51
|
CD166(pos) subpopulation from differentiated human ES and iPS cells support repair of acute lung injury. Mol Ther 2012; 20:2335-46. [PMID: 22968480 DOI: 10.1038/mt.2012.182] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Previous efforts to derive lung progenitor cells from human embryonic stem (hES) cells using embryoid body formation or stromal feeder cocultures had been limited by low efficiencies. Here, we report a step-wise differentiation method to drive both hES and induced pluripotent stem (iPS) cells toward the lung lineage. Our data demonstrated a 30% efficiency in generating lung epithelial cells (LECs) that expresses various distal lung markers. Further enrichment of lung progenitor cells using a stem cell marker, CD166 before transplantation into bleomycin-injured NOD/SCID mice resulted in enhanced survivability of mice and improved lung pulmonary functions. Immunohistochemistry of lung sections from surviving mice further confirmed the specific engraftment of transplanted cells in the damaged lung. These cells were shown to express surfactant protein C, a specific marker for distal lung progenitor in the alveoli. Our study has therefore demonstrated the proof-of-concept of using iPS cells for the repair of acute lung injury, demonstrating the potential usefulness of using patient's own iPS cells to prevent immune rejection which arise from allogenic transplantation.
Collapse
|
52
|
van der Meer AD, van den Berg A. Organs-on-chips: breaking the in vitro impasse. Integr Biol (Camb) 2012; 4:461-70. [PMID: 22388577 DOI: 10.1039/c2ib00176d] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In vitro models of biological tissues are indispensable tools for unraveling human physiology and pathogenesis. They usually consist of a single layer of a single cell type, which makes them robust and suitable for parallelized research. However, due to their simplicity, in vitro models are also less valid as true reflections of the complex biological tissues of the human body. Even though the realism of the models can be increased by including more cell types, this will inevitably lead to a decrease in robustness and throughput. The constant trade-off between realism and simplicity has led to an impasse in the development of new in vitro models. Organs-on-chips, a class of microengineered in vitro tissue models, have the potential to break the in vitro impasse. These models combine an artificially engineered, physiologically realistic cell culture microenvironment with the potential for parallelization and increased throughput. They are robust, because the engineered physiological, organ-level features such as tissue organization, geometry, soluble gradients and mechanical stimulation are well-defined and controlled. Moreover, their microfluidic properties and integrated sensors pave the way for high-throughput studies. In this review, we define the in vitro impasse, we explain why organs-on-chips have the potential to break the impasse and we formulate a view on the future of the field. We focus on the design philosophy of organs-on-chips, the integration of technology and biology and on how to connect to the potential end-users.
Collapse
Affiliation(s)
- Andries D van der Meer
- BIOS/Lab on a Chip group, MESA+ Institute for Nanotechnology, University of Twente, The Netherlands
| | | |
Collapse
|
53
|
Ishikawa T, Banas A, Teratani T, Iwaguro H, Ochiya T. Regenerative Cells for Transplantation in Hepatic Failure. Cell Transplant 2012. [DOI: 10.3727/096368911x605286b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Human embryonic stem (ES) cells and induced pluripotent stem (iPS) cells have an enormous potential; however, their potential clinical application is being arrested due to various limitations such as teratoma formation followed by tumorigenesis, emergent usage, and the quality control of cells, as well as safety issues regarding long-term culture are also delaying their clinical application. In addition, human ES cells have two crucial issues: immunogenicity and ethical issues associated with their clinical application. The efficient generation of human iPS cells requires gene transfer, yet the mechanism underlying pluripotent stem cell induction has not yet been fully elucidated. Otherwise, although human adult regenerative cells including mesenchymal stem cells have a limited capacity for differentiation, they are nevertheless promising candidates for tissue regeneration in a clinical setting. This review highlights the use of regenerative cells for transplantation in hepatic failure.
Collapse
Affiliation(s)
- Tetsuya Ishikawa
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Agnieszka Banas
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Takumi Teratani
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Hideki Iwaguro
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
54
|
Allameh A, Kazemnejad S. Safety evaluation of stem cells used for clinical cell therapy in chronic liver diseases; with emphasize on biochemical markers. Clin Biochem 2012; 45:385-96. [PMID: 22306885 DOI: 10.1016/j.clinbiochem.2012.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 01/16/2012] [Accepted: 01/18/2012] [Indexed: 12/11/2022]
Abstract
There are several issues to be considered to reduce the risk of rejection and minimize side effects associated with liver cell transplantation in chronic liver diseases. The source and the condition of stem cell proliferation and differentiation ex vivo and the transplantation protocols are important safety considerations for cell based therapy. The biochemical and molecular markers are important tools for safety evaluation of different processes of cell expansion and transplantation. Studies show that hepatocytes differentiated from adult and embryonic stem cells exhibit biochemical and metabolic properties resembling mature hepatocytes. Therefore these assays can help to assess the biological and metabolic performance of hepatocytes and progenitor stem cells. The assays also help in testing the contribution of transplanted hepatocytes in improving the repair and function of damaged liver in the recipient. Here we review the biochemical and metabolic markers, which are implicated in evaluation of safety issues of stem cells used for therapeutic purposes in chronic liver diseases and regeneration of damaged liver. We also highlight application of biochemical tests for assessment of liver cell transplantation.
Collapse
Affiliation(s)
- Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, PO Box 14115-111, Tehran, Islamic Republic of Iran.
| | | |
Collapse
|
55
|
Sison-Young RLC, Kia R, Heslop J, Kelly L, Rowe C, Cross MJ, Kitteringham NR, Hanley N, Park BK, Goldring CEP. Human pluripotent stem cells for modeling toxicity. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 63:207-256. [PMID: 22776643 DOI: 10.1016/b978-0-12-398339-8.00006-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The development of xenobiotics, driven by the demand for therapeutic, domestic and industrial uses continues to grow. However, along with this increasing demand is the risk of xenobiotic-induced toxicity. Currently, safety screening of xenobiotics uses a plethora of animal and in vitro model systems which have over the decades proven useful during compound development and for application in mechanistic studies of xenobiotic-induced toxicity. However, these assessments have proven to be animal-intensive and costly. More importantly, the prevalence of xenobiotic-induced toxicity is still significantly high, causing patient morbidity and mortality, and a costly impediment during drug development. This suggests that the current models for drug safety screening are not reliable in toxicity prediction, and the results not easily translatable to the clinic due to insensitive assays that do not recapitulate fully the complex phenotype of a functional cell type in vivo. Recent advances in the field of stem cell research have potentially allowed for a readily available source of metabolically competent cells for toxicity studies, derived using human pluripotent stem cells harnessed from embryos or reprogrammed from mature somatic cells. Pluripotent stem cell-derived cell types also allow for potential disease modeling in vitro for the purposes of drug toxicology and safety pharmacology, making this model possibly more predictive of drug toxicity compared with existing models. This article will review the advances and challenges of using human pluripotent stem cells for modeling metabolism and toxicity, and offer some perspectives as to where its future may lie.
Collapse
Affiliation(s)
- R L C Sison-Young
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Han S, Bourdon A, Hamou W, Dziedzic N, Goldman O, Gouon-Evans V. Generation of functional hepatic cells from pluripotent stem cells. ACTA ACUST UNITED AC 2012; Suppl 10:1-7. [PMID: 25364624 DOI: 10.4172/2157-7633.s10-008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liver diseases affect millions of people worldwide, especially in developing country. According to the American Liver Foundation, nearly 1 in every 10 Americans suffers from some form of liver disease. Even though, the liver has great ability to self-repair, in end-stage liver diseases including fibrosis, cirrhosis, and liver cancer induced by viral hepatitis and drugs, the liver regenerative capacity is exhausted. The only successful treatment for chronic liver failure is the whole liver transplantation. More recently, some clinical trials using hepatocyte transplantation have shown some clinical improvement for metabolic liver diseases and acute liver failure. However, the shortage of donor livers remains a life-threatening challenge in liver disease patients. To overcome the scarcity of donor livers, hepatocytes generated from embryonic stem cell or induced pluripotent stem cell differentiation cultures could provide an unlimited supply of such cells for transplantation. This review provides an updated summary of hepatic differentiation protocols published so far, with a characterization of the hepatic cells generated in vitro and their ability to regenerate damaged livers in vivo following transplantation in pre-clinical liver deficient mouse models.
