatic metabolism regulated by the circadian clock. Our findings have implications for reducing pyrrolizidine alkaloid-induced toxicity via a chronotherapeutic approach.
Collapse
Affiliation(s)
- Li Guo
- Department of Critical Care Medicine, Zhongshan Torch Development Zone Hospital, Zhongshan, China
| | - Li Zhang
- Jinan University, Guangzhou, China
| | | | - Pei Yu
- Jinan University, Guangzhou, China
| | - Zhigang Wang
- Department of Intensive Care Unit, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Danyi Lu
- Jinan University, College of Pharmacy, Guangzhou, China
| | - Min Chen
- Jinan University, Guangzhou, China
| | - Baojian Wu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
Cai X, Li H, Kaplan DE. Murine hepatoblast-derived liver tumors resembling human combined hepatocellular-cholangiocarcinoma with stem cell features. Cell Biosci 2020; 10:38. [PMID: 32190288 PMCID: PMC7071781 DOI: 10.1186/s13578-020-00395-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Combined hepatocellular-cholangiocarcinoma (CHC) is a primary hepatic malignancy with heterogeneously combined histological features of putative hepatic progenitor cells (HPC) origin. We describe a mouse model that exhibits the heterogenous histological and phenotypic finding similar to human CHC. METHODS We injected hepatoblasts isolated from p53-/- C57BL/6 mice into syngeneic wild-type pre-conditioned C57BL/6 mice. We confirmed that p53-/- murine hepatoblasts act as tumor-initiating cells (TICs) that generate CHC both in situ and within metastases. For comparative pathological study, 8 human cases of CHC with stem cell features were recruited by immunohistochemistry and multicolor fluorescence immunostaining. RESULTS We identified corresponding areas in murine tumors matching each WHO criteria-described subtype of human CHC. In both murine and human tumors, HPC-like cells in tumor nests and associated stem cell features/traits are suggested histologically to be the progenitor origin of the cancer. CONCLUSIONS The pathological characteristics of murine tumors recapitulate human CHC with stem cell features. These data provide additional comparative pathological evidence that CHC with stem cell features originate from HPCs and validate a model to study this cancer type in vivo.
Collapse
Affiliation(s)
- Xiong Cai
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, 3400 Civic Center Drive, PCAM GI 7S, Philadelphia, PA 19104-6145 USA
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, 430022 China
| | - Heli Li
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - David E. Kaplan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, 3400 Civic Center Drive, PCAM GI 7S, Philadelphia, PA 19104-6145 USA
| |
Collapse
|
3
|
Czekaj P, Król M, Limanówka Ł, Michalik M, Lorek K, Gramignoli R. Assessment of animal experimental models of toxic liver injury in the context of their potential application as preclinical models for cell therapy. Eur J Pharmacol 2019; 861:172597. [PMID: 31408648 DOI: 10.1016/j.ejphar.2019.172597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/04/2019] [Accepted: 08/08/2019] [Indexed: 02/06/2023]
Abstract
Preclinical animal models allow to study development and progression of several diseases, including liver disorders. These studies, for ethical reasons and medical limits, are impossible to carry out in human patients. At the same time, such experimental models constitute an important source of knowledge on pathomechanisms for drug- and virus-induced hepatotoxicity, both acute and chronic. Carbon tetrachloride, D-Galactosamine, and retrorsine are xenobiotics that can be used in immunocompetent animal models of hepatotoxicity, where chemical-intoxicated livers present histological features representative of human viruses-related infection. A prolonged derangement into liver architecture and functions commonly lead to cirrhosis, eventually resulting in hepatocellular carcinoma. In human, orthotopic liver transplantation commonly resolve most the problems related to cirrhosis. However, the shortage of donors does not allow all the patients in the waiting list to receive an organ on time. A promising alternative treatment for acute and chronic liver disease has been advised in liver cell transplantation, but the limited availability of hepatocytes for clinical approaches, in addition to the immunosuppressant regiment required to sustain cellular long-term engraftment have been encouraging the use of alternative cell sources. A recent effective source of stem cells have been recently identified in the human amnion membrane. Human amnion epithelial cells (hAEC) have been preclinically tested and proven sufficient to rescue immunocompetent rodents lethally intoxicated with drugs. The adoption of therapeutic procedures based on hAEC transplant in immunocompetent recipients affected by liver diseases, as well as patients with immune-related disorders, may constitute a successful new alternative therapy in regenerative medicine.
Collapse
Affiliation(s)
- Piotr Czekaj
- Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland.
| | - Mateusz Król
- Students Scientific Society, Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland.
| | - Łukasz Limanówka
- Students Scientific Society, Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland
| | - Marcin Michalik
- Students Scientific Society, Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland
| | - Katarzyna Lorek
- Students Scientific Society, Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland
| | - Roberto Gramignoli
- Department of Laboratory Medicine (LABMED), H5, Division of Pathology, Karolinska Institutet, Alfred Nobels Allé 8, 14152, Huddinge, Sweden.
| |
Collapse
|
4
|
Alwahsh SM, Rashidi H, Hay DC. Liver cell therapy: is this the end of the beginning? Cell Mol Life Sci 2018; 75:1307-1324. [PMID: 29181772 PMCID: PMC5852182 DOI: 10.1007/s00018-017-2713-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/08/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022]
Abstract
The prevalence of liver diseases is increasing globally. Orthotopic liver transplantation is widely used to treat liver disease upon organ failure. The complexity of this procedure and finite numbers of healthy organ donors have prompted research into alternative therapeutic options to treat liver disease. This includes the transplantation of liver cells to promote regeneration. While successful, the routine supply of good quality human liver cells is limited. Therefore, renewable and scalable sources of these cells are sought. Liver progenitor and pluripotent stem cells offer potential cell sources that could be used clinically. This review discusses recent approaches in liver cell transplantation and requirements to improve the process, with the ultimate goal being efficient organ regeneration. We also discuss the potential off-target effects of cell-based therapies, and the advantages and drawbacks of current pre-clinical animal models used to study organ senescence, repopulation and regeneration.
Collapse
Affiliation(s)
- Salamah M Alwahsh
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| | - Hassan Rashidi
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
5
|
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
|
6
|
Yuan RH, Ogawa A, Ogawa E, Neufeld D, Zhu L, Shafritz DA. p27Kip1 Inactivation Provides a Proliferative Advantage to Transplanted Hepatocytes in DPPIV/Rag2 Double Knockout Mice after Repeated Host Liver Injury. Cell Transplant 2017; 12:907-19. [PMID: 14763511 DOI: 10.3727/000000003771000147] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Studies were conducted to develop a new DPPIV–/–/Rag2–/– mouse model for hepatocyte transplantation by allogeneic and xenogeneic cells and to compare the proliferative capacity of p27 null hepatocytes versus normal hepatocytes in this system. Dipeptidyl peptidase IV (DPPIV) gene knockout mice, in which wild-type (wt) DPPIV+ donor hepatocytes can be readily identified by enzyme histochemistry, were bred with Rag2 null mice to prepare immunotolerant DPPIV–/–/Rag2–/– double knockout mice. DPPIV–/–/Rag–/– mice were transplanted with wt hepatocytes or p27 null mouse hepatocytes, which show enhanced cell cycle activity due to disruption of the Kip1 cyclin kinase inhibitor gene, and liver repopulation was assessed under nonproliferative versus proliferative experimental conditions. After their initial engraftment, transplanted wt hepatocytes did not proliferate in untreated livers or increase significantly in response to an acute liver regenerative stimulus. p27 null hepatocytes engrafted with the same efficiency as wt hepatocytes, but showed a noticeable, although not statistically significant, increase in proliferation in response to partial hepatectomy or acute CCl4 administration. Repeated treatments with CCl4 substantially increased proliferation and liver repopulation by p27 null hepatocytes but not by wt hepatocytes. These results suggest that p27 gene inactivation does not overcome proliferative restrictions imposed on hepatocytes by the normal liver, but that after repeated episodes of toxic liver injury, the augmented proliferative capacity of p27 null hepatocytes leads to significant liver repopulation compared with wt hepatocytes. These properties of p27-deficient hepatocytes could prove useful as a target for liver repopulation in patients with intermittent or a low level of chronic liver injury.
Collapse
Affiliation(s)
- Ray-Hwang Yuan
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | |
Collapse
|
7
|
Hamooda M. Hepatocyte transplantation in children with liver cell failure. Electron Physician 2016; 8:3096-3101. [PMID: 27957309 PMCID: PMC5133034 DOI: 10.19082/3096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/07/2016] [Indexed: 12/22/2022] Open
Abstract
Patients with hepatic failure and liver-based metabolic disorders require management which is both costly and complex. Hepatocyte transplantation has been very encouraging as an alternative to organ transplantation for liver disease treatment, and studies in rodents, show that transplants involving isolated liver cells can reverse hepatic failure, and correct various metabolic deficiencies of the liver. This 2016 review is based on a literature search using PubMed including original articles, reviews, cases and clinical guidelines. The search terms were “hepatocyte transplantation”, “liver transplantation”, “liver cell failure”, “metabolic liver disorders”, “orthotropic liver transplantation”, “hepatocytes” and “stem cell transplantation”. The goal of this review is to summarize the significance of hepatocyte transplantation, the sources of hepatocytes and the barriers of hepatocyte transplantation using a detailed review of literature. Our review shows that treatment of patients with liver disease by hepatocyte transplantation has expanded exponentially, especially for patients suffering from liver-based metabolic disorders. Once hepatocyte transplantation has been shown to effectively replace organ transplantation for a portion of patients with life-threatening liver metabolic diseases and those with liver failure it will make cell therapy effective and available for a broad population of patients with liver disorders.
Collapse
Affiliation(s)
- Mohamed Hamooda
- MRCPCH, Paediatrics Specialty Registrar, West Yorkshire and the Humber, United Kingdom
| |
Collapse
|
8
|
Liver-specific knockout of arginase-1 leads to a profound phenotype similar to inducible whole body arginase-1 deficiency. Mol Genet Metab Rep 2016; 9:54-60. [PMID: 27761413 PMCID: PMC5065044 DOI: 10.1016/j.ymgmr.2016.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/05/2016] [Indexed: 12/27/2022] Open
Abstract
Arginase-1 (Arg1) converts arginine to urea and ornithine in the distal step of the urea cycle in liver. We previously generated a tamoxifen-inducible Arg1 deficient mouse model (Arg1-Cre) that disrupts Arg1 expression throughout the whole body and leads to lethality ≈ 2 weeks after gene disruption. Here, we evaluate if liver-selective Arg1 loss is sufficient to recapitulate the phenotype observed in global Arg1 knockout mice, as well as to gauge the effectiveness of gene delivery or hepatocyte transplantation to rescue the phenotype. Liver-selective Arg1 deletion was induced by using an adeno-associated viral (AAV)-thyroxine binding globulin (TBG) promoter-Cre recombinase vector administered to Arg1 “floxed” mice; Arg1fl/fl). An AAV vector expressing an Arg1-enhanced green fluorescent protein (Arg1-eGFP) transgene was used for gene delivery, while intrasplenic injection of wild-type (WT) C57BL/6 hepatocytes after partial hepatectomy was used for cell delivery to “rescue” tamoxifen-treated Arg1-Cre mice. The results indicate that liver-selective loss of Arg1 (> 90% deficient) leads to a phenotype resembling the whole body knockout of Arg1 with lethality ≈ 3 weeks after Cre-induced gene disruption. Delivery of Arg1-eGFP AAV rescues more than half of Arg1 global knockout male mice (survival > 4 months) but a significant proportion still succumb to the enzyme deficiency even though liver expression and enzyme activity of the fusion protein reach levels observed in WT animals. Significant Arg1 enzyme activity from engrafted WT hepatocytes into knockout livers can be achieved but not sufficient for rescuing the lethal phenotype. This raises a conundrum relating to liver-specific expression of Arg1. On the one hand, loss of expression in this organ appears to be both necessary and sufficient to explain the lethal phenotype of the genetic disorder in mice. On the other hand, gene and cell-directed therapies suggest that rescue of extra-hepatic Arg1 expression may also be necessary for disease correction. Further studies are needed in order to illuminate the detailed mechanisms for pathogenesis of Arg1-deficiency.
