1
|
Anand AC, Acharya SK. The Story of Ammonia in Liver Disease: An Unraveling Continuum. J Clin Exp Hepatol 2024; 14:101361. [PMID: 38444405 PMCID: PMC10910335 DOI: 10.1016/j.jceh.2024.101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/03/2024] [Indexed: 03/07/2024] Open
Abstract
Hyperammonemia and liver disease are closely linked. Most of the ammonia in our body is produced by transamination and deamination activities involving amino acid, purine, pyrimidines, and biogenic amines, and from the intestine by bacterial splitting of urea. The only way of excretion from the body is by hepatic conversion of ammonia to urea. Hyperammonemia is associated with widespread toxicities such as cerebral edema, hepatic encephalopathy, immune dysfunction, promoting fibrosis, and carcinogenesis. Over the past two decades, it has been increasingly utilized for prognostication of cirrhosis, acute liver failure as well as acute on chronic liver failure. The laboratory assessment of hyperammonemia has certain limitations, despite which its value in the assessment of various forms of liver disease cannot be negated. It may soon become an important tool to make therapeutic decisions about the use of prophylactic and definitive treatment in various forms of liver disease.
Collapse
|
2
|
Huang C, Zhao Y, Ye Q, Gleason J, Rousseva V, Stout B, Lin S, Hariri R, Zhang X, He S. Characterization of CRISPR/Cas9-edited human placental allogenic stromal cells with low tissue factor expression and reduced thrombotic effects. Cytotherapy 2023; 25:1265-1270.e2. [PMID: 37256239 DOI: 10.1016/j.jcyt.2023.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/21/2023] [Accepted: 04/28/2023] [Indexed: 06/01/2023]
Abstract
The tissue factor (TF/CD142) expressed by mesenchymal stromal cells (MSCs) has been regarded as a safety concern in clinical applications as it may trigger thrombosis when MSCs administered intravenously. Human placental allogenic stromal cells (ASCs) are culture-expanded, undifferentiated MSC-like cells derived from full-term postpartum placenta and possess immunomodulatory and pro-angiogenic activities, however, express TF. Here we performed CRISPR/Cas9-mediated TF gene knock out (TFKO) in ASCs, leading to significantly lower TF expression, activity and thrombotic effects. ASCs' characteristics including expansion, expression of phenotypic markers and secretory profile remained unchanged in edited cells, and their immunomodulatory activities, which are functionally relevant to therapeutic applications, were not affected upon TFKO. Taken together, this study provides a feasible strategy which could improve the clinical safety features of MSC-based cell therapy by CRISRP/Cas9-mediated TF gene knock out.
Collapse
Affiliation(s)
| | | | - Qian Ye
- Celularity Inc., Florham Park, New Jersey, USA
| | | | | | | | | | | | | | - Shuyang He
- Celularity Inc., Florham Park, New Jersey, USA
| |
Collapse
|
3
|
Al Reza H, Okabe R, Takebe T. Organoid transplant approaches for the liver. Transpl Int 2021; 34:2031-2045. [PMID: 34614263 PMCID: PMC8602742 DOI: 10.1111/tri.14128] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/13/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Organoid technology is a state-of-the-art cell culture tool that has revolutionized study of development, regeneration, and diseases. Human liver organoids (HLOs) are now derived from either adult stem/progenitors or pluripotent stem cells (PSCs), emulating cellular diversity and structural symphony akin to the human liver. With the rapid rise in decompensated liver disease conditions only treated by liver transplant therapy, HLOs represent an alternate source for transplantation to address the ongoing shortage of grafts. Although ongoing advancements in bioengineering technology have moved the organoid transplant approach to the next level, sustained survival of the transplanted tissue still eludes us toward functional organ replacement. Herein, we review the development of HLOs and discuss promises and challenges on organoid transplant approaches.
Collapse
Affiliation(s)
- Hasan Al Reza
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229-3039, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | - Ryo Okabe
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanori Takebe
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229-3039, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229-3039, USA
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229-3039, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Communication Design Center, Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Japan
| |
Collapse
|
4
|
Tang Y, Kong Y. Hereditary tyrosinemia type Ⅰ: newborn screening, diagnosis and treatment. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:514-523. [PMID: 34704422 PMCID: PMC8777462 DOI: 10.3724/zdxbyxb-2021-0255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/17/2021] [Indexed: 11/25/2022]
Abstract
Hereditary tyrosinemia type Ⅰ (HT-1) is a severe autosomal recessive inherited metabolic disease. Due to the deficiency of fumarylacetoacetase hydrolase (FAH), the toxic metabolites are accumulated in the body, resulting in severe liver dysfunction, renal tubular dysfunctions, neurological crises, and the increased risk of hepatocellular carcinoma. Clinical symptoms typically begin at after the birth; the prognosis of patients is poor if they are not treated timely. Succinylacetone is a specific and sensitive marker for HT-1, and the screening in newborns can make early diagnosis of HT-1 at the asymptomatic stage. The diagnosis of HT-1 can be confirmed based on the characteristic biochemical findings and molecular testing of mutations in both alleles of gene. Combined treatment with nitisinone and a low tyrosine diet may significantly improve outcomes for patients. Liver transplantation is an effective treatment in cases where nitisinone is not available. Some novel HT-1 treatments are in clinical trials, including enzyme replacement therapy, hepatocyte transplantation and gene-targeted therapy.