Collapse
Affiliation(s)
- Songyan Han
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA
| | - Alice Bourdon
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA
| | - Wissam Hamou
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA
| | - Noelle Dziedzic
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA
| | - Orit Goldman
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA
| | - Valerie Gouon-Evans
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA
| |
Collapse
|
57
|
Involvement of histone acetylation of Sox17 and Foxa2 promoters during mouse definitive endoderm differentiation revealed by microRNA profiling. PLoS One 2011; 6:e27965. [PMID: 22132182 PMCID: PMC3223193 DOI: 10.1371/journal.pone.0027965] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Accepted: 10/28/2011] [Indexed: 11/19/2022] Open
Abstract
Generation of hepatocyte from embryonic stem cells (ESCs) holds great promise for hepatocyte replacement therapy to treat liver diseases. Achieving high efficiency of directed differentiation of ESCs to hepatocyte is of critical importance. Previously, Wnt3a has been reported to promote Activin A-induced human definitive endoderm (DE) differentiation, the early stage of hepatocyte differentiation. However, the underlying molecular mechanisms are not clear. Growing evidence demonstrated that microRNAs (miRNAs) are key regulators involved in various important biological processes including the regulation of stem cell differentiation. In the present study, we profiled genome wide miRNA expression during Wnt3a and Activin A induced mouse DE differentiation. We uncovered distinct miRNA expression patterns during DE differentiation with the identification of a subset of miRNAs whose expression is synergistically regulated by Wnt3a/Activin A treatment at different stages of DE differentiation. Forced expression of a pool of such synergistically regulated miRNAs alone could partially promote DE differentiation, indicating a regulatory role of them. Using TargetScan and GeneGO pathway analyses, the synergistically regulated miRNAs are predicted to regulate key pathways involved in DE differentiation; among them includes the regulation of histone acetylation. Consistently, Wnt3a and Activin A treatment increased global histone acetylation which can be partially mimicked by over expression of the pooled miRNAs. Chromatin IP (ChIP) experiments demonstrated that the promoter regions of Sox17 and Foxa2 are subjected to histone acetylation regulation. Administration of Hdac inhibitors greatly augmented DE differentiation. Our data uncovered a novel epigenetic mechanism of Wnt3a and Activin A induced DE differentiation, whereby the treatment of growth factors induced histone acetylation at least in part by the regulation of miRNA expression.
Collapse
|
58
|
Hwang Y, Phadke A, Varghese S. Engineered microenvironments for self-renewal and musculoskeletal differentiation of stem cells. Regen Med 2011; 6:505-24. [PMID: 21749208 DOI: 10.2217/rme.11.38] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stem cells hold great promise for therapies aimed at regenerating damaged tissue, drug screening and studying in vitro models of human disease. However, many challenges remain before these applications can become a reality. One such challenge is developing chemically defined and scalable culture conditions for derivation and expansion of clinically viable human pluripotent stem cells, as well as controlling their differentiation with high specificity. Interaction of stem cells with their extracellular microenvironment plays an important role in determining their differentiation commitment and functions. Regenerative medicine approaches integrating cell-matrix and cell-cell interactions, and soluble factors could lead to development of robust microenvironments to control various cellular responses. Indeed, several of these recent developments have provided significant insight into the design of microenvironments that can elicit the targeted cellular response. In this article, we will focus on some of these developments with an emphasis on matrix-mediated expansion of human pluripotent stem cells while maintaining their pluripotency. We will also discuss the role of matrix-based cues and cell-cell interactions in the form of soluble signals in directing stem cell differentiation into musculoskeletal lineages.
Collapse
Affiliation(s)
- Yongsung Hwang
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0412, USA
| | | | | |
Collapse
|
59
|
Kunisada Y, Shoji M, Hosoya M. A gene expression-based screening system for compounds influencing differentiation of mouse embryonic stem cells. ACTA ACUST UNITED AC 2011; 17:140-51. [PMID: 21965112 DOI: 10.1177/1087057111422101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Low molecular weight compounds have been shown to be useful for controlling stem cells, and various high-throughput screening systems have been developed for identifying compounds that regulate the differentiation of stem cells. However, the effects of such compounds on stem cell differentiation are usually evaluated by assessing a single parameter, which is insufficient for proper monitoring of the cellular status. In this study, to classify a number of compounds, the authors established a gene expression-based screening system using mouse embryonic stem (ES) cells that monitored multiple parameters. ES cells were differentiated into three germ layers by embryoid body formation and then treated with the test compounds. Next, cellular changes were assessed by analyzing the expression of multiple genes with the multiplex quantitative reverse transcriptase polymerase chain reaction. By screening a library of pharmacologically active compounds with this system, the authors were able to classify 52 compounds that influenced the gene expression profile of ES cells. They also found that some compounds identified by screening could enhance osteogenic or adipogenic differentiation of human mesenchymal stem cells. These results indicate that the screening system is effective for identifying compounds involved in regulating the differentiation of both ES cells and adult stem cells.
Collapse
Affiliation(s)
- Yuya Kunisada
- Biology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | | | | |
Collapse
|
60
|
Yang DW, Yao P. Cell transplantation for hepatic disease: current research status. Shijie Huaren Xiaohua Zazhi 2011; 19:1720-1725. [DOI: 10.11569/wcjd.v19.i16.1720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [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
Cell transplantation is a promising way to restore liver function. Treatment of end-stage liver disease with stem cells, especially bone marrow stem cells, has attracted wild attention. There is ongoing research to use mature hepatocytes, liver progenitor cells, bone marrow stem cells and embryonic stem cells to restore liver function in patient with hepatic disease. Here we review the current research status of cell transplantation for hepatic disease in terms of cell biology, animal models and clinical trials.
Collapse
|
61
|
Thumanu K, Tanthanuch W, Ye D, Sangmalee A, Lorthongpanich C, Parnpai R, Heraud P. Spectroscopic signature of mouse embryonic stem cell-derived hepatocytes using synchrotron Fourier transform infrared microspectroscopy. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:057005. [PMID: 21639583 DOI: 10.1117/1.3580253] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Stem cell-based therapy for liver regeneration has been proposed to overcome the persistent shortage in the supply of suitable donor organs. A requirement for this to succeed is to find a rapid method to detect functional hepatocytes, differentiated from embryonic stem cells. We propose Fourier transform infrared (FTIR) microspectroscopy as a versatile method to identify the early and last stages of the differentiation process leading to the formation of hepatocytes. Using synchrotron-FTIR microspectroscopy, the means of identifying hepatocytes at the single-cell level is possible and explored. Principal component analysis and subsequent partial least-squares (PLS) discriminant analysis is applied to distinguish endoderm induction from hepatic progenitor cells and matured hepatocyte-like cells. The data are well modeled by PLS with endoderm induction, hepatic progenitor cells, and mature hepatocyte-like cells able to be discriminated with very high sensitivity and specificity. This method provides a practical tool to monitor endoderm induction and has the potential to be applied for quality control of cell differentiation leading to hepatocyte formation.
Collapse
Affiliation(s)
- Kanjana Thumanu
- Synchrotron Light Research Institute, Muang, Nakhon Ratchasima 30000, Thailand
| | | | | | | | | | | | | |
Collapse
|
62
|
Iwamuro M, Shahid JM, Yamamoto K, Kobayashif N. Prospects for Induced Phiripotent Stem Cell-Derived Hepatocytes in Cell Therapy. CELL MEDICINE 2011; 2:1-8. [PMID: 26998398 DOI: 10.3727/215517911x575975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Induced pluripotent stem (iPS) cells, first established in 2006, have the same characteristics of self-renew-ability and pluripotency as embryonic stem (ES) cells. iPS cells are inducible from patient-specific somatic cells; therefore, they hold significant advantages for overcoming immunological rejection as well as the ethical issues associated with the derivation of ES cells from embryos. Generation of patient-derived hepatocytes by iPS technology and their use in cell transplantation therapy for patients with liver disease is quite attractive. Here, we discuss recent advances and challenges in hepatocyte differentiation from iPS cells and their utility in cell therapy.
