1
|
Tupone MG, d'Angelo M, Castelli V, Catanesi M, Benedetti E, Cimini A. A State-of-the-Art of Functional Scaffolds for 3D Nervous Tissue Regeneration. Front Bioeng Biotechnol 2021; 9:639765. [PMID: 33816451 PMCID: PMC8012845 DOI: 10.3389/fbioe.2021.639765] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/26/2021] [Indexed: 12/15/2022] Open
Abstract
Exploring and developing multifunctional intelligent biomaterials is crucial to improve next-generation therapies in tissue engineering and regenerative medicine. Recent findings show how distinct characteristics of in situ microenvironment can be mimicked by using different biomaterials. In vivo tissue architecture is characterized by the interconnection between cells and specific components of the extracellular matrix (ECM). Last evidence shows the importance of the structure and composition of the ECM in the development of cellular and molecular techniques, to achieve the best biodegradable and bioactive biomaterial compatible to human physiology. Such biomaterials provide specialized bioactive signals to regulate the surrounding biological habitat, through the progression of wound healing and biomaterial integration. The connection between stem cells and biomaterials stimulate the occurrence of specific modifications in terms of cell properties and fate, influencing then processes such as self-renewal, cell adhesion and differentiation. Recent studies in the field of tissue engineering and regenerative medicine have shown to deal with a broad area of applications, offering the most efficient and suitable strategies to neural repair and regeneration, drawing attention towards the potential use of biomaterials as 3D tools for in vitro neurodevelopment of tissue models, both in physiological and pathological conditions. In this direction, there are several tools supporting cell regeneration, which associate cytokines and other soluble factors delivery through the scaffold, and different approaches considering the features of the biomaterials, for an increased functionalization of the scaffold and for a better promotion of neural proliferation and cells-ECM interplay. In fact, 3D scaffolds need to ensure a progressive and regular delivery of cytokines, growth factors, or biomolecules, and moreover they should serve as a guide and support for injured tissues. It is also possible to create scaffolds with different layers, each one possessing different physical and biochemical aspects, able to provide at the same time organization, support and maintenance of the specific cell phenotype and diversified ECM morphogenesis. Our review summarizes the most recent advancements in functional materials, which are crucial to achieve the best performance and at the same time, to overcome the current limitations in tissue engineering and nervous tissue regeneration.
Collapse
Affiliation(s)
- Maria Grazia Tupone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Center for Microscopy, University of L'Aquila, L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA, United States
| |
Collapse
|
2
|
Zakeri N, Mirdamadi ES, Kalhori D, Solati-Hashjin M. Signaling molecules orchestrating liver regenerative medicine. J Tissue Eng Regen Med 2020; 14:1715-1737. [PMID: 33043611 DOI: 10.1002/term.3135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/06/2020] [Accepted: 09/09/2020] [Indexed: 12/19/2022]
Abstract
The liver is in charge of more than 500 functions in the human body, which any damage and failure to the liver can significantly compromise human life. Numerous studies are being carried out in regenerative medicine, as a potential driving force, toward alleviating the need for liver donors and fabrication of a 3D-engineered transplantable hepatic tissue. Liver tissue engineering brings three main factors of cells, extracellular matrix (ECM), and signaling molecules together, while each of these three factors tries to mimic the physiological state of the tissue to direct tissue regeneration. Signaling molecules play a crucial role in directing tissue fabrication in liver tissue engineering. When mimicking the natural in vivo process of regeneration, it is tightly associated with three main phases of differentiation, proliferation (progression), and tissue maturation through vascularization while directing each of these phases is highly regulated by the specific signaling molecules. The understanding of how these signaling molecules guide the dynamic behavior of regeneration would be a tool for further tailoring of bioengineered systems to help the liver regeneration with many cellular, molecular, and tissue-level functions. Hence, the signaling molecules come to aid all these phases for further improvements toward the clinical use of liver tissue engineering as the goal.
Collapse
Affiliation(s)
- Nima Zakeri
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Elnaz Sadat Mirdamadi
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Dianoosh Kalhori
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Mehran Solati-Hashjin
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
3
|
Impact of Three-Dimentional Culture Systems on Hepatic Differentiation of Puripotent Stem Cells and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018. [PMID: 30357683 DOI: 10.1007/978-981-13-0947-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Generation of functional hepatocytes from human pluripotent stem cells (hPSCs) is a vital tool to produce large amounts of human hepatocytes, which hold a great promise for biomedical and regenerative medicine applications. Despite a tremendous progress in developing the differentiation protocols recapitulating the developmental signalling and stages, these resulting hepatocytes from hPSCs yet achieve maturation and functionality comparable to those primary hepatocytes. The absence of 3D milieu in the culture and differentiation of these hepatocytes may account for this, at least partly, thus developing an optimal 3D culture could be a step forward to achieve this aim. Hence, review focuses on current development of 3D culture systems for hepatic differentiation and maturation and the future perspectives of its application.
Collapse
|
4
|
Yamamoto H, Tonello JM, Sambuichi T, Kawabe Y, Ito A, Kamihira M. Characterization of genetically engineered mouse hepatoma cells with inducible liver functions by overexpression of liver-enriched transcription factors. J Biosci Bioeng 2017; 125:131-139. [PMID: 28847578 DOI: 10.1016/j.jbiosc.2017.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 11/24/2022]
Abstract
New cell sources for the research and therapy of organ failure could significantly alleviate the shortage of donor livers that are available to patients who suffer from liver disease. Liver carcinoma derived cells, or hepatoma cells, are the ideal cells for developing bioartificial liver systems. Such cancerous liver cells are easy to prepare in large quantities and can be maintained over long periods under standard culture conditions, unlike primary hepatocytes. However, hepatoma cells possess only a fraction of the functions of primary hepatocytes. In a previous study, by transducing cells with liver-enriched transcription factors that could be inducibly overexpressed-hepatocyte nuclear factor (HNF)1α, HNF1β, HNF3β [FOXA2], HNF4α, HNF6, CCAAT/enhancer binding protein (C/EBP)α, C/EBPβ and C/EBPγ-we created mouse hepatoma cells with high liver-specific gene expression called the Hepa/8F5 cell line. In the present study, we performed functional and genetic analyses to characterize the Hepa/8F5 cell line. Further, in three-dimensional cultures, the function of these cells improved significantly compared to parental cells. Ultimately, these cells might become a new resource that can be used in basic and applied hepatic research.