Collapse
Affiliation(s)
- Yue Tang
- Department of Newborn Screening, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100020, China
| | - Yuanyuan Kong
- Department of Newborn Screening, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100020, China
| |
Collapse
|
5
|
Tsuchida T, Murata S, Hasegawa S, Mikami S, Enosawa S, Hsu HC, Fukuda A, Okamoto S, Mori A, Matsuo M, Kawakatsu Y, Matsunari H, Nakano K, Nagashima H, Taniguchi H. Investigation of Clinical Safety of Human iPS Cell-Derived Liver Organoid Transplantation to Infantile Patients in Porcine Model. Cell Transplant 2021; 29:963689720964384. [PMID: 33103476 PMCID: PMC7784600 DOI: 10.1177/0963689720964384] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transplantation of liver organoids has been investigated as a treatment alternative to liver transplantation for chronic liver disease. Transportal approach can be considered as a method of delivering organoids to the liver. It is important to set the allowable organoid amount and verify translocation by intraportal transplantation. We first examined the transplantation tolerance and translocation of porcine fetal liver-derived allogeneic organoids using piglets. Fetal liver-derived organoids generated from the Kusabira Orange-transduced pig were transplanted to the 10-day-old piglet liver through the left branch of the portal vein. All recipients survived without any observable adverse events. In contrast, both local and main portal pressures increased transiently during transplantation. In necropsy samples, Kusabira Orange-positive donor cells were detected primarily in the target lobe of the liver and partly in other areas, including the lungs and brain. As we confirmed the transplantation allowance by porcine fetal liver-derived organoids, we performed intraportal transplantation of human-induced pluripotent stem cell (iPSC)-derived liver organoid, which we plan to use in clinical trials, and portal pressure and translocation were investigated. Human iPSC-derived liver organoids were transplanted into the same 10-day-old piglet. Portal hypertension and translocation of human iPSC-derived liver organoids to the lungs were observed in one of two transplanted animals. Translocation occurred in the piglet in which patent ductus venosus (PDV) was observed. Therefore, a 28-day-old piglet capable of surgically ligating PDV was used, and after the PDV was ligated, human iPSC-derived liver organoids with the amount of which is scheduled in clinical trials were transplanted. This procedure inhibited the translocation of human iPSC-derived liver organoids to extrahepatic sites without no portal hypertension. In conclusion, human iPSC-derived liver organoids can be safely transplanted through the portal vein. Ligation of the ductus venosus prior to transplantation was effective in inhibiting extrahepatic translocation in newborns and infants.
Collapse
Affiliation(s)
- Tomonori Tsuchida
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Soichiro Murata
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shunsuke Hasegawa
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoshi Mikami
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shin Enosawa
- Division for Advanced Medical Sciences, National Center for Child Health and Development, Tokyo, Japan
| | - Huai-Che Hsu
- Division for Advanced Medical Sciences, National Center for Child Health and Development, Tokyo, Japan
| | - Akinari Fukuda
- Department of Transplantation Surgery, Organ Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akihiro Mori
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Megumi Matsuo
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yumi Kawakatsu
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hitomi Matsunari
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Kazuaki Nakano
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Hiroshi Nagashima
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Division of Regenerative Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Immuno-comparative screening of adult-derived human liver stem/progenitor cells for immune-inflammatory-associated molecules. Inflamm Res 2021; 70:229-239. [PMID: 33404674 DOI: 10.1007/s00011-020-01428-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 11/04/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE One of the main challenges in liver cell therapy is the replacement of damaged cells and the induction of a tolerogenic microenvironment to promote graft acceptance by the recipient. Adult-derived human liver stem/progenitor cells (ADHLSCs) are currently evaluated at the clinical levels as a promising pro-regenerative and immune-modulatory tool. The expression profile of several immunological molecules may influence the local immune-inflammatory response and, therefore, modulate the tissue healing process. To increase the quality and safety of ADHLSCs before transplantation requires an appropriate analysis and characterization of their pattern expression of immune-inflammatory-associated molecules. METHODS The expression of 27 molecules belonging to T-cell co-stimulatory pathway, CD47 partners, Ikaros family, CD300 family and TNF family were analyzed using flow cytometry. We compared their expression profiles to PBMCs, hepatocytes and ADHLSCs in both expansion and after hepatogenic differentiation culture conditions. RESULTS This original immuno-comparative screening revealed that liver cell populations do not constitutively present significant immunological pattern compared to PBMCs. Moreover, our findings highlight that neither the expansion nor the hepatogenic differentiation induces the expression of immune-inflammatory molecules. The detailed expression characteristics (percentage of positive cells and median fluorescence intensity) of each molecule were analyzed and presented. CONCLUSION By analyzing 27 relevant molecules, our immuno-comparative screening demonstrates that ADHLSCs keep a non-immunogenic profile independent of their expansion or hepatogenic differentiation state. Accordingly, the immunological profile of ADHLSCs seems to support their safe and efficient use in liver tissue therapeutic repair strategy.