Collapse
|
9
|
Yang Y, Lin X, Lu X, Luo G, Zeng T, Tang J, Jiang F, Li L, Cui X, Huang W, Hou G, Chen X, Ouyang Q, Tang S, Sun H, Chen L, Gonzalez FJ, Wu M, Cong W, Chen L, Wang H. Interferon-microRNA signalling drives liver precancerous lesion formation and hepatocarcinogenesis. Gut 2016; 65:1186-201. [PMID: 26860770 PMCID: PMC6624432 DOI: 10.1136/gutjnl-2015-310318] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 01/12/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Precancerous lesion, a well-established histopathologically premalignant tissue with the highest risk for tumourigenesis, develops preferentially from activation of DNA damage checkpoint and persistent inflammation. However, little is known about the mechanisms by which precancerous lesions are initiated and their physiological significance. DESIGN Laser capture microdissection was used to acquire matched normal liver, precancerous lesion and tumour tissues. miR-484(-/-), Ifnar1(-/-) and Tgfbr2(△hep) mice were employed to determine the critical role of the interferon (IFN)-microRNA pathway in precancerous lesion formation and tumourigenesis. RNA immunoprecipitation (RIP), pull-down and chromatin immunoprecipitation (ChIP) assays were applied to explore the underlying mechanisms. RESULTS miR-484 is highly expressed in over 88% liver samples clinically. DEN-induced precancerous lesions and hepatocellular carcinoma were dramatically impaired in miR-484(-/-) mice. Mechanistically, ectopic expression of miR-484 initiates tumourigenesis and cell malignant transformation through synergistic activation of the transforming growth factor-β/Gli and nuclear factor-κB/type I IFN pathways. Specific acetylation of H3K27 is indispensable for basal IFN-induced continuous transcription of miR-484 and cell transformation. Convincingly, formation of precancerous lesions were significantly attenuated in both Tgfbr2(△hep) and Ifnar1(-/-) mice. CONCLUSIONS These findings demonstrate a new protumourigenic axis involving type I IFN-microRNA signalling, providing a potential therapeutic strategy to manipulate or reverse liver precancerous lesions and tumourigenesis.
Collapse
Affiliation(s)
- Yingcheng Yang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China,Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Ximeng Lin
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China,Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Xinyuan Lu
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Guijuan Luo
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China,National Center for Liver Cancer, Shanghai, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Tang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China,National Center for Liver Cancer, Shanghai, China
| | - Feng Jiang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Liang Li
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China,National Center for Liver Cancer, Shanghai, China
| | - Xiuliang Cui
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China,National Center for Liver Cancer, Shanghai, China
| | - Wentao Huang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Guojun Hou
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Xin Chen
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Qing Ouyang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Shanhua Tang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Huanlin Sun
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mengchao Wu
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Wenming Cong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Lei Chen
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China,National Center for Liver Cancer, Shanghai, China,Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hongyang Wang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China,National Center for Liver Cancer, Shanghai, China,State Key Laboratory for Oncogenes and Related Genes, Cancer Institute of RenJi Hospital, Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|
10
|
Yanagida A, Mizuno N, Yamazaki Y, Kato-Itoh M, Umino A, Sato H, Ito K, Yamaguchi T, Nakauchi H, Kamiya A. Investigation of bipotent differentiation of hepatoblasts using inducible diphtheria toxin receptor-transgenic mice. Hepatol Res 2016; 46:816-28. [PMID: 26584962 DOI: 10.1111/hepr.12622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 02/08/2023]
Abstract
AIM Hepatic progenitor cells, called hepatoblasts, are highly proliferative and exhibit bipotential differentiation into hepatocytes and cholangiocytes in the fetal liver. Thus, they are the ideal source for transplantation therapy. Although several studies have been performed in vitro, the molecular mechanisms regulating hepatoblast differentiation in vivo following transplantation remain poorly understood. The aim of this study was to investigate an in vivo model to analyze hepatoblast bipotency and proliferative ability. METHODS Hepatic transplantation model using Cre-inducible diphtheria toxin receptor-transgenic mice (iDTR), and albafpCre mice expressing Cre under the control of albumin and α-fetoprotein (AFP) regulatory elements were established. Fresh hepatoblasts were transplanted into diphtheria toxin (DT)-injected iDTRalbafpCre mice and we analyzed their differentiation and proliferation abilities by immunostaining and gene expression profiles. RESULTS Fresh hepatoblasts transplanted into DT-injected iDTRalbafpCre mice engrafted and differentiated into both hepatocytes and cholangiocytes. Additionally, the number of engrafted hepatoblast-derived hepatocytes increased following partial hepatectomy and serial DT injections. Expression levels of hepatic functional genes in transplanted hepatoblast-derived hepatocytes were similar to that of normal hepatocytes. CONCLUSION In our iDTRalbafpCre transplantation model, fresh hepatoblasts could differentiate into hepatocytes and cholangiocytes. In addition, these donor cells were induced to proliferate by the following liver injury stimulation. This result suggests that this model is valuable for investigating hepatoblast differentiation pathways in vivo.
Collapse
Affiliation(s)
- Ayaka Yanagida
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Naoak Mizuno
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yuji Yamazaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Megumi Kato-Itoh
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ayumi Umino
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hideyuki Sato
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Keiichi Ito
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, USA
| | - Tomoyuki Yamaguchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA
| | - Akihide Kamiya
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
11
|
Abstract
Hepatic progenitor cells isolated from fetal liver can be transplanted into recipient mice to reconstitute an organ system. Retroviral infection can be used to introduce putative oncogenes or short-hairpin RNAs (shRNAs) targeting putative tumor suppressors into the cells; reconstituted mice then develop livers that are chimeric for cells bearing a specific alteration. This approach has the advantage of examining tumorigenesis on a largely wild-type background (if only a subset of cells are infected), a situation that more accurately parallels the human situation. Additionally, tumor development occurs within the appropriate native microenvironment. Here, we describe the isolation of hepatic progenitor cells, as well as the reconstitution and tumor monitoring of recipient mice.
Collapse
Affiliation(s)
- Lars Zender
- Division of Molecular Oncology of Solid Tumors, Department of Internal Medicine I, University Hospital, 72076 Tübingen, Germany
| | - Michael Hemann
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
12
|
Li J, Chanrion M, Sawey E, Wang T, Chow E, Tward A, Su Y, Xue W, Lucito R, Zender L, Lowe SW, Bishop JM, Powers S. Reciprocal interaction of Wnt and RXR-α pathways in hepatocyte development and hepatocellular carcinoma. PLoS One 2015; 10:e0118480. [PMID: 25738607 PMCID: PMC4349704 DOI: 10.1371/journal.pone.0118480] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 01/14/2015] [Indexed: 11/29/2022] Open
Abstract
Genomic analysis of human hepatocellular carcinoma (HCC) is potentially confounded by the differentiation state of the hepatic cell-of-origin. Here we integrated genomic analysis of mouse HCC (with defined cell-of-origin) along with normal development. We found a major shift in expression of Wnt and RXR-α pathway genes (up and down, respectively) coincident with the transition from hepatoblasts to hepatocytes. A combined Wnt and RXR-α gene signature categorized HCCs into two subtypes (high Wnt, low RXR-α and low Wnt, high RXR-α), which matched cell-of-origin in mouse models and the differentiation state of human HCC. Suppression of RXR-α levels in hepatocytes increased Wnt signaling and enhanced tumorigenicity, whereas ligand activation of RXR-α achieved the opposite. These results corroborate that there are two main HCC subtypes that correspond to the degree of hepatocyte differentation and that RXR-α, in part via Wnt signaling, plays a key functional role in the hepatocyte-like subtype and potentially could serve as a selective therapeutic target.
Collapse
Affiliation(s)
- Jinyu Li
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Maia Chanrion
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Eric Sawey
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Tim Wang
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Edward Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Aaron Tward
- G. W. Hooper Foundation and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, United States of America
| | - Yi Su
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Wen Xue
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, United States of America
| | - Robert Lucito
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Lars Zender
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, United States of America
| | - Scott W. Lowe
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, United States of America
| | - J. Michael Bishop
- G. W. Hooper Foundation and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, United States of America
| | - Scott Powers
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, United States of America
- * E-mail:
| |
Collapse
|
13
|
Huch M, Gehart H, van Boxtel R, Hamer K, Blokzijl F, Verstegen MMA, Ellis E, van Wenum M, Fuchs SA, de Ligt J, van de Wetering M, Sasaki N, Boers SJ, Kemperman H, de Jonge J, Ijzermans JNM, Nieuwenhuis EES, Hoekstra R, Strom S, Vries RRG, van der Laan LJW, Cuppen E, Clevers H. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell 2015; 160:299-312. [PMID: 25533785 PMCID: PMC4313365 DOI: 10.1016/j.cell.2014.11.050] [Citation(s) in RCA: 1114] [Impact Index Per Article: 111.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 09/21/2014] [Accepted: 11/21/2014] [Indexed: 02/08/2023]
Abstract
Despite the enormous replication potential of the human liver, there are currently no culture systems available that sustain hepatocyte replication and/or function in vitro. We have shown previously that single mouse Lgr5+ liver stem cells can be expanded as epithelial organoids in vitro and can be differentiated into functional hepatocytes in vitro and in vivo. We now describe conditions allowing long-term expansion of adult bile duct-derived bipotent progenitor cells from human liver. The expanded cells are highly stable at the chromosome and structural level, while single base changes occur at very low rates. The cells can readily be converted into functional hepatocytes in vitro and upon transplantation in vivo. Organoids from α1-antitrypsin deficiency and Alagille syndrome patients mirror the in vivo pathology. Clonal long-term expansion of primary adult liver stem cells opens up experimental avenues for disease modeling, toxicology studies, regenerative medicine, and gene therapy.