Collapse
Affiliation(s)
- Masaya Iwamuro
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences , Okayama , Japan
| | - Javed M Shahid
- † Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences , Okayama , Japan
| | - Kazuhide Yamamoto
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences , Okayama , Japan
| | - Naoya Kobayashif
- † Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences , Okayama , Japan
| |
Collapse
|
63
|
Soto-Gutierrez A, Basma H, Navarro-Alvarez N, Uygun BE, Yarmush ML, Kobayashi N, Fox IJ. Differentiating stem cells into liver. Biotechnol Genet Eng Rev 2011; 25:149-63. [PMID: 21412354 DOI: 10.5661/bger-25-149] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Research involving differentiated embryonic stem (ES) cells may revolutionize the study of liver disease, improve the drug discovery process, and assist in the development of stem-cell-based clinical therapies. Generation of ES cell-derived hepatic tissue has benefited from an understanding of the cytokines, growth factors and biochemical compounds that are essential in liver development, and this knowledge has been used to mimic some aspects of embryonic development in vitro. Although great progress has been made in differentiating human ES cells into liver cells, current protocols have not yet produced cells with the phenotype of a mature hepatocyte. There is a significant need to formally establish criteria that would define what constitutes a functional human stem cell-derived hepatocyte. Here, we explore current challenges and future opportunities in development and use of ES cell-derived liver cells. ES-derived hepatocytes could be used to better understand liver biology, begin the process of "personalizing" health care, and to treat some forms of liver disease.
Collapse
Affiliation(s)
- Alejandroo Soto-Gutierrez
- Center for Engineering in Medicine and Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and the Shriners Hospitals for Children, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
64
|
Takata A, Otsuka M, Kogiso T, Kojima K, Yoshikawa T, Tateishi R, Kato N, Shiina S, Yoshida H, Omata M, Koike K. Direct differentiation of hepatic cells from human induced pluripotent stem cells using a limited number of cytokines. Hepatol Int 2011; 5:890-8. [PMID: 21484132 DOI: 10.1007/s12072-011-9251-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Accepted: 01/05/2011] [Indexed: 01/02/2023]
Abstract
PURPOSE Development of improved protocols for differentiating induced pluripotent stem (iPS) cells into hepatic cells is an important step toward their use in the field of hepatology. Specifically, the number of different cytokines should be reduced to limit undesired effects and to reduce the cost of the process. In this report, we describe a simple method for directing human iPS cells to differentiate into hepatic cells using only two cytokines and a short incubation time. METHODS A two-step protocol for differentiating iPS cells into hepatic cells was developed. A high dose of activin A was applied for 3 days to induce definitive endoderm formation. Subsequently, cells were treated with hepatocyte growth factor (HGF) for 5 days to generate hepatic cells. Differentiation was confirmed by immunostaining for differentiation markers. Albumin mRNA levels in differentiated hepatic cells generated using a previously tested three-step protocol that uses activin A, fibroblast growth factor (FGF)/bone morphogenetic protein (BMP), and HGF, and our new protocol were compared to determine the efficiency of differentiation. RESULTS Our two-step protocol induced the differentiation of iPS cells into hepatic cells and required a shorter differentiation period than the previous three-step protocol. The differentiation efficiencies of the two protocols were comparable and the induced hepatic cells were functional. CONCLUSIONS Developing efficient induction and culture methods to generate more highly matured hepatocytes is essential for regenerative cell-based therapies. Our protocol provides a simple, cost-effective, and time-saving approach for generating hepatic cells from iPS cells.
Collapse
Affiliation(s)
- Akemi Takata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Tomomi Kogiso
- Department of Medicine and Institute of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kentaro Kojima
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Yoshikawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryosuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoya Kato
- Global COE Program, Centre for Education and Research for the Advanced Genome-Based Medicine, Unit of Disease Control Genome Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuichiro Shiina
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruhiko Yoshida
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masao Omata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
65
|
Inamura M, Kawabata K, Takayama K, Tashiro K, Sakurai F, Katayama K, Toyoda M, Akutsu H, Miyagawa Y, Okita H, Kiyokawa N, Umezawa A, Hayakawa T, Furue MK, Mizuguchi H. Efficient generation of hepatoblasts from human ES cells and iPS cells by transient overexpression of homeobox gene HEX. Mol Ther 2010; 19:400-7. [PMID: 21102561 PMCID: PMC3034848 DOI: 10.1038/mt.2010.241] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have the potential to differentiate into all cell lineages, including hepatocytes, in vitro. Induced hepatocytes have a wide range of potential application in biomedical research, drug discovery, and the treatment of liver disease. However, the existing protocols for hepatic differentiation of PSCs are not very efficient. In this study, we developed an efficient method to induce hepatoblasts, which are progenitors of hepatocytes, from human ESCs and iPSCs by overexpression of the HEX gene, which is a homeotic gene and also essential for hepatic differentiation, using a HEX-expressing adenovirus (Ad) vector under serum/feeder cell-free chemically defined conditions. Ad-HEX-transduced cells expressed α-fetoprotein (AFP) at day 9 and then expressed albumin (ALB) at day 12. Furthermore, the Ad-HEX-transduced cells derived from human iPSCs also produced several cytochrome P450 (CYP) isozymes, and these P450 isozymes were capable of converting the substrates to metabolites and responding to the chemical stimulation. Our differentiation protocol using Ad vector-mediated transient HEX transduction under chemically defined conditions efficiently generates hepatoblasts from human ESCs and iPSCs. Thus, our methods would be useful for not only drug screening but also therapeutic applications.
Collapse
Affiliation(s)
- Mitsuru Inamura
- Department of Biochemistry and Molecular Biology, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Saxena AK. Congenital Anomalies of Soft Tissues: Birth Defects Depending on Tissue Engineering Solutions and Present Advances in Regenerative Medicine. TISSUE ENGINEERING PART B-REVIEWS 2010; 16:455-66. [DOI: 10.1089/ten.teb.2009.0700] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Amulya K. Saxena
- Department of Pediatric and Adolescent Surgery, Medical University of Graz, Graz, Austria
| |
Collapse
|
67
|
Huang HP, Yu CY, Chen HF, Chen PH, Chuang CY, Lin SJ, Huang ST, Chan WH, Ueng TH, Ho HN, Kuo HC. Factors from human embryonic stem cell-derived fibroblast-like cells promote topology-dependent hepatic differentiation in primate embryonic and induced pluripotent stem cells. J Biol Chem 2010; 285:33510-33519. [PMID: 20720011 DOI: 10.1074/jbc.m110.122093] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The future clinical use of embryonic stem cell (ESC)-based hepatocyte replacement therapy depends on the development of an efficient procedure for differentiation of hepatocytes from ESCs. Here we report that a high density of human ESC-derived fibroblast-like cells (hESdFs) supported the efficient generation of hepatocyte-like cells with functional and mature hepatic phenotypes from primate ESCs and human induced pluripotent stem cells. Molecular and immunocytochemistry analyses revealed that hESdFs caused a rapid loss of pluripotency and induced a sequential endoderm-to-hepatocyte differentiation in the central area of ESC colonies. Knockdown experiments demonstrated that pluripotent stem cells were directed toward endodermal and hepatic lineages by FGF2 and activin A secreted from hESdFs. Furthermore, we found that the central region of ESC colonies was essential for the hepatic endoderm-specific differentiation, because its removal caused a complete disruption of endodermal differentiation. In conclusion, we describe a novel in vitro differentiation model and show that hESdF-secreted factors act in concert with regional features of ESC colonies to induce robust hepatic endoderm differentiation in primate pluripotent stem cells.