Collapse
Affiliation(s)
- Hideaki Yamamoto
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Jane Marie Tonello
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takanori Sambuichi
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Akira Ito
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masamichi Kamihira
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| |
Collapse
|
5
|
CÉLULAS MADRE: FUNDAMENTOS Y REVISIÓN DE LA EXPERIENCIA CLÍNICA EN ENFERMEDADES HEPÁTICAS. REVISTA MÉDICA CLÍNICA LAS CONDES 2017. [DOI: 10.1016/j.rmclc.2017.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
6
|
Zakikhan K, Pournasr B, Vosough M, Nassiri-Asl M. In Vitro Generated Hepatocyte-Like Cells: A Novel Tool in Regenerative Medicine and Drug Discovery. CELL JOURNAL 2017; 19:204-217. [PMID: 28670513 PMCID: PMC5412779 DOI: 10.22074/cellj.2016.4362] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/05/2016] [Indexed: 12/19/2022]
Abstract
Hepatocyte-like cells (HLCs) are generated from either various human pluripotent stem
cells (hPSCs) including induced pluripotent stem cells (iPSCs) and embryonic stem cells
(ESCs), or direct cell conversion, mesenchymal stem cells as well as other stem cells like
gestational tissues. They provide potential cell sources for biomedical applications. Liver
transplantation is the gold standard treatment for the patients with end stage liver disease,
but there are many obstacles limiting this process, like insufficient number of donated
healthy livers. Meanwhile, the number of patients receiving a liver organ transplant for
a better life is increasing. In this regard, HLCs may provide an adequate cell source to
overcome these shortages. New molecular engineering approaches such as CRISPR/
Cas system applying in iPSCs technology provide the basic principles of gene correction
for monogenic inherited metabolic liver diseases, as another application of HLCs. It has
been shown that HLCs could replace primary human hepatocytes in drug discovery and
hepatotoxicity tests. However, generation of fully functional HLCs is still a big challenge;
several research groups have been trying to improve current differentiation protocols to
achieve better HLCs according to morphology and function of cells. Large-scale generation
of functional HLCs in bioreactors could make a new opportunity in producing enough
hepatocytes for treating end-stage liver patients as well as other biomedical applications
such as drug studies. In this review, regarding the biomedical value of HLCs, we focus
on the current and efficient approaches for generating hepatocyte-like cells in vitro and
discuss about their applications in regenerative medicine and drug discovery.
Collapse
Affiliation(s)
- Kobra Zakikhan
- Cellular and Molecular Research Center, Department of Molecular Medicine, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Behshad Pournasr
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marjan Nassiri-Asl
- Cellular and Molecular Research Center, Department of Molecular Medicine, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.,Cellular and Molecular Research Center, Department of Pharmacology, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
7
|
Human Cord Blood-Derived CD133 +/C-Kit +/Lin - Cells Have Bipotential Ability to Differentiate into Mesenchymal Stem Cells and Outgrowth Endothelial Cells. Stem Cells Int 2016; 2016:7162160. [PMID: 28074098 PMCID: PMC5203918 DOI: 10.1155/2016/7162160] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023] Open
Abstract
Recent evidence suggests that mononuclear cells (MNCs) derived from bone marrow and cord blood can differentiate into mesenchymal stem cells (MSCs) or outgrowth endothelial cells (OECs). However, controversy exists as to whether MNCs have the pluripotent capacity to differentiate into MSCs or OECs or are a mixture of cell lineage-determined progenitors of MSCs or OECs. Here, using CD133+/C-kit+/Lin− mononuclear cells (CKL− cells) isolated from human umbilical cord blood using magnetic cell sorting, we characterized the potency of MNC differentiation. We first found that CKL− cells cultured with conditioned medium of OECs or MSCs differentiated into OECs or MSCs and this differentiation was also induced by cell-to-cell contact. When we cultured single CKL− cells on OEC- or MSC-conditioned medium, the cells differentiated morphologically and genetically into OEC- or MSC-like cells, respectively. Moreover, we confirmed that OECs or MSCs differentiated from CKL− cells had the ability to form capillary-like structures in Matrigel and differentiate into osteoblasts, chondrocytes, and adipocytes. Finally, using microarray analysis, we identified specific factors of OECs or MSCs that could potentially be involved in the differentiation fate of CKL− cells. Together, these results suggest that cord blood-derived CKL− cells possess at least bipotential differentiation capacity toward MSCs or OECs.
Collapse
|
8
|
Chaudhari P, Prasad N, Tian L, Jang YY. Determination of Functional Activity of Human iPSC-Derived Hepatocytes by Measurement of CYP Metabolism. Methods Mol Biol 2016; 1357:383-94. [PMID: 25410290 DOI: 10.1007/7651_2014_145] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The advent of induced pluripotent stem cell (iPSC) technology has enabled the modeling of an array of specific human disease phenotypes, aiding in the increasingly important and indispensable understanding of disease progression and pathogenesis. Pluripotent stem cell-derived hepatocytes present a new avenue for drug screening and personalized drug testing toward precision medicine. CYP450 microsomal enzymes play a critical role in drug metabolism. Hence, CYP activity measurement of iPSC-derived hepatocytes is a vital prerequisite, to ensure metabolic functionality before proceeding to drug testing. Herein, we describe the protocol for measurement of different CYP450 enzyme activities in human iPSC-derived hepatocytes.
Collapse
Affiliation(s)
- Pooja Chaudhari
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Neha Prasad
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Lipeng Tian
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yoon-Young Jang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
9
|
Liu K, Yu C, Xie M, Li K, Ding S. Chemical Modulation of Cell Fate in Stem Cell Therapeutics and Regenerative Medicine. Cell Chem Biol 2016; 23:893-916. [PMID: 27524294 DOI: 10.1016/j.chembiol.2016.07.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 12/19/2022]
Abstract
Regenerative medicine aims to repair and regenerate injured tissues and restore their impaired functions. Recent developments in stem cell biology have attracted significant interest in their applications in regenerative medicine. Chemical approaches using small molecules have yielded exciting results in induction and differentiation of pluripotent stem cells, lineage conversion of somatic cells, and ex vivo as well as in vivo modulation of adult stem cells. In this review, we discuss recent progress, new insights, and future challenges of the chemical approaches in stem cell biology and regenerative medicine.
Collapse
Affiliation(s)
- Kai Liu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Chen Yu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Min Xie
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ke Li
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sheng Ding
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
10
|
Sanal MG. Cell therapy from bench to bedside: Hepatocytes from fibroblasts - the truth and myth of transdifferentiation. World J Gastroenterol 2015; 21:6427-6433. [PMID: 26074681 PMCID: PMC4458753 DOI: 10.3748/wjg.v21.i21.6427] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/24/2015] [Accepted: 05/07/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocyte transplantation is an alternative to liver transplantation in certain disorders such as inherited liver diseases and liver failure. It is a relatively less complicated surgical procedure, and has the advantage that it can be repeated several times if unsuccessful. Another advantage is that hepatocytes can be isolated from partly damaged livers which are not suitable for liver transplantation. Despite these advantages hepatocyte transplantation is less popular. Important issues are poor engraftment of the transplanted cells and the scarcity of donor hepatocytes. Generation of “hepatocyte like cells”/iHeps from embryonic stem cells (ES) and induced pluripotent stem cells (iPSCs) by directed differentiation is an emerging solution to the latter issue. Direct conversation or trans-differentiation of fibroblasts to “hepatocyte like cells” is another way which is, being explored. However this method has several inherent and technical disadvantages compared to the directed differentiation from ES or iPSC. There are several methods claiming to be “highly efficient” for generating “highly functional”“hepatocyte like cells”. Currently different groups are working independently and coming up with differentiation protocols and each group claiming an advantage for their protocol. Directed differentiation protocols need to be designed, compared, analyzed and tweaked systematically and logically than empirically. There is a need for a well-coordinated global initiative comparable to the Human Genome Project to achieve this goal in the near future.