Collapse
Affiliation(s)
- Meritxell Huch
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands.
| | - Helmuth Gehart
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Ruben van Boxtel
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Karien Hamer
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Francis Blokzijl
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC-University Medical Center, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Ewa Ellis
- Unit for Transplantation Surgery, Department of CLINTEC, Karolinska Institute, Karolinska University Hospital Huddinge, Hälsovägen, Flemingsberg, SE-141 86 Stockholm, Sweden
| | - Martien van Wenum
- Surgical Laboratory, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Sabine A Fuchs
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Joep de Ligt
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Marc van de Wetering
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands; Hubrecht Organoid Technology (HUB), Uppsalalaan 8, 3584CT, Utrecht, the Netherlands
| | - Nobuo Sasaki
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Susanne J Boers
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Hans Kemperman
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Jeroen de Jonge
- Department of Surgery, Erasmus MC-University Medical Center, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Jan N M Ijzermans
- Department of Surgery, Erasmus MC-University Medical Center, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Edward E S Nieuwenhuis
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Ruurdtje Hoekstra
- Surgical Laboratory, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Stephen Strom
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Alfred Nobels Alle 8, F 56 141-86 Stockholm, Sweden
| | - Robert R G Vries
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands; Hubrecht Organoid Technology (HUB), Uppsalalaan 8, 3584CT, Utrecht, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Edwin Cuppen
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Hans Clevers
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, CancerGenomics.nl, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands.
| |
Collapse
|
14
|
Nagahama Y, Sone M, Chen X, Okada Y, Yamamoto M, Xin B, Matsuo Y, Komatsu M, Suzuki A, Enomoto K, Nishikawa Y. Contributions of hepatocytes and bile ductular cells in ductular reactions and remodeling of the biliary system after chronic liver injury. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3001-12. [PMID: 25193593 DOI: 10.1016/j.ajpath.2014.07.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 07/01/2014] [Accepted: 07/10/2014] [Indexed: 12/18/2022]
Abstract
Mature hepatocytes are suggested to possess a capacity for bile ductular transdifferentiation, but whether and how hepatocytes contribute to ductular reaction in chronic liver diseases has not been elucidated. We examined whether mouse hepatocytes can transdifferentiate into bile ductular cells in vitro, using a three-dimensional collagen gel culture method, and in vivo, using a liver repopulation model in which β-galactosidase-positive hepatocytes from Alb-Cre × ROSA26R mice were transplanted into the liver of wild-type mice. We further examined the relative contribution of intrinsic hepatocytes in ductular reaction in a hepatocyte lineage-tracing model using Mx1-Cre × ROSA26R mice treated with polyinosinic-polycytidylic acid. Within collagen gels, hepatocytes exhibited branching morphogenesis associated with the emergence of bile duct-like phenotype. In the liver repopulation model, many β-galactosidase-positive, hepatocyte-derived bile ductular structures were identified; these markedly increased after liver injury. In Mx1-Cre × ROSA26R mice, relatively minor but significant contributions of hepatocyte-derived bile ductules were observed in both periportal and centrilobular ductular reaction. As the centrilobular ductular reaction progressed, the portal ducts or ductules migrated toward the injured area and joined with hepatocyte-derived ductules, leaving the portal tract without biliary structures. We conclude that hepatocytes and bile ducts or ductules are important sources of ductular reaction and that the intrahepatic biliary system undergoes remarkable remodeling in response to chronic liver injury.
Collapse
Affiliation(s)
- Yasuharu Nagahama
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan; Fujii Memorial Research Institute, Otsuka Pharmaceutical Co., Ltd., Otsu, Japan
| | - Masayuki Sone
- Fujii Memorial Research Institute, Otsuka Pharmaceutical Co., Ltd., Otsu, Japan
| | - Xi Chen
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yoko Okada
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Masahiro Yamamoto
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Bing Xin
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yasuhiro Matsuo
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Mikiko Komatsu
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Akira Suzuki
- Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.
| |
Collapse
|
15
|
Navarro-Alvarez N, Yang YG. Lack of CD47 on donor hepatocytes promotes innate immune cell activation and graft loss: a potential barrier to hepatocyte xenotransplantation. Cell Transplant 2014; 23:345-354. [PMID: 23394628 PMCID: PMC3751988 DOI: 10.3727/096368913x663604] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We have previously shown that interspecies incompatibility of CD47 plays an important role in triggering rejection of xenogeneic hematopoietic cells by macrophages. However, whether CD47 incompatibility also induces rejection of nonhematopoietic cellular xenografts remains unknown. Herein, we have addressed this question in a mouse model of hepatocyte transplantation in which CD47(-/-) hepatocytes were used to resemble xenografts for CD47 incompatibility. We show that intrasplenic transplantation of CD47(-/-), but not wild-type (WT) hepatocytes, into partially hepatectomized syngeneic WT mice resulted in a rapid increase in Mac-1(+) cells with an activation phenotype (i.e., Mac-1(+)CD14(+) and Mac-1(+)CD16/32(high)), compared to nontransplant controls. In addition, CD47(-/-) hepatocytes were more severely damaged than WT hepatocytes as indicated by the greater AST and ALT serum levels in these mice. Furthermore, long-term donor hepatocyte survival and liver repopulation were observed in mice receiving WT hepatocytes, whereas CD47(-/-) hepatocytes were completely rejected within 2 weeks. These results suggest that CD47 on donor hepatocytes prevents recipient myeloid innate immune cell activation, hence aiding in graft survival after hepatocyte transplantation. Thus, CD47 incompatibility is likely to present an additional barrier to hepatocyte xenotransplantation.
Collapse
Affiliation(s)
- Nalu Navarro-Alvarez
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Yong-Guang Yang
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
16
|
Li YH, Kan WLT, Li N, Lin G. Assessment of pyrrolizidine alkaloid-induced toxicity in an in vitro screening model. JOURNAL OF ETHNOPHARMACOLOGY 2013; 150:560-567. [PMID: 24045176 DOI: 10.1016/j.jep.2013.09.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/30/2013] [Accepted: 09/05/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pyrrolizidine alkaloids (PAs) are a group of heterocyclic phytotoxins present in a wide range of plants. The consumption of PA-containing medicinal herbs or PA-contaminated foodstuffs has long been reported to cause human hepatotoxicity. However, the degrees of hepatotoxicity of different PAs are unknown, which makes it difficult to determine a universal threshold of toxic dose of individual PAs for safe regulation of PA-containing natural products. The aim of the present study is to develop a simple and convenient in vitro model to assess the hepatotoxicity of different PAs. MATERIAL AND METHODS Six common cytotoxicity assays were used to evaluate the hepatotoxicity of different PAs in human hepatocellular carcinoma HepG2 cells. RESULTS The combination of MTT and bromodeoxyuridine incorporation (BrdU) assays demonstrated to be a suitable method to evaluate the toxic potencies of various PAs in HepG2 cells, and the results indicated that otonecine-type PA (clivorine: IC₂₀=0.013 ± 0.004 mM (MTT), 0.066 ± 0.031 mM (BrdU)) exhibited significantly higher cytotoxic and anti-proliferative effects than retronecine-type PA (retrorsine: IC₂₀=0.27 ± 0.07 mM (MTT), 0.19 ± 0.03 mM (BrdU)). While as expected, the known less toxic platyphylline-type PA (platyphylline: IC₂₀=0.85 ± 0.11 mM (MTT), 1.01 ± 0.40 mM (BrdU)) exhibited significantly less toxicity. The different cytotoxic and anti-proliferative potencies of various PAs in the same retronecine-type could also be discriminated by using the combined MTT and BrdU assays. In addition, the developed assays were further utilized to test alkaloid extract of Gynura segetum, a senecionine and seneciphylline-containing herb, the overall cytotoxicity of two PAs in the extract was comparable to that of these two PAs tested individually. CONCLUSION Using the developed in vitro model, the cytotoxicity of different PAs and the extract of a PA-containing herb were investigated in parallel in one system, and their different hepatotoxic potencies were determined and directly compared for the first time. The results suggested that the developed model has a great potential to be applied for the quick screening of the toxicity of PAs and PA-containing natural products.
Collapse
Affiliation(s)
- Yan Hong Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| | | | | | | |
Collapse
|
17
|
He G, Dhar D, Nakagawa H, Font-Burgada J, Ogata H, Jiang Y, Shalapour S, Seki E, Yost SE, Jepsen K, Frazer KA, Harismendy O, Hatziapostolou M, Iliopoulos D, Suetsugu A, Hoffman RM, Tateishi R, Koike K, Karin M. Identification of liver cancer progenitors whose malignant progression depends on autocrine IL-6 signaling. Cell 2013; 155:384-96. [PMID: 24120137 PMCID: PMC4015514 DOI: 10.1016/j.cell.2013.09.031] [Citation(s) in RCA: 366] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 06/04/2013] [Accepted: 09/19/2013] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a slowly developing malignancy postulated to evolve from premalignant lesions in chronically damaged livers. However, it was never established that premalignant lesions actually contain tumor progenitors that give rise to cancer. Here, we describe isolation and characterization of HCC progenitor cells (HcPCs) from different mouse HCC models. Unlike fully malignant HCC, HcPCs give rise to cancer only when introduced into a liver undergoing chronic damage and compensatory proliferation. Although HcPCs exhibit a similar transcriptomic profile to bipotential hepatobiliary progenitors, the latter do not give rise to tumors. Cells resembling HcPCs reside within dysplastic lesions that appear several months before HCC nodules. Unlike early hepatocarcinogenesis, which depends on paracrine IL-6 production by inflammatory cells, due to upregulation of LIN28 expression, HcPCs had acquired autocrine IL-6 signaling that stimulates their in vivo growth and malignant progression. This may be a general mechanism that drives other IL-6-producing malignancies.
Collapse
Affiliation(s)
- Guobin He
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Debanjan Dhar
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Hayato Nakagawa
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- Department of Gastroenterology, University of Tokyo, Tokyo 113-8655, Japan
| | - Joan Font-Burgada
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Hisanobu Ogata
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuhong Jiang
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Shabnam Shalapour
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Ekihiro Seki
- Department of Medicine, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Shawn E. Yost
- Bioinformatics Graduate Program, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- Rady’s Children’s Hospital and Department of Pediatrics, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Kristen Jepsen
- Rady’s Children’s Hospital and Department of Pediatrics, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Kelly A. Frazer
- Rady’s Children’s Hospital and Department of Pediatrics, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- Moores UCSD Cancer Center, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- Clinical and Translational Research Institute, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- Institute for Genomic Medicine, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Olivier Harismendy
- Rady’s Children’s Hospital and Department of Pediatrics, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- Moores UCSD Cancer Center, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- Clinical and Translational Research Institute, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Maria Hatziapostolou
- Center for Systems Biomedicine, Division of Digestive Diseases and Institute for Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Dimitrios Iliopoulos
- Center for Systems Biomedicine, Division of Digestive Diseases and Institute for Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Atsushi Suetsugu
- Department of Surgery, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- AntiCancer, Inc., San Diego, CA 92111, USA
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- AntiCancer, Inc., San Diego, CA 92111, USA
| | - Ryosuke Tateishi
- Department of Gastroenterology, University of Tokyo, Tokyo 113-8655, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, University of Tokyo, Tokyo 113-8655, Japan
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
- Moores UCSD Cancer Center, University of California San Diego, School of Medicine, 9500 Gilman Drive, San Diego, CA 92093, USA
| |
Collapse
|
18
|
Jorns C, Takahashi T, Callaghan E, Zemack H, Larsson L, Nowak G, Parini P, Ericzon BG, Ellis E. Serum Apolipoprotein E as a Marker to Monitor Graft Function After Hepatocyte Transplantation in a Clinically Relevant Mouse Model. Transplant Proc 2013; 45:1780-6. [DOI: 10.1016/j.transproceed.2013.01.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 01/15/2013] [Indexed: 10/26/2022]
|
19
|
Ma X, Duan Y, Tschudy-Seney B, Roll G, Behbahan IS, Ahuja TP, Tolstikov V, Wang C, McGee J, Khoobyari S, Nolta JA, Willenbring H, Zern MA. Highly efficient differentiation of functional hepatocytes from human induced pluripotent stem cells. Stem Cells Transl Med 2013; 2:409-19. [PMID: 23681950 DOI: 10.5966/sctm.2012-0160] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) hold great potential for use in regenerative medicine, novel drug development, and disease progression/developmental studies. Here, we report highly efficient differentiation of hiPSCs toward a relatively homogeneous population of functional hepatocytes. hiPSC-derived hepatocytes (hiHs) not only showed a high expression of hepatocyte-specific proteins and liver-specific functions, but they also developed a functional biotransformation system including phase I and II metabolizing enzymes and phase III transporters. Nuclear receptors, which are critical for regulating the expression of metabolizing enzymes, were also expressed in hiHs. hiHs also responded to different compounds/inducers of cytochrome P450 as mature hepatocytes do. To follow up on this observation, we analyzed the drug metabolizing capacity of hiHs in real time using a novel ultra performance liquid chromatography-tandem mass spectrometry. We found that, like freshly isolated primary human hepatocytes, the seven major metabolic pathways of the drug bufuralol were found in hiHs. In addition, transplanted hiHs engrafted, integrated, and proliferated in livers of an immune-deficient mouse model, and secreted human albumin, indicating that hiHs also function in vivo. In conclusion, we have generated a method for the efficient generation of hepatocytes from induced pluripotent stem cells in vitro and in vivo, and it appears that the cells function similarly to primary human hepatocytes, including developing a complete metabolic function. These results represent a significant step toward using patient/disease-specific hepatocytes for cell-based therapeutics as well as for pharmacology and toxicology studies.