Collapse
|
7
|
Najimi M. Cell- and Stem Cell-Based Therapies for Liver Defects: Recent Advances and Future Strategies. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
8
|
Meneghel J, Kilbride P, Morris GJ. Cryopreservation as a Key Element in the Successful Delivery of Cell-Based Therapies-A Review. Front Med (Lausanne) 2020; 7:592242. [PMID: 33324662 PMCID: PMC7727450 DOI: 10.3389/fmed.2020.592242] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022] Open
Abstract
Cryopreservation is a key enabling technology in regenerative medicine that provides stable and secure extended cell storage for primary tissue isolates and constructs and prepared cell preparations. The essential detail of the process as it can be applied to cell-based therapies is set out in this review, covering tissue and cell isolation, cryoprotection, cooling and freezing, frozen storage and transport, thawing, and recovery. The aim is to provide clinical scientists with an overview of the benefits and difficulties associated with cryopreservation to assist them with problem resolution in their routine work, or to enable them to consider future involvement in cryopreservative procedures. It is also intended to facilitate networking between clinicians and cryo-researchers to review difficulties and problems to advance protocol optimization and innovative design.
Collapse
Affiliation(s)
- Julie Meneghel
- Asymptote, Cytiva, Danaher Corporation, Cambridge, United Kingdom
| | - Peter Kilbride
- Asymptote, Cytiva, Danaher Corporation, Cambridge, United Kingdom
| | | |
Collapse
|
9
|
El-Kehdy H, Najar M, De Kock J, Agha DM, Rogiers V, Merimi M, Lagneaux L, Sokal EM, Najimi M. Inflammation Differentially Modulates the Biological Features of Adult Derived Human Liver Stem/Progenitor Cells. Cells 2020; 9:cells9071640. [PMID: 32650454 PMCID: PMC7408415 DOI: 10.3390/cells9071640] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
The progression of mesenchymal stem cell-based therapy from concept to cure closely depends on the optimization of conditions that allow a better survival and favor the cells to achieve efficient liver regeneration. We have previously demonstrated that adult-derived human liver stem/progenitor cells (ADHLSC) display significant features that support their clinical development. The current work aims at studying the impact of a sustained pro-inflammatory environment on the principal biological features of ADHLSC in vitro. METHODS: ADHLSC from passages 4–7 were exposed to a cocktail of inflammatory cytokines for 24 h and 9 days and subsequently analyzed for their viability, expression, and secretion profiles by using flow cytometry, RT-qPCR, and antibody array assay. The impact of inflammation on the hepatocytic differentiation potential of ADHLSC was also evaluated. RESULTS: ADHLSC treated with a pro-inflammatory cocktail displayed significant decrease of cell yield at both times of treatment while cell mortality was observed at 9 days post-priming. After 24 h, no significant changes in the immuno-phenotype of ADHLSC expression profile could be noticed while after 9 days, the expression profile of relevant markers has changed both in the basal conditions and after inflammation treatment. Inflammation cocktail enhanced the release of IL-6, IL-8, CCL5, monocyte-chemo-attractant protein-2 and 3, CXCL1/GRO, and CXCL5/ENA78. Furthermore, while IP-10 secretion was increased after 24 h priming, granulocyte macrophage colony-stimulating factor enhanced secretion was noticed after 9 days treatment. Finally, priming of ADHLSC did not affect their potential to differentiate into hepatocyte-like cells. CONCLUSION: These results indicate that ADHLSCs are highly sensitive to inflammation and respond to such signals by adjusting their gene and protein expression. Accordingly, monitoring the inflammatory status of patients at the time of cell transplantation, will certainly help in enhancing ADHLSC safety and efficiency.
Collapse
Affiliation(s)
- Hoda El-Kehdy
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (H.E.-K.); (E.M.S.)
| | - Mehdi Najar
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada;
| | - Joery De Kock
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (J.D.K.); (V.R.)
| | - Douaa Moussa Agha
- Laboratory of Experimental Hematology (HEMEXP), Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium; (D.M.A.); (M.M.)
| | - Vera Rogiers
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (J.D.K.); (V.R.)
| | - Makram Merimi
- Laboratory of Experimental Hematology (HEMEXP), Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium; (D.M.A.); (M.M.)
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy (LCCT), Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
| | - Etienne M. Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (H.E.-K.); (E.M.S.)