Collapse
|
11
|
Stecklum M, Wulf-Goldenberg A, Purfürst B, Siegert A, Keil M, Eckert K, Fichtner I. Cell differentiation mediated by co-culture of human umbilical cord blood stem cells with murine hepatic cells. In Vitro Cell Dev Biol Anim 2015; 51:183-91. [PMID: 25270685 DOI: 10.1007/s11626-014-9817-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 08/28/2014] [Indexed: 12/27/2022]
Abstract
In the present study, purified human cord blood stem cells were co-cultivated with murine hepatic alpha mouse liver 12 (AML12) cells to compare the effect on endodermal stem cell differentiation by either direct cell-cell interaction or by soluble factors in conditioned hepatic cell medium. With that approach, we want to mimic in vitro the situation of preclinical transplantation experiments using human cells in mice. Cord blood stem cells, cultivated with hepatic conditioned medium, showed a low endodermal differentiation but an increased connexin 32 (Cx32) and Cx43, and cytokeratin 8 (CK8) and CK19 expression was monitored by reverse transcription polymerase chain reaction (RT-PCR). Microarray profiling indicated that in cultivated cord blood cells, 604 genes were upregulated 2-fold, with the highest expression for epithelial CK19 and epithelial cadherin (E-cadherin). On ultrastructural level, there were no major changes in the cellular morphology, except a higher presence of phago(ly)some-like structures observed. Direct co-culture of AML12 cells with cord blood cells led to less incisive differentiation with increased sex-determining region Y-box 17 (SOX17), Cx32 and Cx43, as well as epithelial CK8 and CK19 expressions. On ultrastructural level, tight cell contacts along the plasma membranes were revealed. FACS analysis in co-cultivated cells quantified dye exchange on low level, as also proved by time relapse video-imaging of labelled cells. Modulators of gap junction formation influenced dye transfer between the co-cultured cells, whereby retinoic acid increased and 3-heptanol reduced the dye transfer. The study indicated that the cell-co-cultured model of human umbilical cord blood cells and murine AML12 cells may be a suitable approach to study some aspects of endodermal/hepatic cell differentiation induction.
Collapse
Affiliation(s)
- Maria Stecklum
- Max Delbrück Center for Molecular Medicine, Berlin-Buch, Robert-Rössle-Str. 10, 13125, Berlin, Germany,
| | | | | | | | | | | | | |
Collapse
|
12
|
Gouliarmou V, Pelkonen O, Coecke S. Differentiation-Promoting Medium Additives for Hepatocyte Cultivation and Cryopreservation. Methods Mol Biol 2015; 1250:143-159. [PMID: 26272140 DOI: 10.1007/978-1-4939-2074-7_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Isolated primary hepatocytes are considered as the reference system for in vitro hepatic methods. Following the isolation of primary hepatocytes from liver tissue, an unfavorable process named dedifferentiation is initiated leading to the attenuation of the hepatocellular phenotype both at the morphological and functional level. Freshly isolated hepatocytes can be used immediately or can be cryopreserved for future purposes. Currently, a number of antidedifferentiation strategies exist to extend the life span of isolated hepatocytes. The addition of differentiation-promoting compounds to the hepatocyte culture medium is the oldest and simplest antidedifferentiation approach applied. In the present chapter, the most commonly used medium additives for cultivation and cryopreservation of primary hepatocytes are reviewed.
Collapse
Affiliation(s)
- Varvara Gouliarmou
- EURL ECVAM, Systems Toxicology Unit, Institute for Health and Consumer Protection, European Commission, Joint Research Center, Via Fermi 2749, Ispra, 21027, Italy
| | | | | |
Collapse
|
13
|
Yao R, Wang J, Li X, Jung Jung D, Qi H, Kee KK, Du Y. Hepatic differentiation of human embryonic stem cells as microscaled multilayered colonies leading to enhanced homogeneity and maturation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:4311-4323. [PMID: 25059765 DOI: 10.1002/smll.201401040] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/28/2014] [Indexed: 06/03/2023]
Abstract
Directed differentiation of human embryonic stem cells (hESCs) towards hepatocyte-like cells on planar tissue culture plates has been extensively investigated with great promise to provide alternative cell sources for drug metabolism/toxicity testing. Recently, hepatic differentiation of hESCs in 3D configuration with better mimicry of embryonic liver development represents incremental efforts to improve the differentiation efficiency and cellular maturation. However, most of the present 3D differentiation configurations involved interruptive operations during the multi-staged differentiation process, which might impose unwanted influence on cellular differentiation. Most of the current researches resulted in generation of hepatocytes with high expression of AFP, which is minimally expressed in primary hepatocytes. Here, off-the-shelf micro-stencil arrays are developed to generate adherent multilayered colonies composed of hESCs-derived cells. Uninterrupted cellular differentiation and proliferation is achieved to recapitulate the continuous and multi-stage liver development. Compared with conventional 2D format, the micro-scaled multilayered colonies with uniform and defined sizes constrained within the microwells are composed of more homogenous and mature hepatocyte-like cells with significantly lowered AFP expression and elevated hepatic functions. The multilayered colonies as novel 3D configuration for hepatic differentiation of hESCs represent a significant step toward efficient generation of functional hepatocytes for regenerative medicine and drug discovery.
Collapse
Affiliation(s)
- Rui Yao
- Biofabrication Center, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China; Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Yin CH, Chen W, Hsiao CC, Kuo CY, Chen CL, Wu WT. Production of Mouse Embryoid Bodies with Hepatic Differentiation Potential by Stirred Tank Bioreactor. Biosci Biotechnol Biochem 2014; 71:728-34. [PMID: 17341832 DOI: 10.1271/bbb.60568] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Embryonic stem (ES) cells can differentiate into functional hepatic lineage cells, which can potentially be used in biomedicine. To obtain hepatic lineage cells from ES cells, embryoid bodies (EBs) must be formed. In this study, we developed an EB formation system using a spinner flask for mass production of EBs. ES cells were inoculated into the spinner flask, where they formed EBs within 4 d. The EBs were then transferred into an attached culture for hepatic differentiation. To verify the hepatic lineage cells, albumin secretion and hepatic-specific gene expression were examined. We found that EBs formed by either the spinner flask or hanging drops exhibited similar albumin secretion potential and hepatic-specific gene expression. We conclude that the spinner flask method can be used to produce mouse EBs that can be used to mass produce hepatic lineage cells for use in biomedicine.
Collapse
Affiliation(s)
- Chih-Hsiu Yin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | | | | | |
Collapse
|
15
|
Picanço-Castro V, Moreira LF, Kashima S, Covas DT. Can pluripotent stem cells be used in cell-based therapy? Cell Reprogram 2014; 16:98-107. [PMID: 24606201 DOI: 10.1089/cell.2013.0072] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pluripotent stem cells, both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have the ability to differentiate into several cell types that can be used in drug testing and also in the study and treatment of diseases. These cells can be differentiated by in vitro systems, which may serve as models for human diseases and for cell transplantation. In this review, we address the pluripotent cell types, how to obtain and characterize these cells, and differentiation assays. We also focus on the potential of these cells in clinical trials, and we describe the clinical trials that are underway.