Collapse
Affiliation(s)
- Xiaocui Ma
- Department of Internal Medicine, University of California, Sacramento, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wan Z, Zhang XG, Liu ZW, Lv Y. Therapeutic liver repopulation for metabolic liver diseases: Advances from bench to bedside. Hepatol Res 2013; 43:122-130. [PMID: 22971121 DOI: 10.1111/j.1872-034x.2012.01081.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 07/20/2012] [Accepted: 07/23/2012] [Indexed: 12/14/2022]
Abstract
Metabolic liver diseases are characterized by inherited defects in hepatic enzymes or other proteins with metabolic functions. Therapeutic liver repopulation (TLR), an approach of massive liver replacement by transplanted normal hepatocytes, could be used to provide the missing metabolic function elegantly. However, partial and transient correction of the underlying metabolic defects due to very few integrated donor cell mass remains the major obstacle for the effective and widespread use of this approach. Little engraftment and proliferation insufficiency lead to the poor outcome. This article reviews the advances in the mechanisms of initial engraftment and selective proliferation and suggests some effective treatment strategies, from pharmacological preconditioning to stem cell transplantation, to optimize liver repopulation with liver cell transplantation. Enhancing cell viability and plating efficiency, increasing sinusoidal spaces, regulation of sinusoidal endothelial cell barrier and controlling inflammatory reaction may promote initial cell engraftment. Liver-directed irradiation, reversible portal vein embolization and fetal liver stem/progenitor cell transplantation induce preferential proliferation of donor cells substantially without severe side-effects. Furthermore, it seems better to use combined approaches to achieve a high level of liver repopulation for the management of metabolic liver diseases.
Collapse
Affiliation(s)
- Zhen Wan
- Hepatobiliary Surgery; Institute of Advanced Surgical Techniques and Tissue Engineering Research, Xi'an Jiaotong University, Xi'an, China
| | | | | | | |
Collapse
|
21
|
Iwamoto T, Terai S, Mizunaga Y, Yamamoto N, Omori K, Uchida K, Yamasaki T, Fujii Y, Nishina H, Sakaida I. Splenectomy enhances the anti-fibrotic effect of bone marrow cell infusion and improves liver function in cirrhotic mice and patients. J Gastroenterol 2012; 47:300-12. [PMID: 22065159 DOI: 10.1007/s00535-011-0486-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 09/11/2011] [Indexed: 02/04/2023]
Abstract
BACKGROUND In 2003, we initiated a clinical trial to examine autologous bone marrow cell infusion (ABMi) therapy for cirrhotic patients and reported the clinical effect of the therapy. To analyze how splenectomy may potentiate the effects of bone marrow cell infusion on cirrhosis, we performed a mouse study and a clinical trial on patients with cirrhosis. METHODS In mice, we analyzed the effect of splenectomy on bone marrow cell infusion in four experimental groups (group A, splenectomy + bone marrow cell infusion + CCl(4); group B, sham operation + bone marrow cell infusion + CCl(4); group C, splenectomy + CCl(4); group D, sham operation + CCl(4)). In clinical, we compared the effect of splenectomy on ABMi therapy. RESULTS We observed significantly increased average serum albumin levels and higher expression of green fluorescent protein (GFP), matrix metalloproteinase 9 (MMP9), and proliferating cell nuclear antigen in the livers of group A. We observed MMP9/GFP double-positive cells in the cirrhotic livers. A significant decrease in the liver fibrosis areas was observed in group A. Splenectomy enhanced the repopulation of bone marrow cells into the cirrhotic liver and improved the liver microenvironment via expression of MMP9 secreted from repopulating GFP-positive cells. Next, we performed a clinical trial to compare the effect of splenectomy on the efficacy of ABMi therapy. Cirrhotic patients who underwent splenectomy before ABMi therapy tended to have a greater improvement in liver function. CONCLUSION ABMi therapy with splenectomy may be an effective therapeutic modality for cirrhosis.
Collapse
Affiliation(s)
- Takuya Iwamoto
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
The nude mouse as model for liver deficiency study and treatment and xenotransplantation. Int J Hepatol 2012; 2012:140147. [PMID: 23193481 PMCID: PMC3502033 DOI: 10.1155/2012/140147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 08/13/2012] [Accepted: 09/07/2012] [Indexed: 02/06/2023] Open
Abstract
We aimed at reviewing the various uses of Nude mouse for the development of liver deficiency models and evaluation of efficacy of hepatic cell xenotransplantation. The first part records the large range of liver deficiency models that can be developed in Nude mice: surgical partial hepatectomy, acute toxic liver deficiency, chronic cirrhosis, and transgenic liver injury. The second part tackles the outcome of rat hepatocyte as well as human cell transplantation, both mature hepatocyte and hepatic progenitor, into Nude mouse submitted to liver injury. Results are discussed and compared to other available immunodeficient mouse models. The issue of humanized liver creation is also addressed. Altogether, these results show that Nude mouse appears to be a suitable small animal model to expand our insight into liver cell engraftment and regeneration.
Collapse
|
23
|
Fetal liver cell transplantation as a potential alternative to whole liver transplantation? J Gastroenterol 2011; 46:953-65. [PMID: 21698354 DOI: 10.1007/s00535-011-0427-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 05/02/2011] [Indexed: 02/04/2023]
Abstract
Because organ shortage is the fundamental limitation of whole liver transplantation, novel therapeutic options, especially the possibility of restoring liver function through cell transplantation, are urgently needed to treat end-stage liver diseases. Groundbreaking in vivo studies have shown that transplanted hepatocytes are capable of repopulating the rodent liver. The two best studied models are the urokinase plasminogen activator (uPA) transgenic mouse and the fumarylacetoacetate hydrolase (FAH)-deficient mouse, in which genetic modifications of the recipient liver provide a tissue environment in which there is extensive liver injury and selection pressure favoring the proliferation and survival of transplanted hepatocytes. Because transplanted hepatocytes do not significantly repopulate the (near-)normal liver, attention has been focused on finding alternative cell types, such as stem or progenitor cells, that have a higher proliferative potential than hepatocytes. Several sources of stem cells or stem-like cells have been identified and their potential to repopulate the recipient liver has been evaluated in certain liver injury models. However, rat fetal liver stem/progenitor cells (FLSPCs) are the only cells identified to date that can effectively repopulate the (near-)normal liver, are morphologically and functionally fully integrated into the recipient liver, and remain viable long-term. Even though primary human fetal liver cells are not likely to be routinely used for clinical liver cell repopulation in the future, using or engineering candidate cells exhibiting the characteristics of FLSPCs suggests a new direction in developing cell transplantation strategies for therapeutic liver replacement. This review will give a brief overview concerning the existing animal models and cell sources that have been used to restore normal liver structure and function, and will focus specifically on the potential of FLSPCs to repopulate the liver.
Collapse
|
24
|
Shafritz DA, Oertel M. Model systems and experimental conditions that lead to effective repopulation of the liver by transplanted cells. Int J Biochem Cell Biol 2011; 43:198-213. [PMID: 20080205 PMCID: PMC2907475 DOI: 10.1016/j.biocel.2010.01.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 12/22/2009] [Accepted: 01/07/2010] [Indexed: 12/26/2022]
Abstract
In recent years, there has been substantial progress in transplanting cells into the liver with the ultimate goal of restoring liver mass and function in both inherited and acquired liver diseases. The basis for considering that this might be feasible is that the liver is a highly regenerative organ. After massive liver injury or surgical removal of two-thirds or more of the liver tissue, the organ can restore its mass with completely normal morphologic structure and function. It has also been found under highly selective conditions that transplanted hepatocytes can fully repopulate the liver and cure a metabolic disorder or deficiency state. Fetal liver cells can also substantially repopulate the normal liver, and it is hoped in the future that effective repopulation will be achievable with cultured cells or cell lines, pluripotent stem cells from other somatic tissues, embryonic stem cells, or induced pluripotent stem cells, which can now be generated in vitro by a variety of methods. The purpose of this review is to present the major systems that have been used for liver repopulation, the variables involved in obtaining successful repopulation and what has been achieved in these various systems to date with different cell types.
Collapse
Affiliation(s)
- David A Shafritz
- Marion Bessin Liver Research Center, Department of Medicine and Division of Hepatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | |
Collapse
|
25
|
Soltys KA, Soto-Gutiérrez A, Nagaya M, Baskin KM, Deutsch M, Ito R, Shneider BL, Squires R, Vockley J, Guha C, Roy-Chowdhury J, Strom SC, Platt JL, Fox IJ. Barriers to the successful treatment of liver disease by hepatocyte transplantation. J Hepatol 2010; 53:769-774. [PMID: 20667616 PMCID: PMC2930077 DOI: 10.1016/j.jhep.2010.05.010] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 05/24/2010] [Accepted: 05/28/2010] [Indexed: 12/11/2022]
Abstract
Management of patients with hepatic failure and liver-based metabolic disorders is complex and expensive. Hepatic failure results in impaired coagulation, altered consciousness and cerebral function, a heightened risk of multiple organ system failure, and sepsis [1]. Such manifold problems are only treatable today and for the foreseeable future by transplantation. In fact, whole or auxiliary partial liver transplantation is often the only available treatment option for severe, even if transient, hepatic failure. Patients with life-threatening liver-based metabolic disorders similarly require organ transplantation even though their metabolic diseases are typically the result of a single enzyme deficiency, and the liver otherwise functions normally. For all of the benefits it may confer, liver transplantation is not an ideal therapy, even for severe hepatic failure. More than 17,000 patients currently await liver transplantation in the United States, a number that seriously underestimates the number of patients that need treatment [2], as it has been estimated that more than a million patients could benefit from transplantation [3]. Unfortunately, use of whole liver transplantation to treat these disorders is limited by a severe shortage of donors and by the risks to the recipient associated with major surgery [4].