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (H.E.-K.); (E.M.S.)
- Correspondence:
| |
Collapse
|
10
|
Tricot T, De Boeck J, Verfaillie C. Alternative Cell Sources for Liver Parenchyma Repopulation: Where Do We Stand? Cells 2020; 9:E566. [PMID: 32121068 PMCID: PMC7140465 DOI: 10.3390/cells9030566] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/20/2020] [Accepted: 02/22/2020] [Indexed: 12/28/2022] Open
Abstract
Acute and chronic liver failure is a highly prevalent medical condition with high morbidity and mortality. Currently, the therapy is orthotopic liver transplantation. However, in some instances, chiefly in the setting of metabolic diseases, transplantation of individual cells, specifically functional hepatocytes, can be an acceptable alternative. The gold standard for this therapy is the use of primary human hepatocytes, isolated from livers that are not suitable for whole organ transplantations. Unfortunately, primary human hepatocytes are scarcely available, which has led to the evaluation of alternative sources of functional hepatocytes. In this review, we will compare the ability of most of these candidate alternative cell sources to engraft and repopulate the liver of preclinical animal models with the repopulation ability found with primary human hepatocytes. We will discuss the current shortcomings of the different cell types, and some of the next steps that we believe need to be taken to create alternative hepatocyte progeny capable of regenerating the failing liver.
Collapse
|
11
|
Improved in vivo efficacy of clinical-grade cryopreserved human hepatocytes in mice with acute liver failure. Cytotherapy 2020; 22:114-121. [PMID: 31987755 DOI: 10.1016/j.jcyt.2019.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/04/2019] [Accepted: 12/08/2019] [Indexed: 12/21/2022]
Abstract
Clinical hepatocyte transplantation short-term efficacy has been demonstrated; however, some major limitations, mainly due to the shortage of organs, the lack of quality of isolated cells and the low cell engraftment after transplantation, should be solved for increasing its efficacy in clinical applications. Cellular stress during isolation causes an unpredictable loss of attachment ability of the cells, which can be aggravated by cryopreservation and thawing. In this work, we focused on the use of a Good Manufacturing Practice (GMP) solution compared with the standard cryopreservation medium, the University of Wisconsin medium, for the purpose of improving the functional quality of cells and their ability to engraft in vivo, with the idea of establishing a biobank of cryopreserved human hepatocytes available for their clinical use. We evaluated not only cell viability but also specific hepatic function indicators of the functional performance of the cells such as attachment efficiency, ureogenic capability, phase I and II enzymes activities and the expression of specific adhesion molecules in vitro. Additionally, we also assessed and compared the in vivo efficacy of human hepatocytes cryopreserved in different media in an animal model of acute liver failure. Human hepatocytes cryopreserved in the new GMP solution offered better in vitro and in vivo functionality compared with those cryopreserved in the standard medium. Overall, the results indicate that the new tested GMP solution maintains better hepatic functions and, most importantly, shows better results in vivo, which could imply an increase in long-term efficacy when used in patients.
Collapse
|
12
|
Tsuchida T, Murata S, Matsuki K, Mori A, Matsuo M, Mikami S, Okamoto S, Ueno Y, Tadokoro T, Zheng YW, Taniguchi H. The Regenerative Effect of Portal Vein Injection of Liver Organoids by Retrorsine/Partial Hepatectomy in Rats. Int J Mol Sci 2019; 21:ijms21010178. [PMID: 31887985 PMCID: PMC6981799 DOI: 10.3390/ijms21010178] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022] Open
Abstract
In this study, we reveal that liver organoid transplantation through the portal vein is a safe and effective method for the treatment of chronic liver damage. The liver organoids significantly reconstituted the hepatocytes; hence, the liver was significantly enlarged in this group, compared to the monolayer cell transplantation group in the retrorsine/partial hepatectomy (RS/PH) model. In the liver organoid transplantation group, the bile ducts were located in the donor area and connected to the recipient bile ducts. Thus, the rate of bile reconstruction in the liver was significantly higher compared to that in the monolayer group. By transplanting liver organoids, we saw a level of 70% replacement of the damaged liver. Consequently, in the transplantation group, diminished ductular reaction and a decrease of placental glutathione S-transferase (GST-p) precancerous lesions were observed. After trans-portal injection, the human induced pluripotent stem cell (hiPSC)-derived liver organoids revealed no translocation outside the liver; in contrast, the monolayer cells had spread to the lungs. The hiPSC-derived liver organoids were attached to the liver in the immunodeficient RS/PH rats. This study clearly demonstrates that liver organoid transplantation through the portal vein is a safe and effective method for the treatment of chronic liver damage in rats.