Collapse
|
16
|
Streckfuss-Bömeke K, Jende J, Cheng IF, Hasenfuss G, Guan K. Efficient generation of hepatic cells from multipotent adult mouse germ-line stem cells using an OP9 co-culture system. Cell Reprogram 2013; 16:65-76. [PMID: 24380658 DOI: 10.1089/cell.2013.0057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
On the basis of their self-renewal capacity and their ability to differentiate into derivatives of all three germ layers, germ line-derived multipotent adult stem cells (maGSCs) from mouse testis might serve as one of preferable sources for pluripotent stem cells in regenerative medicine. In our study, we aimed for an efficient hepatic differentiation protocol that is applicable for both maGSCs and embryonic stem cells (ESCs). We attempted to accomplish this goal by using a new established co-culture system with OP9 stroma cells for direct differentiation of maGSCs and ESCs into hepatic cells. We found that the hepatic differentiation of maGSCs was induced by the OP9 co-culture system in comparison to the gelatin culture. Furthermore, we showed that the combination of OP9 co-culture with activin A resulted in the increased expression of endodermal and early hepatic markers Gata4, Sox17, Foxa2, Hnf4, Afp, and Ttr compared to differentiated cells on gelatin or on OP9 alone. Moreover, the hepatic progenitors were capable of differentiating further into mature hepatic cells, demonstrated by the expression of liver-specific markers Aat, Alb, Tdo2, Krt18, Krt8, Krt19, Cps1, Sek, Cyp7a1, Otc, and Pah. A high percentage of maGSC-derived hepatic progenitors (51% AFP- and 61% DLK1-positive) and mature hepatic-like cells (26% ALB-positive) were achieved using this OP9 co-culture system. These generated hepatic cells successfully demonstrated in vitro functions associated with mature hepatocytes, including albumin and urea secretion, glycogen storage, and uptake of low-density lipoprotein. The established co-culture system for maGSCs into functional hepatic cells might serve as a suitable model to delineate the differentiation process for the generation of high numbers of mature hepatocytes in humans without genetic manipulations and make germ line-derived stem cells a potential autologous and alternative cell source for hepatic transplants in metabolic liver disorders.
Collapse
Affiliation(s)
- Katrin Streckfuss-Bömeke
- 1 Department of Cardiology and Pneumology, Georg-August-University of Göttingen , 37075, Göttingen, Germany
| | | | | | | | | |
Collapse
|
17
|
Sun P, Zhou X, Farnworth SL, Patel AH, Hay DC. Modeling human liver biology using stem cell-derived hepatocytes. Int J Mol Sci 2013; 14:22011-21. [PMID: 24201130 PMCID: PMC3856048 DOI: 10.3390/ijms141122011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 02/05/2023] Open
Abstract
Stem cell-derived hepatocytes represent promising models to study human liver biology and disease. This concise review discusses the recent progresses in the field, with a focus on human liver disease, drug metabolism and virus infection.
Collapse
Affiliation(s)
- Pingnan Sun
- Shantou University Medical College, Shantou 515041, China; E-Mails: (P.S.); (X.Z.)
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK; E-Mail:
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK; E-Mail:
| | - Xiaoling Zhou
- Shantou University Medical College, Shantou 515041, China; E-Mails: (P.S.); (X.Z.)
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK; E-Mail:
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK; E-Mail:
| | - Sarah L. Farnworth
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK; E-Mail:
| | - Arvind H. Patel
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK; E-Mail:
| | - David C. Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +44-131-651-9549; Fax: +44-131-651-9501
| |
Collapse
|
18
|
Zhang Z, Gong Y, Guo Y, Hai Y, Yang H, Yang S, Liu Y, Ma M, Liu L, Li Z, Gao WQ, He Z. Direct transdifferentiation of spermatogonial stem cells to morphological, phenotypic and functional hepatocyte-like cells via the ERK1/2 and Smad2/3 signaling pathways and the inactivation of cyclin A, cyclin B and cyclin E. Cell Commun Signal 2013; 11:67. [PMID: 24047406 PMCID: PMC3848919 DOI: 10.1186/1478-811x-11-67] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 09/03/2013] [Indexed: 01/04/2023] Open
Abstract
Background Severe shortage of liver donors and hepatocytes highlights urgent requirement of extra-liver and stem cell source of hepatocytes for treating liver-related diseases. Here we hypothesized that spermatogonial stem cells (SSCs) can directly transdifferentiate to hepatic stem-like cells capable of differentiating into mature hepatocyte-like cells in vitro without an intervening pluripotent state. Results SSCs first changed into hepatic stem-like cells since they resembled hepatic oval cells in morphology and expressed Ck8, Ck18, Ck7, Ck19, OV6, and albumin. Importantly, they co-expressed CK8 and CK19 but not ES cell markers. Hepatic stem-like cells derived from SSCs could differentiate into small hepatocytes based upon their morphological features and expression of numerous hepatic cell markers but lacking of bile epithelial cell hallmarks. Small hepatocytes were further coaxed to differentiate into mature hepatocyte-like cells, as identified by their morphological traits and strong expression of Ck8, Ck18, Cyp7a1, Hnf3b, Alb, Tat, Ttr, albumin, and CYP1A2 but not Ck7 or CK19. Notably, these differentiated cells acquired functional attributes of hepatocyte-like cells because they secreted albumin, synthesized urea, and uptake and released indocyanine green. Moreover, phosphorylation of ERK1/2 and Smad2/3 rather than Akt was activated in hepatic stem cells and mature hepatocytes. Additionally, cyclin A, cyclin B and cyclin E transcripts and proteins but not cyclin D1 or CDK1 and CDK2 transcripts or proteins were reduced in mature hepatocyte-like cells or hepatic stem-like cells derived from SSCs compared to SSCs. Conclusions SSCs can transdifferentiate to hepatic stem-like cells capable of differentiating into cells with morphological, phenotypic and functional characteristics of mature hepatocytes via the activation of ERK1/2 and Smad2/3 signaling pathways and the inactivation of cyclin A, cyclin B and cyclin E. This study thus provides an invaluable source of mature hepatocytes for treating liver-related diseases and drug toxicity screening and offers novel insights into mechanisms of liver development and cell reprogramming.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- Renji Hospital, Stem Cell Research Center, Shanghai Jiao Tong University School of Medicine, 1630 Dongfang Road, Shanghai 200127, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Katsuda T, Teratani T, Chowdhury MM, Ochiya T, Sakai Y. Hypoxia efficiently induces differentiation of mouse embryonic stem cells into endodermal and hepatic progenitor cells. Biochem Eng J 2013. [DOI: 10.1016/j.bej.2013.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
20
|
Alaimo G, Cozzoli E, Marfe G, Esposito L, Ranalli M, Hmada D, Giordano A, Gambacurta A. Blood-derived stem cells (BDSCs) plasticity: in vitro hepatic differentiation. J Cell Physiol 2013; 228:1249-54. [PMID: 23139117 DOI: 10.1002/jcp.24279] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 10/29/2012] [Indexed: 12/19/2022]
Abstract
The limited availability of hepatic tissue suitable for the treatment of liver disease and drug research encourages the generation of hepatic-like cells from alternative sources as support for the regenerative medicine. Human blood derived stem cells (BDSCs) express surface markers and genes characteristic of pluripotent stem cells and have the ability to differentiate into different cell types, including tissues of endodermal origin (i.e., liver). Therefore they can represent a valuable source of hepatocytes for medicine. In this investigation, we exploited a fast hepatic differentiation protocol to generate hepatocyte-like cells from human BDSCs using only hepatocyte growth factor (HGF) and fibroblast growth factor-4 (FGF-4) as growth factors. The resulting cell population exhibited hepatic cell-like morphology and it was characterized with a variety of biological endpoint analyses. Here, we demonstrate how human BDSCs can be reprogrammed in hepatocyte-like cells by morphological, functional analysis, reverse transcriptase (RT)-PCR, and Western Blot assay. This study defines a fast and easy reprogramming strategy that facilitates the differentiation of human BDSCs along a hepatic lineage and provides a framework for a helpful source in the stem cells therapy and liver disorders.