Collapse
Affiliation(s)
- Kyle A. Soltys
- Thomas E. Starzl Transplant Institute, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA
| | - Alejandro Soto-Gutiérrez
- Department of Surgery, and McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Masaki Nagaya
- Department of Surgery, and McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kevin M. Baskin
- Division of Vascular and Interventional Radiology, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA
| | - Melvin Deutsch
- Department of Radiation Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ryotaro Ito
- Department of Surgery, and McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Benjamin L. Shneider
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA
| | - Robert Squires
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA
| | - Jerry Vockley
- Departments of Pediatrics and Human Genetics, University of Pittsburgh School of Medicine and Department of Medical Genetics, Children’s Hospital of Pittsburgh of UPMC
| | - Chandan Guha
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY
| | - Jayanta Roy-Chowdhury
- Department of Medicine (Hepatology Division) and Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY
| | - Stephen C. Strom
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh PA 15261, USA
| | - Jeffrey L. Platt
- Departments of Surgery and Microbiology and Immunology, University of Michigan, Ann Arbor MI 48109, USA
| | - Ira J. Fox
- Department of Surgery, and McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
26
|
Abstract
The liver has an extraordinary faculty to regenerate. Hepatocytes are highly differentiated cells that, despite a resting G0 state in the normal quiescent liver, can re-enter the cell cycle to reconstitute the organ after an injury. However, the first cell therapy approaches trying to harness this specific characteristic of the hepatocytes came up against the competition with resident hepatocytes in the ability to proliferate. This review will describe the different rodent models that have been developed in the last 15 years to demonstrate the concept of liver repopulation with transplanted cells harbouring a selective advantage over resident hepatocytes. Examples will then be given to show how these models demonstrated the therapeutic efficiency of cell transplantation in specific disorders. The transplantation of human hepatocytes into some of these mouse models led to the creation of humanized livers. These humanized mice provide a powerful tool to study the physiopathology of human hepatotropic pathogens and to develop drugs against them. Finally, emphasis will be placed on the role of these rodent models in the demonstration of the hepatocytic potential of stem cells.
Collapse
|
27
|
Tokai H, Kawashita Y, Ito Y, Yamanouchi K, Takatsuki M, Eguchi S, Tajima Y, Kanematsu T. Efficacy and limitation of bone marrow transplantation in the treatment of acute and subacute liver failure in rats. Hepatol Res 2009; 39:1137-43. [PMID: 19619255 DOI: 10.1111/j.1872-034x.2009.00556.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM Recent reports have shown that bone marrow cells (BMC) retain the potential to differentiate into hepatocytes. Thus, the BMC have been recognized as an attractive source for liver regenerative medicine. However, it has not been clarified whether BMC transplantation can be used to treat liver damage in vivo. In the present study, we explored whether BMC possess therapeutic potential to treat acute and/or subacute liver failure. METHODS Fulminant hepatic failure (FHF) was induced by 70% hepatectomy with ligation of the right lobe pedicle (24% liver mass), followed by transplantation of BMC into the spleen. Dipeptidyl peptidase IV-positive (DPPIV(+)) BMC were then transplanted into DPPIV-negative (DPPIV(-)) recipients following hepatic irradiation (HIR) in which 70% of the liver was resected and the remnant liver irradiated. RESULTS There was no benefit of BMC transplantation towards survival in the FHF model. DPPIV(+) hepatocytes appeared in the liver tissues of the DPPIV(-) HIR model rats, but DPPIV(+) hepatocytes replaced less than 13% of the recipient liver. CONCLUSION BMC transplantation may have limitations in the treatment of fulminant or acute liver failure because they do not have sufficient time to develop into functional hepatocytes. Preparative HIR may be beneficial in help to convert the transplanted BMC into host hepatocytes, and provide a survival benefit. Although, However, the precise mechanism warrants further studies.
Collapse
Affiliation(s)
- Hirotaka Tokai
- Department of Surgery, Graduate School of Biochemical Sciences, Nagasaki University, Nagasaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Duncan AW, Dorrell C, Grompe M. Stem cells and liver regeneration. Gastroenterology 2009; 137:466-81. [PMID: 19470389 PMCID: PMC3136245 DOI: 10.1053/j.gastro.2009.05.044] [Citation(s) in RCA: 412] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 04/13/2009] [Accepted: 05/11/2009] [Indexed: 12/16/2022]
Abstract
One of the defining features of the liver is the capacity to maintain a constant size despite injury. Although the precise molecular signals involved in the maintenance of liver size are not completely known, it is clear that the liver delicately balances regeneration with overgrowth. Mammals, for example, can survive surgical removal of up to 75% of the total liver mass. Within 1 week after liver resection, the total number of liver cells is restored. Moreover, liver overgrowth can be induced by a variety of signals, including hepatocyte growth factor or peroxisome proliferators; the liver quickly returns to its normal size when the proliferative signal is removed. The extent to which liver stem cells mediate liver regeneration has been hotly debated. One of the primary reasons for this controversy is the use of multiple definitions for the hepatic stem cell. Definitions for the liver stem cell include the following: (1) cells responsible for normal tissue turnover, (2) cells that give rise to regeneration after partial hepatectomy, (3) cells responsible for progenitor-dependent regeneration, (4) cells that produce hepatocyte and bile duct epithelial phenotypes in vitro, and (5) transplantable liver-repopulating cells. This review will consider liver stem cells in the context of each definition.
Collapse
Affiliation(s)
- Andrew W Duncan
- Oregon Stem Cell Center, Oregon Health & Science University, Portland, Oregon 97239-3098, USA
| | | | | |
Collapse
|
29
|
Skvorak KJ, Paul HS, Dorko K, Marongiu F, Ellis E, Chace D, Ferguson C, Gibson KM, Homanics GE, Strom SC. Hepatocyte transplantation improves phenotype and extends survival in a murine model of intermediate maple syrup urine disease. Mol Ther 2009; 17:1266-73. [PMID: 19436271 DOI: 10.1038/mt.2009.99] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Maple syrup urine disease (MSUD; OMIM 248600) is an inborn error of metabolism of the branched chain alpha-ketoacid dehydrogenase (BCKDH) complex that is treated primarily by dietary manipulation of branched-chain amino acids (BCAA). Dietary restriction is lifelong and compliance is difficult. Liver transplantation significantly improves outcomes; however, alternative therapies are needed. To test novel therapies such as hepatocyte transplantation (HTx), we previously created a murine model of intermediate MSUD (iMSUD), which closely mimics human iMSUD. LacZ-positive murine donor hepatocytes were harvested and directly injected (10(5) cells/50 microl) into liver of iMSUD mice (two injections at 1-10 days of age). Donor hepatocytes engrafted into iMSUD recipient liver, increased liver BCKDH activity, improved blood total BCAA/alanine ratio, increased body weight at weaning, and extended the lifespan of HTx-treated iMSUD mice compared to phosphate-buffered saline (PBS)-treated and untreated iMSUD mice. Based on these data demonstrating partial metabolic correction of iMSUD in a murine model, coupled to the fact that multiple transplants are possible to enhance these results, we suggest that HTx represents a promising therapeutic intervention for MSUD that warrants further investigation.
Collapse
Affiliation(s)
- Kristen J Skvorak
- Department of Human Genetics, University of Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Basma H, Soto-Gutiérrez A, Yannam GR, Liu L, Ito R, Yamamoto T, Ellis E, Carson SD, Sato S, Chen Y, Muirhead D, Navarro-Alvarez N, Wong RJ, Roy-Chowdhury J, Platt JL, Mercer DF, Miller JD, Strom SC, Kobayashi N, Fox IJ. Differentiation and transplantation of human embryonic stem cell-derived hepatocytes. Gastroenterology 2009; 136:990-9. [PMID: 19026649 PMCID: PMC2732349 DOI: 10.1053/j.gastro.2008.10.047] [Citation(s) in RCA: 363] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 10/17/2008] [Accepted: 10/23/2008] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS The ability to obtain unlimited numbers of human hepatocytes would improve the development of cell-based therapies for liver diseases, facilitate the study of liver biology, and improve the early stages of drug discovery. Embryonic stem cells are pluripotent, potentially can differentiate into any cell type, and therefore could be developed as a source of human hepatocytes. METHODS To generate human hepatocytes, human embryonic stem cells were differentiated by sequential culture in fibroblast growth factor 2 and human activin-A, hepatocyte growth factor, and dexamethasone. Functional hepatocytes were isolated by sorting for surface asialoglycoprotein-receptor expression. Characterization was performed by real-time polymerase chain reaction, immunohistochemistry, immunoblot, functional assays, and transplantation. RESULTS Embryonic stem cell-derived hepatocytes expressed liver-specific genes, but not genes representing other lineages, secreted functional human liver-specific proteins similar to those of primary human hepatocytes, and showed human hepatocyte cytochrome P450 metabolic activity. Serum from rodents given injections of embryonic stem cell-derived hepatocytes contained significant amounts of human albumin and alpha1-antitrypsin. Colonies of cytokeratin-18 and human albumin-expressing cells were present in the livers of recipient animals. CONCLUSIONS Human embryonic stem cells can be differentiated into cells with many characteristics of primary human hepatocytes. Hepatocyte-like cells can be enriched and recovered based on asialoglycoprotein-receptor expression and potentially could be used in drug discovery research and developed as therapeutics.
Collapse
Affiliation(s)
- Hesham Basma
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Hepatocyte transplantation has therapeutic potential for multiple hepatic and extrahepatic disorders with genetic or acquired basis. To demonstrate whether cell populations of interest will be effective for clinical applications, it is first necessary to characterize their properties in animal systems. Demonstrating the potential of cells to engraft and proliferate is a critical part of this characterization. Similarly, for stem/progenitor cells, demonstrating the capacity to differentiate along appropriate lineages and generate mature cells that can engraft and proliferate is essential. In various animal models, preconditioning of recipients prior to cell transplantation has been necessary to improve engraftment of cells, to stimulate proliferation of engrafted cells, and to induce extensive repopulation of the host liver by transplanted cells. Although this is an area of active investigation, effective preconditioning protocols should alter the hepatic microenvironment, such that transplanted cells can obtain selective advantages for engrafting and proliferating in the liver. Use of such experimental systems in animals will help generate further strategies for liver repopulation and thereby advance clinical applications of liver cell therapy.