Collapse
Affiliation(s)
- Tomonori Tsuchida
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Soichiro Murata
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Correspondence: (S.M.); (H.T.); Tel./Fax: +81-45-787-8963 (S.M. & H.T.)
| | - Koichiro Matsuki
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Akihiro Mori
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Megumi Matsuo
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Satoshi Mikami
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Yasuharu Ueno
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Tomomi Tadokoro
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Yun-Wen Zheng
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Department of Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Correspondence: (S.M.); (H.T.); Tel./Fax: +81-45-787-8963 (S.M. & H.T.)
| |
Collapse
|
13
|
Natural Flavonol, Myricetin, Enhances the Function and Survival of Cryopreserved Hepatocytes In Vitro and In Vivo. Int J Mol Sci 2019; 20:ijms20246123. [PMID: 31817281 PMCID: PMC6940939 DOI: 10.3390/ijms20246123] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/28/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022] Open
Abstract
To improve the therapeutic potential of hepatocyte transplantation, the effects of the mitogen-activated protein kinase kinase 4 (MKK4) inhibitor, myricetin (3,3′,4′,5,5′,7-hexahydroxylflavone) were examined using porcine and human hepatocytes in vitro and in vivo. Hepatocytes were cultured, showing the typical morphology of hepatic parenchymal cell under 1–10 µmol/L of myricetin, keeping hepatocyte specific gene expression, and ammonia removal activity. After injecting the hepatocytes into neonatal Severe combined immunodeficiency (SCID) mouse livers, cell colony formation was found at 10–15 weeks after transplantation. The human albumin levels in the sera of engrafted mice were significantly higher in the recipients of myricetin-treated cells than non-treated cells, corresponding to the size of the colonies. In terms of therapeutic efficacy, the injection of myricetin-treated hepatocytes significantly prolonged the survival of ornithine transcarbamylase-deficient SCID mice from 32 days (non-transplant control) to 54 days. Biochemically, the phosphorylation of MKK4 was inhibited in the myricetin-treated hepatocytes. These findings suggest that myricetin has a potentially therapeutic benefit that regulates hepatocyte function and survival, thereby treating liver failure.
Collapse
|
14
|
Human Hepatocytes and Differentiated Adult-Derived Human Liver Stem/Progenitor Cells Display In Vitro Immunosuppressive Properties Mediated, at Least in Part, through the Nonclassical HLA Class I Molecule HLA-G. J Immunol Res 2019; 2019:8250584. [PMID: 31612154 PMCID: PMC6757295 DOI: 10.1155/2019/8250584] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/08/2019] [Indexed: 12/25/2022] Open
Abstract
One of the main challenges in liver cell therapy (LCT) is the induction of a tolerogenic microenvironment to promote graft acceptance in the recipient. Little is known about the immunomodulatory potential of the hepatic cells used in liver cell therapy. In this work, we wanted to evaluate the immunosuppressive properties of human hepatocytes and adult-derived human liver stem/progenitor cells (ADHLSCs), as well as the potential involvement of the immunomodulatory molecule HLA-G. We demonstrated that both cell types were capable of inhibiting the proliferative response of PBMCs to an allogenic stimulus and that the immune inhibitory potential of ADHLSCs, although lower than that of hepatocytes, increased after hepatogenic differentiation. We demonstrated that liver cells express HLA-G and that the immune inhibition pattern was clearly associated to its expression. Interestingly, HLA-G expression increased after the third step of differentiation, wherein oncostatin M (OSM) was added. A 48 hr treatment with OSM was sufficient to induce HLA-G expression in ADHLSCs and result in immune inhibition. Surprisingly, blocking HLA-G partially reversed the immune inhibition mediated by hepatocytes and differentiated ADHLSCs, but not that of undifferentiated ADHLSCs, suggesting that additional immune inhibitory mechanisms may be used by these cells. In conclusion, we demonstrated that both hepatocytes and ADHLSCs present immunomodulatory properties mediated, at least in part, through HLA-G, which can be upregulated following hepatogenic differentiation or liver cell pretreatment with OSM. These observations open up new perspectives for the induction of tolerance following LCT and for potential therapeutic applications of these liver cells.
Collapse
|
15
|
Abstract
Alagille syndrome is a complex multisystem autosomal dominant disorder with a wide variability in penetrance of clinical features. A majority of patients have pathogenic mutations in either the JAG1 gene, encoding a Notch pathway ligand, or the receptor NOTCH2. No genotype-phenotype correlations have been found in any organ system. Liver disease is a major cause of morbidity in this population, whereas cardiac and vascular involvement accounts for most of the mortality. Current therapies are supportive, but the future is promising for the development of targeted interventions to augment Notch pathway signaling in involved tissues.