Collapse
Affiliation(s)
- Giorgia Alaimo
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Maruyama J, Matsunaga T, Yamaori S, Sakamoto S, Kamada N, Nakamura K, Kikuchi S, Ohmori S. Differentiation of Monkey Embryonic Stem Cells to Hepatocytes by Feeder-Free Dispersion Culture and Expression Analyses of Cytochrome P450 Enzymes Responsible for Drug Metabolism. Biol Pharm Bull 2013; 36:292-8. [DOI: 10.1248/bpb.b12-00866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Junya Maruyama
- Department of Pharmacy, Shinshu University Hospital, 3–1–1 Asahi, Matsumoto 390–8621, Japan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Li YS, Harn HJ, Hsieh DK, Wen TC, Subeq YM, Sun LY, Lin SZ, Chiou TW. Cells and materials for liver tissue engineering. Cell Transplant 2012; 22:685-700. [PMID: 23127824 DOI: 10.3727/096368912x655163] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Liver transplantation is currently the most efficacious treatment for end-stage liver diseases. However, one main problem with liver transplantation is the limited number of donor organs that are available. Therefore, liver tissue engineering based on cell transplantation that combines materials to mimic the liver is under investigation with the goal of restoring normal liver functions. Tissue engineering aims to mimic the interactions among cells with a scaffold. Particular materials or a matrix serve as a scaffold and provide a three-dimensional environment for cell proliferation and interaction. Moreover, the scaffold plays a role in regulating cell maturation and function via these interactions. In cultures of hepatic lineage cells, regulation of cell proliferation and specific function using biocompatible synthetic, biodegradable bioderived matrices, protein-coated materials, surface-modified nanofibers, and decellularized biomatrix has been demonstrated. Furthermore, beneficial effects of addition of growth factor cocktails to a flow bioreactor or coculture system on cell viability and function have been observed. In addition, a system for growing stem cells, liver progenitor cells, and primary hepatocytes for transplantation into animal models was developed, which produces hepatic lineage cells that are functional and that show long-term proliferation following transplantation. The major limitation of cells proliferated with matrix-based transplantation systems is the high initial cell loss and dysfunction, which may be due to the absence of blood flow and the changes in nutrients. Thus, the development of vascular-like scaffold structures, the formation of functional bile ducts, and the maintenance of complex metabolic functions remain as major problems in hepatic tissue engineering and will need to be addressed to enable further advances toward clinical applications.
Collapse
Affiliation(s)
- Yuan-Sheng Li
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Generation, characterization and potential therapeutic applications of mature and functional hepatocytes from stem cells. J Cell Physiol 2012; 228:298-305. [DOI: 10.1002/jcp.24150] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
24
|
Orman MA, Mattick J, Androulakis IP, Berthiaume F, Ierapetritou MG. Stoichiometry based steady-state hepatic flux analysis: computational and experimental aspects. Metabolites 2012; 2:268-91. [PMID: 24957379 PMCID: PMC3901202 DOI: 10.3390/metabo2010268] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 03/05/2012] [Accepted: 03/06/2012] [Indexed: 11/16/2022] Open
Abstract
: The liver has many complex physiological functions, including lipid, protein and carbohydrate metabolism, as well as bile and urea production. It detoxifies toxic substances and medicinal products. It also plays a key role in the onset and maintenance of abnormal metabolic patterns associated with various disease states, such as burns, infections and major traumas. Liver cells have been commonly used in in vitro experiments to elucidate the toxic effects of drugs and metabolic changes caused by aberrant metabolic conditions, and to improve the functions of existing systems, such as bioartificial liver. More recently, isolated liver perfusion systems have been increasingly used to characterize intrinsic metabolic changes in the liver caused by various perturbations, including systemic injury, hepatotoxin exposure and warm ischemia. Metabolic engineering tools have been widely applied to these systems to identify metabolic flux distributions using metabolic flux analysis or flux balance analysis and to characterize the topology of the networks using metabolic pathway analysis. In this context, hepatic metabolic models, together with experimental methodologies where hepatocytes or perfused livers are mainly investigated, are described in detail in this review. The challenges and opportunities are also discussed extensively.
Collapse
Affiliation(s)
- Mehmet A Orman
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - John Mattick
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ioannis P Androulakis
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Marianthi G Ierapetritou
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
25
|
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
|
26
|
Transcriptional expression profile of cultured human embryonic stem cells in vitro and in vivo. In Vitro Cell Dev Biol Anim 2012; 48:165-74. [PMID: 22311374 DOI: 10.1007/s11626-012-9487-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 01/13/2012] [Indexed: 01/23/2023]
Abstract
The aims of this study were to analyze the spontaneous differentiation of human embryonic stem cells in vitro and in vivo and to investigate the influence of in vitro partial differentiation on in vivo teratoma formation in immunodeficient mice. Standardized methods are needed for long-term cultivation of undifferentiated stem cells and the multilineage cells that spontaneously differentiate from them. Accordingly, SA002 human embryonic stem cells were cultured on irradiated mouse embryonic fibroblasts cells, on irradiated human foreskin fibroblasts, or were cultured feeder-free using matrigel. Expression of marker protein transcripts was analyzed in undifferentiated and differentiated stem cells using real-time PCR, and both types of stem cells were transplanted subcutaneously into immunodeficient NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice to test for teratoma formation. Teratoma histology and expression profiles were subsequently characterized. Cells cultured using different conditions and morphologically undifferentiated cells had comparable marker expression profiles, showing high expression levels of markers for pluripotency and low-to-moderate expression levels of germ layer markers. Cells showing spontaneous differentiation that were cultured in feeder-free conditions in the absence of basic fibroblast growth factor demonstrated slight upregulation of sex determining region Y-box 17, connexin 32, and albumin expression at early time points, as well as expression of octamer-binding transcription factor 4, proteoglycan epitopes on podocalyxin (Trafalgar), and alkaline phosphatase. At later time points, expression of hepatocyte nuclear factor-3-beta, and hepatocyte nuclear factor-4-alpha and alpha fetoprotein was upregulated, whereas beta-3-tubulin, chemokine receptor, nestin, sex-determining region Y-box 17, and connexin 32 were downregulated. Expression of pluripotency markers remained high, and hematopoetic markers were not expressed. SA002 cells that showed spontaneous partial differentiation in vitro had a low teratoma formation capacity in vivo. Cells that were partially differentiated led to slower growing teratomas with more uniform histology.