Collapse
Affiliation(s)
- Hsiang-Po Huang
- From the Divisions of Medical Research, Taipei 10002, Taiwan
| | - Chun-Ying Yu
- Reproductive Endocrinology and Infertility, Taipei 10002, Taiwan
| | - Hsin-Fu Chen
- Reproductive Endocrinology and Infertility, Taipei 10002, Taiwan; Institute of Clinical Genomics, Taipei 10617, Taiwan
| | - Pin-Hsun Chen
- From the Divisions of Medical Research, Taipei 10002, Taiwan
| | | | - Sung-Jan Lin
- Departments of Dermatology, Taipei 10002, Taiwan; Biomedical Engineering, Taipei 10617, Taiwan
| | - Shih-Tsung Huang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11574, Taiwan
| | - Wei-Hung Chan
- Anesthesiology, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Tzuu-Huei Ueng
- Institute of Toxicology, National Taiwan University, Taipei 10617, Taiwan
| | - Hong-Nerng Ho
- Reproductive Endocrinology and Infertility, Taipei 10002, Taiwan; Institute of Clinical Genomics, Taipei 10617, Taiwan
| | - Hung-Chih Kuo
- Genomics Research Center, Taipei 11574, Taiwan; Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11574, Taiwan; Institute of Clinical Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
68
|
Zheng MH, Ye C, Braddock M, Chen YP. Liver tissue engineering: promises and prospects of new technology. Cytotherapy 2010; 12:349-60. [PMID: 20053145 DOI: 10.3109/14653240903479655] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Today, many patients suffer from acute liver failure and hepatoma. This is an area of high unmet clinical need as these conditions are associated with very high mortality. There is an urgent need to develop techniques that will enable liver tissue engineering or generate a bioartificial liver, which will maintain or improve liver function or offer the possibility of liver replacement. Liver tissue engineering is an innovative way of constructing an implantable liver and has the potential to alleviate the shortage of organ donors for orthotopic liver transplantation. In this review we describe, from an engineering perspective, progress in the field of liver tissue engineering, including three main aspects involving cell sources, scaffolds and vascularization.
Collapse
Affiliation(s)
- Ming-Hua Zheng
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical College, Wenzhou, China
| | | | | | | |
Collapse
|
69
|
Baxter MA, Rowe C, Alder J, Harrison S, Hanley KP, Park BK, Kitteringham NR, Goldring CE, Hanley NA. Generating hepatic cell lineages from pluripotent stem cells for drug toxicity screening. Stem Cell Res 2010; 5:4-22. [PMID: 20483202 PMCID: PMC3556810 DOI: 10.1016/j.scr.2010.02.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 02/24/2010] [Accepted: 02/25/2010] [Indexed: 02/06/2023] Open
Abstract
Hepatotoxicity is an enormous and increasing problem for the pharmaceutical industry. Early detection of problems during the drug discovery pathway is advantageous to minimize costs and improve patient safety. However, current cellular models are sub-optimal. This review addresses the potential use of pluripotent stem cells in the generation of hepatic cell lineages. It begins by highlighting the scale of the problem faced by the pharmaceutical industry, the precise nature of drug-induced liver injury and where in the drug discovery pathway the need for additional cell models arises. Current research is discussed, mainly for generating hepatocyte-like cells rather than other liver cell-types. In addition, an effort is made to identify where some of the major barriers remain in translating what is currently hypothesis-driven laboratory research into meaningful platform technologies for the pharmaceutical industry.
Collapse
Affiliation(s)
- Melissa A. Baxter
- Endocrinology & Diabetes, School of Biomedicine, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Cliff Rowe
- Endocrinology & Diabetes, School of Biomedicine, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Jane Alder
- School of Pharmacy and Pharmaceutical Science, University of Central Lancashire, Preston PR1 2HE, UK
| | - Sean Harrison
- Endocrinology & Diabetes, School of Biomedicine, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Karen Piper Hanley
- Endocrinology & Diabetes, School of Biomedicine, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - B. Kevin Park
- MRC Centre for Drug Safety Science, Department of Pharmacology & Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool L69 3GE, UK
| | - Neil R. Kitteringham
- MRC Centre for Drug Safety Science, Department of Pharmacology & Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool L69 3GE, UK
| | - Chris E. Goldring
- MRC Centre for Drug Safety Science, Department of Pharmacology & Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool L69 3GE, UK
| | - Neil A. Hanley
- Endocrinology & Diabetes, School of Biomedicine, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
70
|
Soto-Gutierrez A, Navarro-Alvarez N, Yagi H, Nahmias Y, Yarmush ML, Kobayashi N. Engineering of an hepatic organoid to develop liver assist devices. Cell Transplant 2010; 19:815-22. [PMID: 20573303 DOI: 10.3727/096368910x508933] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cell-based technologies to support/restore liver function represent one of the most promising opportunities in the treatment of acute liver failure. However, the understanding of the constituent cell types that interact to achieve liver-specific structure and function has not been achieved in the development of liver assist devices (LADs). Here we show that hepatocytes migrated toward and adhered and formed sinusoids-like structures in conjunction with liver nonparenchymal cells, and that this liver organoid formed sophisticated tissue after 7 days in an implanted LAD in rodents. Hepatocytes only or in combination with human nonparenchymal liver cell lines (endothelial, cholangiocytes, and stellate cells) were cultured in Matrigel. Ultrastructural analysis showed that the hepatocyte-decorated endothelial vascular structures resemble in vivo sinusoids containing plate-like structures, bile canaliculi, and lumen. The sinusoid-like structures retained albumin secretion and drug metabolism capabilities. In addition, LADs containing cocultures of human liver nonparenchymal cells were transplanted in animals for a week; the liver tissue formed sophisticated structures resembling the liver. These results demonstrate the importance of nonparenchymal cells in the cellular composition of LADs. The novelty of the culture's sinusoid-like organization and function strongly support the integration of liver nonparenchymal units into hepatocyte coculture-based LADs as a potential destination therapy for liver failure.
Collapse
Affiliation(s)
- Alejandro Soto-Gutierrez
- Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan.
| | | | | | | | | | | |
Collapse
|
71
|
Hybrid cells differentiate to hepatic lineage cells and repair oxidative damage. Cell Mol Biol Lett 2010; 15:451-72. [PMID: 20563703 PMCID: PMC6275737 DOI: 10.2478/s11658-010-0018-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 05/26/2010] [Indexed: 02/07/2023] Open
Abstract
Hybrid cells derived from stem cells play an important role in organogenesis, tissue regeneration and cancer formation. However, the fate of hybrid cells and their range of function are poorly understood. Fusing stem cells and somatic cells induces somatic cell reprogramming, and the resulting hybrid cells are embryonic stem cell-like cells. Therefore, we hypothesize that fusion-induced hybrid cells may behave like ES cells in certain microenvironments. In this study, human hepatic cells were induced to apoptosis with H(2)O(2), and then co-cultured with hybrid cells that had been derived from mouse ES cells and human hepatic cells using a transwell. After co-culturing, the degree of apoptosis was evaluated using Annexin-V/PI double-staining analysis, flow cytometry and Western-blot. We observed that H(2)O(2)-induced cell apoptosis was inhibited by co-culture. In addition, the activity of injury-related enzymes (GSH-Px, LDH and SOD) and the level of albumin release in the co-culture system trended toward the level of normal undamaged hepatic cells. The stably increased levels of secretion of ALB in the co-culture system also confirmed that co-culture with hybrid cells helped in recovery from injury. The fate of the hybrid cells was studied by analyzing their gene expression and protein expression profiles. The results of RT-PCR indicated that during co-culturing, like ES cells, hybrid cells differentiated into hepatic lineage cells. Hybrid cells transcripted genes from both parental cell genomes. Via immunocytochemical analysis, hepatic directional differentiation of the hybrid cells was also confirmed. After injecting the hybrid cells into the mouse liver, the GFP-labeled transplanted cells were distributed in the hepatic lobules and engrafted into the liver structure. This research expands the knowledge of fusion-related events and the possible function of hybrid cells. Moreover, it could indicate a new route of differentiation from pluripotent cells to tissue-specific cells via conditional co-culture.