Collapse
Affiliation(s)
- Ellen Mitchell
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Melissa Gilbert
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Kathleen M Loomes
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Demaret T, Varma S, Stephenne X, Smets F, Scheers I, Wanders R, Van Maldergem L, Reding R, Sokal E. Living-donor liver transplantation for mild Zellweger spectrum disorder: Up to 17 years follow-up. Pediatr Transplant 2018; 22:e13112. [PMID: 29453832 DOI: 10.1111/petr.13112] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2017] [Indexed: 12/12/2022]
Abstract
Mild Zellweger spectrum disorder, also described as Infantile Refsum disease, is attributable to mutations in PEX genes. Its clinical course is characterized by progressive hearing and vision loss, and neurodevelopmental regression. Supportive management is currently considered the standard of care, as no treatment has shown clinical benefits. LT was shown to correct levels of circulating toxic metabolites, partly responsible for chronic neurological impairment. Of three patients having undergone LT for mild ZSD, one died after LT, while the other two displayed significant neurodevelopmental improvement on both the long-term (17 years post-LT) and short-term (9 months post-LT) follow-up. We documented a sustained improvement of biochemical functions, with a complete normalization of plasma phytanic, pristanic, and pipecolic acid levels. This was associated with stabilization of hearing and visual functions, and improved neurodevelopmental status, which has enabled the older patient to lead a relatively autonomous lifestyle on the long term. The psychomotor acquisitions have been markedly improved as compared to their affected siblings, who did not undergo LT and exhibited a poor neurological outcome with severe disabilities. We speculate that LT performed before the onset of severe sensorineural defects in mild ZSD enables partial metabolic remission and improved long-term clinical outcomes.
Collapse
Affiliation(s)
- Tanguy Demaret
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Sharat Varma
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Xavier Stephenne
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Françoise Smets
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Isabelle Scheers
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Ronald Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, the Netherlands
| | - Lionel Van Maldergem
- Centre de Génétique Humaine, Centre Hospitalier Régional Universitaire de Besançon, Université de Franche-Comté, Besançon, France
| | - Raymond Reding
- Unité de Chirurgie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Etienne Sokal
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
17
|
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: 7.8] [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
|
18
|
Anderson TN, Zarrinpar A. Hepatocyte transplantation: past efforts, current technology, and future expansion of therapeutic potential. J Surg Res 2018; 226:48-55. [PMID: 29661288 DOI: 10.1016/j.jss.2018.01.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 11/16/2017] [Accepted: 01/17/2018] [Indexed: 12/16/2022]
Abstract
Hepatic cell transplantation (HCT) continues to garner interest as an alternative to orthotopic liver transplantation and the attendant donor shortage. When compared with solid organ transplantation, advantages of cell transplantation include the potential to treat more patients with a considerably less invasive procedure, the ability to utilize organs otherwise unsuitable for transplant, and leaving the native organ in situ with the potential for regeneration. While studies date back to the early 1960s, advancement of clinical application has been slow due in part to limitations of suitable tissue supplies and reproducible robust techniques. Compared with orthotopic liver transplantation, there are fewer absolute contraindications for donor selection. And, current techniques used to harvest, isolate, store, and even transfuse cells vary little between institutions. Significant variation is seen due to a lack of consensus with maintenance therapy. Although the ideal recipient has not been clearly identified, the most significant results have been demonstrated with correction of congenital metabolic liver disorders, with a few trials examining its utility in cirrhotics and more recently acute liver failure. The most exciting new topic of discussion examines techniques to improve engraftment, with many such as ischemic preconditioning and nonselective partial embolization (microbead therapy), while not yet used in HCT study, showing promise in solid organ research. Advancements in HCT, although slow in progress, have great potential in the ability to alleviate the burden faced in solid organ transplantation and possibly become a long-term viable option, beyond that of a bridge or salvage therapy.
Collapse
Affiliation(s)
- Tiffany N Anderson
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Ali Zarrinpar
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida.
| |
Collapse
|
19
|
Oeller M, Laner-Plamberger S, Hochmann S, Ketterl N, Feichtner M, Brachtl G, Hochreiter A, Scharler C, Bieler L, Romanelli P, Couillard-Despres S, Russe E, Schallmoser K, Strunk D. Selection of Tissue Factor-Deficient Cell Transplants as a Novel Strategy for Improving Hemocompatibility of Human Bone Marrow Stromal Cells. Am J Cancer Res 2018; 8:1421-1434. [PMID: 29507631 PMCID: PMC5835947 DOI: 10.7150/thno.21906] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 12/09/2017] [Indexed: 12/14/2022] Open
Abstract
Intravascular transplantation of tissue factor (TF)-bearing cells elicits an instant blood-mediated inflammatory reaction (IBMIR) resulting in thrombotic complications and reduced engraftment. Here we studied the hemocompatibility of commonly used human white adipose tissue (WAT), umbilical cord (UC) and bone marrow stromal cells (BMSC) and devised a possible strategy for safe and efficient stromal cell transplantation. Methods: Stromal cell identity, purity, and TF expression was tested by RTQ-PCR, flow cytometry and immunohistochemistry. Pro-coagulant activity and fibrin clot formation/stabilization was measured In Vitro by viscoelastic rotational plasma-thromboelastometry and in vivo by injecting sorted human stromal cells intravenously into rats. The impact of TF was verified in factor VII-deficient plasma and by sort-depleting TF/CD142+ BMSC. Results: We found significantly less TF expression by a subpopulation of BMSC corresponding to reduced pro-coagulant activity. UC and WAT stroma showed broad TF expression and durable clotting. Higher cell numbers significantly increased clot formation partially dependent on coagulation factor VII. Depleting the TF/CD142+ subpopulation significantly ameliorated BMSC's hemocompatibility without affecting immunomodulation. TF-deficient BMSC did not produce thromboembolism in vivo, comparing favorably to massive intravascular thrombosis induction by TF-expressing stromal cells. Conclusion: We demonstrate that plasma-based thromboelastometry provides a reliable tool to detect pro-coagulant activity of therapeutic cells. Selecting TF-deficient BMSC is a novel strategy for improving cell therapy applicability by reducing cell dose-dependent IBMIR risk. The particularly strong pro-coagulant activity of UC and WAT preparations sounds an additional note of caution regarding uncritical systemic application of stromal cells, particularly from non-hematopoietic extravascular sources.