Collapse
|
27
|
Insulin-like growth factor 1 (IGF-I) improves hepatic differentiation of human bone marrow-derived mesenchymal stem cells. Cell Biol Int 2012; 35:1169-76. [PMID: 21910691 DOI: 10.1042/cbi20110016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The ability of MSCs (mesenchymal stem cells) to differentiate between other cell types makes these cells an attractive therapeutic tool for cell transplantation. This project was designed to improve transdifferentiation of human MSCs into liver cells using IGF-I (insulin-like growth factor 1) which, despite its important role in liver development, has not been used for in vitro hepatic differentiation. In the present study, the MSCs derived from healthy human bone marrow samples were cultured and characterized by immunophenotyping and differentiation potential into osteoblast and adipocytes. Transdifferentiation into hepatocyte-like cells was performed in the presence/absence of IGF-I in combination with predefined hepatic differentiation cocktail. To evaluate transdifferentiation, morphological features, immuno-cytochemical staining of specific biological markers and hepatic functions were assessed. Morphological assessment and evaluation of glycogen content, albumin and AFP (α-feto protein) expression as well as albumin and urea secretion revealed statistically significant difference between experimental groups compared with the control. Morphology and function (albumin secretion) of IGF-I-treated cells were significantly better than IGF-I-free experimental group. To the best of our knowledge, our study is the first to demonstrate that the combination of IGF-I with the predefined hepatic differentiation cocktail will significantly improve the morphological features of the differentiated cells and albumin secretion.
Collapse
|
28
|
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
|
29
|
Vasanthan KS, Subramanian A, Krishnan UM, Sethuraman S. Role of biomaterials, therapeutic molecules and cells for hepatic tissue engineering. Biotechnol Adv 2012; 30:742-52. [PMID: 22265845 DOI: 10.1016/j.biotechadv.2012.01.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 12/28/2011] [Accepted: 01/05/2012] [Indexed: 12/18/2022]
Abstract
Current liver transplantation strategies face severe shortcomings owing to scarcity of donors, immunogenicity, prohibitive costs and poor survival rates. Due to the lengthy list of patients requiring transplant, high mortality rates are observed during the endless waiting period. Tissue engineering could be an alternative strategy to regenerate the damaged liver and improve the survival and quality of life of the patient. The development of an ideal scaffold for liver tissue engineering depends on the nature of the scaffold, its architecture and the presence of growth factors and recognition motifs. Biomimetic scaffolds can simulate the native extracellular matrix for the culture of hepatocytes to enable them to exhibit their functionality both in vitro and in vivo. This review highlights the physiology and pathophysiology of liver, the current treatment strategies, use of various scaffolds, incorporation of adhesion motifs, growth factors and stem cells that can stabilize and maintain hepatocyte cultures for a long period.
Collapse
|
30
|
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
|
31
|
Kawabata K, Inamura M, Mizuguchi H. Efficient hepatic differentiation from human iPS cells by gene transfer. Methods Mol Biol 2012; 826:115-24. [PMID: 22167644 DOI: 10.1007/978-1-61779-468-1_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Establishment of protocols for the differentiation of hepatic cells from human embryonic stem (ES) and induced pluripotent stem (iPS) cells could contribute to regenerative cell therapies or drug discovery and development. However, the differentiation efficiency of endoderm-derived cells, such as hepatic cells, from human ES and iPS cells is poor because hepatic cells are differentiated via multiple lineages including endodermal cells, hepatic progenitor cells, and mature hepatocytes. We show here the protocols for efficient hepatic differentiation from human ES and iPS cells by adenovirus vector-mediated gene transfer.
Collapse
Affiliation(s)
- Kenji Kawabata
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | | | | |
Collapse
|
32
|
Ayatollahi M, Soleimani M, Tabei SZ, Kabir Salmani M. Hepatogenic differentiation of mesenchymal stem cells induced by insulin like growth factor-I. World J Stem Cells 2011; 3:113-21. [PMID: 22224170 PMCID: PMC3251745 DOI: 10.4252/wjsc.v3.i12.113] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 10/30/2011] [Accepted: 11/07/2011] [Indexed: 02/06/2023] Open
Abstract
AIM To improve hepatic differentiation of human mesenchymal stem cell (MSC) using insulin growth factor 1 (IGF-I), which has important role in liver development, hepatocyte differentiation and function. METHODS Bone marrow of healthy donors was aspirated from the iliac crest. The adherent cells expanded rapidly and were maintained with periodic passages until a relatively homogeneous population was established. The identification of these cells was carried out by immunophenotype analysis and differentiation potential into osteocytes and adipocytes. To effectively induce hepatic differentiation, we designed a protocol based on a combination of IGF-I and liver specific factors (hepatocyte growth factor, oncostatin M and dexamethasone). Morphological features, hepatic functions and cytological staining were assessed to evaluate transdifferentiation of human marrow-derived MSCs. RESULTS Flow cytometric analysis and the differentiation potential into osteoblasts and adipocytes showed that more than 90% of human MSCs which were isolated and expanded were positive by specific markers and functional tests. Morphological assessment and evaluation of glycogen storage, albumin and α-feto protein expression, as well as albumin and urea secretion revealed a statistically significant difference between the experimental groups and control. CONCLUSION In vitro differentiated MSCs using IGF-I were able to display advanced liver metabolic functions, supporting the possibility of developing them as potential alternatives to primary hepatocytes.
Collapse
Affiliation(s)
- Maryam Ayatollahi
- Maryam Ayatollahi, Transplant Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | |
Collapse
|
33
|
New approaches in the differentiation of human embryonic stem cells and induced pluripotent stem cells toward hepatocytes. Stem Cell Rev Rep 2011; 7:748-59. [PMID: 21336836 PMCID: PMC3137783 DOI: 10.1007/s12015-010-9216-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Orthotropic liver transplantation is the only established treatment for end-stage liver diseases. Utilization of hepatocyte transplantation and bio-artificial liver devices as alternative therapeutic approaches requires an unlimited source of hepatocytes. Stem cells, especially embryonic stem cells, possessing the ability to produce functional hepatocytes for clinical applications and drug development, may provide the answer to this problem. New discoveries in the mechanisms of liver development and the emergence of induced pluripotent stem cells in 2006 have provided novel insights into hepatocyte differentiation and the use of stem cells for therapeutic applications. This review is aimed towards providing scientists and physicians with the latest advancements in this rapidly progressing field.
Collapse
|
34
|
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
|
35
|
Wobus AM, Löser P. Present state and future perspectives of using pluripotent stem cells in toxicology research. Arch Toxicol 2011; 85:79-117. [PMID: 21225242 PMCID: PMC3026927 DOI: 10.1007/s00204-010-0641-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 12/21/2010] [Indexed: 02/08/2023]
Abstract
The use of novel drugs and chemicals requires reliable data on their potential toxic effects on humans. Current test systems are mainly based on animals or in vitro–cultured animal-derived cells and do not or not sufficiently mirror the situation in humans. Therefore, in vitro models based on human pluripotent stem cells (hPSCs) have become an attractive alternative. The article summarizes the characteristics of pluripotent stem cells, including embryonic carcinoma and embryonic germ cells, and discusses the potential of pluripotent stem cells for safety pharmacology and toxicology. Special attention is directed to the potential application of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) for the assessment of developmental toxicology as well as cardio- and hepatotoxicology. With respect to embryotoxicology, recent achievements of the embryonic stem cell test (EST) are described and current limitations as well as prospects of embryotoxicity studies using pluripotent stem cells are discussed. Furthermore, recent efforts to establish hPSC-based cell models for testing cardio- and hepatotoxicity are presented. In this context, methods for differentiation and selection of cardiac and hepatic cells from hPSCs are summarized, requirements and implications with respect to the use of these cells in safety pharmacology and toxicology are presented, and future challenges and perspectives of using hPSCs are discussed.