Collapse
|
72
|
Wu Y, Ge C, Zeng W, Zhang C. Induced multilineage differentiation of chicken embryonic germ cells via embryoid body formation. Stem Cells Dev 2010; 19:195-202. [PMID: 19548770 DOI: 10.1089/scd.2008.0383] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Although the pluripotent and proliferative capacity of embryonic germ (EG) cells is thought to be equivalent to that of embryonic stem (ES) cells, there has been far less attention focused on the potential use of EG cells for applications in developing novel strategies of tissue transplantation in the treatment of degenerative diseases. In this study, EG cells were derived from primordial germ cells (PGCs) of genital ridges of 4-day-old chicken embryos. These cells satisfied the criteria previously used for defining chicken EG cells by using the expression of markers characteristic to ES cells. When injected subcutaneously, chicken EG cells could form teratomas that enable differentiation into a wide range of tissue types of all three primary cell lineages including neural cells, cartilage, forming bone, adipocytes, blood vessels, smooth muscle, and secretory epithelia in the recipients. Furthermore, cells in embryoid bodies (EBs) expressed lineage-specific markers of three germ layers and could be induced to differentiate into more advanced stages of various committed cell types, including dopamine and cholinergic neurons, astrocytes, oligodendrocytes, adipocytes, and hepatocytes, which were demonstrated by immunocytochemical staining or RT-PCR analysis. These findings support the multilineage differentiation capability of chicken pluripotent EG cells, thus confirming the presumption that chicken embryos may be used as a potential model for better understanding the mechanisms of tissue-specific differentiation and regeneration that will help to devise strategies based on the transplantation of stem cell-derived tissues for restoring function to damaged or diseased tissues.
Collapse
Affiliation(s)
- Yanqun Wu
- Key Laboratory of Animal Epidemic Etiology & Immunological Prevention of the Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | | | | | | |
Collapse
|
73
|
Iwamuro M, Komaki T, Kubota Y, Seita M, Kawamoto H, Yuasa T, Shahid JM, Hassan RARA, Hassan WARA, Nakaji S, Nishikawa Y, Kondo E, Yamamoto K, Kobayashi N. Comparative Analysis of Endoderm Formation Efficiency between Mouse ES Cells and iPS Cells. Cell Transplant 2010; 19:831-9. [PMID: 20955658 DOI: 10.3727/096368910x508951] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Definitive endoderm (DE) derived from stem cells holds potential to differentiate into hepatocytes. Stem cell therapy using those cells has potential for a treatment of liver disease. To date, various ways of inducing hepatocytes from embryonic stem (ES) cells have been reported by researchers. However, it has not been proved enough that induced pluripotent stem (iPS) cells behave in the same manner as ES cells in endoderm differentiation. The purpose of this study was to establish an efficient method to induce DE from iPS cells, through comparatively analyzing the efficacy of endoderm formation from mouse ES cells. Furthermore, the efficiency of a serum-free medium in the differentiation into DE was investigated. Mouse ES cells and iPS cells were floated in culture medium for 2 or 5 days and embryoid bodies (EB) were formed. Subsequently, DE was induced with 100 ng/ml activin A and 100 ng/ml basic fibroblast growth factor (bFGF). RT-PCR and real-time PCR analyses were carried out at each step to determine the gene expression of EB markers. The difference in cellular proliferation between serum-containing and serum-free media was examined by an MTS assay in EB and DE induction. iPS cells showed the paralleled mRNA expression to ES cells in each step of differentiation into EB, but the levels of expression of Sox17 and Foxa2 were relatively higher in ES cell-derived DE, whereas Cxcr4 expression was higher in iPS cell-derived DE. The utilization of serum-free medium for iPS cells showed significantly favorable cellular proliferation during EB formation and subsequent DE induction. Forming EB for 5 days and subsequently DE induction with activin A and bFGF with serum-free medium was an appropriate protocol in iPS cells. This may represent an important step for generating hepatocytes from iPS cells for the development of cell therapy.
Collapse
Affiliation(s)
- Masaya Iwamuro
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | | | - Yasuhiro Kubota
- Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Masayuki Seita
- Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hironobu Kawamoto
- Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Takeshi Yuasa
- Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Javed M. Shahid
- Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Reham A. R. A. Hassan
- Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Wael A. R. A. Hassan
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Shuhei Nakaji
- Department of Biomedical Engineering, School of Engineering, Okayama University of Science, Okayama, Japan
| | - Yuriko Nishikawa
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Eisaku Kondo
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhide Yamamoto
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Naoya Kobayashi
- Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
74
|
Balasubramanian S, Babai N, Chaudhuri A, Qiu F, Bhattacharya S, Dave BJ, Parameswaran S, Carson SD, Thoreson WB, Sharp JG, Rao M, Ahmad I. Non cell-autonomous reprogramming of adult ocular progenitors: generation of pluripotent stem cells without exogenous transcription factors. Stem Cells 2010; 27:3053-62. [PMID: 19859985 DOI: 10.1002/stem.242] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Direct reprogramming of differentiated cells to induced pluripotent stem (iPS) cells by ectopic expression of defined transcription factors (TFs) represents a significant breakthrough towards the use of stem cells in regenerative medicine (Takahashi and Yamanaka Cell 2006;126:663-676). However, the virus-mediated expression of exogenous transcription factors could be potentially harmful and, therefore, represents a barrier to the clinical use of iPS cells. Several approaches, ranging from plasmid-mediated TF expression to introduction of recombinant TFs (Yamanaka Cell 2009;137:13-17; Zhou, Wu, Joo et al. Cell Stem Cell 2009;4:381-384), have been reported to address the risk associated with viral integration. We describe an alternative strategy of reprogramming somatic progenitors entirely through the recruitment of endogenous genes without the introduction of genetic materials or exogenous factors. To this end, we reprogrammed accessible and renewable progenitors from the limbal epithelium of adult rat eye by microenvironment-based induction of endogenous iPS cell genes. Non cell-autonomous reprogramming generates cells that are pluripotent and capable of differentiating into functional neurons, cardiomyocytes, and hepatocytes, which may facilitate autologous cell therapy to treat degenerative diseases.
Collapse
Affiliation(s)
- Sudha Balasubramanian
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Fukumitsu K, Ishii T, Yasuchika K, Amagai Y, Kawamura-Saito M, Kawamoto T, Kawase E, Suemori H, Nakatsuji N, Ikai I, Uemoto S. Establishment of a cell line derived from a mouse fetal liver that has the characteristic to promote the hepatic maturation of mouse embryonic stem cells by a coculture method. Tissue Eng Part A 2010; 15:3847-56. [PMID: 19558317 DOI: 10.1089/ten.tea.2009.0357] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Stromal cells residing in murine fetal livers have the ability to promote the hepatic maturation of murine embryonic stem cells (ESCs) and hepatic progenitor cells (HPCs) 3848 in vitro. These stromal cells were isolated as the CD49f(+/-)CD45(-)Thy1(+)gp38(+) cell fraction. The present study established a murine fetal liver stromal cell line that induced hepatic maturation in mouse ESCs and HPCs. A transgene containing a temperature-sensitive SV40 large T antigen was transfected into the primary fetal liver stromal cells. These immortalized cells, which were named as the gp38-positive and Thy1-positive murine liver stromal (MLSgt) cells, induced both mouse ESCs and HPCs to differentiate into mature hepatocyte-like cells using a coculture method. Since MLSgt is not a cloned cell line, one clone, MLSgt20, was selected as a line with the characteristic to induce hepatic differentiation, which was comparable to its parental stromal cells. The ESC-derived endoderm cells cocultured with the MLSgt20 cells expressed mature hepatocyte-specific gene markers, including glucose-6-phosphatase, tyrosine aminotransferase, tryptophan 2,3-dioxgenase, and cytochrome P450 (CYP1a1, Cyp1b1, Cyp1a2, and Cyp3a11). In addition, these cells also exhibited hepatic functions, such as glycogen storage and ammonia metabolism. Transmission electron microscopy showed that the cocultured ESCs expressed the morphologic features of mature hepatocytes. In conclusion, a cell line was established that has the characteristic to promote the hepatic maturation of mouse ESCs and HPCs by a coculture method.