Collapse
|
20
|
The Pharmabiotic Approach to Treat Hyperammonemia. Nutrients 2018; 10:nu10020140. [PMID: 29382084 PMCID: PMC5852716 DOI: 10.3390/nu10020140] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 12/12/2022] Open
Abstract
Ammonia is constantly produced as a metabolic waste from amino acid catabolism in mammals. Ammonia, the toxic waste metabolite, is resolved in the liver where the urea cycle converts free ammonia to urea. Liver malfunctions cause hyperammonemia that leads to central nervous system (CNS) dysfunctions, such as brain edema, convulsions, and coma. The current treatments for hyperammonemia, such as antibiotics or lactulose, are designed to decrease the intestinal production of ammonia and/or its absorption into the body and are not effective, besides being often accompanied by side effects. In recent years, increasing evidence has shown that modifications of the gut microbiota could be used to treat hyperammonemia. Considering the role of the gut microbiota and the physiological characteristics of the intestine, the removal of ammonia from the intestine by modulating the gut microbiota would be an ideal approach to treat hyperammonemia. In this review, we discuss the significance of hyperammonemia and its related diseases and the efficacy of the current management methods for hyperammonemia to understand the mechanism of ammonia transport in the human body. The possibility to use the gut microbiota as pharmabiotics to treat hyperammonemia and its related diseases is also explored.
Collapse
|
21
|
Immuno-biological comparison of hepatic stellate cells in a reverted and activated state. Biomed Pharmacother 2017; 98:52-62. [PMID: 29245066 DOI: 10.1016/j.biopha.2017.12.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/29/2017] [Accepted: 12/07/2017] [Indexed: 12/20/2022] Open
Abstract
Human hepatic stellate cells (HSCs) demonstrated great immunological plasticity with important consequences for liver cell therapy. Activated HSCs (aHSCs) are in vitro reverted (rHSCs) to a quiescent-like phenotype with potential benefit to reduce liver fibrosis. The goal of this study is to establish and compare the immunological profile of activated and in vitro reverted HSCs and to investigate the impact of inflammatory priming on the immunobiology of both HSCs populations. The distribution of inflammatory primed activated and reverted HSCs across the different phases of the cell cycle is assessed by flow cytometry. In addition, Flow analysis was done to assess the expression level of neuronal, endothelial and stromal markers, cell adhesion molecules, human leucocyte antigens, co-stimulatory molecules, immunoregulatory molecules and natural killer ligands. Our results showed that the cell cycle distribution of both HSCs populations is significantly modulated by inflammation. Accordingly, activated HSC that were in G1 phase switch to S- and G2 phases when exposed to inflammation, while reverted HSCs mostly redistribute into sub-G0 phase. In a HSC state dependent manner, inflammatory priming modulated the expression of the stromal marker CD90, biological receptors (CD95 and CD200R), cell adhesion molecules (CD29, CD54, CD58, CD106 and CD166), human leucocyte antigen HLA-G, co-stimulatory molecules (CD40 and CD252), as well as the immunoregulatory molecules (CD200 and CD274). In conclusion, the immunologic profile of HSCs is significantly modulated by their activation state and inflammation and is important for the development of novel HSC liver cell-based therapy.