Collapse
Affiliation(s)
- Anna M Wobus
- In Vitro Differentiation Group, Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Corrensstr. 3, 06466 Gatersleben, Germany.
| | | |
Collapse
|
36
|
Nishiofuku M, Yoshikawa M, Ouji Y, Saito K, Moriya K, Ishizaka S, Nishimura F, Matsuda R, Yamada S, Fukui H. Modulated differentiation of embryonic stem cells into hepatocyte-like cells by coculture with hepatic stellate cells. J Biosci Bioeng 2011; 111:71-7. [DOI: 10.1016/j.jbiosc.2010.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 07/27/2010] [Accepted: 08/09/2010] [Indexed: 12/29/2022]
|
37
|
Vazin T, Freed WJ. Human embryonic stem cells: derivation, culture, and differentiation: a review. Restor Neurol Neurosci 2010; 28:589-603. [PMID: 20714081 DOI: 10.3233/rnn-2010-0543] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The greatest therapeutic promise of human embryonic stem cells (hESC) is to generate specialized cells to replace damaged tissue in patients suffering from various degenerative diseases. However, the signaling mechanisms involved in lineage restriction of ESC to adopt various cellular phenotypes are still under investigation. Furthermore, for progression of hESC-based therapies towards clinical applications, appropriate culture conditions must be developed to generate genetically stable homogenous populations of cells, to hinder possible adverse effects following transplantation. Other critical challenges that must be addressed for successful cell implantation include problems related to survival and functional efficacy of the grafted cells. This review initially describes the derivation of hESC and focuses on recent advances in generation, characterization, and maintenance of these cells. We also give an overview of original and emerging differentiation strategies used to convert hESC to different cell types. Finally, we will discuss transplantation studies of hESC-derived cells with respect to safety and functional recovery.
Collapse
Affiliation(s)
- Tandis Vazin
- Development and Plasticity Section, Cellular Neurobiology Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, USA.
| | | |
Collapse
|
38
|
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
|
39
|
Synnergren J, Heins N, Brolén G, Eriksson G, Lindahl A, Hyllner J, Olsson B, Sartipy P, Björquist P. Transcriptional profiling of human embryonic stem cells differentiating to definitive and primitive endoderm and further toward the hepatic lineage. Stem Cells Dev 2010; 19:961-78. [PMID: 19757991 DOI: 10.1089/scd.2009.0220] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Human embryonic stem cells (hESC) can differentiate into a variety of specialized cell types, and they constitute a useful model system to study embryonic development in vitro. In order to fully utilize the potential of these cells, the mechanisms that regulate the developmental processes of specific lineage differentiation need to be better defined. The aim of this study was to explore the molecular program involved in the differentiation of hESC toward definitive endoderm (DE) and further into the hepatic lineage, and to compare that with primitive endoderm (PrE) differentiation. To that end, we applied two protocols: a specific DE differentiation protocol and an intrinsic differentiation protocol that mainly mediates PrE formation. We collected hESC, hESC-derived DE, DE-derived hepatocyte-progenitors (DE-Prog), DE-derived hepatocyte-like cells (DE-Hep), and the corresponding PrE derivatives. The samples were analyzed using microarrays, and we identified sets of genes that were exclusively up-regulated in DE derivatives (compared to PrE derivatives) at discrete developmental stages. We also investigated known protein interactions among the set of up-regulated genes in DE-Hep. The results demonstrate important differences between DE and PrE differentiation on the transcriptional level. In particular, our results identify a unique molecular program, exclusively activated during development of DE and the subsequent differentiation of DE toward the hepatic lineage. We identified key genes and pathways of potential importance for future efforts to improve hepatic differentiation from hESC. These results reveal new opportunities for rational design of specific interventions with the purpose of generating enriched populations of DE derivatives, including functional hepatocytes.
Collapse
Affiliation(s)
- Jane Synnergren
- School of Life Sciences, University of Skövde , Skövde, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Greenhough S, Medine CN, Hay DC. Pluripotent stem cell derived hepatocyte like cells and their potential in toxicity screening. Toxicology 2010; 278:250-5. [PMID: 20674645 DOI: 10.1016/j.tox.2010.07.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 07/07/2010] [Accepted: 07/21/2010] [Indexed: 12/29/2022]
Abstract
Despite considerable progress in modelling human liver toxicity, the requirement still exists for efficient, predictive and cost effective in vitro models to reduce attrition during drug development. Thousands of compounds fail in this process, with hepatotoxicity being one of the significant causes of failure. The cost of clinical studies is substantial, therefore it is essential that toxicological screening is performed early on in the drug development process. Human hepatocytes represent the gold standard model for evaluating drug toxicity, but are a limited resource. Current alternative models are based on immortalised cell lines and animal tissue, but these are limited by poor function, exhibit species variability and show instability in culture. Pluripotent stem cells are an attractive alternative as they are capable of self-renewal and differentiation to all three germ layers, and thereby represent a potentially inexhaustible source of somatic cells. The differentiation of human embryonic stem cells and induced pluripotent stem cells to functional hepatocyte like cells has recently been reported. Further development of this technology could lead to the scalable production of hepatocyte like cells for liver toxicity screening and clinical therapies. Additionally, induced pluripotent stem cell derived hepatocyte like cells may permit in vitro modelling of gene polymorphisms and genetic diseases.
Collapse
Affiliation(s)
- Sebastian Greenhough
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, 49 Little France Crescent, Edinburgh EH164SB, Scotland, UK
| | | | | |
Collapse
|
41
|
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
|
42
|
JIN LF, JI SH, GUO XY, WANG XH, JI WZ. Induction of Rhesus Monkey Embryonic Stem Cells into Hepatocyte-like Cells by a Three-step Method. Zool Res 2010. [DOI: 10.3724/sp.j.1141.2009.05509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
43
|
Tanase JI, Mitani T, Udagawa K, Nishikawa JI, Ohyama T. Competence of an artificial bent DNA as a transcriptional activator in mouse ES cells. Mol Biol Rep 2010; 38:37-47. [PMID: 20306228 DOI: 10.1007/s11033-010-0075-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 03/05/2010] [Indexed: 11/29/2022]
Abstract
Curved DNA structures with a left-handed superhelical conformation can activate eukaryotic transcription. However, their potency in transgene activation in embryonic stem (ES) cells has not been examined. T20 is an artificial curved DNA of 180 bp that serves as a transcriptional activator. We investigated the effect of T20 on transcription in mouse ES cell lines or hepatocytes differentiated from them. We established 10 sets of cell lines each harboring a single copy of the reporter construct. Each set comprised a T20-harboring cell line and a T20-less control cell line. Analyses showed that in ES cells and in hepatocytes originating from these cells, T20 both activated and repressed transcription in a manner that was dependent on the locus of reporter. The present and previous studies strongly suggest that in cells that have a strict gene regulation system, transcriptional activation by T20 occurs only in a transcriptionally active locus in the genome.