Collapse
Affiliation(s)
- Ken Fukumitsu
- Department of Surgery, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Li W, Wang D, Qin J, Liu C, Zhang Q, Zhang X, Yu X, Lahn BT, Mao FF, Xiang AP. Generation of functional hepatocytes from mouse induced pluripotent stem cells. J Cell Physiol 2010; 222:492-501. [PMID: 20020528 DOI: 10.1002/jcp.22000] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Induced pluripotent stem cells are derived from somatic cells by forced expression of several transcriptional factors. Induced pluripotent stem cells resemble embryonic stem cells in many aspects, such as the expression of certain stem cell markers, chromatin methylation patterns, embryoid body formation and teratoma formation. Therefore, induced pluripotent stem cells provide a powerful tool for study of developmental biology and unlimited resources for transplantation therapy. Here we reported the successful induction of mouse induced pluripotent stem cells and a simple and efficient process for generation of functional hepatocytes from mouse induced pluripotent stem cells by sequential addition of inducing factors. These induced pluripotent stem cell-derived hepatocytes, just as mouse embryonic stem cell-derived hepatocytes, expressed hepatic lineage markers including CK7, CK8, CK18, CK19, alpha-fetoprotein, albumin, Cyp7a1, and exhibited functional hepatic characteristics, including glycogen storage, indocyanine green (ICG) uptake and release, low-density lipoprotein (LDL) uptake and urea secretion. Although we observed some variations in the efficiency of hepatic differentiation between induced pluripotent stem cells and common mouse embryonic stem cell lines, our results indicate that mouse induced pluripotent stem cells can efficiently differentiate into functional hepatocytes in vitro, which may be helpful for the study of liver development and regenerative medicine.
Collapse
Affiliation(s)
- Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-Sen University, Guangzhou 510080, P.R.China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Navarro-Alvarez N, Soto-Gutierrez A, Kobayashi N. Hepatic stem cells and liver development. Methods Mol Biol 2010; 640:181-236. [PMID: 20645053 DOI: 10.1007/978-1-60761-688-7_10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The liver consists of many cell types with specialized functions. Hepatocytes are one of the main players in the organ and therefore are the most vulnerable cells to damage. Since they are not everlasting cells, they need to be replenished throughout life. Although the capacity of hepatocytes to contribute to their own maintenance has long been recognized, recent studies have indicated the presence of both intrahepatic and extrahepatic stem/progenitor cell populations that serve to maintain the normal organ and to regenerate damaged parenchyma in response to a variety of insults.The intrahepatic compartment most likely derives primarily from the biliary tree, particularly the most proximal branches, i.e. the canals of Hering and smallest ductules. The extrahepatic compartment is at least in part derived from diverse populations of cells from the bone marrow. Embryonic stem cells (ES's) are considered as a part of the extrahepatic compartment. Due to their pluripotent capabilities, ES cell-derived cells form a potential future source of hepatocytes, to replace or restore hepatic tissues that have been damaged by disease or injury. Progressing knowledge about stem cells in the liver would allow a better understanding of the mechanisms of hepatic homeostasis and regeneration. Although a human stem cell-derived cell type equivalent to primary hepatocytes does not yet exist, the promising results obtained with extrahepatic stem cells would open the way to cell-based therapy for liver diseases.
Collapse
Affiliation(s)
- Nalu Navarro-Alvarez
- Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | |
Collapse
|
78
|
Ochiya T, Yamamoto Y, Banas A. Commitment of stem cells into functional hepatocytes. Differentiation 2009; 79:65-73. [PMID: 19883970 DOI: 10.1016/j.diff.2009.10.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 07/14/2009] [Accepted: 10/09/2009] [Indexed: 01/05/2023]
Abstract
Liver transplants represent the only way to treat patients suffering from terminal liver failure, but they are associated with numerous problems, including a chronic shortage of donors, high cost, rejection, and side effects for the donor. It is anticipated that regenerative medicine will provide an alternative to liver transplants for such patients. Regenerative medicine refers to the academic field of eliciting the inherent capacity of organisms for self-regeneration to the greatest possible extent in order to develop new methods of treatment for intractable disorders. From this perspective, much is expected from the use of human embryonic stem cells (ES cells) or induced pluripotent stem cells (iPS cells), and the vigorous development of technology to induce the differentiation of such stem cells into cells possessing hepatic functions is underway. Clinical applications of these human stem cells, however, have yet to reach even the earliest stages of implementation. Facing off against these versatile ES cells are stem cells derived from somatic cells present within organisms, which are attracting attention owing to their superiority in terms of ethics and safety, with many research institutes now in the process of elucidating the details of stem cell separation and identification as well as their plasticity and pluripotency. Bone marrow cells are the best-known somatic-cell-derived stem cells, but the use of mesenchymal stem cells (MSCs) found in adipose tissue has also recently attracted attention. This paper will review the differentiation ability and mechanisms of these various stem cell types to hepatocytes and their application to liver regeneration and the future outlook.
Collapse
Affiliation(s)
- Takahiro Ochiya
- National Cancer Center Research Institute, Tsukiji, Chuoku, Tokyo, Japan.
| | | | | |
Collapse
|
79
|
Lee JY, Tuleuova N, Jones CN, Ramanculov E, Zern MA, Revzin A. Directing hepatic differentiation of embryonic stem cells with protein microarray-based co-cultures. Integr Biol (Camb) 2009; 1:460-8. [PMID: 20023756 DOI: 10.1039/b905757a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Embryonic stem cells hold considerable promise in tissue engineering and regenerative medicine as a source of tissue-specific cells. However, realizing this promise requires novel methods for guiding lineage-specific differentiation of stem cells. In this study, we developed a micropatterned co-culture platform for stimulating hepatic differentiation of mouse embryonic stem cells (mESCs). Studies of mESC and hepatic cell adhesion preferences revealed that mESCs required fibronectin for attachment, while hepatic cells (HepG2) preferred collagen (I) substrate and did not adhere to fibronectin. Printing columns of collagen (I) and fibronectin spots (300 microm diameter), followed by sequential seeding of the two cell types, allowed the positioning of clusters of mESCs adjacent to groups of hepatic cells within the same microarray. These micropatterned co-cultures were maintained for up to two weeks in hepatic differentiation media supplemented. To examine the differentiation, mESCs were selectively extracted from the co-culture using laser microdissection and analyzed using real-time reverse transcriptase (RT)-polymerase chain reaction (PCR). These analyses revealed that mESCs co-cultured with HepG2 cells showed a decrease in pluripotency gene expression concomitant with up-regulation of endodermal genes. In addition, the co-culture format induced a significant increase in the expression of liver genes compared to mESCs cultured alone. In conclusion, micropatterned co-cultures of mESCs and hepatic cells showed a significant promise in driving stem cell differentiation towards hepatic phenotype. In the future, this cell culture platform will be further enhanced to enable efficient conversion of mouse and human ESCs to hepatocytes.
Collapse
Affiliation(s)
- Ji Youn Lee
- Department of Biomedical Engineering, University of California, Davis, 451 East Health Sciences St. #2619, Davis, CA, USA
| | | | | | | | | | | |
Collapse
|
80
|
Momose Y, Matsunaga T, Murai K, Takezawa T, Ohmori S. Differentiation of monkey embryonic stem cells into hepatocytes and mRNA expression of cytochrome p450 enzymes responsible for drug metabolism: comparison of embryoid body formation conditions and matrices. Biol Pharm Bull 2009; 32:619-26. [PMID: 19336894 DOI: 10.1248/bpb.32.619] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated the effects of embryoid body (EB) forming conditions on the expression of hepatocyte marker genes such as alpha-fetoprotein, albumin and CYP7A1 in cells cultured on Matrigel-coated plates for 15 d. The expression levels of hepatocyte marker genes in the cells cultured for 2 d for EB formation from cynomolgus monkey embryonic stem (cmES) cells was higher than those in cells cultured for 5 d. However, the fragment-size of cmES colonies did not markedly affect the expression levels. The expression levels of hepatocyte marker genes, and CYP1A1 and CYP2C43 in cells cultured on Matrigel were considerably higher than those on Matrigel reduced and collagen I. CYP1A1 and CYP3A8 mRNAs were significantly induced by 3-methylcholanthrene and rifampicin, respectively. However, CYP2C43 and CYP2D17 were not induced by these compounds. These results suggested that the differentiation into hepatocytes is affected by the incubation period for EB formation, and that Matrigel successfully promoted in vitro differentiation of cmES cells to hepatocytes.