Collapse
|
22
|
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: 0.9] [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
|
23
|
Najimi M, Berardis S, El-Kehdy H, Rosseels V, Evraerts J, Lombard C, El Taghdouini A, Henriet P, van Grunsven L, Sokal EM. Human liver mesenchymal stem/progenitor cells inhibit hepatic stellate cell activation: in vitro and in vivo evaluation. Stem Cell Res Ther 2017; 8:131. [PMID: 28583205 PMCID: PMC5460523 DOI: 10.1186/s13287-017-0575-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 12/27/2022] Open
Abstract
Background Progressive liver fibrosis leads to cirrhosis and end-stage liver disease. This disease is a consequence of strong interactions between matrix-producing hepatic stellate cells (HSCs) and resident and infiltrating immune cell populations. Accumulated experimental evidence supports the involvement of adult-derived human liver mesenchymal stem/progenitor cells (ADHLSCs) in liver regeneration. The aim of the present study was to evaluate the influence of ADHLSCs on HSCs, both in vitro and in vivo. Methods Activated human HSCs were co-cultured with ADHLSCs or ADHLSC-conditioned culture medium. The characteristics of the activated human HSCs were assessed by microscopy and biochemical assays, whereas proliferation was analyzed using flow cytometry and immunocytochemistry. The secretion profile of activated HSCs was evaluated by ELISA and Luminex. ADHLSCs were transplanted into a juvenile rat model of fibrosis established after co-administration of phenobarbital and CCl4. Results When co-cultured with ADHLSCs or conditioned medium, the proliferation of HSCs was inhibited, beginning at 24 h and for up to 7 days. The HSCs were blocked in G0/G1 phase, and showed decreased Ki-67 positivity. Pro-collagen I production was reduced, while secretion of HGF, IL-6, MMP1, and MMP2 was enhanced. Neutralization of HGF partially blocked the inhibitory effect of ADHLSCs on the proliferation and secretion profile of HSCs. Repeated intrahepatic transplantation of cryopreserved/thawed ADHLSCs without immunosuppression inhibited the expression of markers of liver fibrosis in 6 out of 11 rats, as compared to their expression in the vehicle-transplanted group. Conclusions These data provide evidence for a direct inhibitory effect of ADHLSCs on activated HSCs, which supports their development for the treatment of liver fibrosis. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0575-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mustapha Najimi
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Avenue Mounier, 52, 1200, Brussels, Belgium.
| | - Silvia Berardis
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Avenue Mounier, 52, 1200, Brussels, Belgium
| | - Hoda El-Kehdy
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Avenue Mounier, 52, 1200, Brussels, Belgium
| | - Valérie Rosseels
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Avenue Mounier, 52, 1200, Brussels, Belgium
| | - Jonathan Evraerts
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Avenue Mounier, 52, 1200, Brussels, Belgium
| | - Catherine Lombard
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Avenue Mounier, 52, 1200, Brussels, Belgium
| | - Adil El Taghdouini
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Avenue Mounier, 52, 1200, Brussels, Belgium
| | - Patrick Henriet
- Cell Biology Unit, de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, 1200, Brussels, Belgium
| | - Leo van Grunsven
- Liver Cell Biology Lab, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Etienne Marc Sokal
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Avenue Mounier, 52, 1200, Brussels, Belgium
| |
Collapse
|
24
|
Fanti M, Gramignoli R, Serra M, Cadoni E, Strom SC, Marongiu F. Differentiation of amniotic epithelial cells into various liver cell types and potential therapeutic applications. Placenta 2017; 59:139-145. [PMID: 28411944 DOI: 10.1016/j.placenta.2017.03.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/25/2017] [Accepted: 03/29/2017] [Indexed: 02/07/2023]
Abstract
The aim of Regenerative Medicine is to replace or regenerate human cells, tissues or organs in order to restore normal function. Among all organs, the liver is endowed with remarkable regenerative capacity. Nonetheless, there are conditions in which this ability is impaired, and the use of isolated cells, including stem cells, is being considered as a possible therapeutic tool for the management of chronic hepatic disease. Placenta holds great promise for the field of regenerative medicine. It has long been used for the treatment of skin lesions and in ophthalmology, due to its ability to modulate inflammation and promote healing. More recently, cells isolated from the amniotic membrane are being considered as a possible resource for tissue regeneration, including in the context liver disease. Two cell types can be easily isolated from human amnion: epithelial cells (hAEC) and mesenchymal stromal cells (hAMSC). However only the first cell population has been demonstrated to be a possible source of proficient hepatic cells. This review will summarize current knowledge on the differentiation of hAEC into liver cells and their potential therapeutic application.
Collapse
Affiliation(s)
- Maura Fanti
- University of Cagliari, Department of Biomedical Sciences, Unit of Experimental Medicine, Cagliari, Italy
| | - Roberto Gramignoli
- Karolinska Institutet, Department of Laboratory Medicine, Division of Pathology, Stockholm, Sweden
| | - Monica Serra
- University of Cagliari, Department of Biomedical Sciences, Unit of Experimental Medicine, Cagliari, Italy
| | - Erika Cadoni
- University of Cagliari, Department of Biomedical Sciences, Unit of Experimental Medicine, Cagliari, Italy
| | - Stephen C Strom
- Karolinska Institutet, Department of Laboratory Medicine, Division of Pathology, Stockholm, Sweden
| | - Fabio Marongiu
- University of Cagliari, Department of Biomedical Sciences, Unit of Experimental Medicine, Cagliari, Italy.
| |
Collapse
|