Collapse
Affiliation(s)
- Jun-ichi Tanase
- Department of Biology, Faculty of Education and Integrated Arts and Sciences, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | | | | | | | | |
Collapse
|
44
|
Three-dimensional culture of human embryonic stem cell derived hepatic endoderm and its role in bioartificial liver construction. J Biomed Biotechnol 2010; 2010:236147. [PMID: 20169088 PMCID: PMC2821762 DOI: 10.1155/2010/236147] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Accepted: 12/03/2009] [Indexed: 12/11/2022] Open
Abstract
The liver carries out a range of functions essential for bodily homeostasis. The impairment of liver functions has serious implications and is responsible for high rates of patient morbidity and mortality. Presently, liver transplantation remains the only effective treatment, but donor availability is a major limitation. Therefore, artificial and bioartificial liver devices have been developed to bridge patients to liver transplantation. Existing support devices improve hepatic encephalopathy to a certain extent; however their usage is associated with side effects. The major hindrance in the development of bioartificial liver devices and cellular therapies is the limited availability of human hepatocytes. Moreover, primary hepatocytes are difficult to maintain and lose hepatic identity and function over time even with sophisticated tissue culture media. To overcome this limitation, renewable cell sources are being explored. Human embryonic stem cells are one such cellular resource and have been shown to generate a reliable and reproducible supply of human hepatic endoderm. Therefore, the use of human embryonic stem cell-derived hepatic endoderm in combination with tissue engineering has the potential to pave the way for the development of novel bioartificial liver devices and predictive drug toxicity assays.
Collapse
|
45
|
Abstract
Human embryonic stem cells (HESCs) are pluripotent cells having a self-renewal capacity. These unique characteristics of HESCs allow them to be an unlimited source of cells that was shown to differentiate into many cell types, among them hepatocytes. The creation of hepatocytes in culture will allow us to further understand the mechanisms involved in the embryogenesis of hepatocytes in humans and to study pathologies related to aberrant differentiation of these cells. The resultant hepatocytes may serve as a source of cells for transplantation and as cells for toxicological studies and drug screening. In the past 10 years, since the derivation of HESCs, various protocols for the differentiation of HESCs to hepatic-like cells were published. In this chapter we detail our protocol for differentiating HESCs into hepatic-like cells through embryoid bodies. We further describe the method for the genetic labeling of the hepatic-like cells derived from the HESCs and their isolation by fluorescence-activated cell sorter. We also summarize the published protocols for differentiation of HESCs into hepatic-like cells.
Collapse
Affiliation(s)
- Neta Lavon
- Department of Genetics, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
46
|
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
|
47
|
Meyburg J, Hoffmann GF. Liver, liver cell and stem cell transplantation for the treatment of urea cycle defects. Mol Genet Metab 2010; 100 Suppl 1:S77-83. [PMID: 20156696 DOI: 10.1016/j.ymgme.2010.01.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 01/13/2010] [Accepted: 01/13/2010] [Indexed: 12/23/2022]
Abstract
Despite advances in pharmacological therapy of urea cycle disorders (UCDs), the overall long-term prognosis is poor, especially for neonatal manifestations. Transplantation of liver tissue or isolated cells appears suitable for transfer of the missing enzyme. Liver transplantation (LT) for UCDs has an excellent 5-year survival rate of approximately 90% and is the only way to completely cure the disease. However, major neurological damage can only be prevented if the operation is performed during the first months of life. Unfortunately, such early LTs have a substantial risk for peri- and postoperative complications, mostly caused by a relatively large liver graft. Liver cell transplantation (LCT) is less invasive than LT, but has still to be regarded as an experimental therapy with about 100 patients treated since its first use in 1993. UCDs are a model disease for LCT, because of the poor prognosis, mainly hepatic enzyme defects, and excellent outcome after LT. So far, 10 children underwent LCT for UCDs with very few technical complications and encouraging clinical results. A first prospective study on its use in severe neonatal UCDs has recently started. However, availability of hepatocytes is limited by the scarcity of donor livers; therefore the use of stem cells is under investigation. Several different cell types may be regarded as liver stem cells, and in vivo transformation into hepatocyte-like cells has been shown in animal studies. However, a clear proof of principle in animal models of human metabolic disease is still missing, which is the prerequisite for clinical application in humans.
Collapse
Affiliation(s)
- Jochen Meyburg
- Department of General Pediatrics, University Children's Hospital, Heidelberg, Germany.
| | | |
Collapse
|
48
|
Abstract
Increasingly, research suggests that for certain systems, animal models are insufficient for human toxicology testing. The development of robust, in vitro models of human toxicity is required to decrease our dependence on potentially misleading in vivo animal studies. A critical development in human toxicology testing is the use of human primary hepatocytes to model processes that occur in the intact liver. However, in order to serve as an appropriate model, primary hepatocytes must be maintained in such a way that they persist in their differentiated state. While many hepatocyte culture methods exist, the two-dimensional collagen "sandwich" system combined with a serum-free medium, supplemented with physiological glucocorticoid concentrations, appears to robustly maintain hepatocyte character. Studies in rat and human hepatocytes have shown that when cultured under these conditions, hepatocytes maintain many markers of differentiation including morphology, expression of plasma proteins, hepatic nuclear factors, phase I and II metabolic enzymes. Functionally, these culture conditions also preserve hepatic stress response pathways, such as the SAPK and MAPK pathways, as well as prototypical xenobiotic induction responses. This chapter will briefly review culture methodologies but will primarily focus on hallmark hepatocyte structural, expression and functional markers that characterize the differentiation status of the hepatocyte.
Collapse
Affiliation(s)
- Katy M Olsavsky Goyak
- Center for Molecular Toxicology & Carcinogenesis and Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | | | | |
Collapse
|
49
|
Microarray Analysis of Differentiation of Mouse Embryonic Stem Cells Into Hepatocyte-like Cells*. PROG BIOCHEM BIOPHYS 2009. [DOI: 10.3724/sp.j.1206.2008.00010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Sharma NS, Wallenstein EJ, Novik E, Maguire T, Schloss R, Yarmush ML. Enrichment of hepatocyte-like cells with upregulated metabolic and differentiated function derived from embryonic stem cells using S-NitrosoAcetylPenicillamine. Tissue Eng Part C Methods 2009; 15:297-306. [PMID: 19196121 DOI: 10.1089/ten.tec.2008.0303] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The generation of a large number of fully functional hepatocytes from a renewable cell source can provide an unlimited resource for bioartificial liver devices and cell replacement therapies. We have established a directed differentiation system using sodium butyrate treatment to generate an enriched population of hepatocyte-like cells from embryonic stem cells. A metabolic analysis of the hepatocyte populations revealed glycolytic and mitochondrial phenotypes similar to mouse hepatoma cells, implying that these cells represent an immature hepatocyte phenotype. To mediate further differentiation, S-NitrosoAcetylPenicillamine (SNAP), a nitric oxide donor, was utilized to induce mitochondrial development in the precursor populations. A comparative analysis of the different treated populations showed that 500microM SNAP treatment resulted in the generation of an enriched population of metabolically mature hepatocyte-like cells with increased differentiated function. Specifically, 500microM SNAP treatment increased glucose consumption, lactate production rates, mitochondrial mass, and potential as compared to untreated populations. In addition, functional analysis revealed that intracellular albumin content, urea secretion rates, and cytochrome P450 7a1 promoter activity were increased in the treated population. The methodology described here to generate an enriched population of metabolically and functionally mature hepatocyte-like cells may have potential implications in drug discovery and regenerative medicine.
Collapse
Affiliation(s)
- Nripen S Sharma
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | | | | | | | | | | |
Collapse
|