Collapse
Affiliation(s)
- Yao-Ming Wu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | | |
Collapse
|
32
|
Willenbring H, Sharma AD, Vogel A, Lee AY, Rothfuss A, Wang Z, Finegold M, Grompe M. Loss of p21 permits carcinogenesis from chronically damaged liver and kidney epithelial cells despite unchecked apoptosis. Cancer Cell 2008; 14:59-67. [PMID: 18598944 PMCID: PMC2526059 DOI: 10.1016/j.ccr.2008.05.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Revised: 03/02/2008] [Accepted: 05/14/2008] [Indexed: 01/28/2023]
Abstract
Accumulation of toxic metabolites in hereditary tyrosinemia type I (HT1) patients leads to chronic DNA damage and the highest risk for hepatocellular carcinomas (HCCs) of any human disease. Here we show that hepatocytes of HT1 mice exhibit a profound cell-cycle arrest that, despite concomitant apoptosis resistance, causes mortality from impaired liver regeneration. However, additional loss of p21 in HT1 mice restores the proliferative capabilities of hepatocytes and renal proximal tubular cells. This growth response compensates cell loss due to uninhibited apoptosis and enables animal survival but rapidly leads to HCCs, renal cysts, and renal carcinomas. Thus, p21's antiproliferative function is indispensable for the suppression of carcinogenesis from chronically injured liver and renal epithelial cells and cannot be compensated by apoptosis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Cycle
- Cell Proliferation/drug effects
- Cyclin-Dependent Kinase Inhibitor p21/deficiency
- Cyclin-Dependent Kinase Inhibitor p21/genetics
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
- Cyclohexanones/pharmacology
- Disease Models, Animal
- Enzyme Inhibitors/pharmacology
- Hepatectomy
- Hepatocytes/drug effects
- Hepatocytes/enzymology
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Hydrolases/genetics
- Hydrolases/metabolism
- Kidney Diseases, Cystic/etiology
- Kidney Diseases, Cystic/metabolism
- Kidney Diseases, Cystic/pathology
- Kidney Neoplasms/etiology
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Liver Neoplasms/etiology
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Regeneration/drug effects
- Mice
- Mice, Knockout
- Neoplasms/etiology
- Neoplasms/metabolism
- Neoplasms/pathology
- Nitrobenzoates/pharmacology
- Tyrosinemias/complications
- Tyrosinemias/genetics
- Tyrosinemias/metabolism
- Tyrosinemias/pathology
- Tyrosinemias/physiopathology
Collapse
Affiliation(s)
- Holger Willenbring
- Institute for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Vidal I, Blanchard N, Alexandre E, Gandillet A, Chenard-Neu MP, Staedtler F, Schumacher M, Bachellier P, Jaeck D, Firat H, Heyd B, Richert L. Improved Xenogenic Hepatocyte Implantation into Nude Mouse Liver Parenchyma with Acute Liver Failure when Followed by Repeated Anti-Fas Antibody (Jo2) Treatment. Cell Transplant 2008; 17:507-24. [DOI: 10.3727/096368908785096051] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Hepatocyte transplantation is a promising therapy for acute liver failure in humans. Recently, we succeeded in inducing various acute and chronic liver failures in nude mice. Engraftment of transplanted xenogeneic rat hepatocytes, visualized in the host liver by anti-MHC class I immunohistochemistry, revealed that liver repopulation was limited, and equivalent in nude mice with and without acute liver failure. In the present study, acute liver failure was induced in nude mice by a single injection of sublethal anti-Fas antibody Jo2, followed 24 h later by rat hepatocyte transplantation and than by a weekly repeated injection of Jo2. Rat hepatocyte engraftment into the recipient liver parenchyma 3 weeks following hepatocyte transplantation was about sevenfold increased when nude mice were subsequently subjected to weekly repeated Jo2 injection. Genomic analysis of these mice showed an overall transcriptome profile of upregulation of cellular cycle blocking transcripts, activation of liver injury inducing IFN-γ/STAT1 pathway, and circadian transcript signature of antiproliferative cell status compared to mice submitted to hepatocyte transplantation only. The findings of the present study suggest that the induction of cell proliferation blockade in recipient livers could promote sufficient engraftment of transplanted hepatocytes to allow transient or definitive treatment of liver failure in humans.
Collapse
Affiliation(s)
- Isabelle Vidal
- EA 3921, IFR 133, Faculté de Médecine et de Pharmacie, Besançon, France
- Laboratoire de Chirurgie Expérimentale, Fondation Transplantation, Strasbourg, France
| | - Nadège Blanchard
- Laboratoire de Chirurgie Expérimentale, Fondation Transplantation, Strasbourg, France
- KaLy-Cell, Témis Innovation, Besançon, France
| | - Eliane Alexandre
- Laboratoire de Chirurgie Expérimentale, Fondation Transplantation, Strasbourg, France
| | - Arnaud Gandillet
- Laboratoire de Chirurgie Expérimentale, Fondation Transplantation, Strasbourg, France
| | | | - Frank Staedtler
- Novartis Pharma AG, Biomarker Development, Basel, Switzerland
| | | | - Philippe Bachellier
- Laboratoire de Chirurgie Expérimentale, Fondation Transplantation, Strasbourg, France
- Centre de Chirurgie Viscérale et de Transplantation, Hôpital de Hautepierre, Strasbourg, France
| | - Daniel Jaeck
- Laboratoire de Chirurgie Expérimentale, Fondation Transplantation, Strasbourg, France
- Centre de Chirurgie Viscérale et de Transplantation, Hôpital de Hautepierre, Strasbourg, France
| | | | - Bruno Heyd
- EA 3921, IFR 133, Faculté de Médecine et de Pharmacie, Besançon, France
- Service de Chirurgie Digestive et Vasculaire, Hôpital Jean Minjoz, Besançon, France
| | - Lysiane Richert
- EA 3921, IFR 133, Faculté de Médecine et de Pharmacie, Besançon, France
- Laboratoire de Chirurgie Expérimentale, Fondation Transplantation, Strasbourg, France
- KaLy-Cell, Témis Innovation, Besançon, France
| |
Collapse
|
34
|
Agarwal S, Holton KL, Lanza R. Efficient differentiation of functional hepatocytes from human embryonic stem cells. Stem Cells 2008; 26:1117-27. [PMID: 18292207 DOI: 10.1634/stemcells.2007-1102] [Citation(s) in RCA: 270] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Differentiation of human embryonic stem cells (hESCs) to specific functional cell types can be achieved using methods that mimic in vivo embryonic developmental programs. Current protocols for generating hepatocytes from hESCs are hampered by inefficient differentiation procedures that lead to low yields and large cellular heterogeneity. We report here a robust and highly efficient process for the generation of high-purity (70%) hepatocyte cultures from hESCs that parallels sequential hepatic development in vivo. Highly enriched populations of definitive endoderm were generated from hESCs and then induced to differentiate along the hepatic lineage by the sequential addition of inducing factors implicated in physiological hepatogenesis. The differentiation process was largely uniform, with cell cultures progressively expressing increasing numbers of hepatic lineage markers, including GATA4, HNF4alpha, alpha-fetoprotein, CD26, albumin, alpha-1-antitrypsin, Cyp7A1, and Cyp3A4. The hepatocytes exhibited functional hepatic characteristics, such as glycogen storage, indocyanine green uptake and release, and albumin secretion. In a mouse model of acute liver injury, the hESC-derived definitive endoderm differentiated into hepatocytes and repopulated the damaged liver. The methodology described here represents a significant step toward the efficient generation of hepatocytes for use in regenerative medicine and drug discovery.
Collapse
Affiliation(s)
- Sadhana Agarwal
- Advanced Cell Technology, 381 Plantation Street, Worcester, Massachusetts 01605, USA
| | | | | |
Collapse
|
35
|
Lin H, Mao Q, Wang YM, Jiang L. Proliferation of L02 human hepatocytes in tolerized genetically immunocompetent rats. World J Gastroenterol 2008; 14:2329-37. [PMID: 18416458 PMCID: PMC2705086 DOI: 10.3748/wjg.14.2329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether human hepatocytes could proliferate after transplantation to normal immunocompetent rats treated with 2-acetaminofluorene or Retrorsine and partial hepatectomy.
METHODS: L02 hepatocyte-tolerant Sprague-Dawley rats were injected with Retrorsine, 2-acetaminofluorene or normal saline. L02 hepatocytes were then transplanted via the spleen. Human albumin and its mRNA, specific proliferating cell nuclear antigen (PCNA), L02 hepatocyte dynamic distribution, number density and area density of PCNA-positive cells in the liver were determined.
RESULTS: All the examined indicators were not significantly different between the rats treated with 2-acetaminofluorene and normal saline, which was not the case with rats treated with Retrorsine. A dynamic distribution of L02 hepatocytes in the rat liver was detected from wk 1 to mo 6 after transplantation in the Retrorsine group and from wk 1 to 10 in the 2-acetaminofluorene group. Human albumin and its mRNA were detected from wk 2 to mo 6 in the Retrorsine group and from wk 1 to 8 in the 2-acetaminofluorene group. Specific human PCNA was detected in the rat liver from wk 2 to mo 6 in the Retrorsine group and from wk 2 to 6 in the 2-acetaminofluorene group. Human albumin and its mRNA contents as well as the number of PCNA positive cells reached a peak at wk 4.
CONCLUSION: L02 human hepatocytes could not proliferate significiantly after transplantation to the normal, immunocompetent rats treated with 2-acetaminofluorene. L02 human hepatocytes can survive for 10 wk after transplantation and express human albumin for 8 wk. L02 human hepatocytes can proliferate and express human albumin for 6 mo after transplantation to the rats treated with Retrorsine. The chimeric L02 human hepatocytes, which then underwent transplantation into tolerant rats, were normal in morphogenesis, biochemistry and function.
Collapse
|
36
|
Wu YM, Joseph B, Berishvili E, Kumaran V, Gupta S. Hepatocyte transplantation and drug-induced perturbations in liver cell compartments. Hepatology 2008; 47:279-87. [PMID: 17935178 DOI: 10.1002/hep.21937] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
UNLABELLED The potential for organ damage after using drugs or chemicals is a critical issue in medicine. To delineate mechanisms of drug-induced hepatic injury, we used transplanted cells as reporters in dipeptidyl peptidase IV-deficient mice. These mice were given phenytoin and rifampicin for 3 days, after which monocrotaline was given followed 1 day later by intrasplenic transplantation of healthy C57BL/6 mouse hepatocytes. We examined endothelial and hepatic damage by serologic or tissue studies and assessed changes in transplanted cell engraftment and liver repopulation by histochemical staining for dipeptidyl peptidase IV. Monocrotaline caused denudation of the hepatic sinusoidal endothelium and increased serum hyaluronic acid levels, along with superior transplanted cell engraftment. Together, phenytoin, rifampicin, and monocrotaline caused further endothelial damage, reflected by greater improvement in cell engraftment. Phenytoin, rifampicin, and monocrotaline produced injury in hepatocytes that was not apparent after conventional tissue studies. This led to transplanted cell proliferation and extensive liver repopulation over several weeks, which was more efficient in males compared with females, including greater induction by phenytoin and rifampicin of cytochrome P450 3A4 isoform that converts monocrotaline to toxic intermediates. Through this and other possible mechanisms, monocrotaline-induced injury in the endothelial compartment was retargeted to simultaneously involve hepatocytes over the long term. Moreover, after this hepatic injury, native liver cells were more susceptible to additional pro-oxidant injury through thyroid hormone, which accelerated the kinetics of liver repopulation. CONCLUSION Transplanted reporter cells will be useful for obtaining insights into homeostatic mechanisms involving liver cell compartments, whereas targeted injury in hepatic endothelial and parenchymal cells with suitable drugs will also help advance liver cell therapy.
Collapse
Affiliation(s)
- Yao-Ming Wu
- Marion Bessin Liver Research Center, Diabetes Center, Cancer Research Center, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | |
Collapse
|
37
|
Improvement of the survival rate by fetal liver cell transplantation in a mice lethal liver failure model. Transplantation 2007; 84:1233-9. [PMID: 18049107 DOI: 10.1097/01.tp.0000287967.54222.4d] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The use of cell transplantation as an alternative therapy for orthotopic liver transplantation has been widely anticipated due to a chronic donor shortage. We previously reported the method used to enrich hepatic progenitor cells (HPCs) forming cell aggregations. In this study, we transplanted HPCs into the liver injury model mice to determine whether HPC transplantation may improve the liver dysfunction. METHODS We obtained donor cells from E13.5 fetal livers of green fluorescent protein (GFP) transgenic mice. We transplanted GFP-positive fetal liver cells into the transgenic mice which express diphtheria toxin (DT) receptors under the control of an albumin enhancer/promoter. Subsequently, we induced selective liver injury to recipient mice by DT administration. We then evaluated the engraftment of the transplanted cells and their effect on survivorship. RESULTS The low dose of DT induced sublethal liver injury and the high dose of DT was lethal to the liver injury model mice. The transplanted GFP-positive cells were engrafted into the recipient livers and expressed albumin, resembling mature hepatocytes. They continued to proliferate, forming clusters. The survival rate at 25 days after transplantation of the cell-transplanted group (8 of 20; 40.0%) was improved significantly (P=0.0047) in comparison to that of the sham-operated group (0 of 20; 0%). CONCLUSIONS The transplanted cells were engrafted and repopulated the liver of recipient mice, resulting in the improvement of the survival rate of the liver injury model mice. We therefore propose that HPCs are a desirable cell source for cell transplantation.