Collapse
|
81
|
Zhu S, Wurdak H, Wang J, Lyssiotis CA, Peters EC, Cho CY, Wu X, Schultz PG. A Small Molecule Primes Embryonic Stem Cells for Differentiation. Cell Stem Cell 2009; 4:416-26. [DOI: 10.1016/j.stem.2009.04.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 02/24/2009] [Accepted: 04/01/2009] [Indexed: 12/25/2022]
|
82
|
Novik EI, Barminko J, Maguire TJ, Sharma N, Wallenstein EJ, Schloss RS, Yarmush ML. Augmentation of EB-directed hepatocyte-specific function via collagen sandwich and SNAP. Biotechnol Prog 2009; 24:1132-41. [PMID: 19194923 DOI: 10.1002/btpr.41] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The development of implantable engineered liver tissue constructs and ex vivo hepatocyte-based therapeutic devices are limited by an inadequate hepatocyte cell source. In our previous studies, embryoid body (EB)-mediated stem cell differentiation spontaneously yielded populations of hepatocyte lineage cells expressing mature hepatocyte markers such as albumin (ALB) and cytokeratin-18 (CK18). However, these cultures neither yielded a homogenous hepatocyte lineage population nor exhibited detoxification function typical of a more mature hepatocyte lineage cell. In this study, secondary culture configurations were used to study the effects of collagen sandwich culture and oncostatin-M (OSM) or S-nitroso-N-acetylpenicillamine (SNAP) supplementation of EB-derived hepatocyte-lineage cell function. Quantitative immunofluorescence and secreted protein analyses were used to provide insights into the long-term maintenance and augmentation of existing functions. The results of these studies suggest that SNAP, independent of the collagen supplementation, maintained the highest levels of ALB expression, however, mature liver-specific CK18 was only expressed in the presence of gel sandwich culture supplemented with SNAP. In addition, albumin secretion and cytochrome P450 detoxification studies indicated that this condition was the best for the augmentation of hepatocyte-like function. Maintenance and augmentation of hepatocyte-like cells isolated from heterogeneous EB cell populations will be a critical step in generating large numbers of functional differentiated cells for therapeutic use.
Collapse
Affiliation(s)
- Eric I Novik
- Dept of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | |
Collapse
|
83
|
Current world literature. Curr Opin Organ Transplant 2009; 14:103-11. [PMID: 19337155 DOI: 10.1097/mot.0b013e328323ad31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
84
|
Abstract
PURPOSE OF REVIEW Cell transplantation to restore liver function as an alternative to whole liver transplantation has thus far not been successful in humans. RECENT FINDINGS Adult mature hepatocytes and various populations of liver progenitors and stem cells are being studied for their regenerative capabilities. Hepatocyte transplantation to treat metabolic deficiencies has shown promising early improvement in liver function; however, long-term success has not been achieved. Liver progenitor cells can now be identified and were shown to be capable to differentiate into a hepatocyte-like phenotype. Despite evidence of mesenchymal stem cell fusion in animal models of liver regeneration, encouraging results were seen in a small group of patients receiving autologous transplantation of CD133 mesenchymal stem cells to repopulate the liver after extensive hepatectomy for liver masses. Ethical issues, availability, potential rejection and limited understanding of the totipotent capabilities of embryonic stem cells are the limitations that prevent their use for restoration of liver function. The effectiveness of embryonic stem cells to support liver function has been proven with their application in the bioartificial liver model in rodents. SUMMARY There is ongoing research to restore liver function in cell biology, animal models and clinical trials using mature hepatocytes, liver progenitor cells, mesenchymal stem cells and embryonic stem cells.
Collapse
Affiliation(s)
- Tanya R Flohr
- Department of Surgery, University of Virginia Health Sciences Center, Charlottesville, Virginia 22908, USA
| | | | | | | |
Collapse
|
85
|
Abstract
Embryonic stem cells (ESCs) can give rise to any adult cell type and thus offer enormous potential for regenerative medicine and drug discovery. Molecular biomarkers serve as valuable tools to classify and isolate ESCs and to monitor their differentiation state by antibody-based techniques. A number of biomarkers, such as certain cell surface antigens, are used to assign pluripotent ESCs; however, accumulating evidence suggests that ESCs are heterogeneous in morphology, phenotype and function, and are thereby classified into subpopulations characterized by multiple sets of molecular biomarkers. Biomarker discovery is also important for ESC biology to elucidate the molecular mechanisms that regulate pluripotency and differentiation. This review summarizes studies of ESC biomarker discovery. "Genome-wide" expression profiling of ESC mRNAs and proteins and direct analyses of the cell surface subproteome have demonstrated that ESCs express a diverse range of biomarkers, cell surface antigens, and signaling molecules found in different cell lineages, as well as a number of key molecules that assure "stemness". Clearly, future quantitative proteomics approaches will enhance our knowledge of the stage- and lineage-specific expression of the proteome and its temporal changes upon differentiation, and provide a more detailed view of nascent and clonally amplified ESCs.
Collapse
Affiliation(s)
- Kohji Nagano
- Department of Pharmaceutical Technology, Research Division, Chugai Pharmaceutical Co., Ltd., Kanagawa, Japan
| | | | | |
Collapse
|
86
|
Moore RN, Dasgupta A, Rajaei N, Yarmush ML, Toner M, Larue L, Moghe PV. Enhanced differentiation of embryonic stem cells using co-cultivation with hepatocytes. Biotechnol Bioeng 2008; 101:1332-43. [PMID: 18571804 DOI: 10.1002/bit.21987] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We examined the effects of co-cultivated hepatocytes on the hepatospecific differentiation of murine embryonic stem (ES) cells. Utilizing an established mouse ES cell line expressing high or low levels of E-cadherin, that we have previously shown to be responsive to hepatotrophic growth factor stimulation (Dasgupta et al., 2005. Biotechnol Bioeng 92(3):257-266), we compared co-cultures of cadherin-expressing ES (CE-ES) cells with cultured rat hepatocytes, allowing for either paracrine interactions (indirect co-cultures) or both juxtacrine and paracrine interactions (direct co-cultures, random and patterned). Hepatospecific differentiation of ES cells was evaluated in terms of hepatic-like cuboidal morphology, heightened gene expression of late maturation marker, glucose-6-phosphatase in relation to early marker, alpha-fetoprotein (AFP), and the intracellular localization of albumin. Hepatocytes co-cultured with growth factor primed CE-ES cells markedly enhanced ES cell differentiation toward the hepatic lineage, an effect that was reversed through E-cadherin blockage and inhibited in control ES cells with reduced cadherin expression. Comparison of single ES cell cultures versus co-cultures show that direct contact co-cultures of hepatocytes and CE-ES cells maximally promoted ES cell commitment towards hepatodifferentiation, suggesting cooperative effects of cadherin-based juxtacrine and paracrine interactions. In contrast, E-cadherin deficient mouse ES (CD-ES) cells co-cultured with hepatocytes failed to show increased G6P expression, confirming the role of E-cadherin expression. To establish whether albumin expression in CE-ES cells was spatially regulated by co-cultured hepatocytes, we co-cultivated CE-ES cells around micropatterned, pre-differentiated rat hepatocytes. Albumin localization was enhanced "globally" within CE-ES cell colonies and was inhibited through E-cadherin antibody blockage in all but an interfacial band of ES cells. Thus, stem cell based cadherin presentation may be an effective tool to induce hepatotrophic differentiation by leveraging both distal/paracrine and contact/juxtacrine interactions with primary cells of the liver.
Collapse
Affiliation(s)
- Rebecca N Moore
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | | | | | | | | | | | | |
Collapse
|
87
|
Jinno S, Moeller HC, Chen CL, Rajalingam B, Chung BG, Dokmeci MR, Khademhosseini A. Microfabricated multilayer parylene-C stencils for the generation of patterned dynamic co-cultures. J Biomed Mater Res A 2008; 86:278-88. [PMID: 18442109 DOI: 10.1002/jbm.a.32030] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Co-culturing different cell types can be useful to engineer a more in vivo-like microenvironment for cells in culture. Recent approaches to generating cellular co-cultures have used microfabrication technologies to regulate the degree of cell-cell contact between different cell types. However, these approaches are often limited to the co-culture of only two cell types in static cultures. The dynamic aspect of cell-cell interaction, however, is a key regulator of many biological processes such as early development, stem cell differentiation, and tissue regeneration. In this study, we describe a micropatterning technique based on microfabricated multilayer parylene-C stencils and demonstrate the potential of parylene-C technology for co-patterning of proteins and cells with the ability to generate a series of at least five temporally controlled patterned co-cultures. We generated dynamic co-cultures of murine embryonic stem cells in culture with various secondary cell types that could be sequentially introduced and removed from the co-cultures. Our studies suggested that dynamic co-cultures generated by using parylene-C stencils may be applicable in studies investigating cellular interactions in controlled microenvironments such as studies of ES cell differentiation, wound healing and development.