Collapse
|
38
|
Krishnan A, Viker K, Rietema H, Telgenkamp M, Knudsen B, Charlton M. Prolonged engraftment of human hepatocytes in mice transgenic for the deleted form of human hepatocyte growth factor. Hepatol Res 2007; 37:854-62. [PMID: 17573952 DOI: 10.1111/j.1872-034x.2007.00139.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIM Small animal models chimeric for human hepatocytes have provided valuable insights into the biology of hepatotropic viral infection and provided a platform for the study of therapeutic agents. Existing models of human hepatocyte transplantation are limited by phenotypic fragility and impaired immunity. We hypothesized that mice transgenic for human hepatocyte growth factor (HGF), a potent human hepatocyte mitogen, would engraft human hepatocytes in the absence of immunodeficiency. METHODS A plasmid construct containing the 2.3 kb coding region of the 723 amino acid isoform of HGF cDNA under the transcriptional control of the mouse albumin promoter/enhancer was used to generate transgenic mice. Cryopreserved human hepatocytes were transplanted into nine transgenic and six non-transgenic mice. Engraftment of human hepatocytes was followed for a period of 12 weeks by immunoblotting for human albumin in mouse serum samples. RESULTS In six out of the nine transgenic mice, abundance of human albumin, following an initial decline, increased andpeaked at > 70 days post transplantation, demonstrating sustained engraftment of transplanted human hepatocytes. In all the non-transgenic mice, post-transplant human albumin levels declined sequentially without evidence of sustained engraftment. Immunostaining of mouse liver sections indicated the presence of human hepatocytes adjacent to clusters of non-staining murine hepatocytes. CONCLUSION These results demonstrate that sustained engraftment of human hepatocytes in mice is facilitated by expression of the human dHGF transgene. Human hepatocyte engraftment in this model has been achieved on an immunocompetent strain background and merits further study as a candidate for the study of hepatotropic viral infections.
Collapse
|
39
|
Caballero M, Lightfoot HM, Lapaglia M, Pleasant A, Hatada S, Cairns BA, Fair JH. Detection and characterization of hepatic engraftment of embryonic stem derived cells by fluorescent stereomicroscopy. J Surg Res 2007; 141:134-40. [PMID: 17543343 PMCID: PMC2082135 DOI: 10.1016/j.jss.2006.04.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2005] [Revised: 11/14/2005] [Accepted: 04/10/2006] [Indexed: 12/16/2022]
Abstract
BACKGROUND Embryonic stem (ES) cells have been investigated as a potential replacement therapy for failed organs, such as the liver. However, detection of hepatic engraftment from candidate stem cells has been difficult due to low engraftment efficiency. Previous detection methods required that the graft be processed by molecular and/or immunohistochemical techniques, limiting further functional studies. This study evaluated the use of three-dimensional fluorescent stereomicroscopy for gross detection of ES cell derived hepatic engraftment. MATERIAL AND METHODS Murine ES cells expressing the enhanced green fluorescence protein (EGFP) underwent directed endodermal lineage differentiation. Three days after two thirds partial hepatectomy, cells were injected into the liver parenchyma, and livers were harvested at 10 to 20 d and examined by fluorescence stereomicroscopy with a GFP2 long pass filter (100447084; Leica Microsystems AG, Wetzlar, Germany). The sensitivity and reliability of the test was evaluated using quantitative polymerase chain reaction (q-PCR) to assay for the presence of EGFP mRNA in the tissue. RESULTS Fluorescent microscopy detected EGFP-positive cells engrafted with normal histology in 5 of 11 specimens. EGFP mRNA was confirmed in all five specimens by q-PCR. Only one of the 11 specimens was negative by fluorescence stereomicroscopy and positive by q-PCR, P < 0.02, Fisher's exact test. CONCLUSION Utilization of three-dimensional stereomicroscopy with a GFP2 long pass filter is a powerful and fast screening tool for GFP-ES derived hepatic engraftment.
Collapse
Affiliation(s)
- Montserrat Caballero
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7211, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
Cubero FJ, Maganto P, Mula N, Ortiz A, Barrutia MG, Codesal FJ, Arahuetes RM. Functional response of hepatocytes transplanted into Gunn rats stimulated with thyroid hormone. Dig Dis Sci 2007; 52:210-6. [PMID: 17160715 DOI: 10.1007/s10620-006-9614-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Accepted: 09/14/2006] [Indexed: 12/09/2022]
Abstract
In the attempt to translate laboratory studies into clinical practice, the small number of cells that can be transplanted is currently a problem to be solved. The aim of this work is to study the functional response of intrasplenically transplanted syngeneic rat adult and fetal hepatocytes to a proliferative stimulus, 3,5,3'-triiodothyronine. Total serum bilirubin significantly decreased from 7 to 90 days after fetal hepatocyte transplantation and from 24 hr to 30 days after adult hepatocyte transplantation. Concomitant with these changes, bile conjugated bilirubin increased from 7 to 90 days after fetal and from 24 hr to 30 days after adult hepatocyte transplantation. In both cases, administration of thyroid hormone enhances this effect. We conclude that although adult and fetal hepatocytes correct the hyperbilirubinemia, fetal cells function longer than adult hepatocytes. Thyroid hormone is a powerful stimulator of function of hepatocytes since it improves both adult and fetal response.
Collapse
Affiliation(s)
- Francisco J Cubero
- Servicio de Cirugía Experimental, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
41
|
Gouon-Evans V, Boussemart L, Gadue P, Nierhoff D, Koehler CI, Kubo A, Shafritz DA, Keller G. BMP-4 is required for hepatic specification of mouse embryonic stem cell-derived definitive endoderm. Nat Biotechnol 2006; 24:1402-11. [PMID: 17086172 DOI: 10.1038/nbt1258] [Citation(s) in RCA: 309] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Accepted: 09/27/2006] [Indexed: 01/26/2023]
Abstract
When differentiated in the presence of activin A in serum-free conditions, mouse embryonic stem cells efficiently generate an endoderm progenitor population defined by the coexpression of either Brachyury, Foxa2 and c-Kit, or c-Kit and Cxcr4. Specification of these progenitors with bone morphogenetic protein-4 in combination with basic fibroblast growth factor and activin A results in the development of hepatic populations highly enriched (45-70%) for cells that express the alpha-fetoprotein and albumin proteins. These cells also express transcripts of Afp, Alb1, Tat, Cps1, Cyp7a1 and Cyp3a11; they secrete albumin, store glycogen, show ultrastructural characteristics of mature hepatocytes, and are able to integrate into and proliferate in injured livers in vivo and mature into hepatocytes expressing dipeptidyl peptidase IV or fumarylacetoacetate hydrolase. Together, these findings establish a developmental pathway in embryonic stem cell differentiation cultures that leads to efficient generation of cells with an immature hepatocytic phenotype.
Collapse
Affiliation(s)
- Valerie Gouon-Evans
- Department of Gene and Cell Medicine, Black Family Stem Cell Institute, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, New York 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Zender L, Xue W, Cordón-Cardo C, Hannon GJ, Lucito R, Powers S, Flemming P, Spector MS, Lowe SW. Generation and analysis of genetically defined liver carcinomas derived from bipotential liver progenitors. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2006; 70:251-61. [PMID: 16869761 PMCID: PMC4595853 DOI: 10.1101/sqb.2005.70.059] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Hepatocellular carcinoma is a chemoresistant cancer and a leading cause of cancer mortality; however, the molecular mechanisms responsible for the aggressive nature of this disease are poorly understood. In this study, we developed a new liver cancer mouse model that is based on the ex vivo genetic manipulation of embryonic liver progenitor cells (hepatoblasts). After retroviral gene transfer of oncogenes or short hairpin RNAs targeting tumor suppressor genes, genetically altered liver progenitor cells are seeded into the liver of otherwise normal recipient mice. We show that histopathology of the engineered liver carcinomas reveals features of the human disease. Furthermore, representational oligonucleotide microarray analysis (ROMA) of murine liver tumors initiated by two defined genetic hits revealed spontaneously acquired genetic alterations that are characteristic for human hepatocellular carcinoma. This model provides a powerful platform for applications like cancer gene discovery or high-throughput preclinical drug testing.
Collapse
Affiliation(s)
- L Zender
- Cold Spring Harbor Laboratory, New York 11724, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zender L, Spector MS, Xue W, Flemming P, Cordon-Cardo C, Silke J, Fan ST, Luk JM, Wigler M, Hannon GJ, Mu D, Lucito R, Powers S, Lowe SW. Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach. Cell 2006; 125:1253-67. [PMID: 16814713 PMCID: PMC3026384 DOI: 10.1016/j.cell.2006.05.030] [Citation(s) in RCA: 903] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2005] [Revised: 04/25/2006] [Accepted: 05/26/2006] [Indexed: 12/12/2022]
Abstract
The heterogeneity and instability of human tumors hamper straightforward identification of cancer-causing mutations through genomic approaches alone. Herein we describe a mouse model of liver cancer initiated from progenitor cells harboring defined cancer-predisposing lesions. Genome-wide analyses of tumors in this mouse model and in human hepatocellular carcinomas revealed a recurrent amplification at mouse chromosome 9qA1, the syntenic region of human chromosome 11q22. Gene-expression analyses delineated cIAP1, a known inhibitor of apoptosis, and Yap, a transcription factor, as candidate oncogenes in the amplicon. In the genetic context of their amplification, both cIAP1 and Yap accelerated tumorigenesis and were required to sustain rapid growth of amplicon-containing tumors. Furthermore, cIAP1 and Yap cooperated to promote tumorigenesis. Our results establish a tractable model of liver cancer, identify two oncogenes that cooperate by virtue of their coamplification in the same genomic locus, and suggest an efficient strategy for the annotation of human cancer genes.
Collapse
Affiliation(s)
- Lars Zender
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Mona S. Spector
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Wen Xue
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Peer Flemming
- Department of Pathology, Hannover Medical School, 30625 Hannover, Germany
| | - Carlos Cordon-Cardo
- Division of Molecular Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Sheung-Tat Fan
- Department of Surgery, University of Hong Kong, Hong Kong, China
| | - John M. Luk
- Department of Surgery, University of Hong Kong, Hong Kong, China
| | - Michael Wigler
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Gregory J. Hannon
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Howard Hughes Medical Institute, Cold Spring Harbor, NY 11724, USA
| | - David Mu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Robert Lucito
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Scott Powers
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Scott W. Lowe
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Howard Hughes Medical Institute, Cold Spring Harbor, NY 11724, USA
| |
Collapse
|
44
|
Dan YY, Riehle KJ, Lazaro C, Teoh N, Haque J, Campbell JS, Fausto N. Isolation of multipotent progenitor cells from human fetal liver capable of differentiating into liver and mesenchymal lineages. Proc Natl Acad Sci U S A 2006; 103:9912-7. [PMID: 16782807 PMCID: PMC1502553 DOI: 10.1073/pnas.0603824103] [Citation(s) in RCA: 236] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Little is known about the differentiation capabilities of nonhematopoietic cells of the human fetal liver. We report the isolation and characterization of a human fetal liver multipotent progenitor cell (hFLMPC) population capable of differentiating into liver and mesenchymal cell lineages. Human fetal livers (74-108 days of gestation) were dissociated and maintained in culture. We treated the colonies with geneticin and mechanically isolated hFLMPCs, which were kept in an undifferentiated state by culturing on feeder layers. We derived daughter colonies by serial dilution, verifying monoclonality using the Humara assay. hFLMPCs, which have been maintained in culture for up to 100 population doublings, have a high self-renewal capability with a doubling time of 46 h. The immunophenotype is: CD34+, CD90+, c-kit+, EPCAM+, c-met+, SSEA-4+, CK18+, CK19+, albumin-, alpha-fetoprotein-, CD44h+, and vimentin+. Passage 1 (P1) and P10 cells have identical morphology, immunophenotype, telomere length, and differentiation capacity. Placed in appropriate media, hFLMPCs differentiate into hepatocytes and bile duct cells, as well as into fat, bone, cartilage, and endothelial cells. Our results suggest that hFLMPCs are mesenchymal-epithelial transitional cells, probably derived from mesendoderm. hFLMPCs survive and differentiate into functional hepatocytes in vivo when transplanted into animal models of liver disease. hFLMPCs are a valuable tool for the study of human liver development, liver injury, and hepatic repopulation.
Collapse
Affiliation(s)
| | - K. J. Riehle
- Departments of *Pathology and
- Surgery, University of Washington, Seattle, WA 98115
| | | | - N. Teoh
- Departments of *Pathology and
| | | | | | - N. Fausto
- Departments of *Pathology and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
45
|
Abstract
AIM: To study the effect of retrorsine on mouse he-patocyte proliferation.
METHODS: Mice and rats were treated respectively with two injections of retrorsine (as retrosine-treated group) or saline (as non-treated group) at 2 wk intervals. They received a single injection of carbon tetrachloride (CCl4) 4 wk later. On d 0, 1, 2, 3, 4, 6, 15 after CCl4 administration, the animals were killed and their livers were excised. Hematoxylin and eosin (HE) staining and Ki-67 antibody immunohistochemical analysis of liver samples were used to evaluate the pathological changes and hepatocyte proliferation.
RESULTS: In rats treated with retrorsine and CCl4, the liver displayed obvious megalocytosis, proliferation of mild bile duct, small hepatocyte-forming nodule, which were not found in liver samples from non-treated group. However, in mice treated with retrorsine combined with CCl4, the liver displayed hepatocyte degeneration and necrosis in perivenous areas. There was no obvious difference between retrorsine-treated group and non-treated group. Ki-67 immunohistochemical analysis showed that in rats treated with retrorsine, the positive hepatocytes mainly found in small hepatocyte nodules, were obviously less than those in non-treated group. The mice treated with retrorsine showed that the number of Ki-67 positive hepatocytes was very high and more than that in non-treated group.
CONCLUSION: Retrorsine has no effect on mouse hepatocyte proliferation.
Collapse
Affiliation(s)
- Xiao-Fei Zhou
- Center for Developmental Biology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | | | | | | |
Collapse
|
46
|
Kawashita Y, Guha C, Yamanouchi K, Ito Y, Kamohara Y, Kanematsu T. Liver repopulation: a new concept of hepatocyte transplantation. Surg Today 2006; 35:705-10. [PMID: 16133662 DOI: 10.1007/s00595-005-3024-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2004] [Accepted: 11/16/2004] [Indexed: 11/26/2022]
Abstract
Hepatocyte transplantation has been recognized as an alternative strategy for organ transplantation because the supply of donor livers is limited. However, in conventional hepatocyte transplantation, only 1%-10% of the liver replaced with transplanted hepatocytes. Recently a novel concept termed "liver repopulation" has been established, where the whole recipient liver can be replaced by a small number of donor hepatocytes. To induce liver repopulation, growth advantage of the donor hepatocytes against the host liver seems to be required according to the data of previous studies. Additionally, various cell sources, including bone marrow cells and other stem cells, could potentially be used as donor cells for liver repopulation. In this article, we discuss recent progress and future perspectives of this emerging technology.
Collapse
Affiliation(s)
- Yujo Kawashita
- Department of Transplantation, Digestive Surgery, Nagasaki University Graduate School of Biomedical Science, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Gandillet A, Vidal I, Alexandre E, Audet M, Chenard-Neu MP, Stutzmann J, Heyd B, Jaeck D, Richert L. Experimental models of acute and chronic liver failure in nude mice to study hepatocyte transplantation. Cell Transplant 2005; 14:277-90. [PMID: 16052909 DOI: 10.3727/000000005783983061] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although hepatocyte transplantation is a promising therapy for acute liver failure in human, there is still a lack of animal models suffering from hepatic injury in which the benefits of hepatocyte transplantation could be evaluated solely, without the bias caused by immunosuppression. As a consequence, the aim of the study was first to develop reproducible models of partial hepatectomy and of thioacetamide (TA)- or Jo2-induced acute liver failure in nude mice. Chronic liver disease was also investigated by repeated injections of sublethal doses of thioacetamide. Survival rates, routine histologic observations, alanin aminotransferase sera content, Ki67, and caspase 3 immunodetection were investigated both after 40% partial hepatectomy and after toxic-induced damages. Liver injuries were more severe and/or precocious in nude mice than in Balb/c mice for a given treatment with a maximum of acute injury obtained 24 h after single toxic injection, and were found to be transitory and reversible within 10 days. Toxics induced apoptosis followed by necrosis, confirming recent published data. Onset of fibrosis leading to reproducible chronic cirrhosis in nude mice correlated with increasing number of Ki67-positive cells, indicating that high levels of cell proliferation occurred. Chronic cirrhosis progressively reversed to fibrosis when the treatment ceased. Preliminary results demonstrated that engrafted xenogeneic hepatocytes could be detected in the host liver by anti-MHC class I immunohistochemistry. Fractions enriched in 2n or 4n hepatocytes by cell sorting using a flow cytometer were equivalent to the unpurified fraction in terms of engraftment in control nude mice or in nude mice subjected to PH. However, in mice suffering from liver injury 24 h after Jo2 or TA treatment, the engraftment of 2n hepatocytes was about twice that of an unpurified hepatocyte population or of a population enriched in 4n hepatocytes.
Collapse
Affiliation(s)
- Amaud Gandillet
- Laboratoire de Chirurgie Expérimentale, Fondation Transplantation, 67200 Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kim KS, Joseph B, Inada M, Gupta S. Regulation of Hepatocyte Engraftment and Proliferation after Cytotoxic Drug-Induced Perturbation of the Rat Liver. Transplantation 2005; 80:653-9. [PMID: 16177641 DOI: 10.1097/01.tp.0000173382.11916.bf] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Perturbations in specific liver cell compartments benefit transplanted cell engraftment and/or proliferation. We analyzed whether cytotoxic drugs interfering with the integrity of genomic DNA or cell division could be useful for liver cell transplantation. METHODS We used dipeptidyl peptidase IV deficient (DPPIV-) rats as recipients of syngeneic F344 rat hepatocytes. Rats were pretreated with doxorubicin, irinotecan, or vincristine prior to cell transplantation and synergistic liver perturbations were induced by drug administration followed by partial hepatectomy or carbon tetrachloride treatments. Transplanted cells were identified by DPPIV histochemistry and cell engraftment and proliferation were analyzed morphometrically. Perturbations in endothelial, Kupffer cell, and hepatocyte compartments were analyzed by electron microscopy, carbon incorporation, and blood tests, respectively. RESULTS Cell engraftment was improved in rats treated with doxorubicin but not with irinotecan or vincristine. Doxorubicin disrupted endothelial cells for up to seven days without causing Kupffer cell or hepatocellular toxicity. Neither doxorubicin nor vincristine induced liver repopulation in animals up to three months, including after partial hepatectomy or carbon tetrachloride-induced additional liver injury. CONCLUSIONS Doxorubicin-induced hepatic endothelial damage enhanced cell engraftment, which should be useful in cell therapy strategies.
Collapse
Affiliation(s)
- Kyung Sik Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
49
|
Witek RP, Fisher SH, Petersen BE. Monocrotaline, an alternative to retrorsine-based hepatocyte transplantation in rodents. Cell Transplant 2005; 14:41-7. [PMID: 15789661 DOI: 10.3727/000000005783983278] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Retrorsine has been used extensively to inhibit proliferation of resident hepatocytes in various transplantation models. Here we report a successful alternative to currently unavailable retrorsine that can be used in cellular transplantation models. Based on structural and molecular similarities, we investigate the use of monocrotaline (MCT) in cell transplantation studies in rodents. In this study, MCT was given to rats intraperitoneally in two injections 2 weeks apart. Two weeks after the final injection, a partial hepatectomy followed by splenic hepatocyte transplantation was performed. The results indicate that MCT, at two doses of 30 mg/kg, highly enhances liver repopulation by donor hepatocytes following partial hepatectomy and produces 15.3 +/- 4.9% liver repopulation within the first 6 weeks following transplantation. Additionally, we tested the effectiveness of MCT in a murine model. Using two injections of 50 mg/kg each, given 2 weeks apart, hepatocyte proliferation in the native liver was inhibited and subsequent oval cell transplants engrafted at 18 +/- 21.3% after 16 weeks posttransplantation. In conclusion, MCT can be used as an effective selective pressure for donor hepatocytes in cell transplantation to the liver in rodents.
Collapse
Affiliation(s)
- Rafal P Witek
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610-0275, USA
| | | | | |
Collapse
|
50
|
Li WL, Su J, Yao YC, Tao XR, Yan YB, Yu HY, Wang XM, Li JX, Yang YJ, Lau JTY, Hu YP. Isolation and characterization of bipotent liver progenitor cells from adult mouse. Stem Cells 2005; 24:322-32. [PMID: 16109753 DOI: 10.1634/stemcells.2005-0108] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Liver progenitor cells have drawn a great deal of attention both for their therapeutic potential and for their usefulness in exploring the molecular events surrounding liver development and regeneration. Despite the intensive studies on liver progenitors from rats, equivalent progenitor cells derived from mice are relatively rare. We used retrosine treatment followed by partial hepatectomy to elicit liver progenitors in mice. From these animals showing prominent ductular reactions, mouse-derived liver progenitor cell lines (LEPCs) were isolated by single-cell cloning. Phenotypic and lineage profiling of the LEPC clones were performed using immunochemistry, reverse transcription-polymerase chain reaction, and a dual-color system comprising the reporter EGFP under the control of the cytokeratin 19 promoter and the DsRed reporter under the control of the albumin promoter. LEPCs expressed liver progenitor cell markers. LEPCs also expressed some markers shared by bone marrow-derived hematopoietic stem cells c-Kit and Thy-1 but not CD34 and CD45. When cultured as aggregates in Matrigel, LEPCs differentiated into hepatocyte upon treatment with 50 ng/ml epithelial growth factor or differentiated into biliary lineage cells upon treatment with 20 ng/ml hepatocyte growth factor. In the presence of 2% dimethyl sulfoxide and 2% Matrigel, LEPCs acquired predominantly bile lineage phenotypes, with occasional patches of cells exhibiting hepatocyte phenotypes. Upon transplantation into CCl4-injured-liver, LEPCs engrafted into liver parenchyma and differentiated into hepatocytes. Considering the amenability of the mouse to genetic manipulation, these mouse-derived LEPCs may be useful tools as in vitro models to study molecular events in liver development and regeneration and can shed light in studying the therapy potential of liver stem cells.
Collapse
Affiliation(s)
- Wen-Lin Li
- Department of Cell Biology, Second Military Medical University, Xiangyin Road 800, Shanghai 200433, P.R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|