Collapse
Affiliation(s)
- Satoshi Jinno
- Department of Medicine, Center for Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
88
|
Abstract
The promise of liver stem cells lie in their potential to provide a continual and readily available source of liver cells that can be used for gene therapy, cellular transplant, bioartificial liver-assisted devices, drug toxicology testing and use as an in vitro model to understand the developmental biology of the liver. Both the rodent and human embryonic stem cell, bone marrow hematopoietic stem cell, mesenchymal stem cell, umbilical cord blood cell, fetal liver progenitor cell, adult liver progenitor cell as well as the mature hepatocyte have been reported to be capable of self-renewal, giving rise to daughter hepatocytes both in vivo and in vitro. These cells can repopulate livers in animal models of liver injury and seemingly improve liver function. However, significant challenges still exist before these cells can be used in humans. These include lack of consensus in immunophenotype of liver progenitor cells, uncertainty of the physiological role of reported candidate stem/progenitor cell, practicality in obtaining sufficient quantity of cells for clinical use and concerns over ethics, long-term efficacy and safety. Current molecular techniques of stem cell identification are confounded by cell fusion, horizontal gene transfer, incomplete differentiation and fetal microchimerism. Reports of stem cell transplantation and phase 1 trials of bone marrow transplantation in humans for liver diseases are exciting but require more robust verification. We review the evidence for various candidate stem cells, human clinical trials reported to date and highlight the challenges facing clinicians in their quest to use liver stem cells to save lives.
Collapse
Affiliation(s)
- Yock Young Dan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, National University Hospital, Singapore.
| | | |
Collapse
|
89
|
Hoganson DM, Pryor HI, Vacanti JP. Tissue engineering and organ structure: a vascularized approach to liver and lung. Pediatr Res 2008; 63:520-6. [PMID: 18427297 DOI: 10.1203/01.pdr.0000305879.38476.0c] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Over the past two decades, great strides have been made in the field of tissue engineering. Many of the initial attempts to develop an engineered tissue construct were based on the concept of seeding cells onto an avascular scaffold. Using advanced manufacturing technologies, the creation of a preformed vascular scaffold has become a reality. This article discusses some of the issues surrounding the development of such a vascular scaffold. We then examine of the challenges associated with applying this scaffold technology to two vital organ constructs: liver and lung.
Collapse
Affiliation(s)
- David M Hoganson
- Department of Pediatric Surgery, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|
90
|
Derive and conquer: sourcing and differentiating stem cells for therapeutic applications. Nat Rev Drug Discov 2008; 7:131-42. [PMID: 18079756 DOI: 10.1038/nrd2403] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although great progress has been made in the isolation and culture of stem cells, the future of stem-cell-based therapies and their productive use in drug discovery and regenerative medicine depends on two key factors: finding reliable sources of multipotent and pluripotent cells and the ability to control their differentiation to generate desired derivatives. It is essential for clinical applications to establish reliable sources of pathogen-free human embryonic stem cells (ESCs) and develop suitable differentiation techniques. Here, we address some of the problems associated with the sourcing of human ESCs and discuss the current status of stem-cell differentiation technology.
Collapse
|
91
|
Pan RL, Chen Y, Pan RL, Chen Y, Xiang LX, Shao JZ, Dong XJ, Zhang GR. Fetal liver-conditioned medium induces hepatic specification from mouse bone marrow mesenchymal stromal cells: a novel strategy for hepatic transdifferentiation. Cytotherapy 2008; 10:668-75. [DOI: 10.1080/14653240802360704] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
92
|
Yamanaka S, Li J, Kania G, Elliott S, Wersto RP, Van Eyk J, Wobus AM, Boheler KR. Pluripotency of embryonic stem cells. Cell Tissue Res 2007; 331:5-22. [PMID: 18026755 DOI: 10.1007/s00441-007-0520-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Accepted: 09/18/2007] [Indexed: 12/27/2022]
Abstract
Embryonic stem (ES) cells derived from pre-implantation embryos have the potential to differentiate into any cell type derived from the three germ layers of ectoderm (epidermal tissues and nerves), mesoderm (muscle, bone, blood), and endoderm (liver, pancreas, gastrointestinal tract, lungs), including fetal and adult cells. Alone, these cells do not develop into a viable fetus or adult animal because they do not retain the potential to contribute to extraembryonic tissue, and in vitro, they lack spatial and temporal signaling cues essential to normal in vivo development. The basis of pluripotentiality resides in conserved regulatory networks composed of numerous transcription factors and multiple signaling cascades. Together, these regulatory networks maintain ES cells in a pluripotent and undifferentiated form; however, alterations in the stoichiometry of these signals promote differentiation. By taking advantage of this differentiation capacity in vitro, ES cells have clearly been shown to possess the potential to generate multipotent stem and progenitor cells capable of differentiating into a limited number of cell fates. These latter types of cells may prove to be therapeutically viable, but perhaps more importantly, the studies of these cells have led to a greater understanding of mammalian development.
Collapse
Affiliation(s)
- Satoshi Yamanaka
- Laboratory of Cardiovascular Sciences, Gerontology Research Center, National Institute on Aging, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | |
Collapse
|
93
|
|
94
|
Cezar GG. Can human embryonic stem cells contribute to the discovery of safer and more effective drugs? Curr Opin Chem Biol 2007; 11:405-9. [PMID: 17662644 DOI: 10.1016/j.cbpa.2007.05.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Accepted: 05/28/2007] [Indexed: 11/24/2022]
Abstract
Few scientific achievements have received such irresistible attention from scientists, clinicians, and the general public as the ability of human embryonic stem (hES) cells to differentiate into functional cell types for regenerative medicine. The most immediate benefit of neurons, cardiomyocytes, and insulin-secreting cells derived from hES cells, however, may reside in their application in drug discovery and toxicology. The availability of renewable human cells with functional similarities to their in vivo counterparts is the first landmark for a new generation of cell-based assays. The development of cell-based assays using human cells that are physiological targets of drug activity will increase the robustness of target validation and efficacy, high-throughput screening (HTS), structure-activity relationship (SAR), and should introduce safer drugs into clinical trials and the marketplace. The pluripotency of embryonic stem cells, that is, the capacity to generate multiple cell types, is a novel path for the discovery of 'regenerative drugs', the pursuit of small molecules that promote tissue repair (neurogenesis, cardiogenesis) or proliferation of resident stem cells in different organs, thus creating drugs that work by a novel mechanism.
Collapse
Affiliation(s)
- Gabriela Gebrin Cezar
- Department of Animal Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, WI 53706, United States.
| |
Collapse
|
95
|
Banas A, Yamamoto Y, Teratani T, Ochiya T. Stem cell plasticity: Learning from hepatogenic differentiation strategies. Dev Dyn 2007; 236:3228-41. [PMID: 17907200 DOI: 10.1002/dvdy.21330] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Many studies on stem cell plasticity are challenging the concept that stem cells contain an intrinsically predefined, unidirectional differentiation program. This means that the developmental fate of a stem cell is dependent on the general potential of the cell (pre-determined stem cell fate) as well as on microenvironmental cues, such as stimuli from growth factors (stem cell niche). Here, we reviewed reports that examined the hepatocyte differentiation ability of stem cells from two different sources: embryonic stem cells and adult stem cells. All of those stem cells revealed the ability to give rise to hepatocyte-like cells using different induction strategies. However, it is still not clear which of those stem cells would be the best source for hepatocyte replacement or which would be the best protocol. We herein present the current knowledge regarding available protocols and factors used in order to obtain functional hepatocytes from stem cells.
Collapse
Affiliation(s)
- Agnieszka Banas
- Section for Studies on Metastasis, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | |
Collapse
|